link
stringlengths
57
71
question
stringlengths
428
3.74k
opa
stringlengths
6
200
opb
stringlengths
6
213
opc
stringlengths
3
177
opd
stringlengths
4
183
diagnosis
stringlengths
6
302
answer_idx
stringclasses
4 values
answer
stringlengths
3
213
explanation
stringlengths
1.52k
8.11k
field
stringclasses
10 values
case
stringlengths
402
3.73k
clinical_question
stringclasses
15 values
normalized_question
stringclasses
4 values
opa_shuffled
stringlengths
6
174
opb_shuffled
stringlengths
4
213
opc_shuffled
stringlengths
3
170
opd_shuffled
stringlengths
6
193
answer_idx_shuffled
stringclasses
4 values
test_image
int64
0
1
test_lab
int64
0
1
test_other
int64
0
1
figure
int64
0
1
gender
stringclasses
3 values
pregnancy
int64
0
1
woman_health
int64
0
1
age
float64
0
95
age_group
stringclasses
10 values
ethnicity
stringclasses
17 values
case_id
int64
1
1.52k
version
stringclasses
7 values
https://jamanetwork.com/journals/jamadermatology/fullarticle/2811083
A man in his 30s with AIDS presented with acute-onset painful scattered umbilicated papulopustules and ovoid ulcerated plaques with elevated, pink borders on the face, trunk, and extremities (Figure, A). The patient also had a new-onset cough but was afebrile and denied other systemic symptoms. Due to his significant immunocompromise, the clinical presentation was highly suspicious for infection. For rapid bedside differentiation of multiple infectious etiologies, a Tzanck smear was performed by scraping the base of an ulcerated lesion and inner aspect of a pseudopustule and scraping its base with a #15 blade. These contents were placed on a glass slide, fixed, and stained with Wright-Giemsa and subsequently Papanicolaou staining to further characterize the changes seen.A, Clinical image demonstrating papulopustules and ovoid ulcerated plaques with elevated, pink borders on the elbows. B, Tzanck smear using Wright-Giemsa staining of specimen demonstrating ballooning of keratinocytes and peripheralization of nuclear material (original magnification ×20). What Is Your Diagnosis?
Herpes simplex virus
Histoplasmosis
Molluscum contagiosum
Mpox
D. Mpox
D
Mpox
The photographs demonstrate a Tzanck smear using Wright-Giemsa stain (Figure, B) with large ballooning keratinocytes with peripheral nuclear staining seen in a patient with subsequently polymerase chain reaction (PCR)-confirmed mpox. Mpox is a member of the pox virus family, which presents with nonspecific symptoms such as fever followed by characteristic skin changes including macules, papules, vesicles, and pustules progressing with cephalocaudal spread or in the genital and/or gluteal region in the most recent outbreak. On histopathologic findings, mpox features vary depending on the stage of the lesion.1,2 Early lesions tend to demonstrate ballooning degeneration, whereas later lesions have few viable keratinocytes.2 The viral cytopathic changes, most often seen in pseudopustular lesions, include multinucleated keratinocytes with ground glass appearance and nuclear contents pushed to the periphery, resulting in a basophilic halo.2 The lack of a true pustule with collections of neutrophils in the stratum corneum have led some to describe these lesions as pseudopustular.Tzanck smear is an inexpensive, rapid, and simple technique to perform cytologic analysis from skin. Initially described in 1947 by Arnault Tzanck for diagnosing vesiculobullous disorders, it is commonly used in diagnosing herpetic infections, which have characteristic features of acantholysis, acute inflammation, and keratinocytic ballooning and specific nuclear changes of molding, multinucleation, and chromatin margination.3 The findings of acantholysis and nuclear cytopathic changes are similarly seen in histopathologic examination of herpes simplex virus (HSV). Herpetic infections may present with clustered vesicles with a pink inflammatory rim, although more verrucous lesions have been described in immunosuppressed patients.In the Tzanck smear from a lesion of mpox in this patient (Figure, B), enlarged keratinocytes were seen with expanded cytoplasm and nuclear material pushed to the periphery, and the nuclear changes associated with HSV were all absent. The observed difference in cytologic features including the nuclear material rimming or pushed to the periphery of the cells is due to the location of viral replication; herpes viruses replicate in the nucleus, whereas mpox replicates in the cytoplasm.4 Although not part of routine practice, Papanicolaou staining was performed in this case to confirm infected cells and provide additional nuclear detail because Tzanck smears in patients with mpox were not previously reported. The findings highlighted the peripheralization of nuclear material in cells with eosinophilic staining, confirming the keratinocyte origin to the cells. A background inflammatory infiltrate was noted and can be seen in most infections.Tzanck smear can also be used to diagnose other viral and fungal infections. Molluscum contagiosum, which presents with multiple umbilicated papules, demonstrates the characteristic large (30-40 µm) ovoid homogenous basophilic staining keratinocytes called Henderson-Patterson bodies that correspond to the identically named intracytoplasmic inclusions seen on histopathologic findings.5,6 Using bedside diagnostics (including Wright Giemsa staining), blastomycosis is diagnosed with 8- to 15-µm spores with broad-based budding, similar in appearance to the organism in histologic examination.5 Histoplasmosis has small (2 to 3 µm) intracellular organisms in macrophages with artifactual clearing (pseudocapsule).5 Both of these fungal infections may manifest clinically as nonhealing ulcerations, verrucous plaques, or multiple papules or nodules and can histologically demonstrate pseudoepitheliomatous hyperplasia with characteristic organisms. The use of the Tzanck smear in diagnosis of any infection depends on the timing of the lesion and is most sensitive with vesicular or pustular lesions.6 Although we only tested 1 stage of lesions, we hypothesize this test will similarly be more sensitive for earlier lesions in patients with mpox.Tzanck smear is a useful technique that can be used to differentiate mpox from other infections at the bedside or in remote settings where PCR or additional culture techniques are not easily accessible. Infection prevention and control precautions are important to help prevent additional spread of this virus with sharp injuries, and caution with unroofing of the lesions is critical to avoid additional spread.7 Knowledge of the cytologic differences of mpox compared with other viral infections is important in the early recognition, diagnosis, and treatment of these patients.
Dermatology
A man in his 30s with AIDS presented with acute-onset painful scattered umbilicated papulopustules and ovoid ulcerated plaques with elevated, pink borders on the face, trunk, and extremities (Figure, A). The patient also had a new-onset cough but was afebrile and denied other systemic symptoms. Due to his significant immunocompromise, the clinical presentation was highly suspicious for infection. For rapid bedside differentiation of multiple infectious etiologies, a Tzanck smear was performed by scraping the base of an ulcerated lesion and inner aspect of a pseudopustule and scraping its base with a #15 blade. These contents were placed on a glass slide, fixed, and stained with Wright-Giemsa and subsequently Papanicolaou staining to further characterize the changes seen.A, Clinical image demonstrating papulopustules and ovoid ulcerated plaques with elevated, pink borders on the elbows. B, Tzanck smear using Wright-Giemsa staining of specimen demonstrating ballooning of keratinocytes and peripheralization of nuclear material (original magnification ×20).
what is your diagnosis?
What is your diagnosis?
Herpes simplex virus
Mpox
Histoplasmosis
Molluscum contagiosum
b
0
1
0
1
male
0
0
35
31-40
null
1
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2811083
A man in his 30s with AIDS presented with acute-onset painful scattered umbilicated papulopustules and ovoid ulcerated plaques with elevated, pink borders on the face, trunk, and extremities (Figure, A). The patient also had a new-onset cough but was afebrile and denied other systemic symptoms. Due to his significant immunocompromise, the clinical presentation was highly suspicious for infection. For rapid bedside differentiation of multiple infectious etiologies, a Tzanck smear was performed by scraping the base of an ulcerated lesion and inner aspect of a pseudopustule and scraping its base with a #15 blade. These contents were placed on a glass slide, fixed, and stained with Wright-Giemsa and subsequently Papanicolaou staining to further characterize the changes seen.A, Clinical image demonstrating papulopustules and ovoid ulcerated plaques with elevated, pink borders on the elbows. B, Tzanck smear using Wright-Giemsa staining of specimen demonstrating ballooning of keratinocytes and peripheralization of nuclear material (original magnification ×20). What Is Your Diagnosis?
Herpes simplex virus
Histoplasmosis
Molluscum contagiosum
Mpox
D. Mpox
D
Mpox
The photographs demonstrate a Tzanck smear using Wright-Giemsa stain (Figure, B) with large ballooning keratinocytes with peripheral nuclear staining seen in a patient with subsequently polymerase chain reaction (PCR)-confirmed mpox. Mpox is a member of the pox virus family, which presents with nonspecific symptoms such as fever followed by characteristic skin changes including macules, papules, vesicles, and pustules progressing with cephalocaudal spread or in the genital and/or gluteal region in the most recent outbreak. On histopathologic findings, mpox features vary depending on the stage of the lesion.1,2 Early lesions tend to demonstrate ballooning degeneration, whereas later lesions have few viable keratinocytes.2 The viral cytopathic changes, most often seen in pseudopustular lesions, include multinucleated keratinocytes with ground glass appearance and nuclear contents pushed to the periphery, resulting in a basophilic halo.2 The lack of a true pustule with collections of neutrophils in the stratum corneum have led some to describe these lesions as pseudopustular.Tzanck smear is an inexpensive, rapid, and simple technique to perform cytologic analysis from skin. Initially described in 1947 by Arnault Tzanck for diagnosing vesiculobullous disorders, it is commonly used in diagnosing herpetic infections, which have characteristic features of acantholysis, acute inflammation, and keratinocytic ballooning and specific nuclear changes of molding, multinucleation, and chromatin margination.3 The findings of acantholysis and nuclear cytopathic changes are similarly seen in histopathologic examination of herpes simplex virus (HSV). Herpetic infections may present with clustered vesicles with a pink inflammatory rim, although more verrucous lesions have been described in immunosuppressed patients.In the Tzanck smear from a lesion of mpox in this patient (Figure, B), enlarged keratinocytes were seen with expanded cytoplasm and nuclear material pushed to the periphery, and the nuclear changes associated with HSV were all absent. The observed difference in cytologic features including the nuclear material rimming or pushed to the periphery of the cells is due to the location of viral replication; herpes viruses replicate in the nucleus, whereas mpox replicates in the cytoplasm.4 Although not part of routine practice, Papanicolaou staining was performed in this case to confirm infected cells and provide additional nuclear detail because Tzanck smears in patients with mpox were not previously reported. The findings highlighted the peripheralization of nuclear material in cells with eosinophilic staining, confirming the keratinocyte origin to the cells. A background inflammatory infiltrate was noted and can be seen in most infections.Tzanck smear can also be used to diagnose other viral and fungal infections. Molluscum contagiosum, which presents with multiple umbilicated papules, demonstrates the characteristic large (30-40 µm) ovoid homogenous basophilic staining keratinocytes called Henderson-Patterson bodies that correspond to the identically named intracytoplasmic inclusions seen on histopathologic findings.5,6 Using bedside diagnostics (including Wright Giemsa staining), blastomycosis is diagnosed with 8- to 15-µm spores with broad-based budding, similar in appearance to the organism in histologic examination.5 Histoplasmosis has small (2 to 3 µm) intracellular organisms in macrophages with artifactual clearing (pseudocapsule).5 Both of these fungal infections may manifest clinically as nonhealing ulcerations, verrucous plaques, or multiple papules or nodules and can histologically demonstrate pseudoepitheliomatous hyperplasia with characteristic organisms. The use of the Tzanck smear in diagnosis of any infection depends on the timing of the lesion and is most sensitive with vesicular or pustular lesions.6 Although we only tested 1 stage of lesions, we hypothesize this test will similarly be more sensitive for earlier lesions in patients with mpox.Tzanck smear is a useful technique that can be used to differentiate mpox from other infections at the bedside or in remote settings where PCR or additional culture techniques are not easily accessible. Infection prevention and control precautions are important to help prevent additional spread of this virus with sharp injuries, and caution with unroofing of the lesions is critical to avoid additional spread.7 Knowledge of the cytologic differences of mpox compared with other viral infections is important in the early recognition, diagnosis, and treatment of these patients.
Dermatology
A woman in her 30s with AIDS presented with acute-onset painful scattered umbilicated papulopustules and ovoid ulcerated plaques with elevated, pink borders on the face, trunk, and extremities (Figure, A). The woman also had a new-onset cough but was afebrile and denied other systemic symptoms. Due to her significant immunocompromise, the clinical presentation was highly suspicious for infection. For rapid bedside differentiation of multiple infectious etiologies, a Tzanck smear was performed by scraping the base of an ulcerated lesion and inner aspect of a pseudopustule and scraping its base with a #15 blade. These contents were placed on a glass slide, fixed, and stained with Wright-Giemsa and subsequently Papanicolaou staining to further characterize the changes seen.A, Clinical image demonstrating papulopustules and ovoid ulcerated plaques with elevated, pink borders on the elbows. B, Tzanck smear using Wright-Giemsa staining of specimen demonstrating ballooning of keratinocytes and peripheralization of nuclear material (original magnification ×20).
what is your diagnosis?
What is your diagnosis?
Herpes simplex virus
Mpox
Histoplasmosis
Molluscum contagiosum
b
0
1
0
1
female
0
0
35
31-40
null
1
augmented_female_frommale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2811083
A man in his 30s with AIDS presented with acute-onset painful scattered umbilicated papulopustules and ovoid ulcerated plaques with elevated, pink borders on the face, trunk, and extremities (Figure, A). The patient also had a new-onset cough but was afebrile and denied other systemic symptoms. Due to his significant immunocompromise, the clinical presentation was highly suspicious for infection. For rapid bedside differentiation of multiple infectious etiologies, a Tzanck smear was performed by scraping the base of an ulcerated lesion and inner aspect of a pseudopustule and scraping its base with a #15 blade. These contents were placed on a glass slide, fixed, and stained with Wright-Giemsa and subsequently Papanicolaou staining to further characterize the changes seen.A, Clinical image demonstrating papulopustules and ovoid ulcerated plaques with elevated, pink borders on the elbows. B, Tzanck smear using Wright-Giemsa staining of specimen demonstrating ballooning of keratinocytes and peripheralization of nuclear material (original magnification ×20). What Is Your Diagnosis?
Herpes simplex virus
Histoplasmosis
Molluscum contagiosum
Mpox
D. Mpox
D
Mpox
The photographs demonstrate a Tzanck smear using Wright-Giemsa stain (Figure, B) with large ballooning keratinocytes with peripheral nuclear staining seen in a patient with subsequently polymerase chain reaction (PCR)-confirmed mpox. Mpox is a member of the pox virus family, which presents with nonspecific symptoms such as fever followed by characteristic skin changes including macules, papules, vesicles, and pustules progressing with cephalocaudal spread or in the genital and/or gluteal region in the most recent outbreak. On histopathologic findings, mpox features vary depending on the stage of the lesion.1,2 Early lesions tend to demonstrate ballooning degeneration, whereas later lesions have few viable keratinocytes.2 The viral cytopathic changes, most often seen in pseudopustular lesions, include multinucleated keratinocytes with ground glass appearance and nuclear contents pushed to the periphery, resulting in a basophilic halo.2 The lack of a true pustule with collections of neutrophils in the stratum corneum have led some to describe these lesions as pseudopustular.Tzanck smear is an inexpensive, rapid, and simple technique to perform cytologic analysis from skin. Initially described in 1947 by Arnault Tzanck for diagnosing vesiculobullous disorders, it is commonly used in diagnosing herpetic infections, which have characteristic features of acantholysis, acute inflammation, and keratinocytic ballooning and specific nuclear changes of molding, multinucleation, and chromatin margination.3 The findings of acantholysis and nuclear cytopathic changes are similarly seen in histopathologic examination of herpes simplex virus (HSV). Herpetic infections may present with clustered vesicles with a pink inflammatory rim, although more verrucous lesions have been described in immunosuppressed patients.In the Tzanck smear from a lesion of mpox in this patient (Figure, B), enlarged keratinocytes were seen with expanded cytoplasm and nuclear material pushed to the periphery, and the nuclear changes associated with HSV were all absent. The observed difference in cytologic features including the nuclear material rimming or pushed to the periphery of the cells is due to the location of viral replication; herpes viruses replicate in the nucleus, whereas mpox replicates in the cytoplasm.4 Although not part of routine practice, Papanicolaou staining was performed in this case to confirm infected cells and provide additional nuclear detail because Tzanck smears in patients with mpox were not previously reported. The findings highlighted the peripheralization of nuclear material in cells with eosinophilic staining, confirming the keratinocyte origin to the cells. A background inflammatory infiltrate was noted and can be seen in most infections.Tzanck smear can also be used to diagnose other viral and fungal infections. Molluscum contagiosum, which presents with multiple umbilicated papules, demonstrates the characteristic large (30-40 µm) ovoid homogenous basophilic staining keratinocytes called Henderson-Patterson bodies that correspond to the identically named intracytoplasmic inclusions seen on histopathologic findings.5,6 Using bedside diagnostics (including Wright Giemsa staining), blastomycosis is diagnosed with 8- to 15-µm spores with broad-based budding, similar in appearance to the organism in histologic examination.5 Histoplasmosis has small (2 to 3 µm) intracellular organisms in macrophages with artifactual clearing (pseudocapsule).5 Both of these fungal infections may manifest clinically as nonhealing ulcerations, verrucous plaques, or multiple papules or nodules and can histologically demonstrate pseudoepitheliomatous hyperplasia with characteristic organisms. The use of the Tzanck smear in diagnosis of any infection depends on the timing of the lesion and is most sensitive with vesicular or pustular lesions.6 Although we only tested 1 stage of lesions, we hypothesize this test will similarly be more sensitive for earlier lesions in patients with mpox.Tzanck smear is a useful technique that can be used to differentiate mpox from other infections at the bedside or in remote settings where PCR or additional culture techniques are not easily accessible. Infection prevention and control precautions are important to help prevent additional spread of this virus with sharp injuries, and caution with unroofing of the lesions is critical to avoid additional spread.7 Knowledge of the cytologic differences of mpox compared with other viral infections is important in the early recognition, diagnosis, and treatment of these patients.
Dermatology
A patient in their 30s with AIDS presented with acute-onset painful scattered umbilicated papulopustules and ovoid ulcerated plaques with elevated, pink borders on the face, trunk, and extremities (Figure, A). The patient also had a new-onset cough but was afebrile and denied other systemic symptoms. Due to their significant immunocompromise, the clinical presentation was highly suspicious for infection. For rapid bedside differentiation of multiple infectious etiologies, a Tzanck smear was performed by scraping the base of an ulcerated lesion and inner aspect of a pseudopustule and scraping its base with a #15 blade. These contents were placed on a glass slide, fixed, and stained with Wright-Giemsa and subsequently Papanicolaou staining to further characterize the changes seen.A, Clinical image demonstrating papulopustules and ovoid ulcerated plaques with elevated, pink borders on the elbows. B, Tzanck smear using Wright-Giemsa staining of specimen demonstrating ballooning of keratinocytes and peripheralization of nuclear material (original magnification ×20).
what is your diagnosis?
What is your diagnosis?
Herpes simplex virus
Mpox
Histoplasmosis
Molluscum contagiosum
b
0
1
0
1
neutral
0
0
35
31-40
null
1
augmented_neutral_frommale
https://jamanetwork.com/journals/jama/fullarticle/2810596
An 80-year-old man with stage II bladder carcinoma (T2NXM0) and atrial fibrillation treated with apixaban presented to the emergency department with 1 week of fatigue and 2 days of dyspnea on exertion. One week prior to presentation, he received a fourth cycle of carboplatin/gemcitabine for bladder carcinoma with 6 mg of pegylated granulocyte colony-stimulating factor (G-CSF). The patient reported no anorexia, fever, melena, hematemesis, hematuria, cough, orthopnea, or peripheral edema.His vital signs were normal except for a heart rate of 103/min. His white blood cell count was 22 × 103/μL (reference, 4-11 × 103/μL), increased from 4.8 × 103/μL 8 days prior. His manual differential, which was previously normal, showed 18% bands (0%-10%), 2% metamyelocytes, 7% myelocytes, 7% promyelocytes, and 6% blasts. His hemoglobin level was 5.2 g/dL (reference, 13-17 g/dL), decreased from 7.4 g/dL, and platelets were 25 × 103/μL (reference, 150-420 × 103/μL), decreased from 268 × 103/μL 8 days prior. Ferritin was 1423 ng/mL (reference, 300-400 ng/mL). Mean corpuscular volume, prothrombin time, international normalized ratio, partial thromboplastin time, fibrinogen, haptoglobin, vitamin B12, and methylmalonic acid values were normal, and results of a direct antiglobulin test were negative. A computed tomography (CT) scan of his abdomen and pelvis was normal. He received 2 units of packed red blood cells and was admitted to the hospital. Flow cytometry identified a small population of CD34+/CD117+ cells (Figure).Left, Peripheral blood smear showing normocytic anemia with anisopoikilocytosis and leukocytosis with 6% to 8% blast forms. Right, Flow cytometry of peripheral blood demonstrating a small population of white blood cells that stained positive for CD34 and CD117, which are markers of immature myeloblasts.Esophagogastroduodenoscopy revealed 2 nonbleeding angioectasias in the stomach that were treated with argon plasma coagulation. Three days after admission, his white blood cell count was 27.7 × 103/μL with 4% peripheral blasts, hemoglobin was 7.3 g/dL, and platelet count had increased to 92 × 103/μL without a platelet transfusion.Repeat complete blood cell count with differential in 1 to 2 weeks What Would You Do Next?
Perform a bone marrow biopsy
Prescribe all-trans retinoic acid
Repeat complete blood cell count with differential in 1 to 2 weeks
Start cytoreductive therapy with hydroxyurea
Granulocyte colony-stimulating factor (G-CSF)–induced increase in peripheral blasts
C
Repeat complete blood cell count with differential in 1 to 2 weeks
The key to the correct diagnosis is recognizing that leukocytosis with immature granulocytes and mildly elevated blasts in the peripheral blood can occur after G-CSF administration. Bone marrow biopsy (choice A) is incorrect because his platelet count increased spontaneously 3 days after presentation, making malignant infiltration of his bone marrow unlikely, and recent G-CSF administration can alter bone marrow biopsy results. All-trans retinoic acid and cytoreductive therapy (options B and D) should not be prescribed because the patient did not have biopsy-proven acute promyelocytic leukemia or acute leukemia.Myeloblasts are immature leukocytes that arise from progenitor stem cells in the bone marrow, and their presence in peripheral blood suggests a perturbation of normal hematopoietic differentiation. Myeloblasts are characterized by the presence of CD34 and CD1171 antigens, identified with flow cytometry. A peripheral blood myeloblast count of greater than 20% is diagnostic of acute myeloid leukemia (AML), although AML can occur with a lower myeloblast count.2 Increased peripheral blood myeloblasts with leukopenia may be due to AML, myelodysplastic syndrome (MDS), chemotherapy, or occasionally severe infection. Causes of elevated peripheral blood myeloblasts with leukocytosis include AML, myelodysplastic syndrome, myeloproliferative neoplasms, severe infection, bone marrow damage or infiltration by fibrosis, malignancy or infection,3,4 and administration of G-CSF.G-CSF, which is produced by macrophages, T cells, endothelial cells, and fibroblasts,5 causes proliferation and differentiation of hematopoietic stem cells and progenitor cells, generating polymorphonuclear neutrophils via the precursor stages of myeloblast, promyelocyte, myelocyte, metamyelocyte, and bands. G-CSF therapy stimulates neutrophil production in the bone marrow, causing a rapid increase in peripheral blood neutrophils (emergency granulopoiesis) during severe infections. G-CSF also acts to maintain a neutrophil reserve in the bone marrow and regulates the slow release of neutrophils into the peripheral circulation.3Recombinant human G-CSF (filgrastim) received US Food and Drug Administration (FDA) approval in 1991 for prevention of chemotherapy-induced neutropenia.3,5 In 2002, the FDA approved pegfilgrastim, which is administered once per chemotherapy cycle, with at least 12 days between doses.6,7 G-CSF use is typically recommended during all chemotherapy cycles in which there is greater than 20% risk of febrile neutropenia.7 G-CSF can also be given to prevent infection in patients who experienced neutropenic complications during a prior round of chemotherapy.Patients treated with G-CSF may have peripheral blasts as high as 40% of circulating leukocytes.8 Although the mechanism of action is uncertain, induction of peripheral blasts by G-CSF may be related to differential expression of isoforms or gene variants in the G-CSF receptor. G-CSF–induced transient increase in peripheral blasts, which is most commonly reported in patients with MDS, AML, and other hematologic malignancies, is treated by withholding G-CSF therapy.3,8-10 Peripheral blasts typically resolve 1 to 2 weeks after peak blast count,8,9 at which point G-CSF therapy can be reinitiated with close monitoring of blood counts.After a 3-day hospitalization, the patient had a white blood cell count of 22.7 × 103/μL with 9% bands, 2% metamyelocytes, 2% myelocytes, 2% promyelocytes, and 4% blasts; hemoglobin was 7.3 g/dL, and platelets were 92 × 103/μL. Ten days after hospital discharge, his white blood cell count was 7.4 × 103/μL with a normal differential and no blasts; hemoglobin was 8.9 g/dL, and platelets were 384 × 103/μL. Two months later, he underwent cystectomy and received treatment with nivolumab for 5 months. However, his cancer progressed, and the patient died 5 months after his initial presentation.
General
An 80-year-old man with stage II bladder carcinoma (T2NXM0) and atrial fibrillation treated with apixaban presented to the emergency department with 1 week of fatigue and 2 days of dyspnea on exertion. One week prior to presentation, he received a fourth cycle of carboplatin/gemcitabine for bladder carcinoma with 6 mg of pegylated granulocyte colony-stimulating factor (G-CSF). The patient reported no anorexia, fever, melena, hematemesis, hematuria, cough, orthopnea, or peripheral edema.His vital signs were normal except for a heart rate of 103/min. His white blood cell count was 22 × 103/μL (reference, 4-11 × 103/μL), increased from 4.8 × 103/μL 8 days prior. His manual differential, which was previously normal, showed 18% bands (0%-10%), 2% metamyelocytes, 7% myelocytes, 7% promyelocytes, and 6% blasts. His hemoglobin level was 5.2 g/dL (reference, 13-17 g/dL), decreased from 7.4 g/dL, and platelets were 25 × 103/μL (reference, 150-420 × 103/μL), decreased from 268 × 103/μL 8 days prior. Ferritin was 1423 ng/mL (reference, 300-400 ng/mL). Mean corpuscular volume, prothrombin time, international normalized ratio, partial thromboplastin time, fibrinogen, haptoglobin, vitamin B12, and methylmalonic acid values were normal, and results of a direct antiglobulin test were negative. A computed tomography (CT) scan of his abdomen and pelvis was normal. He received 2 units of packed red blood cells and was admitted to the hospital. Flow cytometry identified a small population of CD34+/CD117+ cells (Figure).Left, Peripheral blood smear showing normocytic anemia with anisopoikilocytosis and leukocytosis with 6% to 8% blast forms. Right, Flow cytometry of peripheral blood demonstrating a small population of white blood cells that stained positive for CD34 and CD117, which are markers of immature myeloblasts.Esophagogastroduodenoscopy revealed 2 nonbleeding angioectasias in the stomach that were treated with argon plasma coagulation. Three days after admission, his white blood cell count was 27.7 × 103/μL with 4% peripheral blasts, hemoglobin was 7.3 g/dL, and platelet count had increased to 92 × 103/μL without a platelet transfusion.Repeat complete blood cell count with differential in 1 to 2 weeks
what would you do next?
What would you do next?
Start cytoreductive therapy with hydroxyurea
Prescribe all-trans retinoic acid
Repeat complete blood cell count with differential in 1 to 2 weeks
Perform a bone marrow biopsy
c
1
1
1
1
male
0
0
80
71-80
White
2
original
https://jamanetwork.com/journals/jama/fullarticle/2810596
An 80-year-old man with stage II bladder carcinoma (T2NXM0) and atrial fibrillation treated with apixaban presented to the emergency department with 1 week of fatigue and 2 days of dyspnea on exertion. One week prior to presentation, he received a fourth cycle of carboplatin/gemcitabine for bladder carcinoma with 6 mg of pegylated granulocyte colony-stimulating factor (G-CSF). The patient reported no anorexia, fever, melena, hematemesis, hematuria, cough, orthopnea, or peripheral edema.His vital signs were normal except for a heart rate of 103/min. His white blood cell count was 22 × 103/μL (reference, 4-11 × 103/μL), increased from 4.8 × 103/μL 8 days prior. His manual differential, which was previously normal, showed 18% bands (0%-10%), 2% metamyelocytes, 7% myelocytes, 7% promyelocytes, and 6% blasts. His hemoglobin level was 5.2 g/dL (reference, 13-17 g/dL), decreased from 7.4 g/dL, and platelets were 25 × 103/μL (reference, 150-420 × 103/μL), decreased from 268 × 103/μL 8 days prior. Ferritin was 1423 ng/mL (reference, 300-400 ng/mL). Mean corpuscular volume, prothrombin time, international normalized ratio, partial thromboplastin time, fibrinogen, haptoglobin, vitamin B12, and methylmalonic acid values were normal, and results of a direct antiglobulin test were negative. A computed tomography (CT) scan of his abdomen and pelvis was normal. He received 2 units of packed red blood cells and was admitted to the hospital. Flow cytometry identified a small population of CD34+/CD117+ cells (Figure).Left, Peripheral blood smear showing normocytic anemia with anisopoikilocytosis and leukocytosis with 6% to 8% blast forms. Right, Flow cytometry of peripheral blood demonstrating a small population of white blood cells that stained positive for CD34 and CD117, which are markers of immature myeloblasts.Esophagogastroduodenoscopy revealed 2 nonbleeding angioectasias in the stomach that were treated with argon plasma coagulation. Three days after admission, his white blood cell count was 27.7 × 103/μL with 4% peripheral blasts, hemoglobin was 7.3 g/dL, and platelet count had increased to 92 × 103/μL without a platelet transfusion.Repeat complete blood cell count with differential in 1 to 2 weeks What Would You Do Next?
Perform a bone marrow biopsy
Prescribe all-trans retinoic acid
Repeat complete blood cell count with differential in 1 to 2 weeks
Start cytoreductive therapy with hydroxyurea
Granulocyte colony-stimulating factor (G-CSF)–induced increase in peripheral blasts
C
Repeat complete blood cell count with differential in 1 to 2 weeks
The key to the correct diagnosis is recognizing that leukocytosis with immature granulocytes and mildly elevated blasts in the peripheral blood can occur after G-CSF administration. Bone marrow biopsy (choice A) is incorrect because his platelet count increased spontaneously 3 days after presentation, making malignant infiltration of his bone marrow unlikely, and recent G-CSF administration can alter bone marrow biopsy results. All-trans retinoic acid and cytoreductive therapy (options B and D) should not be prescribed because the patient did not have biopsy-proven acute promyelocytic leukemia or acute leukemia.Myeloblasts are immature leukocytes that arise from progenitor stem cells in the bone marrow, and their presence in peripheral blood suggests a perturbation of normal hematopoietic differentiation. Myeloblasts are characterized by the presence of CD34 and CD1171 antigens, identified with flow cytometry. A peripheral blood myeloblast count of greater than 20% is diagnostic of acute myeloid leukemia (AML), although AML can occur with a lower myeloblast count.2 Increased peripheral blood myeloblasts with leukopenia may be due to AML, myelodysplastic syndrome (MDS), chemotherapy, or occasionally severe infection. Causes of elevated peripheral blood myeloblasts with leukocytosis include AML, myelodysplastic syndrome, myeloproliferative neoplasms, severe infection, bone marrow damage or infiltration by fibrosis, malignancy or infection,3,4 and administration of G-CSF.G-CSF, which is produced by macrophages, T cells, endothelial cells, and fibroblasts,5 causes proliferation and differentiation of hematopoietic stem cells and progenitor cells, generating polymorphonuclear neutrophils via the precursor stages of myeloblast, promyelocyte, myelocyte, metamyelocyte, and bands. G-CSF therapy stimulates neutrophil production in the bone marrow, causing a rapid increase in peripheral blood neutrophils (emergency granulopoiesis) during severe infections. G-CSF also acts to maintain a neutrophil reserve in the bone marrow and regulates the slow release of neutrophils into the peripheral circulation.3Recombinant human G-CSF (filgrastim) received US Food and Drug Administration (FDA) approval in 1991 for prevention of chemotherapy-induced neutropenia.3,5 In 2002, the FDA approved pegfilgrastim, which is administered once per chemotherapy cycle, with at least 12 days between doses.6,7 G-CSF use is typically recommended during all chemotherapy cycles in which there is greater than 20% risk of febrile neutropenia.7 G-CSF can also be given to prevent infection in patients who experienced neutropenic complications during a prior round of chemotherapy.Patients treated with G-CSF may have peripheral blasts as high as 40% of circulating leukocytes.8 Although the mechanism of action is uncertain, induction of peripheral blasts by G-CSF may be related to differential expression of isoforms or gene variants in the G-CSF receptor. G-CSF–induced transient increase in peripheral blasts, which is most commonly reported in patients with MDS, AML, and other hematologic malignancies, is treated by withholding G-CSF therapy.3,8-10 Peripheral blasts typically resolve 1 to 2 weeks after peak blast count,8,9 at which point G-CSF therapy can be reinitiated with close monitoring of blood counts.After a 3-day hospitalization, the patient had a white blood cell count of 22.7 × 103/μL with 9% bands, 2% metamyelocytes, 2% myelocytes, 2% promyelocytes, and 4% blasts; hemoglobin was 7.3 g/dL, and platelets were 92 × 103/μL. Ten days after hospital discharge, his white blood cell count was 7.4 × 103/μL with a normal differential and no blasts; hemoglobin was 8.9 g/dL, and platelets were 384 × 103/μL. Two months later, he underwent cystectomy and received treatment with nivolumab for 5 months. However, his cancer progressed, and the patient died 5 months after his initial presentation.
General
An 80-year-old woman with stage II bladder carcinoma (T2NXM0) and atrial fibrillation treated with apixaban presented to the emergency department with 1 week of fatigue and 2 days of dyspnea on exertion. One week prior to presentation, she received a fourth cycle of carboplatin/gemcitabine for bladder carcinoma with 6 mg of pegylated granulocyte colony-stimulating factor (G-CSF). The woman reported no anorexia, fever, melena, hematemesis, hematuria, cough, orthopnea, or peripheral edema.Her vital signs were normal except for a heart rate of 103/min. Her white blood cell count was 22 × 103/μL (reference, 4-11 × 103/μL), increased from 4.8 × 103/μL 8 days prior. Her manual differential, which was previously normal, showed 18% bands (0%-10%), 2% metamyelocytes, 7% myelocytes, 7% promyelocytes, and 6% blasts. Her hemoglobin level was 5.2 g/dL (reference, 13-17 g/dL), decreased from 7.4 g/dL, and platelets were 25 × 103/μL (reference, 150-420 × 103/μL), decreased from 268 × 103/μL 8 days prior. Ferritin was 1423 ng/mL (reference, 300-400 ng/mL). Mean corpuscular volume, prothrombin time, international normalized ratio, partial thromboplastin time, fibrinogen, haptoglobin, vitamin B12, and methylmalonic acid values were normal, and results of a direct antiglobulin test were negative. A computed tomography (CT) scan of her abdomen and pelvis was normal. She received 2 units of packed red blood cells and was admitted to the hospital. Flow cytometry identified a small population of CD34+/CD117+ cells (Figure).Left, Peripheral blood smear showing normocytic anemia with anisopoikilocytosis and leukocytosis with 6% to 8% blast forms. Right, Flow cytometry of peripheral blood demonstrating a small population of white blood cells that stained positive for CD34 and CD117, which are markers of immature myeloblasts.Esophagogastroduodenoscopy revealed 2 nonbleeding angioectasias in the stomach that were treated with argon plasma coagulation. Three days after admission, her white blood cell count was 27.7 × 103/μL with 4% peripheral blasts, hemoglobin was 7.3 g/dL, and platelet count had increased to 92 × 103/μL without a platelet transfusion.Repeat complete blood cell count with differential in 1 to 2 weeks
what would you do next?
What would you do next?
Start cytoreductive therapy with hydroxyurea
Prescribe all-trans retinoic acid
Repeat complete blood cell count with differential in 1 to 2 weeks
Perform a bone marrow biopsy
c
1
1
1
1
female
0
0
80
71-80
White
2
augmented_female_frommale
https://jamanetwork.com/journals/jama/fullarticle/2810596
An 80-year-old man with stage II bladder carcinoma (T2NXM0) and atrial fibrillation treated with apixaban presented to the emergency department with 1 week of fatigue and 2 days of dyspnea on exertion. One week prior to presentation, he received a fourth cycle of carboplatin/gemcitabine for bladder carcinoma with 6 mg of pegylated granulocyte colony-stimulating factor (G-CSF). The patient reported no anorexia, fever, melena, hematemesis, hematuria, cough, orthopnea, or peripheral edema.His vital signs were normal except for a heart rate of 103/min. His white blood cell count was 22 × 103/μL (reference, 4-11 × 103/μL), increased from 4.8 × 103/μL 8 days prior. His manual differential, which was previously normal, showed 18% bands (0%-10%), 2% metamyelocytes, 7% myelocytes, 7% promyelocytes, and 6% blasts. His hemoglobin level was 5.2 g/dL (reference, 13-17 g/dL), decreased from 7.4 g/dL, and platelets were 25 × 103/μL (reference, 150-420 × 103/μL), decreased from 268 × 103/μL 8 days prior. Ferritin was 1423 ng/mL (reference, 300-400 ng/mL). Mean corpuscular volume, prothrombin time, international normalized ratio, partial thromboplastin time, fibrinogen, haptoglobin, vitamin B12, and methylmalonic acid values were normal, and results of a direct antiglobulin test were negative. A computed tomography (CT) scan of his abdomen and pelvis was normal. He received 2 units of packed red blood cells and was admitted to the hospital. Flow cytometry identified a small population of CD34+/CD117+ cells (Figure).Left, Peripheral blood smear showing normocytic anemia with anisopoikilocytosis and leukocytosis with 6% to 8% blast forms. Right, Flow cytometry of peripheral blood demonstrating a small population of white blood cells that stained positive for CD34 and CD117, which are markers of immature myeloblasts.Esophagogastroduodenoscopy revealed 2 nonbleeding angioectasias in the stomach that were treated with argon plasma coagulation. Three days after admission, his white blood cell count was 27.7 × 103/μL with 4% peripheral blasts, hemoglobin was 7.3 g/dL, and platelet count had increased to 92 × 103/μL without a platelet transfusion.Repeat complete blood cell count with differential in 1 to 2 weeks What Would You Do Next?
Perform a bone marrow biopsy
Prescribe all-trans retinoic acid
Repeat complete blood cell count with differential in 1 to 2 weeks
Start cytoreductive therapy with hydroxyurea
Granulocyte colony-stimulating factor (G-CSF)–induced increase in peripheral blasts
C
Repeat complete blood cell count with differential in 1 to 2 weeks
The key to the correct diagnosis is recognizing that leukocytosis with immature granulocytes and mildly elevated blasts in the peripheral blood can occur after G-CSF administration. Bone marrow biopsy (choice A) is incorrect because his platelet count increased spontaneously 3 days after presentation, making malignant infiltration of his bone marrow unlikely, and recent G-CSF administration can alter bone marrow biopsy results. All-trans retinoic acid and cytoreductive therapy (options B and D) should not be prescribed because the patient did not have biopsy-proven acute promyelocytic leukemia or acute leukemia.Myeloblasts are immature leukocytes that arise from progenitor stem cells in the bone marrow, and their presence in peripheral blood suggests a perturbation of normal hematopoietic differentiation. Myeloblasts are characterized by the presence of CD34 and CD1171 antigens, identified with flow cytometry. A peripheral blood myeloblast count of greater than 20% is diagnostic of acute myeloid leukemia (AML), although AML can occur with a lower myeloblast count.2 Increased peripheral blood myeloblasts with leukopenia may be due to AML, myelodysplastic syndrome (MDS), chemotherapy, or occasionally severe infection. Causes of elevated peripheral blood myeloblasts with leukocytosis include AML, myelodysplastic syndrome, myeloproliferative neoplasms, severe infection, bone marrow damage or infiltration by fibrosis, malignancy or infection,3,4 and administration of G-CSF.G-CSF, which is produced by macrophages, T cells, endothelial cells, and fibroblasts,5 causes proliferation and differentiation of hematopoietic stem cells and progenitor cells, generating polymorphonuclear neutrophils via the precursor stages of myeloblast, promyelocyte, myelocyte, metamyelocyte, and bands. G-CSF therapy stimulates neutrophil production in the bone marrow, causing a rapid increase in peripheral blood neutrophils (emergency granulopoiesis) during severe infections. G-CSF also acts to maintain a neutrophil reserve in the bone marrow and regulates the slow release of neutrophils into the peripheral circulation.3Recombinant human G-CSF (filgrastim) received US Food and Drug Administration (FDA) approval in 1991 for prevention of chemotherapy-induced neutropenia.3,5 In 2002, the FDA approved pegfilgrastim, which is administered once per chemotherapy cycle, with at least 12 days between doses.6,7 G-CSF use is typically recommended during all chemotherapy cycles in which there is greater than 20% risk of febrile neutropenia.7 G-CSF can also be given to prevent infection in patients who experienced neutropenic complications during a prior round of chemotherapy.Patients treated with G-CSF may have peripheral blasts as high as 40% of circulating leukocytes.8 Although the mechanism of action is uncertain, induction of peripheral blasts by G-CSF may be related to differential expression of isoforms or gene variants in the G-CSF receptor. G-CSF–induced transient increase in peripheral blasts, which is most commonly reported in patients with MDS, AML, and other hematologic malignancies, is treated by withholding G-CSF therapy.3,8-10 Peripheral blasts typically resolve 1 to 2 weeks after peak blast count,8,9 at which point G-CSF therapy can be reinitiated with close monitoring of blood counts.After a 3-day hospitalization, the patient had a white blood cell count of 22.7 × 103/μL with 9% bands, 2% metamyelocytes, 2% myelocytes, 2% promyelocytes, and 4% blasts; hemoglobin was 7.3 g/dL, and platelets were 92 × 103/μL. Ten days after hospital discharge, his white blood cell count was 7.4 × 103/μL with a normal differential and no blasts; hemoglobin was 8.9 g/dL, and platelets were 384 × 103/μL. Two months later, he underwent cystectomy and received treatment with nivolumab for 5 months. However, his cancer progressed, and the patient died 5 months after his initial presentation.
General
An 80-year-old patient with stage II bladder carcinoma (T2NXM0) and atrial fibrillation treated with apixaban presented to the emergency department with 1 week of fatigue and 2 days of dyspnea on exertion. One week prior to presentation, they received a fourth cycle of carboplatin/gemcitabine for bladder carcinoma with 6 mg of pegylated granulocyte colony-stimulating factor (G-CSF). The patient reported no anorexia, fever, melena, hematemesis, hematuria, cough, orthopnea, or peripheral edema.Their vital signs were normal except for a heart rate of 103/min. Their white blood cell count was 22 × 103/μL (reference, 4-11 × 103/μL), increased from 4.8 × 103/μL 8 days prior. Their manual differential, which was previously normal, showed 18% bands (0%-10%), 2% metamyelocytes, 7% myelocytes, 7% promyelocytes, and 6% blasts. Their hemoglobin level was 5.2 g/dL (reference, 13-17 g/dL), decreased from 7.4 g/dL, and platelets were 25 × 103/μL (reference, 150-420 × 103/μL), decreased from 268 × 103/μL 8 days prior. Ferritin was 1423 ng/mL (reference, 300-400 ng/mL). Mean corpuscular volume, prothrombin time, international normalized ratio, partial thromboplastin time, fibrinogen, haptoglobin, vitamin B12, and methylmalonic acid values were normal, and results of a direct antiglobulin test were negative. A computed tomography (CT) scan of their abdomen and pelvis was normal. They received 2 units of packed red blood cells and was admitted to the hospital. Flow cytometry identified a small population of CD34+/CD117+ cells (Figure).Left, Peripheral blood smear showing normocytic anemia with anisopoikilocytosis and leukocytosis with 6% to 8% blast forms. Right, Flow cytometry of peripheral blood demonstrating a small population of white blood cells that stained positive for CD34 and CD117, which are markers of immature myeloblasts.Esophagogastroduodenoscopy revealed 2 nonbleeding angioectasias in the stomach that were treated with argon plasma coagulation. Three days after admission, their white blood cell count was 27.7 × 103/μL with 4% peripheral blasts, hemoglobin was 7.3 g/dL, and platelet count had increased to 92 × 103/μL without a platelet transfusion.Repeat complete blood cell count with differential in 1 to 2 weeks
what would you do next?
What would you do next?
Start cytoreductive therapy with hydroxyurea
Prescribe all-trans retinoic acid
Repeat complete blood cell count with differential in 1 to 2 weeks
Perform a bone marrow biopsy
c
1
1
1
1
neutral
0
0
80
71-80
White
2
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaneurology/fullarticle/2810537
A 68-year-old man presented with progressive right-sided facial numbness and weakness, vision changes, and headaches. His symptoms began 5 years earlier with right temple numbness, which expanded to include the entire right face and mouth. He then developed progressive right facial weakness with a droop, slurred speech, and eyelid ptosis. Finally, binocular vertical and horizontal diplopia and periorbital headaches began. Trials of gabapentin and steroids provided no relief. His history was significant for a left cheek melanoma completely excised years prior, numerous cutaneous squamous and basal cell carcinomas of the bilateral head and neck removed without issue, type 2 diabetes, and hyperlipidemia. His medications included metformin and simvastatin; he had no history of smoking or drinking.On examination, he had right-sided ptosis and severe right facial weakness in a lower motor neuron pattern with temporal wasting. The right pupil was fixed and dilated. There was no afferent pupillary defect. The left pupil was appropriately sized and reactive. His right-sided vision was impaired (20/250) with blurriness. Vertical and horizontal binocular diplopia was present. Right extraocular movements were significantly limited in all directions and painful, and right-sided facial sensation was diminished to pinprick in all cranial nerve (CN) V distributions. His speech was slightly slurred. Left-sided extraocular movements were intact. His left facial sensation was intact to pinprick, and his strength was without deficit. There were no other neurological abnormalities.Initial magnetic resonance images (MRIs) were unremarkable. A biopsy of CN VII was nondiagnostic. Hemoglobin A1c was 6.8%. Tests for infection and autoimmune disease had negative results. Cerebrospinal fluid (CSF) studies showed mildly elevated protein values. Figure 1 shows the MRI at presentation.T1 turbo spin-echo fat-saturated magnetic resonance images of the brain through the skull base at presentation. What Is Your Diagnosis?
Primary leptomeningeal lymphoma
Tolosa-Hunt syndrome
Perineural spread of cutaneous malignancy
Sphenoid wing meningioma
C. Perineural spread of cutaneous malignancy
C
Perineural spread of cutaneous malignancy
The MRI of the brain and orbits revealed asymmetric enhancement of the right CNs V and VII, with denervation atrophy of the masticator space (Figure 2). Biopsy of the infraorbital nerve revealed squamous cell carcinoma, thought to have originated from a cutaneous source given his history. Immunotherapy was initiated with cemiplimab. MRI findings have remained stable over the past 2 years with no progression of disease or worsening symptoms.Same magnetic resonance image as in Figure 1B with an arrowhead denoting asymmetric enhancement of the right maxillary nerve in the foramen rotundum.The onset of multiple cranial neuropathies presents a diagnostic dilemma. Oncological diagnosis is often difficult in the absence of an obvious lesion and normal or equivocal imaging.1 This can lead to diagnostic delays and worse outcomes. This patient was seen by 20 different physicians over 5 years, including general, autonomic, and neuromuscular neurologists and neuro-ophthalmologists, before a diagnosis of malignancy was made. It can take time for perineural invasion (PNI) to become radiographically apparent, and a high index of suspicion for underlying malignancy must be maintained even with initially negative imaging.Primary leptomeningeal lymphoma (A) is a rare manifestation of primary central nervous system lymphoma that can present with multiple cranial neuropathies.2 CSF results are usually abnormal and progression of symptoms more rapid.2 Tolosa-Hunt syndrome (B) can present with painful ophthalmoplegia and multiple cranial neuropathies; however, steroids are typically effective.3 When located along the sphenoid wing, meningiomas (D) can cause multiple progressive cranial neuropathies.4 Typically visible on MRI as extra-axial homogenously enhancing masses,4 no such lesion was observed here. In this patient, the symptomatology, history, and radiographic findings are best explained by PNI of cutaneous squamous cell carcinoma (cSCC), which a biopsy confirmed.One of the most common cancers in the world, cSCC occurs frequently in the head and neck secondary to sun exposure.5 PNI refers to the process of tumor spread into nerves and the space surrounding them.6 An estimated 2% to 6% of patients with cSCC of the head and neck will have PNI.1,6 Cranial nerves V and VII are most commonly involved given their extensive anatomical distributions.1,5,6 Clinically or radiographically apparent PNI confers a poor prognosis with a reported 5-year absolute survival rate of 50%.5Treatment for cSCC of the head and neck includes surgery (if resectable) and high-dose radiotherapy and systemic therapy as appropriate. Immunotherapy with checkpoint inhibition is being used with increasing frequency and notable effectiveness.5-9 Early-phase studies demonstrated impressive response rates near 50% for advanced, unresectable, and metastatic cSCC of the head and neck, earning US Food and Drug Administration approval for 2 checkpoint inhibitors in this setting.7,8 Recently, a landmark phase 2 trial of neoadjuvant cemiplimab for stage II through IV (M0) cSCC before surgery was published, with 51% complete pathological response rates and 68% objective response rates on imaging.9 Similarly, in a recent report of 11 patients with cSCC of the head and neck with clinical PNI treated with checkpoint inhibitors, radiographic disease control was observed in 9 of 11 patients at extended follow-up.5 Many of these responses are durable and curative.The presence of both CN V and VII symptoms in a patient with a history of head and neck cSCC should raise suspicion for perineural recurrence of disease. While initial MRI may be negative, radiographic manifestation of PNI often occurs over time as the disease progresses and symptoms worsen. Expert review by a neuroradiologist can be helpful in identifying this finding earlier in the disease course. Mounting evidence suggests that immunotherapy is a promising treatment option for this population and should be considered as first-line therapy.
Neurology
A 68-year-old man presented with progressive right-sided facial numbness and weakness, vision changes, and headaches. His symptoms began 5 years earlier with right temple numbness, which expanded to include the entire right face and mouth. He then developed progressive right facial weakness with a droop, slurred speech, and eyelid ptosis. Finally, binocular vertical and horizontal diplopia and periorbital headaches began. Trials of gabapentin and steroids provided no relief. His history was significant for a left cheek melanoma completely excised years prior, numerous cutaneous squamous and basal cell carcinomas of the bilateral head and neck removed without issue, type 2 diabetes, and hyperlipidemia. His medications included metformin and simvastatin; he had no history of smoking or drinking.On examination, he had right-sided ptosis and severe right facial weakness in a lower motor neuron pattern with temporal wasting. The right pupil was fixed and dilated. There was no afferent pupillary defect. The left pupil was appropriately sized and reactive. His right-sided vision was impaired (20/250) with blurriness. Vertical and horizontal binocular diplopia was present. Right extraocular movements were significantly limited in all directions and painful, and right-sided facial sensation was diminished to pinprick in all cranial nerve (CN) V distributions. His speech was slightly slurred. Left-sided extraocular movements were intact. His left facial sensation was intact to pinprick, and his strength was without deficit. There were no other neurological abnormalities.Initial magnetic resonance images (MRIs) were unremarkable. A biopsy of CN VII was nondiagnostic. Hemoglobin A1c was 6.8%. Tests for infection and autoimmune disease had negative results. Cerebrospinal fluid (CSF) studies showed mildly elevated protein values. Figure 1 shows the MRI at presentation.T1 turbo spin-echo fat-saturated magnetic resonance images of the brain through the skull base at presentation.
what is your diagnosis?
What is your diagnosis?
Tolosa-Hunt syndrome
Perineural spread of cutaneous malignancy
Sphenoid wing meningioma
Primary leptomeningeal lymphoma
b
1
1
1
1
male
0
0
68
61-70
null
3
original
https://jamanetwork.com/journals/jamaneurology/fullarticle/2810537
A 68-year-old man presented with progressive right-sided facial numbness and weakness, vision changes, and headaches. His symptoms began 5 years earlier with right temple numbness, which expanded to include the entire right face and mouth. He then developed progressive right facial weakness with a droop, slurred speech, and eyelid ptosis. Finally, binocular vertical and horizontal diplopia and periorbital headaches began. Trials of gabapentin and steroids provided no relief. His history was significant for a left cheek melanoma completely excised years prior, numerous cutaneous squamous and basal cell carcinomas of the bilateral head and neck removed without issue, type 2 diabetes, and hyperlipidemia. His medications included metformin and simvastatin; he had no history of smoking or drinking.On examination, he had right-sided ptosis and severe right facial weakness in a lower motor neuron pattern with temporal wasting. The right pupil was fixed and dilated. There was no afferent pupillary defect. The left pupil was appropriately sized and reactive. His right-sided vision was impaired (20/250) with blurriness. Vertical and horizontal binocular diplopia was present. Right extraocular movements were significantly limited in all directions and painful, and right-sided facial sensation was diminished to pinprick in all cranial nerve (CN) V distributions. His speech was slightly slurred. Left-sided extraocular movements were intact. His left facial sensation was intact to pinprick, and his strength was without deficit. There were no other neurological abnormalities.Initial magnetic resonance images (MRIs) were unremarkable. A biopsy of CN VII was nondiagnostic. Hemoglobin A1c was 6.8%. Tests for infection and autoimmune disease had negative results. Cerebrospinal fluid (CSF) studies showed mildly elevated protein values. Figure 1 shows the MRI at presentation.T1 turbo spin-echo fat-saturated magnetic resonance images of the brain through the skull base at presentation. What Is Your Diagnosis?
Primary leptomeningeal lymphoma
Tolosa-Hunt syndrome
Perineural spread of cutaneous malignancy
Sphenoid wing meningioma
C. Perineural spread of cutaneous malignancy
C
Perineural spread of cutaneous malignancy
The MRI of the brain and orbits revealed asymmetric enhancement of the right CNs V and VII, with denervation atrophy of the masticator space (Figure 2). Biopsy of the infraorbital nerve revealed squamous cell carcinoma, thought to have originated from a cutaneous source given his history. Immunotherapy was initiated with cemiplimab. MRI findings have remained stable over the past 2 years with no progression of disease or worsening symptoms.Same magnetic resonance image as in Figure 1B with an arrowhead denoting asymmetric enhancement of the right maxillary nerve in the foramen rotundum.The onset of multiple cranial neuropathies presents a diagnostic dilemma. Oncological diagnosis is often difficult in the absence of an obvious lesion and normal or equivocal imaging.1 This can lead to diagnostic delays and worse outcomes. This patient was seen by 20 different physicians over 5 years, including general, autonomic, and neuromuscular neurologists and neuro-ophthalmologists, before a diagnosis of malignancy was made. It can take time for perineural invasion (PNI) to become radiographically apparent, and a high index of suspicion for underlying malignancy must be maintained even with initially negative imaging.Primary leptomeningeal lymphoma (A) is a rare manifestation of primary central nervous system lymphoma that can present with multiple cranial neuropathies.2 CSF results are usually abnormal and progression of symptoms more rapid.2 Tolosa-Hunt syndrome (B) can present with painful ophthalmoplegia and multiple cranial neuropathies; however, steroids are typically effective.3 When located along the sphenoid wing, meningiomas (D) can cause multiple progressive cranial neuropathies.4 Typically visible on MRI as extra-axial homogenously enhancing masses,4 no such lesion was observed here. In this patient, the symptomatology, history, and radiographic findings are best explained by PNI of cutaneous squamous cell carcinoma (cSCC), which a biopsy confirmed.One of the most common cancers in the world, cSCC occurs frequently in the head and neck secondary to sun exposure.5 PNI refers to the process of tumor spread into nerves and the space surrounding them.6 An estimated 2% to 6% of patients with cSCC of the head and neck will have PNI.1,6 Cranial nerves V and VII are most commonly involved given their extensive anatomical distributions.1,5,6 Clinically or radiographically apparent PNI confers a poor prognosis with a reported 5-year absolute survival rate of 50%.5Treatment for cSCC of the head and neck includes surgery (if resectable) and high-dose radiotherapy and systemic therapy as appropriate. Immunotherapy with checkpoint inhibition is being used with increasing frequency and notable effectiveness.5-9 Early-phase studies demonstrated impressive response rates near 50% for advanced, unresectable, and metastatic cSCC of the head and neck, earning US Food and Drug Administration approval for 2 checkpoint inhibitors in this setting.7,8 Recently, a landmark phase 2 trial of neoadjuvant cemiplimab for stage II through IV (M0) cSCC before surgery was published, with 51% complete pathological response rates and 68% objective response rates on imaging.9 Similarly, in a recent report of 11 patients with cSCC of the head and neck with clinical PNI treated with checkpoint inhibitors, radiographic disease control was observed in 9 of 11 patients at extended follow-up.5 Many of these responses are durable and curative.The presence of both CN V and VII symptoms in a patient with a history of head and neck cSCC should raise suspicion for perineural recurrence of disease. While initial MRI may be negative, radiographic manifestation of PNI often occurs over time as the disease progresses and symptoms worsen. Expert review by a neuroradiologist can be helpful in identifying this finding earlier in the disease course. Mounting evidence suggests that immunotherapy is a promising treatment option for this population and should be considered as first-line therapy.
Neurology
A 68-year-old woman presented with progressive right-sided facial numbness and weakness, vision changes, and headaches. Her symptoms began 5 years earlier with right temple numbness, which expanded to include the entire right face and mouth. She then developed progressive right facial weakness with a droop, slurred speech, and eyelid ptosis. Finally, binocular vertical and horizontal diplopia and periorbital headaches began. Trials of gabapentin and steroids provided no relief. Her history was significant for a left cheek melanoma completely excised years prior, numerous cutaneous squamous and basal cell carcinomas of the bilateral head and neck removed without issue, type 2 diabetes, and hyperlipidemia. Her medications included metformin and simvastatin; she had no history of smoking or drinking.On examination, she had right-sided ptosis and severe right facial weakness in a lower motor neuron pattern with temporal wasting. The right pupil was fixed and dilated. There was no afferent pupillary defect. The left pupil was appropriately sized and reactive. Her right-sided vision was impaired (20/250) with blurriness. Vertical and horizontal binocular diplopia was present. Right extraocular movements were significantly limited in all directions and painful, and right-sided facial sensation was diminished to pinprick in all cranial nerve (CN) V distributions. Her speech was slightly slurred. Left-sided extraocular movements were intact. Her left facial sensation was intact to pinprick, and her strength was without deficit. There were no other neurological abnormalities.Initial magnetic resonance images (MRIs) were unremarkable. A biopsy of CN VII was nondiagnostic. Hemoglobin A1c was 6.8%. Tests for infection and autoimmune disease had negative results. Cerebrospinal fluid (CSF) studies showed mildly elevated protein values. Figure 1 shows the MRI at presentation.T1 turbo spin-echo fat-saturated magnetic resonance images of the brain through the skull base at presentation.
what is your diagnosis?
What is your diagnosis?
Tolosa-Hunt syndrome
Perineural spread of cutaneous malignancy
Sphenoid wing meningioma
Primary leptomeningeal lymphoma
b
1
1
1
1
female
0
0
68
61-70
null
3
augmented_female_frommale
https://jamanetwork.com/journals/jamaneurology/fullarticle/2810537
A 68-year-old man presented with progressive right-sided facial numbness and weakness, vision changes, and headaches. His symptoms began 5 years earlier with right temple numbness, which expanded to include the entire right face and mouth. He then developed progressive right facial weakness with a droop, slurred speech, and eyelid ptosis. Finally, binocular vertical and horizontal diplopia and periorbital headaches began. Trials of gabapentin and steroids provided no relief. His history was significant for a left cheek melanoma completely excised years prior, numerous cutaneous squamous and basal cell carcinomas of the bilateral head and neck removed without issue, type 2 diabetes, and hyperlipidemia. His medications included metformin and simvastatin; he had no history of smoking or drinking.On examination, he had right-sided ptosis and severe right facial weakness in a lower motor neuron pattern with temporal wasting. The right pupil was fixed and dilated. There was no afferent pupillary defect. The left pupil was appropriately sized and reactive. His right-sided vision was impaired (20/250) with blurriness. Vertical and horizontal binocular diplopia was present. Right extraocular movements were significantly limited in all directions and painful, and right-sided facial sensation was diminished to pinprick in all cranial nerve (CN) V distributions. His speech was slightly slurred. Left-sided extraocular movements were intact. His left facial sensation was intact to pinprick, and his strength was without deficit. There were no other neurological abnormalities.Initial magnetic resonance images (MRIs) were unremarkable. A biopsy of CN VII was nondiagnostic. Hemoglobin A1c was 6.8%. Tests for infection and autoimmune disease had negative results. Cerebrospinal fluid (CSF) studies showed mildly elevated protein values. Figure 1 shows the MRI at presentation.T1 turbo spin-echo fat-saturated magnetic resonance images of the brain through the skull base at presentation. What Is Your Diagnosis?
Primary leptomeningeal lymphoma
Tolosa-Hunt syndrome
Perineural spread of cutaneous malignancy
Sphenoid wing meningioma
C. Perineural spread of cutaneous malignancy
C
Perineural spread of cutaneous malignancy
The MRI of the brain and orbits revealed asymmetric enhancement of the right CNs V and VII, with denervation atrophy of the masticator space (Figure 2). Biopsy of the infraorbital nerve revealed squamous cell carcinoma, thought to have originated from a cutaneous source given his history. Immunotherapy was initiated with cemiplimab. MRI findings have remained stable over the past 2 years with no progression of disease or worsening symptoms.Same magnetic resonance image as in Figure 1B with an arrowhead denoting asymmetric enhancement of the right maxillary nerve in the foramen rotundum.The onset of multiple cranial neuropathies presents a diagnostic dilemma. Oncological diagnosis is often difficult in the absence of an obvious lesion and normal or equivocal imaging.1 This can lead to diagnostic delays and worse outcomes. This patient was seen by 20 different physicians over 5 years, including general, autonomic, and neuromuscular neurologists and neuro-ophthalmologists, before a diagnosis of malignancy was made. It can take time for perineural invasion (PNI) to become radiographically apparent, and a high index of suspicion for underlying malignancy must be maintained even with initially negative imaging.Primary leptomeningeal lymphoma (A) is a rare manifestation of primary central nervous system lymphoma that can present with multiple cranial neuropathies.2 CSF results are usually abnormal and progression of symptoms more rapid.2 Tolosa-Hunt syndrome (B) can present with painful ophthalmoplegia and multiple cranial neuropathies; however, steroids are typically effective.3 When located along the sphenoid wing, meningiomas (D) can cause multiple progressive cranial neuropathies.4 Typically visible on MRI as extra-axial homogenously enhancing masses,4 no such lesion was observed here. In this patient, the symptomatology, history, and radiographic findings are best explained by PNI of cutaneous squamous cell carcinoma (cSCC), which a biopsy confirmed.One of the most common cancers in the world, cSCC occurs frequently in the head and neck secondary to sun exposure.5 PNI refers to the process of tumor spread into nerves and the space surrounding them.6 An estimated 2% to 6% of patients with cSCC of the head and neck will have PNI.1,6 Cranial nerves V and VII are most commonly involved given their extensive anatomical distributions.1,5,6 Clinically or radiographically apparent PNI confers a poor prognosis with a reported 5-year absolute survival rate of 50%.5Treatment for cSCC of the head and neck includes surgery (if resectable) and high-dose radiotherapy and systemic therapy as appropriate. Immunotherapy with checkpoint inhibition is being used with increasing frequency and notable effectiveness.5-9 Early-phase studies demonstrated impressive response rates near 50% for advanced, unresectable, and metastatic cSCC of the head and neck, earning US Food and Drug Administration approval for 2 checkpoint inhibitors in this setting.7,8 Recently, a landmark phase 2 trial of neoadjuvant cemiplimab for stage II through IV (M0) cSCC before surgery was published, with 51% complete pathological response rates and 68% objective response rates on imaging.9 Similarly, in a recent report of 11 patients with cSCC of the head and neck with clinical PNI treated with checkpoint inhibitors, radiographic disease control was observed in 9 of 11 patients at extended follow-up.5 Many of these responses are durable and curative.The presence of both CN V and VII symptoms in a patient with a history of head and neck cSCC should raise suspicion for perineural recurrence of disease. While initial MRI may be negative, radiographic manifestation of PNI often occurs over time as the disease progresses and symptoms worsen. Expert review by a neuroradiologist can be helpful in identifying this finding earlier in the disease course. Mounting evidence suggests that immunotherapy is a promising treatment option for this population and should be considered as first-line therapy.
Neurology
A 68-year-old patient presented with progressive right-sided facial numbness and weakness, vision changes, and headaches. Their symptoms began 5 years earlier with right temple numbness, which expanded to include the entire right face and mouth. They then developed progressive right facial weakness with a droop, slurred speech, and eyelid ptosis. Finally, binocular vertical and horizontal diplopia and periorbital headaches began. Trials of gabapentin and steroids provided no relief. Their history was significant for a left cheek melanoma completely excised years prior, numerous cutaneous squamous and basal cell carcinomas of the bilateral head and neck removed without issue, type 2 diabetes, and hyperlipidemia. Their medications included metformin and simvastatin; they had no history of smoking or drinking.On examination, they had right-sided ptosis and severe right facial weakness in a lower motor neuron pattern with temporal wasting. The right pupil was fixed and dilated. There was no afferent pupillary defect. The left pupil was appropriately sized and reactive. Their right-sided vision was impaired (20/250) with blurriness. Vertical and horizontal binocular diplopia was present. Right extraocular movements were significantly limited in all directions and painful, and right-sided facial sensation was diminished to pinprick in all cranial nerve (CN) V distributions. Their speech was slightly slurred. Left-sided extraocular movements were intact. Their left facial sensation was intact to pinprick, and their strength was without deficit. There were no other neurological abnormalities.Initial magnetic resonance images (MRIs) were unremarkable. A biopsy of CN VII was nondiagnostic. Hemoglobin A1c was 6.8%. Tests for infection and autoimmune disease had negative results. Cerebrospinal fluid (CSF) studies showed mildly elevated protein values. Figure 1 shows the MRI at presentation.T1 turbo spin-echo fat-saturated magnetic resonance images of the brain through the skull base at presentation.
what is your diagnosis?
What is your diagnosis?
Tolosa-Hunt syndrome
Perineural spread of cutaneous malignancy
Sphenoid wing meningioma
Primary leptomeningeal lymphoma
b
1
1
1
1
neutral
0
0
68
61-70
null
3
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2810749
A 31-year-old man presented with left cervical and left inguinal masses. He reported intermittent itching and night sweats for 2 years. He denied fever, weight loss, shortness of breath, rashes, diarrhea, and neurological symptoms. On a preemployment evaluation, the patient was told he had a high white blood cell count 2 years ago. On examination, there was left cervical and inguinal lymphadenopathy and no other organomegaly. Complete blood cell count and peripheral blood smear showed marked leukocytosis, with a white blood cell count of 22 340/μL, an absolute neutrophil count of 5360/μL, and 55% eosinophils with an absolute eosinophil count of 12 290/μL (to convert all to cells ×109/L, multiply by 0.001). Vitamin B12 was markedly elevated at more than 4000 pg/mL (to convert to pmol/L, multiply by 0.7378). The erythrocyte sedimentation rate was 5 mm/h. Lactate dehydrogenase was 180 U/L, and alkaline phosphatase was 81 U/L (to convert both to μkat/L, multiply by 0.0167). Evaluations for HIV and hepatitis B and C were all negative. Serum creatinine was 0.76 mg/dL (to convert to μmol/L, multiply by 88.4); alanine aminotransferase and aspartate aminotransferase were 11 U/L and 9.9 U/L, respectively (to convert to μkat/L, multiply by 0.0167); and total bilirubin was 0.35 mg/dL (to convert to μmol/L, multiply by 17.104). Bone marrow biopsy showed hypercellular marrow (cellularity of 100%), myeloid hyperplasia, increased eosinophils with some dysplasia, and a blast count of 2%. Positron emission tomographic–computed tomographic scan showed a left upper cervical lymph node of 2.6 cm and a left inguinal lymph node of 3.1 × 2.3 cm with an standardized uptake value max of 5.7 (Figure, A). Left inguinal lymph node biopsy showed partial involvement by atypical cells with high proliferation index (Ki-67 >95%) that were positive for CD3, CD4, CD8, BCL2, and TDT, suggestive of T-cell lymphoblastic lymphoma/leukemia (Figure, B).A, Positron emission tomographic–computed tomographic (PET/CT) scan of the head and neck at presentation showing a left upper cervical lymph node of 2.6 cm (arrowhead). B, Lymph node biopsy immunohistochemical stain with terminal deoxynucleotidyl transferase. The inset shows interphase fluorescence in situ hybridization for FIP1L1::PDGFRA rearrangement (positive). C, PET/CT 12 weeks after treatment initiation.Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions What Is Your Diagnosis?
Kimura disease
Classic Hodgkin lymphoma
T-cell acute lymphoblastic lymphoma/leukemia
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
D. Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
D
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
The differential diagnoses in young men with eosinophilia and a neck mass can be wide. The typical presentation of Kimura disease is a painless, unilateral swelling of cervical lymph nodes in young adult male patients with peripheral blood eosinophilia and elevated immunoglobulin E levels. Histological evaluation, however, would show hyperplastic reactive follicles with eosinophilic infiltrates in a background of polymorphous inflammatory cells, a picture inconsistent with the presented case. Although Hodgkin lymphoma can present with eosinophilia and neck mass in young adults, the biopsy was inconsistent with the typical morphology of Reed-Sternberg cells in the background of nonneoplastic inflammatory cells. The diagnosis of T-cell acute lymphoblastic lymphoma/leukemia is reasonable given the morphology from lymph node biopsy; however, in the diagnosis hierarchy, myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions (MLN-TK) would supersede other myeloid and lymphoid cancers.1In the fifth edition of the World Health Organization Classification of Haematolymphoid Tumours, MLN-TK refers to BCR::ABL1–negative myeloid or lymphoid neoplasms with gene rearrangements leading to an activated tyrosine-kinase domain that results in cell-signaling dysregulation.1 This includes PDGFRA, PDGFRB, FGFR1, JAK2, and FLT3 rearrangements, in addition to ETV6::ABL1, ETV6::FGFR2, ETV6::LYN, ETV6::NTRK3, RANBP2::ALK, BCR::RET, and FGFR1OP::RET fusions.1 An increase in vitamin B12 is often found in patients with PDGFRA and PDGFRB disease.2,3 Most patients are male with chronic myeloid neoplasms with eosinophilia; however, patients rarely present with acute myeloid or T-cell leukemia or extramedullary disease.2Fluorescence in situ hybridization (FISH) was positive for FIP1L1::PDGFRA (4q12) fusion in 91% of nuclei, while karyotype was normal (46, XY in 18 cells), as a cryptic loss of the CHIC2 gene causes this fusion. Next, generation sequencing panel for myeloid neoplasms was negative for variants in targeted regions of the genes commonly involved in myeloid neoplasms. Combining imatinib with chemotherapy is acceptable in patients with acute leukemia or extramedullary diseases, although monotherapy with imatinib has shown promising results.4 The patient was concerned about fertility, as he and his partner were starting a family, and decided to avoid chemotherapy. The patient was concomitantly initiated on 1 mg/kg of prednisone for 7 days and imatinib at 600 mg daily. The eosinophil counts normalized after 2 days, and bone marrow evaluation after 4 weeks showed resolution of eosinophilic infiltration; however, FISH results were 3% positive for FIP1L1::PDGFRA fusion. Repeat bone marrow evaluation after 8 weeks showed complete resolution of the FISH abnormality, and the imatinib dose was reduced to 400 mg daily. Although studies have confirmed deep and durable remission with a starting dose of imatinib, 100 mg, we elected to start with a high dose given the patient’s young age and blastic presentation.5,6 Follow-up positron emission tomographic–computed tomographic scan 12 weeks after initiation of imatinib was consistent with complete metabolic remission (Figure, C). Complete hematological remission is achieved in more than 95% of patients with imatinib and is typically seen early (within weeks of treatment initiation). Cytogenetic/molecular response can be achieved within 3 to 6 months of treatment initiation.7 In patients who achieved molecular remission, the imatinib dose can be reduced to 100 thrice weekly or weekly.8 Treatment-free remission as the experience with chronic myeloid leukemia has been tried, although with limited experience. The success rate is around 60%, with most patients relapsing within the first 10 months. Reinitiating of imatinib restores hematological remission in almost all patients.9,10The current patient was in complete remission 12 months from his treatment and receiving imatinib. Evaluating patients suspected of MLN-TK is critical, as treatment and prognosis may drastically change.
Oncology
A 31-year-old man presented with left cervical and left inguinal masses. He reported intermittent itching and night sweats for 2 years. He denied fever, weight loss, shortness of breath, rashes, diarrhea, and neurological symptoms. On a preemployment evaluation, the patient was told he had a high white blood cell count 2 years ago. On examination, there was left cervical and inguinal lymphadenopathy and no other organomegaly. Complete blood cell count and peripheral blood smear showed marked leukocytosis, with a white blood cell count of 22 340/μL, an absolute neutrophil count of 5360/μL, and 55% eosinophils with an absolute eosinophil count of 12 290/μL (to convert all to cells ×109/L, multiply by 0.001). Vitamin B12 was markedly elevated at more than 4000 pg/mL (to convert to pmol/L, multiply by 0.7378). The erythrocyte sedimentation rate was 5 mm/h. Lactate dehydrogenase was 180 U/L, and alkaline phosphatase was 81 U/L (to convert both to μkat/L, multiply by 0.0167). Evaluations for HIV and hepatitis B and C were all negative. Serum creatinine was 0.76 mg/dL (to convert to μmol/L, multiply by 88.4); alanine aminotransferase and aspartate aminotransferase were 11 U/L and 9.9 U/L, respectively (to convert to μkat/L, multiply by 0.0167); and total bilirubin was 0.35 mg/dL (to convert to μmol/L, multiply by 17.104). Bone marrow biopsy showed hypercellular marrow (cellularity of 100%), myeloid hyperplasia, increased eosinophils with some dysplasia, and a blast count of 2%. Positron emission tomographic–computed tomographic scan showed a left upper cervical lymph node of 2.6 cm and a left inguinal lymph node of 3.1 × 2.3 cm with an standardized uptake value max of 5.7 (Figure, A). Left inguinal lymph node biopsy showed partial involvement by atypical cells with high proliferation index (Ki-67 >95%) that were positive for CD3, CD4, CD8, BCL2, and TDT, suggestive of T-cell lymphoblastic lymphoma/leukemia (Figure, B).A, Positron emission tomographic–computed tomographic (PET/CT) scan of the head and neck at presentation showing a left upper cervical lymph node of 2.6 cm (arrowhead). B, Lymph node biopsy immunohistochemical stain with terminal deoxynucleotidyl transferase. The inset shows interphase fluorescence in situ hybridization for FIP1L1::PDGFRA rearrangement (positive). C, PET/CT 12 weeks after treatment initiation.Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
what is your diagnosis?
What is your diagnosis?
T-cell acute lymphoblastic lymphoma/leukemia
Kimura disease
Classic Hodgkin lymphoma
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
d
1
1
1
1
male
0
0
31
31-40
White
4
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2810749
A 31-year-old man presented with left cervical and left inguinal masses. He reported intermittent itching and night sweats for 2 years. He denied fever, weight loss, shortness of breath, rashes, diarrhea, and neurological symptoms. On a preemployment evaluation, the patient was told he had a high white blood cell count 2 years ago. On examination, there was left cervical and inguinal lymphadenopathy and no other organomegaly. Complete blood cell count and peripheral blood smear showed marked leukocytosis, with a white blood cell count of 22 340/μL, an absolute neutrophil count of 5360/μL, and 55% eosinophils with an absolute eosinophil count of 12 290/μL (to convert all to cells ×109/L, multiply by 0.001). Vitamin B12 was markedly elevated at more than 4000 pg/mL (to convert to pmol/L, multiply by 0.7378). The erythrocyte sedimentation rate was 5 mm/h. Lactate dehydrogenase was 180 U/L, and alkaline phosphatase was 81 U/L (to convert both to μkat/L, multiply by 0.0167). Evaluations for HIV and hepatitis B and C were all negative. Serum creatinine was 0.76 mg/dL (to convert to μmol/L, multiply by 88.4); alanine aminotransferase and aspartate aminotransferase were 11 U/L and 9.9 U/L, respectively (to convert to μkat/L, multiply by 0.0167); and total bilirubin was 0.35 mg/dL (to convert to μmol/L, multiply by 17.104). Bone marrow biopsy showed hypercellular marrow (cellularity of 100%), myeloid hyperplasia, increased eosinophils with some dysplasia, and a blast count of 2%. Positron emission tomographic–computed tomographic scan showed a left upper cervical lymph node of 2.6 cm and a left inguinal lymph node of 3.1 × 2.3 cm with an standardized uptake value max of 5.7 (Figure, A). Left inguinal lymph node biopsy showed partial involvement by atypical cells with high proliferation index (Ki-67 >95%) that were positive for CD3, CD4, CD8, BCL2, and TDT, suggestive of T-cell lymphoblastic lymphoma/leukemia (Figure, B).A, Positron emission tomographic–computed tomographic (PET/CT) scan of the head and neck at presentation showing a left upper cervical lymph node of 2.6 cm (arrowhead). B, Lymph node biopsy immunohistochemical stain with terminal deoxynucleotidyl transferase. The inset shows interphase fluorescence in situ hybridization for FIP1L1::PDGFRA rearrangement (positive). C, PET/CT 12 weeks after treatment initiation.Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions What Is Your Diagnosis?
Kimura disease
Classic Hodgkin lymphoma
T-cell acute lymphoblastic lymphoma/leukemia
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
D. Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
D
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
The differential diagnoses in young men with eosinophilia and a neck mass can be wide. The typical presentation of Kimura disease is a painless, unilateral swelling of cervical lymph nodes in young adult male patients with peripheral blood eosinophilia and elevated immunoglobulin E levels. Histological evaluation, however, would show hyperplastic reactive follicles with eosinophilic infiltrates in a background of polymorphous inflammatory cells, a picture inconsistent with the presented case. Although Hodgkin lymphoma can present with eosinophilia and neck mass in young adults, the biopsy was inconsistent with the typical morphology of Reed-Sternberg cells in the background of nonneoplastic inflammatory cells. The diagnosis of T-cell acute lymphoblastic lymphoma/leukemia is reasonable given the morphology from lymph node biopsy; however, in the diagnosis hierarchy, myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions (MLN-TK) would supersede other myeloid and lymphoid cancers.1In the fifth edition of the World Health Organization Classification of Haematolymphoid Tumours, MLN-TK refers to BCR::ABL1–negative myeloid or lymphoid neoplasms with gene rearrangements leading to an activated tyrosine-kinase domain that results in cell-signaling dysregulation.1 This includes PDGFRA, PDGFRB, FGFR1, JAK2, and FLT3 rearrangements, in addition to ETV6::ABL1, ETV6::FGFR2, ETV6::LYN, ETV6::NTRK3, RANBP2::ALK, BCR::RET, and FGFR1OP::RET fusions.1 An increase in vitamin B12 is often found in patients with PDGFRA and PDGFRB disease.2,3 Most patients are male with chronic myeloid neoplasms with eosinophilia; however, patients rarely present with acute myeloid or T-cell leukemia or extramedullary disease.2Fluorescence in situ hybridization (FISH) was positive for FIP1L1::PDGFRA (4q12) fusion in 91% of nuclei, while karyotype was normal (46, XY in 18 cells), as a cryptic loss of the CHIC2 gene causes this fusion. Next, generation sequencing panel for myeloid neoplasms was negative for variants in targeted regions of the genes commonly involved in myeloid neoplasms. Combining imatinib with chemotherapy is acceptable in patients with acute leukemia or extramedullary diseases, although monotherapy with imatinib has shown promising results.4 The patient was concerned about fertility, as he and his partner were starting a family, and decided to avoid chemotherapy. The patient was concomitantly initiated on 1 mg/kg of prednisone for 7 days and imatinib at 600 mg daily. The eosinophil counts normalized after 2 days, and bone marrow evaluation after 4 weeks showed resolution of eosinophilic infiltration; however, FISH results were 3% positive for FIP1L1::PDGFRA fusion. Repeat bone marrow evaluation after 8 weeks showed complete resolution of the FISH abnormality, and the imatinib dose was reduced to 400 mg daily. Although studies have confirmed deep and durable remission with a starting dose of imatinib, 100 mg, we elected to start with a high dose given the patient’s young age and blastic presentation.5,6 Follow-up positron emission tomographic–computed tomographic scan 12 weeks after initiation of imatinib was consistent with complete metabolic remission (Figure, C). Complete hematological remission is achieved in more than 95% of patients with imatinib and is typically seen early (within weeks of treatment initiation). Cytogenetic/molecular response can be achieved within 3 to 6 months of treatment initiation.7 In patients who achieved molecular remission, the imatinib dose can be reduced to 100 thrice weekly or weekly.8 Treatment-free remission as the experience with chronic myeloid leukemia has been tried, although with limited experience. The success rate is around 60%, with most patients relapsing within the first 10 months. Reinitiating of imatinib restores hematological remission in almost all patients.9,10The current patient was in complete remission 12 months from his treatment and receiving imatinib. Evaluating patients suspected of MLN-TK is critical, as treatment and prognosis may drastically change.
Oncology
A 31-year-old woman presented with left cervical and left inguinal masses. She reported intermittent itching and night sweats for 2 years. She denied fever, weight loss, shortness of breath, rashes, diarrhea, and neurological symptoms. On a preemployment evaluation, the woman was told she had a high white blood cell count 2 years ago. On examination, there was left cervical and inguinal lymphadenopathy and no other organomegaly. Complete blood cell count and peripheral blood smear showed marked leukocytosis, with a white blood cell count of 22 340/μL, an absolute neutrophil count of 5360/μL, and 55% eosinophils with an absolute eosinophil count of 12 290/μL (to convert all to cells ×109/L, multiply by 0.001). Vitamin B12 was markedly elevated at more than 4000 pg/mL (to convert to pmol/L, multiply by 0.7378). The erythrocyte sedimentation rate was 5 mm/h. Lactate dehydrogenase was 180 U/L, and alkaline phosphatase was 81 U/L (to convert both to μkat/L, multiply by 0.0167). Evaluations for HIV and hepatitis B and C were all negative. Serum creatinine was 0.76 mg/dL (to convert to μmol/L, multiply by 88.4); alanine aminotransferase and aspartate aminotransferase were 11 U/L and 9.9 U/L, respectively (to convert to μkat/L, multiply by 0.0167); and total bilirubin was 0.35 mg/dL (to convert to μmol/L, multiply by 17.104). Bone marrow biopsy showed hypercellular marrow (cellularity of 100%), myeloid hyperplasia, increased eosinophils with some dysplasia, and a blast count of 2%. Positron emission tomographic–computed tomographic scan showed a left upper cervical lymph node of 2.6 cm and a left inguinal lymph node of 3.1 × 2.3 cm with an standardized uptake value max of 5.7 (Figure, A). Left inguinal lymph node biopsy showed partial involvement by atypical cells with high proliferation index (Ki-67 >95%) that were positive for CD3, CD4, CD8, BCL2, and TDT, suggestive of T-cell lymphoblastic lymphoma/leukemia (Figure, B).A, Positron emission tomographic–computed tomographic (PET/CT) scan of the head and neck at presentation showing a left upper cervical lymph node of 2.6 cm (arrowhead). B, Lymph node biopsy immunohistochemical stain with terminal deoxynucleotidyl transferase. The inset shows interphase fluorescence in situ hybridization for FIP1L1::PDGFRA rearrangement (positive). C, PET/CT 12 weeks after treatment initiation.Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
what is your diagnosis?
What is your diagnosis?
T-cell acute lymphoblastic lymphoma/leukemia
Kimura disease
Classic Hodgkin lymphoma
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
d
1
1
1
1
female
0
0
31
31-40
White
4
augmented_female_frommale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2810749
A 31-year-old man presented with left cervical and left inguinal masses. He reported intermittent itching and night sweats for 2 years. He denied fever, weight loss, shortness of breath, rashes, diarrhea, and neurological symptoms. On a preemployment evaluation, the patient was told he had a high white blood cell count 2 years ago. On examination, there was left cervical and inguinal lymphadenopathy and no other organomegaly. Complete blood cell count and peripheral blood smear showed marked leukocytosis, with a white blood cell count of 22 340/μL, an absolute neutrophil count of 5360/μL, and 55% eosinophils with an absolute eosinophil count of 12 290/μL (to convert all to cells ×109/L, multiply by 0.001). Vitamin B12 was markedly elevated at more than 4000 pg/mL (to convert to pmol/L, multiply by 0.7378). The erythrocyte sedimentation rate was 5 mm/h. Lactate dehydrogenase was 180 U/L, and alkaline phosphatase was 81 U/L (to convert both to μkat/L, multiply by 0.0167). Evaluations for HIV and hepatitis B and C were all negative. Serum creatinine was 0.76 mg/dL (to convert to μmol/L, multiply by 88.4); alanine aminotransferase and aspartate aminotransferase were 11 U/L and 9.9 U/L, respectively (to convert to μkat/L, multiply by 0.0167); and total bilirubin was 0.35 mg/dL (to convert to μmol/L, multiply by 17.104). Bone marrow biopsy showed hypercellular marrow (cellularity of 100%), myeloid hyperplasia, increased eosinophils with some dysplasia, and a blast count of 2%. Positron emission tomographic–computed tomographic scan showed a left upper cervical lymph node of 2.6 cm and a left inguinal lymph node of 3.1 × 2.3 cm with an standardized uptake value max of 5.7 (Figure, A). Left inguinal lymph node biopsy showed partial involvement by atypical cells with high proliferation index (Ki-67 >95%) that were positive for CD3, CD4, CD8, BCL2, and TDT, suggestive of T-cell lymphoblastic lymphoma/leukemia (Figure, B).A, Positron emission tomographic–computed tomographic (PET/CT) scan of the head and neck at presentation showing a left upper cervical lymph node of 2.6 cm (arrowhead). B, Lymph node biopsy immunohistochemical stain with terminal deoxynucleotidyl transferase. The inset shows interphase fluorescence in situ hybridization for FIP1L1::PDGFRA rearrangement (positive). C, PET/CT 12 weeks after treatment initiation.Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions What Is Your Diagnosis?
Kimura disease
Classic Hodgkin lymphoma
T-cell acute lymphoblastic lymphoma/leukemia
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
D. Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
D
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
The differential diagnoses in young men with eosinophilia and a neck mass can be wide. The typical presentation of Kimura disease is a painless, unilateral swelling of cervical lymph nodes in young adult male patients with peripheral blood eosinophilia and elevated immunoglobulin E levels. Histological evaluation, however, would show hyperplastic reactive follicles with eosinophilic infiltrates in a background of polymorphous inflammatory cells, a picture inconsistent with the presented case. Although Hodgkin lymphoma can present with eosinophilia and neck mass in young adults, the biopsy was inconsistent with the typical morphology of Reed-Sternberg cells in the background of nonneoplastic inflammatory cells. The diagnosis of T-cell acute lymphoblastic lymphoma/leukemia is reasonable given the morphology from lymph node biopsy; however, in the diagnosis hierarchy, myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions (MLN-TK) would supersede other myeloid and lymphoid cancers.1In the fifth edition of the World Health Organization Classification of Haematolymphoid Tumours, MLN-TK refers to BCR::ABL1–negative myeloid or lymphoid neoplasms with gene rearrangements leading to an activated tyrosine-kinase domain that results in cell-signaling dysregulation.1 This includes PDGFRA, PDGFRB, FGFR1, JAK2, and FLT3 rearrangements, in addition to ETV6::ABL1, ETV6::FGFR2, ETV6::LYN, ETV6::NTRK3, RANBP2::ALK, BCR::RET, and FGFR1OP::RET fusions.1 An increase in vitamin B12 is often found in patients with PDGFRA and PDGFRB disease.2,3 Most patients are male with chronic myeloid neoplasms with eosinophilia; however, patients rarely present with acute myeloid or T-cell leukemia or extramedullary disease.2Fluorescence in situ hybridization (FISH) was positive for FIP1L1::PDGFRA (4q12) fusion in 91% of nuclei, while karyotype was normal (46, XY in 18 cells), as a cryptic loss of the CHIC2 gene causes this fusion. Next, generation sequencing panel for myeloid neoplasms was negative for variants in targeted regions of the genes commonly involved in myeloid neoplasms. Combining imatinib with chemotherapy is acceptable in patients with acute leukemia or extramedullary diseases, although monotherapy with imatinib has shown promising results.4 The patient was concerned about fertility, as he and his partner were starting a family, and decided to avoid chemotherapy. The patient was concomitantly initiated on 1 mg/kg of prednisone for 7 days and imatinib at 600 mg daily. The eosinophil counts normalized after 2 days, and bone marrow evaluation after 4 weeks showed resolution of eosinophilic infiltration; however, FISH results were 3% positive for FIP1L1::PDGFRA fusion. Repeat bone marrow evaluation after 8 weeks showed complete resolution of the FISH abnormality, and the imatinib dose was reduced to 400 mg daily. Although studies have confirmed deep and durable remission with a starting dose of imatinib, 100 mg, we elected to start with a high dose given the patient’s young age and blastic presentation.5,6 Follow-up positron emission tomographic–computed tomographic scan 12 weeks after initiation of imatinib was consistent with complete metabolic remission (Figure, C). Complete hematological remission is achieved in more than 95% of patients with imatinib and is typically seen early (within weeks of treatment initiation). Cytogenetic/molecular response can be achieved within 3 to 6 months of treatment initiation.7 In patients who achieved molecular remission, the imatinib dose can be reduced to 100 thrice weekly or weekly.8 Treatment-free remission as the experience with chronic myeloid leukemia has been tried, although with limited experience. The success rate is around 60%, with most patients relapsing within the first 10 months. Reinitiating of imatinib restores hematological remission in almost all patients.9,10The current patient was in complete remission 12 months from his treatment and receiving imatinib. Evaluating patients suspected of MLN-TK is critical, as treatment and prognosis may drastically change.
Oncology
A 31-year-old patient presented with left cervical and left inguinal masses. They reported intermittent itching and night sweats for 2 years. They denied fever, weight loss, shortness of breath, rashes, diarrhea, and neurological symptoms. On a preemployment evaluation, the patient was told they had a high white blood cell count 2 years ago. On examination, there was left cervical and inguinal lymphadenopathy and no other organomegaly. Complete blood cell count and peripheral blood smear showed marked leukocytosis, with a white blood cell count of 22 340/μL, an absolute neutrophil count of 5360/μL, and 55% eosinophils with an absolute eosinophil count of 12 290/μL (to convert all to cells ×109/L, multiply by 0.001). Vitamin B12 was markedly elevated at more than 4000 pg/mL (to convert to pmol/L, multiply by 0.7378). The erythrocyte sedimentation rate was 5 mm/h. Lactate dehydrogenase was 180 U/L, and alkaline phosphatase was 81 U/L (to convert both to μkat/L, multiply by 0.0167). Evaluations for HIV and hepatitis B and C were all negative. Serum creatinine was 0.76 mg/dL (to convert to μmol/L, multiply by 88.4); alanine aminotransferase and aspartate aminotransferase were 11 U/L and 9.9 U/L, respectively (to convert to μkat/L, multiply by 0.0167); and total bilirubin was 0.35 mg/dL (to convert to μmol/L, multiply by 17.104). Bone marrow biopsy showed hypercellular marrow (cellularity of 100%), myeloid hyperplasia, increased eosinophils with some dysplasia, and a blast count of 2%. Positron emission tomographic–computed tomographic scan showed a left upper cervical lymph node of 2.6 cm and a left inguinal lymph node of 3.1 × 2.3 cm with an standardized uptake value max of 5.7 (Figure, A). Left inguinal lymph node biopsy showed partial involvement by atypical cells with high proliferation index (Ki-67 >95%) that were positive for CD3, CD4, CD8, BCL2, and TDT, suggestive of T-cell lymphoblastic lymphoma/leukemia (Figure, B).A, Positron emission tomographic–computed tomographic (PET/CT) scan of the head and neck at presentation showing a left upper cervical lymph node of 2.6 cm (arrowhead). B, Lymph node biopsy immunohistochemical stain with terminal deoxynucleotidyl transferase. The inset shows interphase fluorescence in situ hybridization for FIP1L1::PDGFRA rearrangement (positive). C, PET/CT 12 weeks after treatment initiation.Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
what is your diagnosis?
What is your diagnosis?
T-cell acute lymphoblastic lymphoma/leukemia
Kimura disease
Classic Hodgkin lymphoma
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions
d
1
1
1
1
neutral
0
0
31
31-40
White
4
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2810850
A 28-year-old woman presented with a 5-day history of painful cervical lymphadenopathy along with fever, nausea, and vomiting. Treatment with antibiotics and corticosteroids was started, without improvement of symptoms. She denied recent travel, sick contacts, or autoimmune disease. Her medical history was significant for α-thalassemia but otherwise noncontributory.On examination, she had tender lymphadenopathy involving right level V. A computed tomography scan of the neck revealed a collection of enlarged, matted nodes without evidence of an abscess. Fine-needle aspiration (FNA) biopsy showed nondiagnostic reactive lymph nodes. She was subsequently admitted to the hospital, where the lymphadenopathy progressed to the left side of the neck and a skin eruption developed over her face, back, and trunk. Results of an infectious disease and rheumatological workup, including bone marrow biopsy, were negative. Given her persistent symptoms, the ear, nose, and throat service consulted for excisional biopsy.On pathologic examination, the lymph nodes showed extensive coagulative necrosis, apoptosis, and karyorrhectic debris (Figure 1). Cellular components included histiocytes and dendritic cells, with absent neutrophils and hematoxylin bodies.Well-defined areas of fibrinoid necrosis in paracortical areas (hematoxylin-eosin, original magnification ×20). What Is Your Diagnosis?
Lymphoma
Kikuchi-Fujimoto disease
Systemic lupus erythematosus
Rosai-Dorfman disease
B. Kikuchi-Fujimoto disease
B
Kikuchi-Fujimoto disease
Common diagnostic considerations of lymphadenopathy alongside recurrent fever in young adults include infection, autoimmune disorders, and malignant neoplasms. Failure of conservative management with antibiotics reduces the likelihood of bacterial infection, whereas lack of cutaneous findings is atypical for a patient with classic systemic lupus erythematosus (SLE) (option C). Furthermore, both FNA and bone marrow biopsy results were not concerning for malignancy. Due to her relatively nonspecific and persistent symptoms, excisional biopsy was performed, giving the final diagnosis of Kikuchi-Fujimoto disease (KFD).Kikuchi-Fujimoto disease (also known as Kikuchi-Fujimoto lymphadenopathy, Kikuchi lymphadenitis, and necrotizing lymphadenitis of unknown etiology) is a benign, self-limiting disease that commonly affects young adults. This rare disease was first reported in Japan in 1972, with the highest prevalence in Asia.1 Patients typically present with painful cervical lymphadenopathy alongside fever and leukopenia.2 It commonly affects women younger than 40 years. Recent studies, however, suggest a more equal distribution among men and women.3Although the etiology is unknown, both autoimmune and viral causes are suspected. Involvement of Epstein-Barr and other herpesviruses is controversial and can be distinguished with periodic acid–Schiff, Grocott methenamine silver, and Ziehl-Nielsen stains. An inappropriate immune reaction has been hypothesized, as KFD is a T-cell–mediated response in those with a specific genetic profile.3 Individuals with KFD have a significantly higher incidence of human leukocyte antigen class II genes (specifically, the DPA1*01 and DPB1*0202 alleles, which are more common in Asian individuals).3 Interestingly, KFD has also occurred in conjunction with SLE, preceding its onset in 30% of cases.4The FNA biopsy is not specific in diagnosing KFD, with an accuracy of 56.7%.5 Therefore, excisional biopsy of the affected lymph nodes is often required. Figure 1 shows a typical necrotic phase of Kikuchi lymphadenitis with well-defined areas of fibrinoid necrosis in paracortical areas of affected lymph nodes. The necrotic foci show abundant karyorrhectic and cellular debris, fibrin deposition, and mononuclear cells (Figure 2). There are no intact neutrophils or eosinophils and only rare plasma cells. Crescentic histiocytes with phagocytized debris are seen along with numerous plasmacytoid dendritic cells/monocytes, macrophages, and activated T cells, which can simulate the appearance of a peripheral T-cell lymphoma. Some cases show prominent xanthomatous inflammation with foamy histiocytes.The necrotic foci show abundant karyorrhectic and cellular debris, fibrin deposition, and mononuclear cells. There are no intact neutrophils or eosinophils and only rare plasma cells. Crescentic histiocytes with phagocytized debris as well as plasmacytoid dendritic cells/monocytes are seen (hematoxylin-eosin, original magnification ×100).This disease is self-limited, lasting 1 to 4 months; although recurrence has been reported, the incidence is 3% to 4%.3 The fatality rate is low and has been reported in only 3 patients. Treatment is largely supportive with analgesic agents, antipyretic agents, and corticosteroids. In patients with recurrent symptoms, low doses of corticosteroids may achieve remission.6The differential diagnosis includes lymphoma with necrosis (option A), cat-scratch disease, and lupus lymphadenitis. Cat-scratch disease is confirmed by serologic testing for Bartonella henselae antibodies.5 Patients with lupus present with lymphadenopathy, fever, skin eruption, and hepatosplenomegaly. In contrast, patients with KFD present with predominantly cervical lymphadenopathy alongside fever and skin eruption.6 Although nonspecific, serologic testing may show leukopenia, anemia, thrombocytopenia, or atypical lymphocytes on peripheral blood smear.At last follow-up, this patient’s fever and lymphadenopathy had resolved, but the skin eruption had progressed to her hands and feet. She is currently receiving prednisone therapy and undergoing an autoimmune workup due to high suspicion of disease progression to SLE or myositis. She will continue to follow up with rheumatology, hematology/oncology, infectious disease, and otolaryngology services.
General
A 28-year-old woman presented with a 5-day history of painful cervical lymphadenopathy along with fever, nausea, and vomiting. Treatment with antibiotics and corticosteroids was started, without improvement of symptoms. She denied recent travel, sick contacts, or autoimmune disease. Her medical history was significant for α-thalassemia but otherwise noncontributory.On examination, she had tender lymphadenopathy involving right level V. A computed tomography scan of the neck revealed a collection of enlarged, matted nodes without evidence of an abscess. Fine-needle aspiration (FNA) biopsy showed nondiagnostic reactive lymph nodes. She was subsequently admitted to the hospital, where the lymphadenopathy progressed to the left side of the neck and a skin eruption developed over her face, back, and trunk. Results of an infectious disease and rheumatological workup, including bone marrow biopsy, were negative. Given her persistent symptoms, the ear, nose, and throat service consulted for excisional biopsy.On pathologic examination, the lymph nodes showed extensive coagulative necrosis, apoptosis, and karyorrhectic debris (Figure 1). Cellular components included histiocytes and dendritic cells, with absent neutrophils and hematoxylin bodies.Well-defined areas of fibrinoid necrosis in paracortical areas (hematoxylin-eosin, original magnification ×20).
what is your diagnosis?
What is your diagnosis?
Lymphoma
Systemic lupus erythematosus
Kikuchi-Fujimoto disease
Rosai-Dorfman disease
c
1
0
1
1
female
0
0
28
21-30
null
5
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2810850
A 28-year-old woman presented with a 5-day history of painful cervical lymphadenopathy along with fever, nausea, and vomiting. Treatment with antibiotics and corticosteroids was started, without improvement of symptoms. She denied recent travel, sick contacts, or autoimmune disease. Her medical history was significant for α-thalassemia but otherwise noncontributory.On examination, she had tender lymphadenopathy involving right level V. A computed tomography scan of the neck revealed a collection of enlarged, matted nodes without evidence of an abscess. Fine-needle aspiration (FNA) biopsy showed nondiagnostic reactive lymph nodes. She was subsequently admitted to the hospital, where the lymphadenopathy progressed to the left side of the neck and a skin eruption developed over her face, back, and trunk. Results of an infectious disease and rheumatological workup, including bone marrow biopsy, were negative. Given her persistent symptoms, the ear, nose, and throat service consulted for excisional biopsy.On pathologic examination, the lymph nodes showed extensive coagulative necrosis, apoptosis, and karyorrhectic debris (Figure 1). Cellular components included histiocytes and dendritic cells, with absent neutrophils and hematoxylin bodies.Well-defined areas of fibrinoid necrosis in paracortical areas (hematoxylin-eosin, original magnification ×20). What Is Your Diagnosis?
Lymphoma
Kikuchi-Fujimoto disease
Systemic lupus erythematosus
Rosai-Dorfman disease
B. Kikuchi-Fujimoto disease
B
Kikuchi-Fujimoto disease
Common diagnostic considerations of lymphadenopathy alongside recurrent fever in young adults include infection, autoimmune disorders, and malignant neoplasms. Failure of conservative management with antibiotics reduces the likelihood of bacterial infection, whereas lack of cutaneous findings is atypical for a patient with classic systemic lupus erythematosus (SLE) (option C). Furthermore, both FNA and bone marrow biopsy results were not concerning for malignancy. Due to her relatively nonspecific and persistent symptoms, excisional biopsy was performed, giving the final diagnosis of Kikuchi-Fujimoto disease (KFD).Kikuchi-Fujimoto disease (also known as Kikuchi-Fujimoto lymphadenopathy, Kikuchi lymphadenitis, and necrotizing lymphadenitis of unknown etiology) is a benign, self-limiting disease that commonly affects young adults. This rare disease was first reported in Japan in 1972, with the highest prevalence in Asia.1 Patients typically present with painful cervical lymphadenopathy alongside fever and leukopenia.2 It commonly affects women younger than 40 years. Recent studies, however, suggest a more equal distribution among men and women.3Although the etiology is unknown, both autoimmune and viral causes are suspected. Involvement of Epstein-Barr and other herpesviruses is controversial and can be distinguished with periodic acid–Schiff, Grocott methenamine silver, and Ziehl-Nielsen stains. An inappropriate immune reaction has been hypothesized, as KFD is a T-cell–mediated response in those with a specific genetic profile.3 Individuals with KFD have a significantly higher incidence of human leukocyte antigen class II genes (specifically, the DPA1*01 and DPB1*0202 alleles, which are more common in Asian individuals).3 Interestingly, KFD has also occurred in conjunction with SLE, preceding its onset in 30% of cases.4The FNA biopsy is not specific in diagnosing KFD, with an accuracy of 56.7%.5 Therefore, excisional biopsy of the affected lymph nodes is often required. Figure 1 shows a typical necrotic phase of Kikuchi lymphadenitis with well-defined areas of fibrinoid necrosis in paracortical areas of affected lymph nodes. The necrotic foci show abundant karyorrhectic and cellular debris, fibrin deposition, and mononuclear cells (Figure 2). There are no intact neutrophils or eosinophils and only rare plasma cells. Crescentic histiocytes with phagocytized debris are seen along with numerous plasmacytoid dendritic cells/monocytes, macrophages, and activated T cells, which can simulate the appearance of a peripheral T-cell lymphoma. Some cases show prominent xanthomatous inflammation with foamy histiocytes.The necrotic foci show abundant karyorrhectic and cellular debris, fibrin deposition, and mononuclear cells. There are no intact neutrophils or eosinophils and only rare plasma cells. Crescentic histiocytes with phagocytized debris as well as plasmacytoid dendritic cells/monocytes are seen (hematoxylin-eosin, original magnification ×100).This disease is self-limited, lasting 1 to 4 months; although recurrence has been reported, the incidence is 3% to 4%.3 The fatality rate is low and has been reported in only 3 patients. Treatment is largely supportive with analgesic agents, antipyretic agents, and corticosteroids. In patients with recurrent symptoms, low doses of corticosteroids may achieve remission.6The differential diagnosis includes lymphoma with necrosis (option A), cat-scratch disease, and lupus lymphadenitis. Cat-scratch disease is confirmed by serologic testing for Bartonella henselae antibodies.5 Patients with lupus present with lymphadenopathy, fever, skin eruption, and hepatosplenomegaly. In contrast, patients with KFD present with predominantly cervical lymphadenopathy alongside fever and skin eruption.6 Although nonspecific, serologic testing may show leukopenia, anemia, thrombocytopenia, or atypical lymphocytes on peripheral blood smear.At last follow-up, this patient’s fever and lymphadenopathy had resolved, but the skin eruption had progressed to her hands and feet. She is currently receiving prednisone therapy and undergoing an autoimmune workup due to high suspicion of disease progression to SLE or myositis. She will continue to follow up with rheumatology, hematology/oncology, infectious disease, and otolaryngology services.
General
A 28-year-old man presented with a 5-day history of painful cervical lymphadenopathy along with fever, nausea, and vomiting. Treatment with antibiotics and corticosteroids was started, without improvement of symptoms. He denied recent travel, sick contacts, or autoimmune disease. Him medical history was significant for α-thalassemia but otherwise noncontributory.On examination, he had tender lymphadenopathy involving right level V. A computed tomography scan of the neck revealed a collection of enlarged, matted nodes without evidence of an abscess. Fine-needle aspiration (FNA) biopsy showed nondiagnostic reactive lymph nodes. He was subsequently admitted to the hospital, where the lymphadenopathy progressed to the left side of the neck and a skin eruption developed over him face, back, and trunk. Results of an infectious disease and rheumatological workup, including bone marrow biopsy, were negative. Given him persistent symptoms, the ear, nose, and throat service consulted for excisional biopsy.On pathologic examination, the lymph nodes showed extensive coagulative necrosis, apoptosis, and karyorrhectic debris (Figure 1). Cellular components included histiocytes and dendritic cells, with absent neutrophils and hematoxylin bodies.Well-defined areas of fibrinoid necrosis in paracortical areas (hematoxylin-eosin, original magnification ×20).
what is your diagnosis?
What is your diagnosis?
Lymphoma
Systemic lupus erythematosus
Kikuchi-Fujimoto disease
Rosai-Dorfman disease
c
1
0
1
1
male
0
0
28
21-30
null
5
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2810850
A 28-year-old woman presented with a 5-day history of painful cervical lymphadenopathy along with fever, nausea, and vomiting. Treatment with antibiotics and corticosteroids was started, without improvement of symptoms. She denied recent travel, sick contacts, or autoimmune disease. Her medical history was significant for α-thalassemia but otherwise noncontributory.On examination, she had tender lymphadenopathy involving right level V. A computed tomography scan of the neck revealed a collection of enlarged, matted nodes without evidence of an abscess. Fine-needle aspiration (FNA) biopsy showed nondiagnostic reactive lymph nodes. She was subsequently admitted to the hospital, where the lymphadenopathy progressed to the left side of the neck and a skin eruption developed over her face, back, and trunk. Results of an infectious disease and rheumatological workup, including bone marrow biopsy, were negative. Given her persistent symptoms, the ear, nose, and throat service consulted for excisional biopsy.On pathologic examination, the lymph nodes showed extensive coagulative necrosis, apoptosis, and karyorrhectic debris (Figure 1). Cellular components included histiocytes and dendritic cells, with absent neutrophils and hematoxylin bodies.Well-defined areas of fibrinoid necrosis in paracortical areas (hematoxylin-eosin, original magnification ×20). What Is Your Diagnosis?
Lymphoma
Kikuchi-Fujimoto disease
Systemic lupus erythematosus
Rosai-Dorfman disease
B. Kikuchi-Fujimoto disease
B
Kikuchi-Fujimoto disease
Common diagnostic considerations of lymphadenopathy alongside recurrent fever in young adults include infection, autoimmune disorders, and malignant neoplasms. Failure of conservative management with antibiotics reduces the likelihood of bacterial infection, whereas lack of cutaneous findings is atypical for a patient with classic systemic lupus erythematosus (SLE) (option C). Furthermore, both FNA and bone marrow biopsy results were not concerning for malignancy. Due to her relatively nonspecific and persistent symptoms, excisional biopsy was performed, giving the final diagnosis of Kikuchi-Fujimoto disease (KFD).Kikuchi-Fujimoto disease (also known as Kikuchi-Fujimoto lymphadenopathy, Kikuchi lymphadenitis, and necrotizing lymphadenitis of unknown etiology) is a benign, self-limiting disease that commonly affects young adults. This rare disease was first reported in Japan in 1972, with the highest prevalence in Asia.1 Patients typically present with painful cervical lymphadenopathy alongside fever and leukopenia.2 It commonly affects women younger than 40 years. Recent studies, however, suggest a more equal distribution among men and women.3Although the etiology is unknown, both autoimmune and viral causes are suspected. Involvement of Epstein-Barr and other herpesviruses is controversial and can be distinguished with periodic acid–Schiff, Grocott methenamine silver, and Ziehl-Nielsen stains. An inappropriate immune reaction has been hypothesized, as KFD is a T-cell–mediated response in those with a specific genetic profile.3 Individuals with KFD have a significantly higher incidence of human leukocyte antigen class II genes (specifically, the DPA1*01 and DPB1*0202 alleles, which are more common in Asian individuals).3 Interestingly, KFD has also occurred in conjunction with SLE, preceding its onset in 30% of cases.4The FNA biopsy is not specific in diagnosing KFD, with an accuracy of 56.7%.5 Therefore, excisional biopsy of the affected lymph nodes is often required. Figure 1 shows a typical necrotic phase of Kikuchi lymphadenitis with well-defined areas of fibrinoid necrosis in paracortical areas of affected lymph nodes. The necrotic foci show abundant karyorrhectic and cellular debris, fibrin deposition, and mononuclear cells (Figure 2). There are no intact neutrophils or eosinophils and only rare plasma cells. Crescentic histiocytes with phagocytized debris are seen along with numerous plasmacytoid dendritic cells/monocytes, macrophages, and activated T cells, which can simulate the appearance of a peripheral T-cell lymphoma. Some cases show prominent xanthomatous inflammation with foamy histiocytes.The necrotic foci show abundant karyorrhectic and cellular debris, fibrin deposition, and mononuclear cells. There are no intact neutrophils or eosinophils and only rare plasma cells. Crescentic histiocytes with phagocytized debris as well as plasmacytoid dendritic cells/monocytes are seen (hematoxylin-eosin, original magnification ×100).This disease is self-limited, lasting 1 to 4 months; although recurrence has been reported, the incidence is 3% to 4%.3 The fatality rate is low and has been reported in only 3 patients. Treatment is largely supportive with analgesic agents, antipyretic agents, and corticosteroids. In patients with recurrent symptoms, low doses of corticosteroids may achieve remission.6The differential diagnosis includes lymphoma with necrosis (option A), cat-scratch disease, and lupus lymphadenitis. Cat-scratch disease is confirmed by serologic testing for Bartonella henselae antibodies.5 Patients with lupus present with lymphadenopathy, fever, skin eruption, and hepatosplenomegaly. In contrast, patients with KFD present with predominantly cervical lymphadenopathy alongside fever and skin eruption.6 Although nonspecific, serologic testing may show leukopenia, anemia, thrombocytopenia, or atypical lymphocytes on peripheral blood smear.At last follow-up, this patient’s fever and lymphadenopathy had resolved, but the skin eruption had progressed to her hands and feet. She is currently receiving prednisone therapy and undergoing an autoimmune workup due to high suspicion of disease progression to SLE or myositis. She will continue to follow up with rheumatology, hematology/oncology, infectious disease, and otolaryngology services.
General
A 28-year-old patient presented with a 5-day history of painful cervical lymphadenopathy along with fever, nausea, and vomiting. Treatment with antibiotics and corticosteroids was started, without improvement of symptoms. They denied recent travel, sick contacts, or autoimmune disease. Them medical history was significant for α-thalassemia but otherwise noncontributory.On examination, they had tender lymphadenopathy involving right level V. A computed tomography scan of the neck revealed a collection of enlarged, matted nodes without evidence of an abscess. Fine-needle aspiration (FNA) biopsy showed nondiagnostic reactive lymph nodes. They was subsequently admitted to the hospital, where the lymphadenopathy progressed to the left side of the neck and a skin eruption developed over them face, back, and trunk. Results of an infectious disease and rheumatological workup, including bone marrow biopsy, were negative. Given them persistent symptoms, the ear, nose, and throat service consulted for excisional biopsy.On pathologic examination, the lymph nodes showed extensive coagulative necrosis, apoptosis, and karyorrhectic debris (Figure 1). Cellular components included histiocytes and dendritic cells, with absent neutrophils and hematoxylin bodies.Well-defined areas of fibrinoid necrosis in paracortical areas (hematoxylin-eosin, original magnification ×20).
what is your diagnosis?
What is your diagnosis?
Lymphoma
Systemic lupus erythematosus
Kikuchi-Fujimoto disease
Rosai-Dorfman disease
c
1
0
1
1
neutral
0
0
28
21-30
null
5
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamacardiology/fullarticle/2810393
A woman in her 90s presented to the emergency department with symptoms of fever, hemoptysis, and syncopal episodes. She had a history of breast cancer and pulmonary nodules with biopsy findings negative for metastatic disease but positive for granulomatous inflammation. Initial vital signs included blood pressure of 103/66 mm Hg, heart rate of 190 bpm, and temperature of 38.7 °C. Electrocardiogram showed monomorphic ventricular tachycardia with a morphology suggesting right ventricular (RV) origin (Figure 1). In addition to receiving intravenous amiodarone hydrochloride, she had correction of mild hypokalemia (3.1 mEq/L [reference range, 3.5-5.2 mEq/L]; to convert to mmol/L, multiply by 1.0). Successful arrhythmia termination occurred shortly thereafter, and her vital signs remained stable. Chest computed tomographic angiography excluded pulmonary embolism but showed pulmonary nodules and mediastinal lymphadenopathy that had progressed compared with her previous computed tomography. An echocardiogram showed severe RV and left ventricular (LV) systolic dysfunction with multiple areas of hypokinesis.Patient electrocardiogram showing monomorphic regular wide complex tachycardia with a left bundle branch block morphology, with a precordial transition at V3 and inferior axis suggesting right ventricular origin. What Would You Do Next?
Cardiac magnetic resonance imaging
Electrophysiology study
Genetic testing
Implantation of cardiac defibrillator
Cardiac sarcoidosis
A
Cardiac magnetic resonance imaging
The patient underwent cardiac magnetic resonance imaging, which demonstrated areas of nonischemic near-transmural late gadolinium enhancement (LGE) of the interventricular septum and epicardium to midmyocardium of the LV basal to midanterior and basal inferior walls extending to the RV walls and septal papillary muscle (Figure 2A). However, T2 mapping revealed only discrete evidence of edema of a small region of the midanterior LV wall. Taken together, these findings raised the possibility of end-stage cardiac sarcoidosis (CS).A, Cardiac magnetic resonance imaging using phase-sensitive inversion recovery with motion correction demonstrates multiple areas of subepicardial, midmyocardial, and transmural and near-transmural late gadolinium enhancement in the left ventricle (pink arrowhead) and extending to the right ventricular wall and septal papillary muscle (blue arrowhead). B, Selected matched contiguous short-axis rubidium-82 (top row) and fluorodexoyglucose F 18 (FDG) positron emission tomographic images (bottom row) demonstrate reduced perfusion in the midseptum with marked FDG uptake in the septum and inferior wall, sparing the lateral wall.Cardiac fluorodeoxyglucose F 18 (FDG) and rubidium-82 positron emission tomography (PET) for CS was obtained with a strict high-fat, low-carbohydrate diet, followed by 12 hours of fasting prior to the examination per American Society of Nuclear Cardiology recommendations.1 Because the patient was hospitalized, dietary compliance was ensured. Furthermore, FDG uptake was segmental and the LV blood pool maximum standard uptake value was only 1.1, demonstrating adequate suppression of normal myocardial glucose utilization. There was an extensive FDG uptake (Figure 2B), with maximum standard uptake value of 10.4 in the LV septum. There was also extracardiac FDG uptake in hilar and mediastinal lymph nodes and in multiple pulmonary nodules. Rubidium-82 PET showed only mildly reduced perfusion in the basal to apical septal wall.This case highlights multiple learning points in the use of multimodality imaging for evaluation of patients with CS and understanding its complex pathophysiology. First, the utility of T2 mapping for detecting myocardial edema, a potential surrogate for active inflammation, needs to be better understood, as this can be challenging and in this case underrepresented the extensive positive FDG uptake seen on cardiac PET. Despite extensive LV and RV LGE and depressed biventricular systolic function, substantial myocardial inflammation in this patient is consistent with active rather than end-stage CS. Second, determining individual patient prognosis and assessment of immunosuppression response in patients with CS using cardiac imaging remains challenging. It has been demonstrated that patients without LGE are at an extremely low risk of arrhythmic events.2,3 However, although there is increased risk of arrhythmic events in patients with cardiac involvement identified by LGE, only a fraction of these patients experience arrhythmic events. Patients with both RV and LV LGE are at elevated risk of cardiovascular events compared with patients with only LV enhancement.2-4 Last, while CS is extremely rare after 70 years of age, as the oldest patient reported in the literature was 80 years old,5 this patient presented with CS in her 90s, reminding us that CS remains on the differential diagnosis in patients with sustained ventricular arrythmias regardless of age.Heart failure guideline–directed medical treatment, oral amiodarone, and immunosuppressive therapy consisting of a high, tapering dose of prednisone with mycophenolate mofetil were started. The patient remained stable with no more arrythmias. After shared decision discussions with the patient, a cardiac defibrillator was not implanted given the patient’s age and out of respect for her wishes. She will undergo follow-up FDG-PET to monitor her response to therapy and disease activity.
Cardiology
A woman in her 90s presented to the emergency department with symptoms of fever, hemoptysis, and syncopal episodes. She had a history of breast cancer and pulmonary nodules with biopsy findings negative for metastatic disease but positive for granulomatous inflammation. Initial vital signs included blood pressure of 103/66 mm Hg, heart rate of 190 bpm, and temperature of 38.7 °C. Electrocardiogram showed monomorphic ventricular tachycardia with a morphology suggesting right ventricular (RV) origin (Figure 1). In addition to receiving intravenous amiodarone hydrochloride, she had correction of mild hypokalemia (3.1 mEq/L [reference range, 3.5-5.2 mEq/L]; to convert to mmol/L, multiply by 1.0). Successful arrhythmia termination occurred shortly thereafter, and her vital signs remained stable. Chest computed tomographic angiography excluded pulmonary embolism but showed pulmonary nodules and mediastinal lymphadenopathy that had progressed compared with her previous computed tomography. An echocardiogram showed severe RV and left ventricular (LV) systolic dysfunction with multiple areas of hypokinesis.Patient electrocardiogram showing monomorphic regular wide complex tachycardia with a left bundle branch block morphology, with a precordial transition at V3 and inferior axis suggesting right ventricular origin.
what would you do next?
What would you do next?
Implantation of cardiac defibrillator
Electrophysiology study
Cardiac magnetic resonance imaging
Genetic testing
c
1
1
1
1
female
0
1
95
91-100
null
6
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2810393
A woman in her 90s presented to the emergency department with symptoms of fever, hemoptysis, and syncopal episodes. She had a history of breast cancer and pulmonary nodules with biopsy findings negative for metastatic disease but positive for granulomatous inflammation. Initial vital signs included blood pressure of 103/66 mm Hg, heart rate of 190 bpm, and temperature of 38.7 °C. Electrocardiogram showed monomorphic ventricular tachycardia with a morphology suggesting right ventricular (RV) origin (Figure 1). In addition to receiving intravenous amiodarone hydrochloride, she had correction of mild hypokalemia (3.1 mEq/L [reference range, 3.5-5.2 mEq/L]; to convert to mmol/L, multiply by 1.0). Successful arrhythmia termination occurred shortly thereafter, and her vital signs remained stable. Chest computed tomographic angiography excluded pulmonary embolism but showed pulmonary nodules and mediastinal lymphadenopathy that had progressed compared with her previous computed tomography. An echocardiogram showed severe RV and left ventricular (LV) systolic dysfunction with multiple areas of hypokinesis.Patient electrocardiogram showing monomorphic regular wide complex tachycardia with a left bundle branch block morphology, with a precordial transition at V3 and inferior axis suggesting right ventricular origin. What Would You Do Next?
Cardiac magnetic resonance imaging
Electrophysiology study
Genetic testing
Implantation of cardiac defibrillator
Cardiac sarcoidosis
A
Cardiac magnetic resonance imaging
The patient underwent cardiac magnetic resonance imaging, which demonstrated areas of nonischemic near-transmural late gadolinium enhancement (LGE) of the interventricular septum and epicardium to midmyocardium of the LV basal to midanterior and basal inferior walls extending to the RV walls and septal papillary muscle (Figure 2A). However, T2 mapping revealed only discrete evidence of edema of a small region of the midanterior LV wall. Taken together, these findings raised the possibility of end-stage cardiac sarcoidosis (CS).A, Cardiac magnetic resonance imaging using phase-sensitive inversion recovery with motion correction demonstrates multiple areas of subepicardial, midmyocardial, and transmural and near-transmural late gadolinium enhancement in the left ventricle (pink arrowhead) and extending to the right ventricular wall and septal papillary muscle (blue arrowhead). B, Selected matched contiguous short-axis rubidium-82 (top row) and fluorodexoyglucose F 18 (FDG) positron emission tomographic images (bottom row) demonstrate reduced perfusion in the midseptum with marked FDG uptake in the septum and inferior wall, sparing the lateral wall.Cardiac fluorodeoxyglucose F 18 (FDG) and rubidium-82 positron emission tomography (PET) for CS was obtained with a strict high-fat, low-carbohydrate diet, followed by 12 hours of fasting prior to the examination per American Society of Nuclear Cardiology recommendations.1 Because the patient was hospitalized, dietary compliance was ensured. Furthermore, FDG uptake was segmental and the LV blood pool maximum standard uptake value was only 1.1, demonstrating adequate suppression of normal myocardial glucose utilization. There was an extensive FDG uptake (Figure 2B), with maximum standard uptake value of 10.4 in the LV septum. There was also extracardiac FDG uptake in hilar and mediastinal lymph nodes and in multiple pulmonary nodules. Rubidium-82 PET showed only mildly reduced perfusion in the basal to apical septal wall.This case highlights multiple learning points in the use of multimodality imaging for evaluation of patients with CS and understanding its complex pathophysiology. First, the utility of T2 mapping for detecting myocardial edema, a potential surrogate for active inflammation, needs to be better understood, as this can be challenging and in this case underrepresented the extensive positive FDG uptake seen on cardiac PET. Despite extensive LV and RV LGE and depressed biventricular systolic function, substantial myocardial inflammation in this patient is consistent with active rather than end-stage CS. Second, determining individual patient prognosis and assessment of immunosuppression response in patients with CS using cardiac imaging remains challenging. It has been demonstrated that patients without LGE are at an extremely low risk of arrhythmic events.2,3 However, although there is increased risk of arrhythmic events in patients with cardiac involvement identified by LGE, only a fraction of these patients experience arrhythmic events. Patients with both RV and LV LGE are at elevated risk of cardiovascular events compared with patients with only LV enhancement.2-4 Last, while CS is extremely rare after 70 years of age, as the oldest patient reported in the literature was 80 years old,5 this patient presented with CS in her 90s, reminding us that CS remains on the differential diagnosis in patients with sustained ventricular arrythmias regardless of age.Heart failure guideline–directed medical treatment, oral amiodarone, and immunosuppressive therapy consisting of a high, tapering dose of prednisone with mycophenolate mofetil were started. The patient remained stable with no more arrythmias. After shared decision discussions with the patient, a cardiac defibrillator was not implanted given the patient’s age and out of respect for her wishes. She will undergo follow-up FDG-PET to monitor her response to therapy and disease activity.
Cardiology
A man in him 90s presented to the emergency department with symptoms of fever, hemoptysis, and syncopal episodes. He had a history of breast cancer and pulmonary nodules with biopsy findings negative for metastatic disease but positive for granulomatous inflammation. Initial vital signs included blood pressure of 103/66 mm Hg, heart rate of 190 bpm, and temperature of 38.7 °C. Electrocardiogram showed monomorphic ventricular tachycardia with a morphology suggesting right ventricular (RV) origin (Figure 1). In addition to receiving intravenous amiodarone hydrochloride, he had correction of mild hypokalemia (3.1 mEq/L [reference range, 3.5-5.2 mEq/L]; to convert to mmol/L, multiply by 1.0). Successful arrhythmia termination occurred shortly thereafter, and him vital signs remained stable. Chest computed tomographic angiography excluded pulmonary embolism but showed pulmonary nodules and mediastinal lymphadenopathy that had progressed compared with him previous computed tomography. An echocardiogram showed severe RV and left ventricular (LV) systolic dysfunction with multiple areas of hypokinesis.Man electrocardiogram showing monomorphic regular wide complex tachycardia with a left bundle branch block morphology, with a precordial transition at V3 and inferior axis suggesting right ventricular origin.
what would you do next?
What would you do next?
Implantation of cardiac defibrillator
Electrophysiology study
Cardiac magnetic resonance imaging
Genetic testing
c
1
1
1
1
male
0
1
95
91-100
null
6
augmented_male_fromfemale
https://jamanetwork.com/journals/jamacardiology/fullarticle/2810393
A woman in her 90s presented to the emergency department with symptoms of fever, hemoptysis, and syncopal episodes. She had a history of breast cancer and pulmonary nodules with biopsy findings negative for metastatic disease but positive for granulomatous inflammation. Initial vital signs included blood pressure of 103/66 mm Hg, heart rate of 190 bpm, and temperature of 38.7 °C. Electrocardiogram showed monomorphic ventricular tachycardia with a morphology suggesting right ventricular (RV) origin (Figure 1). In addition to receiving intravenous amiodarone hydrochloride, she had correction of mild hypokalemia (3.1 mEq/L [reference range, 3.5-5.2 mEq/L]; to convert to mmol/L, multiply by 1.0). Successful arrhythmia termination occurred shortly thereafter, and her vital signs remained stable. Chest computed tomographic angiography excluded pulmonary embolism but showed pulmonary nodules and mediastinal lymphadenopathy that had progressed compared with her previous computed tomography. An echocardiogram showed severe RV and left ventricular (LV) systolic dysfunction with multiple areas of hypokinesis.Patient electrocardiogram showing monomorphic regular wide complex tachycardia with a left bundle branch block morphology, with a precordial transition at V3 and inferior axis suggesting right ventricular origin. What Would You Do Next?
Cardiac magnetic resonance imaging
Electrophysiology study
Genetic testing
Implantation of cardiac defibrillator
Cardiac sarcoidosis
A
Cardiac magnetic resonance imaging
The patient underwent cardiac magnetic resonance imaging, which demonstrated areas of nonischemic near-transmural late gadolinium enhancement (LGE) of the interventricular septum and epicardium to midmyocardium of the LV basal to midanterior and basal inferior walls extending to the RV walls and septal papillary muscle (Figure 2A). However, T2 mapping revealed only discrete evidence of edema of a small region of the midanterior LV wall. Taken together, these findings raised the possibility of end-stage cardiac sarcoidosis (CS).A, Cardiac magnetic resonance imaging using phase-sensitive inversion recovery with motion correction demonstrates multiple areas of subepicardial, midmyocardial, and transmural and near-transmural late gadolinium enhancement in the left ventricle (pink arrowhead) and extending to the right ventricular wall and septal papillary muscle (blue arrowhead). B, Selected matched contiguous short-axis rubidium-82 (top row) and fluorodexoyglucose F 18 (FDG) positron emission tomographic images (bottom row) demonstrate reduced perfusion in the midseptum with marked FDG uptake in the septum and inferior wall, sparing the lateral wall.Cardiac fluorodeoxyglucose F 18 (FDG) and rubidium-82 positron emission tomography (PET) for CS was obtained with a strict high-fat, low-carbohydrate diet, followed by 12 hours of fasting prior to the examination per American Society of Nuclear Cardiology recommendations.1 Because the patient was hospitalized, dietary compliance was ensured. Furthermore, FDG uptake was segmental and the LV blood pool maximum standard uptake value was only 1.1, demonstrating adequate suppression of normal myocardial glucose utilization. There was an extensive FDG uptake (Figure 2B), with maximum standard uptake value of 10.4 in the LV septum. There was also extracardiac FDG uptake in hilar and mediastinal lymph nodes and in multiple pulmonary nodules. Rubidium-82 PET showed only mildly reduced perfusion in the basal to apical septal wall.This case highlights multiple learning points in the use of multimodality imaging for evaluation of patients with CS and understanding its complex pathophysiology. First, the utility of T2 mapping for detecting myocardial edema, a potential surrogate for active inflammation, needs to be better understood, as this can be challenging and in this case underrepresented the extensive positive FDG uptake seen on cardiac PET. Despite extensive LV and RV LGE and depressed biventricular systolic function, substantial myocardial inflammation in this patient is consistent with active rather than end-stage CS. Second, determining individual patient prognosis and assessment of immunosuppression response in patients with CS using cardiac imaging remains challenging. It has been demonstrated that patients without LGE are at an extremely low risk of arrhythmic events.2,3 However, although there is increased risk of arrhythmic events in patients with cardiac involvement identified by LGE, only a fraction of these patients experience arrhythmic events. Patients with both RV and LV LGE are at elevated risk of cardiovascular events compared with patients with only LV enhancement.2-4 Last, while CS is extremely rare after 70 years of age, as the oldest patient reported in the literature was 80 years old,5 this patient presented with CS in her 90s, reminding us that CS remains on the differential diagnosis in patients with sustained ventricular arrythmias regardless of age.Heart failure guideline–directed medical treatment, oral amiodarone, and immunosuppressive therapy consisting of a high, tapering dose of prednisone with mycophenolate mofetil were started. The patient remained stable with no more arrythmias. After shared decision discussions with the patient, a cardiac defibrillator was not implanted given the patient’s age and out of respect for her wishes. She will undergo follow-up FDG-PET to monitor her response to therapy and disease activity.
Cardiology
A patient in them 90s presented to the emergency department with symptoms of fever, hemoptysis, and syncopal episodes. They had a history of breast cancer and pulmonary nodules with biopsy findings negative for metastatic disease but positive for granulomatous inflammation. Initial vital signs included blood pressure of 103/66 mm Hg, heart rate of 190 bpm, and temperature of 38.7 °C. Electrocardiogram showed monomorphic ventricular tachycardia with a morphology suggesting right ventricular (RV) origin (Figure 1). In addition to receiving intravenous amiodarone hydrochloride, they had correction of mild hypokalemia (3.1 mEq/L [reference range, 3.5-5.2 mEq/L]; to convert to mmol/L, multiply by 1.0). Successful arrhythmia termination occurred shortly thereafter, and them vital signs remained stable. Chest computed tomographic angiography excluded pulmonary embolism but showed pulmonary nodules and mediastinal lymphadenopathy that had progressed compared with them previous computed tomography. An echocardiogram showed severe RV and left ventricular (LV) systolic dysfunction with multiple areas of hypokinesis.Patient electrocardiogram showing monomorphic regular wide complex tachycardia with a left bundle branch block morphology, with a precordial transition at V3 and inferior axis suggesting right ventricular origin.
what would you do next?
What would you do next?
Implantation of cardiac defibrillator
Electrophysiology study
Cardiac magnetic resonance imaging
Genetic testing
c
1
1
1
1
neutral
0
1
95
91-100
null
6
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2810455
A 27-year-old woman presented to her primary care physician with primary complaints of headaches and visual-spatial difficulties when driving at night. In addition, the patient described a progressively enlarging lesion on her left scalp, which she had initially noted in high school. She denied any overlying loss of hair, sharp pain, or bleeding from the scalp lesion, and she only reported pain with prolonged pressure. On her scalp examination, a 7-cm ill-defined spongy and pulsatile mass was noted with an overlying gray patch of hair. Magnetic resonance imaging (MRI) of the brain/orbit was initially performed (Figure, A and B), which demonstrated an ill-defined, diffuse, intensely enhancing scalp lesion in the left frontoparietal region with very prominent vascularity and underlying calvarial thickening. No osseous destruction or intracranial extension was present. Subsequent catheter angiography (Figure, C) confirmed the hypervascular mass, supplied via hypertrophied superficial temporal and occipital arteries with marked internal vascularity without direct involvement of the calvarium or intracranial structures. The patient underwent biopsy under general anesthesia; the specimen showed an ill-defined deep dermal/subcutaneous proliferation of spindle cells without significant nuclear pleomorphism or mitotic activity. The lesions stained positive for S100 and SOX10 immunostains.Coronal T2-weighted (A) and postcontrast T1-weighted (B) fat-saturated images show the large left T2 hyperintense scalp lesion (double arrowheads; A and B), with underlying calvarial thickening (asterisk; A and B) without invasion, prominent vascularity (single arrowhead; A), and enhancement (B). Note the preservation of the overlying skin. Catheter angiography (C) shows the hypervascular scalp lesion being supplied by hypertrophied branches arising from the left superficial temporal and occipital arteries, with marked internal vascularity (arrowheads). R indicates right. What Is Your Diagnosis?
Arteriovenous malformation of the scalp
Solitary fibrous tumor
Diffuse-type neurofibroma
Dermatofibrosarcoma protuberans
C. Diffuse-type neurofibroma
C
Diffuse-type neurofibroma
Neurofibromas, which are common in the head and neck, arise from endoneurium and connective tissues of peripheral nerve sheaths. From a morphologic and imaging perspective, there are 3 main types of neurofibromas: localized, diffuse, and plexiform.1 Diffuse neurofibromas are rare and represent less than 5% to 10% of all neurofibromas. The localized type, which is more common (>90%) and is the most well recognized and documented, is usually not a diagnostic conundrum. Plexiform neurofibromas, the other subtype, also have a pathognomonic imaging appearance and are frequently associated with neurofibromatosis 1, with findings of multiple lesions at different sites, which makes the diagnosis easier. Diffuse neurofibromas, however, are frequently solitary and are mostly sporadic, with few reported cases of associated with neurofibromatosis 1 (NF1).2 These lesions are more often seen in children and young adults. They are unusually large and frequently multispatial, with ill-defined margins, which appear to infiltrate and grow around tissues rather than invade them.On imaging, these are frequently plaquelike infiltrative lesions, unlike the more rounded or nodular appearance of localized neurofibromas, with frequent involvement of the subcutaneous/deep fascia both underlying the skin and overlying the musculature. Another characteristic is preservation of the overall morphologic features of the involved tissue space due to its unusual infiltrative pattern.2 A lesser known feature of these lesions is that many also demonstrate marked internal vascularity with prominent flow voids, arterial feeders, and draining veins, which can sometimes mimic vascular malformations.3 On MRI, these are isointense to mildly hyperintense to muscle on T1-weighted imaging and mild to markedly hyperintense on T2-weighted imaging to muscle, with marked enhancement. The lesions are usually solid without any necrotic or cystic changes, calcification, or hemorrhage.Solitary fibrous tumors (option B) (previously referred to as hemangiopericytomas) are rare mesenchymal-origin lesions, usually in older adults. On MRI, these are homogeneous lesions, with usually low to intermediate signals on both T1- and T2-weighted sequences (relative to muscle), with intense enhancement. They can be plaquelike and are frequently hypervascular, with numerous flow voids.4 Rarely, they may have a heterogeneous appearance secondary to internal hemorrhage or necrosis. They frequently mimic meningiomas when they arise intracranially.Scalp arteriovenous malformations (option A) are rare, and they present in children and young adults. On imaging, a predominant vascular lesion is identified with numerous flow voids due to its high flow state. The imaging features can be variable if there are associated abnormalities as well as other low-flow lesions, such as venous or lymphatic malformations. Vascular imaging, such as computed tomographic and magnetic resonance angiography, ultrasonography, and catheter angiography, is frequently done for both diagnosis and treatment planning.5Dermatofibrosarcoma protuberans (option D) are rare superficial lesions of the dermis, usually presenting in the third to fourth decade of life. These are locally aggressive, infiltrative masses that can arise in the head and neck, including the scalp. They present as a painless, slow-growing, nodular mass–like firm lesion. On imaging, these are markedly enhancing lesions, usually T1 hypointense and T2 hyperintense, with no vascular flow voids, underlying osseous invasion, or perineural spread.6 However, underlying calvarial thinning is seen in a few cases.Given the findings of the large scalp neurofibroma and her age, the patient was advised to undergo comprehensive genetic testing for NF1. Although NF1 is autosomal dominant and the patient had a negative family history, almost half of all individuals with NF1 are the first person in their family to have NF1 (de novo variant).7 The patient tested negative for identifiable pathogenic variants in the NF1 or SPRED1 genes, decreasing her risk of having NF1 and confirming that this was likely a sporadic neurofibroma.
General
A 27-year-old woman presented to her primary care physician with primary complaints of headaches and visual-spatial difficulties when driving at night. In addition, the patient described a progressively enlarging lesion on her left scalp, which she had initially noted in high school. She denied any overlying loss of hair, sharp pain, or bleeding from the scalp lesion, and she only reported pain with prolonged pressure. On her scalp examination, a 7-cm ill-defined spongy and pulsatile mass was noted with an overlying gray patch of hair. Magnetic resonance imaging (MRI) of the brain/orbit was initially performed (Figure, A and B), which demonstrated an ill-defined, diffuse, intensely enhancing scalp lesion in the left frontoparietal region with very prominent vascularity and underlying calvarial thickening. No osseous destruction or intracranial extension was present. Subsequent catheter angiography (Figure, C) confirmed the hypervascular mass, supplied via hypertrophied superficial temporal and occipital arteries with marked internal vascularity without direct involvement of the calvarium or intracranial structures. The patient underwent biopsy under general anesthesia; the specimen showed an ill-defined deep dermal/subcutaneous proliferation of spindle cells without significant nuclear pleomorphism or mitotic activity. The lesions stained positive for S100 and SOX10 immunostains.Coronal T2-weighted (A) and postcontrast T1-weighted (B) fat-saturated images show the large left T2 hyperintense scalp lesion (double arrowheads; A and B), with underlying calvarial thickening (asterisk; A and B) without invasion, prominent vascularity (single arrowhead; A), and enhancement (B). Note the preservation of the overlying skin. Catheter angiography (C) shows the hypervascular scalp lesion being supplied by hypertrophied branches arising from the left superficial temporal and occipital arteries, with marked internal vascularity (arrowheads). R indicates right.
what is your diagnosis?
What is your diagnosis?
Diffuse-type neurofibroma
Dermatofibrosarcoma protuberans
Solitary fibrous tumor
Arteriovenous malformation of the scalp
a
1
1
1
1
female
0
0
27
21-30
null
7
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2810455
A 27-year-old woman presented to her primary care physician with primary complaints of headaches and visual-spatial difficulties when driving at night. In addition, the patient described a progressively enlarging lesion on her left scalp, which she had initially noted in high school. She denied any overlying loss of hair, sharp pain, or bleeding from the scalp lesion, and she only reported pain with prolonged pressure. On her scalp examination, a 7-cm ill-defined spongy and pulsatile mass was noted with an overlying gray patch of hair. Magnetic resonance imaging (MRI) of the brain/orbit was initially performed (Figure, A and B), which demonstrated an ill-defined, diffuse, intensely enhancing scalp lesion in the left frontoparietal region with very prominent vascularity and underlying calvarial thickening. No osseous destruction or intracranial extension was present. Subsequent catheter angiography (Figure, C) confirmed the hypervascular mass, supplied via hypertrophied superficial temporal and occipital arteries with marked internal vascularity without direct involvement of the calvarium or intracranial structures. The patient underwent biopsy under general anesthesia; the specimen showed an ill-defined deep dermal/subcutaneous proliferation of spindle cells without significant nuclear pleomorphism or mitotic activity. The lesions stained positive for S100 and SOX10 immunostains.Coronal T2-weighted (A) and postcontrast T1-weighted (B) fat-saturated images show the large left T2 hyperintense scalp lesion (double arrowheads; A and B), with underlying calvarial thickening (asterisk; A and B) without invasion, prominent vascularity (single arrowhead; A), and enhancement (B). Note the preservation of the overlying skin. Catheter angiography (C) shows the hypervascular scalp lesion being supplied by hypertrophied branches arising from the left superficial temporal and occipital arteries, with marked internal vascularity (arrowheads). R indicates right. What Is Your Diagnosis?
Arteriovenous malformation of the scalp
Solitary fibrous tumor
Diffuse-type neurofibroma
Dermatofibrosarcoma protuberans
C. Diffuse-type neurofibroma
C
Diffuse-type neurofibroma
Neurofibromas, which are common in the head and neck, arise from endoneurium and connective tissues of peripheral nerve sheaths. From a morphologic and imaging perspective, there are 3 main types of neurofibromas: localized, diffuse, and plexiform.1 Diffuse neurofibromas are rare and represent less than 5% to 10% of all neurofibromas. The localized type, which is more common (>90%) and is the most well recognized and documented, is usually not a diagnostic conundrum. Plexiform neurofibromas, the other subtype, also have a pathognomonic imaging appearance and are frequently associated with neurofibromatosis 1, with findings of multiple lesions at different sites, which makes the diagnosis easier. Diffuse neurofibromas, however, are frequently solitary and are mostly sporadic, with few reported cases of associated with neurofibromatosis 1 (NF1).2 These lesions are more often seen in children and young adults. They are unusually large and frequently multispatial, with ill-defined margins, which appear to infiltrate and grow around tissues rather than invade them.On imaging, these are frequently plaquelike infiltrative lesions, unlike the more rounded or nodular appearance of localized neurofibromas, with frequent involvement of the subcutaneous/deep fascia both underlying the skin and overlying the musculature. Another characteristic is preservation of the overall morphologic features of the involved tissue space due to its unusual infiltrative pattern.2 A lesser known feature of these lesions is that many also demonstrate marked internal vascularity with prominent flow voids, arterial feeders, and draining veins, which can sometimes mimic vascular malformations.3 On MRI, these are isointense to mildly hyperintense to muscle on T1-weighted imaging and mild to markedly hyperintense on T2-weighted imaging to muscle, with marked enhancement. The lesions are usually solid without any necrotic or cystic changes, calcification, or hemorrhage.Solitary fibrous tumors (option B) (previously referred to as hemangiopericytomas) are rare mesenchymal-origin lesions, usually in older adults. On MRI, these are homogeneous lesions, with usually low to intermediate signals on both T1- and T2-weighted sequences (relative to muscle), with intense enhancement. They can be plaquelike and are frequently hypervascular, with numerous flow voids.4 Rarely, they may have a heterogeneous appearance secondary to internal hemorrhage or necrosis. They frequently mimic meningiomas when they arise intracranially.Scalp arteriovenous malformations (option A) are rare, and they present in children and young adults. On imaging, a predominant vascular lesion is identified with numerous flow voids due to its high flow state. The imaging features can be variable if there are associated abnormalities as well as other low-flow lesions, such as venous or lymphatic malformations. Vascular imaging, such as computed tomographic and magnetic resonance angiography, ultrasonography, and catheter angiography, is frequently done for both diagnosis and treatment planning.5Dermatofibrosarcoma protuberans (option D) are rare superficial lesions of the dermis, usually presenting in the third to fourth decade of life. These are locally aggressive, infiltrative masses that can arise in the head and neck, including the scalp. They present as a painless, slow-growing, nodular mass–like firm lesion. On imaging, these are markedly enhancing lesions, usually T1 hypointense and T2 hyperintense, with no vascular flow voids, underlying osseous invasion, or perineural spread.6 However, underlying calvarial thinning is seen in a few cases.Given the findings of the large scalp neurofibroma and her age, the patient was advised to undergo comprehensive genetic testing for NF1. Although NF1 is autosomal dominant and the patient had a negative family history, almost half of all individuals with NF1 are the first person in their family to have NF1 (de novo variant).7 The patient tested negative for identifiable pathogenic variants in the NF1 or SPRED1 genes, decreasing her risk of having NF1 and confirming that this was likely a sporadic neurofibroma.
General
A 27-year-old man presented to him primary care physician with primary complaints of headaches and visual-spatial difficulties when driving at night. In addition, the man described a progressively enlarging lesion on him left scalp, which he had initially noted in high school. He denied any overlying loss of hair, sharp pain, or bleeding from the scalp lesion, and he only reported pain with prolonged pressure. On him scalp examination, a 7-cm ill-defined spongy and pulsatile mass was noted with an overlying gray patch of hair. Magnetic resonance imaging (MRI) of the brain/orbit was initially performed (Figure, A and B), which demonstrated an ill-defined, diffuse, intensely enhancing scalp lesion in the left frontoparietal region with very prominent vascularity and underlying calvarial thickening. No osseous destruction or intracranial extension was present. Subsequent catheter angiography (Figure, C) confirmed the hypervascular mass, supplied via hypertrophied superficial temporal and occipital arteries with marked internal vascularity without direct involvement of the calvarium or intracranial structures. The man underwent biopsy under general anesthesia; the specimen showed an ill-defined deep dermal/subcutaneous proliferation of spindle cells without significant nuclear pleomorphism or mitotic activity. The lesions stained positive for S100 and SOX10 immunostains.Coronal T2-weighted (A) and postcontrast T1-weighted (B) fat-saturated images show the large left T2 hyperintense scalp lesion (double arrowheads; A and B), with underlying calvarial thickening (asterisk; A and B) without invasion, prominent vascularity (single arrowhead; A), and enhancement (B). Note the preservation of the overlying skin. Catheter angiography (C) shows the hypervascular scalp lesion being supplied by hypertrophied branches arising from the left superficial temporal and occipital arteries, with marked internal vascularity (arrowheads). R indicates right.
what is your diagnosis?
What is your diagnosis?
Diffuse-type neurofibroma
Dermatofibrosarcoma protuberans
Solitary fibrous tumor
Arteriovenous malformation of the scalp
a
1
1
1
1
male
0
0
27
21-30
null
7
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2810455
A 27-year-old woman presented to her primary care physician with primary complaints of headaches and visual-spatial difficulties when driving at night. In addition, the patient described a progressively enlarging lesion on her left scalp, which she had initially noted in high school. She denied any overlying loss of hair, sharp pain, or bleeding from the scalp lesion, and she only reported pain with prolonged pressure. On her scalp examination, a 7-cm ill-defined spongy and pulsatile mass was noted with an overlying gray patch of hair. Magnetic resonance imaging (MRI) of the brain/orbit was initially performed (Figure, A and B), which demonstrated an ill-defined, diffuse, intensely enhancing scalp lesion in the left frontoparietal region with very prominent vascularity and underlying calvarial thickening. No osseous destruction or intracranial extension was present. Subsequent catheter angiography (Figure, C) confirmed the hypervascular mass, supplied via hypertrophied superficial temporal and occipital arteries with marked internal vascularity without direct involvement of the calvarium or intracranial structures. The patient underwent biopsy under general anesthesia; the specimen showed an ill-defined deep dermal/subcutaneous proliferation of spindle cells without significant nuclear pleomorphism or mitotic activity. The lesions stained positive for S100 and SOX10 immunostains.Coronal T2-weighted (A) and postcontrast T1-weighted (B) fat-saturated images show the large left T2 hyperintense scalp lesion (double arrowheads; A and B), with underlying calvarial thickening (asterisk; A and B) without invasion, prominent vascularity (single arrowhead; A), and enhancement (B). Note the preservation of the overlying skin. Catheter angiography (C) shows the hypervascular scalp lesion being supplied by hypertrophied branches arising from the left superficial temporal and occipital arteries, with marked internal vascularity (arrowheads). R indicates right. What Is Your Diagnosis?
Arteriovenous malformation of the scalp
Solitary fibrous tumor
Diffuse-type neurofibroma
Dermatofibrosarcoma protuberans
C. Diffuse-type neurofibroma
C
Diffuse-type neurofibroma
Neurofibromas, which are common in the head and neck, arise from endoneurium and connective tissues of peripheral nerve sheaths. From a morphologic and imaging perspective, there are 3 main types of neurofibromas: localized, diffuse, and plexiform.1 Diffuse neurofibromas are rare and represent less than 5% to 10% of all neurofibromas. The localized type, which is more common (>90%) and is the most well recognized and documented, is usually not a diagnostic conundrum. Plexiform neurofibromas, the other subtype, also have a pathognomonic imaging appearance and are frequently associated with neurofibromatosis 1, with findings of multiple lesions at different sites, which makes the diagnosis easier. Diffuse neurofibromas, however, are frequently solitary and are mostly sporadic, with few reported cases of associated with neurofibromatosis 1 (NF1).2 These lesions are more often seen in children and young adults. They are unusually large and frequently multispatial, with ill-defined margins, which appear to infiltrate and grow around tissues rather than invade them.On imaging, these are frequently plaquelike infiltrative lesions, unlike the more rounded or nodular appearance of localized neurofibromas, with frequent involvement of the subcutaneous/deep fascia both underlying the skin and overlying the musculature. Another characteristic is preservation of the overall morphologic features of the involved tissue space due to its unusual infiltrative pattern.2 A lesser known feature of these lesions is that many also demonstrate marked internal vascularity with prominent flow voids, arterial feeders, and draining veins, which can sometimes mimic vascular malformations.3 On MRI, these are isointense to mildly hyperintense to muscle on T1-weighted imaging and mild to markedly hyperintense on T2-weighted imaging to muscle, with marked enhancement. The lesions are usually solid without any necrotic or cystic changes, calcification, or hemorrhage.Solitary fibrous tumors (option B) (previously referred to as hemangiopericytomas) are rare mesenchymal-origin lesions, usually in older adults. On MRI, these are homogeneous lesions, with usually low to intermediate signals on both T1- and T2-weighted sequences (relative to muscle), with intense enhancement. They can be plaquelike and are frequently hypervascular, with numerous flow voids.4 Rarely, they may have a heterogeneous appearance secondary to internal hemorrhage or necrosis. They frequently mimic meningiomas when they arise intracranially.Scalp arteriovenous malformations (option A) are rare, and they present in children and young adults. On imaging, a predominant vascular lesion is identified with numerous flow voids due to its high flow state. The imaging features can be variable if there are associated abnormalities as well as other low-flow lesions, such as venous or lymphatic malformations. Vascular imaging, such as computed tomographic and magnetic resonance angiography, ultrasonography, and catheter angiography, is frequently done for both diagnosis and treatment planning.5Dermatofibrosarcoma protuberans (option D) are rare superficial lesions of the dermis, usually presenting in the third to fourth decade of life. These are locally aggressive, infiltrative masses that can arise in the head and neck, including the scalp. They present as a painless, slow-growing, nodular mass–like firm lesion. On imaging, these are markedly enhancing lesions, usually T1 hypointense and T2 hyperintense, with no vascular flow voids, underlying osseous invasion, or perineural spread.6 However, underlying calvarial thinning is seen in a few cases.Given the findings of the large scalp neurofibroma and her age, the patient was advised to undergo comprehensive genetic testing for NF1. Although NF1 is autosomal dominant and the patient had a negative family history, almost half of all individuals with NF1 are the first person in their family to have NF1 (de novo variant).7 The patient tested negative for identifiable pathogenic variants in the NF1 or SPRED1 genes, decreasing her risk of having NF1 and confirming that this was likely a sporadic neurofibroma.
General
A 27-year-old patient presented to them primary care physician with primary complaints of headaches and visual-spatial difficulties when driving at night. In addition, the patient described a progressively enlarging lesion on them left scalp, which they had initially noted in high school. They denied any overlying loss of hair, sharp pain, or bleeding from the scalp lesion, and they only reported pain with prolonged pressure. On them scalp examination, a 7-cm ill-defined spongy and pulsatile mass was noted with an overlying gray patch of hair. Magnetic resonance imaging (MRI) of the brain/orbit was initially performed (Figure, A and B), which demonstrated an ill-defined, diffuse, intensely enhancing scalp lesion in the left frontoparietal region with very prominent vascularity and underlying calvarial thickening. No osseous destruction or intracranial extension was present. Subsequent catheter angiography (Figure, C) confirmed the hypervascular mass, supplied via hypertrophied superficial temporal and occipital arteries with marked internal vascularity without direct involvement of the calvarium or intracranial structures. The patient underwent biopsy under general anesthesia; the specimen showed an ill-defined deep dermal/subcutaneous proliferation of spindle cells without significant nuclear pleomorphism or mitotic activity. The lesions stained positive for S100 and SOX10 immunostains.Coronal T2-weighted (A) and postcontrast T1-weighted (B) fat-saturated images show the large left T2 hyperintense scalp lesion (double arrowheads; A and B), with underlying calvarial thickening (asterisk; A and B) without invasion, prominent vascularity (single arrowhead; A), and enhancement (B). Note the preservation of the overlying skin. Catheter angiography (C) shows the hypervascular scalp lesion being supplied by hypertrophied branches arising from the left superficial temporal and occipital arteries, with marked internal vascularity (arrowheads). R indicates right.
what is your diagnosis?
What is your diagnosis?
Diffuse-type neurofibroma
Dermatofibrosarcoma protuberans
Solitary fibrous tumor
Arteriovenous malformation of the scalp
a
1
1
1
1
neutral
0
0
27
21-30
null
7
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2810452
A woman in her 30s presented for evaluation of asymptomatic erythematous scaly plaques over the face and proximal extremities. The lesions started as an erythematous papule on the face, which had progressed to larger plaques within 10 years. There was involvement of the upper respiratory tract, causing palatal perforation and stridor, necessitating a tracheostomy a year ago. In addition, there was history of wheezing and breathlessness. There was no history of preceding trauma, fever, malaise, joint pain, cough, epistaxis, hemoptysis, or hematuria.Examination revealed multiple well-defined brown erythematous scaly plaques with central scarring measuring 2 × 1 to 8 × 7 cm on the face, arm, and thighs. The patient also had saddle nose deformity, perforation of the hard palate, and necrotic plaques on the ear, causing destruction of ear cartilage (Figure, A and B). There was cervical lymphadenopathy: multiple, nontender, discrete nodes, ranging from 1.5 to 2 cm without any surface changes. The results of sensory examination and peripheral nerve examination were within normal limits. Respiratory examination and chest radiography results were normal. Hemogram showed anemia (hemoglobin, 10.1 g/dL [to convert to g/L, multiply by 10.0]); the results of a kidney function test, a liver function test, a fasting blood glucose test, and routine urinalysis were within normal limits. Serologic analysis results for HIV-1/2, antinuclear antibody, and antineutrophil cytoplasmic antibodies were negative. Punch skin biopsy was obtained for histopathologic analysis and culture (Figure, C).A, Multiple well-defined brown erythematous scaly plaques with central scarring, saddle nose deformity, and necrotic lesions on the ear with loss of ear cartilage. B, Perforation of the hard palate. C, Infiltrate of foamy histiocytes and pigmented yeasts without multiaxial septation (Fontana-Masson). What Is Your Diagnosis?
Chromoblastomycosis
Hyalohyphomycosis
Blastomycosis
Phaeohyphomycosis
D. Phaeohyphomycosis
D
Phaeohyphomycosis
Histopathologic results showed pseudoepitheliomatous hyperplasia, parakeratosis, dense dermal infiltrate of histiocytes, foamy histiocytes, neutrophils, and multinucleate giant cells along with pigmented hyphae. Fungal skin culture showed black yeastlike colonies characteristic of Exophiala spinifera species. Based on clinical presentation, histopathologic analysis, and fungal culture, the diagnosis of phaeohyphomycosis was made. The patient will be treated with intravenous amphotericin B, followed by itraconazole.Phaeohyphomycosis is a rare subcutaneous mycosis caused by dematiaceous fungi found in soil, wood, and decomposing vegetation. The disease, caused by dematiaceous fungi, is classified into 2 types based on fungal forms in the tissue: phaeohyphomycosis (presence of yeasts and hyphae) and chromoblastomycosis (muriform bodies). The literal meaning of phaeohyphomycosis is dark fungus with hyphae. The pigment melanin in the fungus is a virulence factor that inhibits phagocytosis by scavenging free radicals produced by the host. The common etiological agents are Exophiala, Aureobasidium, Cladosporium, Alternaria, Phialophora, Wangiella, and Curvularia.1 Phaeohyphomycosis can affect skin, subcutaneous tissue, paranasal sinuses, and, rarely, the central nervous system. Commonly, it presents as asymptomatic cystic swelling on exposed areas, usually after traumatic inoculation involving the hands, feet, and legs. Facial involvement is uncommon and can extend to involve the upper respiratory tract, causing perforation of the palate and destruction of the nasal bridge, leading to mutilation and disfigurement.2-4 Other rare presentations include verrucous plaques, erythematous papules, and noduloulcerative lesions. The diagnosis can be confirmed by demonstration of pigmented septate hyphae with occasional globose swelling on histopathologic analysis and fungal culture. Panfungal real-time polymerase chain reaction is an emerging tool in the diagnosis of phaeohyphomycosis. Treatment with azole antifungal agents (itraconazole, posaconazole, or voriconazole) is the mainstay of treatment. Other treatment options include amphotericin B, terbinafine, flucytosine, and griseofulvin along with physical modalities, such as excision, heat therapy, and cryotherapy.5The common deep mycoses that can cause mutilation include chromoblastomycosis, hyalohyphomycosis, blastomycosis, and phaeohyphomycosis. Both chromoblastomycosis and phaeohyphomycosis are caused by dematiaceous fungi. Chromoblastomycosis (option A) is commonly caused by Fonsecaea pedrosoi, Fonsecaea monophora, and Cladophialophora carrionii and usually presents with localized, verrucous, hyperkeratotic, irregular plaque on distal extremities with scarring.6 Hyalohyphomycosis (option B) is a rare opportunistic fungal infection caused by various fungi, which produce nonpigmented septate hyphae. It has myriad presentations, which include erythematous macules, papules, vesicles, indurated verrucous plaques, necrotic ulcers, and discharging sinus.7 Blastomycosis (option C) is a suppurative fungal infection caused by Blastomyces dermatitidis, endemic in the Ohio and Mississippi River valleys, the Great Lakes, and the Saint Lawrence River. It presents with papulopustular lesions, violaceous nodules, plaques, abscesses, ulcers, and verrucous or crusted lesions on exposed body parts.8 All these infections can have systemic involvement. Chromoblastomycosis involves lungs, lymph nodes, and bone, and hyalohyphomycosis primarily affects paranasal sinuses and lungs. Blastomycosis involves lymph nodes, bone, prostate, and testis, and phaeohyphomycosis commonly involves lungs and heart.All the differential diagnoses show suppurative granuloma on histopathologic analysis; the establishment of the respective fungal form helps in confirming the diagnosis. Chromoblastomycosis is characterized by thick-walled, multicellular, globe-shaped, cigar-colored inclusion bodies with multiaxial septation called sclerotic bodies (Medlar bodies).6 Hyphae in cross-sections in the index case resembled Medlar bodies, and the absence of septation was a clue indicating phaeohyphomycosis.7 Hyalohyphomycosis is characterized by nonpigmented septate hyphae, and culture helps in confirming the diagnosis.8 Large (8-15 μm) thick-walled yeast cells, prominent broad-based budding, and centrally retracted cytoplasm help in the diagnosis of blastomycosis.9Although many clinical differential diagnoses were considered in the case presented, histopathologic analysis and culture played decisive roles in arriving at a diagnosis of phaeohyphomycosis. This case highlights the importance of recognition of rare presentation of deep mycosis and the value of histopathologic analysis and fungal cultures in establishing the right diagnosis.
Dermatology
A woman in her 30s presented for evaluation of asymptomatic erythematous scaly plaques over the face and proximal extremities. The lesions started as an erythematous papule on the face, which had progressed to larger plaques within 10 years. There was involvement of the upper respiratory tract, causing palatal perforation and stridor, necessitating a tracheostomy a year ago. In addition, there was history of wheezing and breathlessness. There was no history of preceding trauma, fever, malaise, joint pain, cough, epistaxis, hemoptysis, or hematuria.Examination revealed multiple well-defined brown erythematous scaly plaques with central scarring measuring 2 × 1 to 8 × 7 cm on the face, arm, and thighs. The patient also had saddle nose deformity, perforation of the hard palate, and necrotic plaques on the ear, causing destruction of ear cartilage (Figure, A and B). There was cervical lymphadenopathy: multiple, nontender, discrete nodes, ranging from 1.5 to 2 cm without any surface changes. The results of sensory examination and peripheral nerve examination were within normal limits. Respiratory examination and chest radiography results were normal. Hemogram showed anemia (hemoglobin, 10.1 g/dL [to convert to g/L, multiply by 10.0]); the results of a kidney function test, a liver function test, a fasting blood glucose test, and routine urinalysis were within normal limits. Serologic analysis results for HIV-1/2, antinuclear antibody, and antineutrophil cytoplasmic antibodies were negative. Punch skin biopsy was obtained for histopathologic analysis and culture (Figure, C).A, Multiple well-defined brown erythematous scaly plaques with central scarring, saddle nose deformity, and necrotic lesions on the ear with loss of ear cartilage. B, Perforation of the hard palate. C, Infiltrate of foamy histiocytes and pigmented yeasts without multiaxial septation (Fontana-Masson).
what is your diagnosis?
What is your diagnosis?
Blastomycosis
Phaeohyphomycosis
Hyalohyphomycosis
Chromoblastomycosis
b
0
1
1
1
female
0
0
35
31-40
null
8
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2810452
A woman in her 30s presented for evaluation of asymptomatic erythematous scaly plaques over the face and proximal extremities. The lesions started as an erythematous papule on the face, which had progressed to larger plaques within 10 years. There was involvement of the upper respiratory tract, causing palatal perforation and stridor, necessitating a tracheostomy a year ago. In addition, there was history of wheezing and breathlessness. There was no history of preceding trauma, fever, malaise, joint pain, cough, epistaxis, hemoptysis, or hematuria.Examination revealed multiple well-defined brown erythematous scaly plaques with central scarring measuring 2 × 1 to 8 × 7 cm on the face, arm, and thighs. The patient also had saddle nose deformity, perforation of the hard palate, and necrotic plaques on the ear, causing destruction of ear cartilage (Figure, A and B). There was cervical lymphadenopathy: multiple, nontender, discrete nodes, ranging from 1.5 to 2 cm without any surface changes. The results of sensory examination and peripheral nerve examination were within normal limits. Respiratory examination and chest radiography results were normal. Hemogram showed anemia (hemoglobin, 10.1 g/dL [to convert to g/L, multiply by 10.0]); the results of a kidney function test, a liver function test, a fasting blood glucose test, and routine urinalysis were within normal limits. Serologic analysis results for HIV-1/2, antinuclear antibody, and antineutrophil cytoplasmic antibodies were negative. Punch skin biopsy was obtained for histopathologic analysis and culture (Figure, C).A, Multiple well-defined brown erythematous scaly plaques with central scarring, saddle nose deformity, and necrotic lesions on the ear with loss of ear cartilage. B, Perforation of the hard palate. C, Infiltrate of foamy histiocytes and pigmented yeasts without multiaxial septation (Fontana-Masson). What Is Your Diagnosis?
Chromoblastomycosis
Hyalohyphomycosis
Blastomycosis
Phaeohyphomycosis
D. Phaeohyphomycosis
D
Phaeohyphomycosis
Histopathologic results showed pseudoepitheliomatous hyperplasia, parakeratosis, dense dermal infiltrate of histiocytes, foamy histiocytes, neutrophils, and multinucleate giant cells along with pigmented hyphae. Fungal skin culture showed black yeastlike colonies characteristic of Exophiala spinifera species. Based on clinical presentation, histopathologic analysis, and fungal culture, the diagnosis of phaeohyphomycosis was made. The patient will be treated with intravenous amphotericin B, followed by itraconazole.Phaeohyphomycosis is a rare subcutaneous mycosis caused by dematiaceous fungi found in soil, wood, and decomposing vegetation. The disease, caused by dematiaceous fungi, is classified into 2 types based on fungal forms in the tissue: phaeohyphomycosis (presence of yeasts and hyphae) and chromoblastomycosis (muriform bodies). The literal meaning of phaeohyphomycosis is dark fungus with hyphae. The pigment melanin in the fungus is a virulence factor that inhibits phagocytosis by scavenging free radicals produced by the host. The common etiological agents are Exophiala, Aureobasidium, Cladosporium, Alternaria, Phialophora, Wangiella, and Curvularia.1 Phaeohyphomycosis can affect skin, subcutaneous tissue, paranasal sinuses, and, rarely, the central nervous system. Commonly, it presents as asymptomatic cystic swelling on exposed areas, usually after traumatic inoculation involving the hands, feet, and legs. Facial involvement is uncommon and can extend to involve the upper respiratory tract, causing perforation of the palate and destruction of the nasal bridge, leading to mutilation and disfigurement.2-4 Other rare presentations include verrucous plaques, erythematous papules, and noduloulcerative lesions. The diagnosis can be confirmed by demonstration of pigmented septate hyphae with occasional globose swelling on histopathologic analysis and fungal culture. Panfungal real-time polymerase chain reaction is an emerging tool in the diagnosis of phaeohyphomycosis. Treatment with azole antifungal agents (itraconazole, posaconazole, or voriconazole) is the mainstay of treatment. Other treatment options include amphotericin B, terbinafine, flucytosine, and griseofulvin along with physical modalities, such as excision, heat therapy, and cryotherapy.5The common deep mycoses that can cause mutilation include chromoblastomycosis, hyalohyphomycosis, blastomycosis, and phaeohyphomycosis. Both chromoblastomycosis and phaeohyphomycosis are caused by dematiaceous fungi. Chromoblastomycosis (option A) is commonly caused by Fonsecaea pedrosoi, Fonsecaea monophora, and Cladophialophora carrionii and usually presents with localized, verrucous, hyperkeratotic, irregular plaque on distal extremities with scarring.6 Hyalohyphomycosis (option B) is a rare opportunistic fungal infection caused by various fungi, which produce nonpigmented septate hyphae. It has myriad presentations, which include erythematous macules, papules, vesicles, indurated verrucous plaques, necrotic ulcers, and discharging sinus.7 Blastomycosis (option C) is a suppurative fungal infection caused by Blastomyces dermatitidis, endemic in the Ohio and Mississippi River valleys, the Great Lakes, and the Saint Lawrence River. It presents with papulopustular lesions, violaceous nodules, plaques, abscesses, ulcers, and verrucous or crusted lesions on exposed body parts.8 All these infections can have systemic involvement. Chromoblastomycosis involves lungs, lymph nodes, and bone, and hyalohyphomycosis primarily affects paranasal sinuses and lungs. Blastomycosis involves lymph nodes, bone, prostate, and testis, and phaeohyphomycosis commonly involves lungs and heart.All the differential diagnoses show suppurative granuloma on histopathologic analysis; the establishment of the respective fungal form helps in confirming the diagnosis. Chromoblastomycosis is characterized by thick-walled, multicellular, globe-shaped, cigar-colored inclusion bodies with multiaxial septation called sclerotic bodies (Medlar bodies).6 Hyphae in cross-sections in the index case resembled Medlar bodies, and the absence of septation was a clue indicating phaeohyphomycosis.7 Hyalohyphomycosis is characterized by nonpigmented septate hyphae, and culture helps in confirming the diagnosis.8 Large (8-15 μm) thick-walled yeast cells, prominent broad-based budding, and centrally retracted cytoplasm help in the diagnosis of blastomycosis.9Although many clinical differential diagnoses were considered in the case presented, histopathologic analysis and culture played decisive roles in arriving at a diagnosis of phaeohyphomycosis. This case highlights the importance of recognition of rare presentation of deep mycosis and the value of histopathologic analysis and fungal cultures in establishing the right diagnosis.
Dermatology
A man in him 30s presented for evaluation of asymptomatic erythematous scaly plaques over the face and proximal extremities. The lesions started as an erythematous papule on the face, which had progressed to larger plaques within 10 years. There was involvement of the upper respiratory tract, causing palatal perforation and stridor, necessitating a tracheostomy a year ago. In addition, there was history of wheezing and breathlessness. There was no history of preceding trauma, fever, malaise, joint pain, cough, epistaxis, hemoptysis, or hematuria.Examination revealed multiple well-defined brown erythematous scaly plaques with central scarring measuring 2 × 1 to 8 × 7 cm on the face, arm, and thighs. The man also had saddle nose deformity, perforation of the hard palate, and necrotic plaques on the ear, causing destruction of ear cartilage (Figure, A and B). There was cervical lymphadenopathy: multiple, nontender, discrete nodes, ranging from 1.5 to 2 cm without any surface changes. The results of sensory examination and peripheral nerve examination were within normal limits. Respiratory examination and chest radiography results were normal. Hemogram showed anemia (hemoglobin, 10.1 g/dL [to convert to g/L, multiply by 10.0]); the results of a kidney function test, a liver function test, a fasting blood glucose test, and routine urinalysis were within normal limits. Serologic analysis results for HIV-1/2, antinuclear antibody, and antineutrophil cytoplasmic antibodies were negative. Punch skin biopsy was obtained for histopathologic analysis and culture (Figure, C).A, Multiple well-defined brown erythematous scaly plaques with central scarring, saddle nose deformity, and necrotic lesions on the ear with loss of ear cartilage. B, Perforation of the hard palate. C, Infiltrate of foamy histiocytes and pigmented yeasts without multiaxial septation (Fontana-Masson).
what is your diagnosis?
What is your diagnosis?
Blastomycosis
Phaeohyphomycosis
Hyalohyphomycosis
Chromoblastomycosis
b
0
1
1
1
male
0
0
35
31-40
null
8
augmented_male_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2810452
A woman in her 30s presented for evaluation of asymptomatic erythematous scaly plaques over the face and proximal extremities. The lesions started as an erythematous papule on the face, which had progressed to larger plaques within 10 years. There was involvement of the upper respiratory tract, causing palatal perforation and stridor, necessitating a tracheostomy a year ago. In addition, there was history of wheezing and breathlessness. There was no history of preceding trauma, fever, malaise, joint pain, cough, epistaxis, hemoptysis, or hematuria.Examination revealed multiple well-defined brown erythematous scaly plaques with central scarring measuring 2 × 1 to 8 × 7 cm on the face, arm, and thighs. The patient also had saddle nose deformity, perforation of the hard palate, and necrotic plaques on the ear, causing destruction of ear cartilage (Figure, A and B). There was cervical lymphadenopathy: multiple, nontender, discrete nodes, ranging from 1.5 to 2 cm without any surface changes. The results of sensory examination and peripheral nerve examination were within normal limits. Respiratory examination and chest radiography results were normal. Hemogram showed anemia (hemoglobin, 10.1 g/dL [to convert to g/L, multiply by 10.0]); the results of a kidney function test, a liver function test, a fasting blood glucose test, and routine urinalysis were within normal limits. Serologic analysis results for HIV-1/2, antinuclear antibody, and antineutrophil cytoplasmic antibodies were negative. Punch skin biopsy was obtained for histopathologic analysis and culture (Figure, C).A, Multiple well-defined brown erythematous scaly plaques with central scarring, saddle nose deformity, and necrotic lesions on the ear with loss of ear cartilage. B, Perforation of the hard palate. C, Infiltrate of foamy histiocytes and pigmented yeasts without multiaxial septation (Fontana-Masson). What Is Your Diagnosis?
Chromoblastomycosis
Hyalohyphomycosis
Blastomycosis
Phaeohyphomycosis
D. Phaeohyphomycosis
D
Phaeohyphomycosis
Histopathologic results showed pseudoepitheliomatous hyperplasia, parakeratosis, dense dermal infiltrate of histiocytes, foamy histiocytes, neutrophils, and multinucleate giant cells along with pigmented hyphae. Fungal skin culture showed black yeastlike colonies characteristic of Exophiala spinifera species. Based on clinical presentation, histopathologic analysis, and fungal culture, the diagnosis of phaeohyphomycosis was made. The patient will be treated with intravenous amphotericin B, followed by itraconazole.Phaeohyphomycosis is a rare subcutaneous mycosis caused by dematiaceous fungi found in soil, wood, and decomposing vegetation. The disease, caused by dematiaceous fungi, is classified into 2 types based on fungal forms in the tissue: phaeohyphomycosis (presence of yeasts and hyphae) and chromoblastomycosis (muriform bodies). The literal meaning of phaeohyphomycosis is dark fungus with hyphae. The pigment melanin in the fungus is a virulence factor that inhibits phagocytosis by scavenging free radicals produced by the host. The common etiological agents are Exophiala, Aureobasidium, Cladosporium, Alternaria, Phialophora, Wangiella, and Curvularia.1 Phaeohyphomycosis can affect skin, subcutaneous tissue, paranasal sinuses, and, rarely, the central nervous system. Commonly, it presents as asymptomatic cystic swelling on exposed areas, usually after traumatic inoculation involving the hands, feet, and legs. Facial involvement is uncommon and can extend to involve the upper respiratory tract, causing perforation of the palate and destruction of the nasal bridge, leading to mutilation and disfigurement.2-4 Other rare presentations include verrucous plaques, erythematous papules, and noduloulcerative lesions. The diagnosis can be confirmed by demonstration of pigmented septate hyphae with occasional globose swelling on histopathologic analysis and fungal culture. Panfungal real-time polymerase chain reaction is an emerging tool in the diagnosis of phaeohyphomycosis. Treatment with azole antifungal agents (itraconazole, posaconazole, or voriconazole) is the mainstay of treatment. Other treatment options include amphotericin B, terbinafine, flucytosine, and griseofulvin along with physical modalities, such as excision, heat therapy, and cryotherapy.5The common deep mycoses that can cause mutilation include chromoblastomycosis, hyalohyphomycosis, blastomycosis, and phaeohyphomycosis. Both chromoblastomycosis and phaeohyphomycosis are caused by dematiaceous fungi. Chromoblastomycosis (option A) is commonly caused by Fonsecaea pedrosoi, Fonsecaea monophora, and Cladophialophora carrionii and usually presents with localized, verrucous, hyperkeratotic, irregular plaque on distal extremities with scarring.6 Hyalohyphomycosis (option B) is a rare opportunistic fungal infection caused by various fungi, which produce nonpigmented septate hyphae. It has myriad presentations, which include erythematous macules, papules, vesicles, indurated verrucous plaques, necrotic ulcers, and discharging sinus.7 Blastomycosis (option C) is a suppurative fungal infection caused by Blastomyces dermatitidis, endemic in the Ohio and Mississippi River valleys, the Great Lakes, and the Saint Lawrence River. It presents with papulopustular lesions, violaceous nodules, plaques, abscesses, ulcers, and verrucous or crusted lesions on exposed body parts.8 All these infections can have systemic involvement. Chromoblastomycosis involves lungs, lymph nodes, and bone, and hyalohyphomycosis primarily affects paranasal sinuses and lungs. Blastomycosis involves lymph nodes, bone, prostate, and testis, and phaeohyphomycosis commonly involves lungs and heart.All the differential diagnoses show suppurative granuloma on histopathologic analysis; the establishment of the respective fungal form helps in confirming the diagnosis. Chromoblastomycosis is characterized by thick-walled, multicellular, globe-shaped, cigar-colored inclusion bodies with multiaxial septation called sclerotic bodies (Medlar bodies).6 Hyphae in cross-sections in the index case resembled Medlar bodies, and the absence of septation was a clue indicating phaeohyphomycosis.7 Hyalohyphomycosis is characterized by nonpigmented septate hyphae, and culture helps in confirming the diagnosis.8 Large (8-15 μm) thick-walled yeast cells, prominent broad-based budding, and centrally retracted cytoplasm help in the diagnosis of blastomycosis.9Although many clinical differential diagnoses were considered in the case presented, histopathologic analysis and culture played decisive roles in arriving at a diagnosis of phaeohyphomycosis. This case highlights the importance of recognition of rare presentation of deep mycosis and the value of histopathologic analysis and fungal cultures in establishing the right diagnosis.
Dermatology
A patient in them 30s presented for evaluation of asymptomatic erythematous scaly plaques over the face and proximal extremities. The lesions started as an erythematous papule on the face, which had progressed to larger plaques within 10 years. There was involvement of the upper respiratory tract, causing palatal perforation and stridor, necessitating a tracheostomy a year ago. In addition, there was history of wheezing and breathlessness. There was no history of preceding trauma, fever, malaise, joint pain, cough, epistaxis, hemoptysis, or hematuria.Examination revealed multiple well-defined brown erythematous scaly plaques with central scarring measuring 2 × 1 to 8 × 7 cm on the face, arm, and thighs. The patient also had saddle nose deformity, perforation of the hard palate, and necrotic plaques on the ear, causing destruction of ear cartilage (Figure, A and B). There was cervical lymphadenopathy: multiple, nontender, discrete nodes, ranging from 1.5 to 2 cm without any surface changes. The results of sensory examination and peripheral nerve examination were within normal limits. Respiratory examination and chest radiography results were normal. Hemogram showed anemia (hemoglobin, 10.1 g/dL [to convert to g/L, multiply by 10.0]); the results of a kidney function test, a liver function test, a fasting blood glucose test, and routine urinalysis were within normal limits. Serologic analysis results for HIV-1/2, antinuclear antibody, and antineutrophil cytoplasmic antibodies were negative. Punch skin biopsy was obtained for histopathologic analysis and culture (Figure, C).A, Multiple well-defined brown erythematous scaly plaques with central scarring, saddle nose deformity, and necrotic lesions on the ear with loss of ear cartilage. B, Perforation of the hard palate. C, Infiltrate of foamy histiocytes and pigmented yeasts without multiaxial septation (Fontana-Masson).
what is your diagnosis?
What is your diagnosis?
Blastomycosis
Phaeohyphomycosis
Hyalohyphomycosis
Chromoblastomycosis
b
0
1
1
1
neutral
0
0
35
31-40
null
8
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2808316
A 64-year-old man with a 2-year history of seizure disorder presented to the emergency department (ED) at an outside hospital with breakthrough seizures. One month prior to his ED visit, he had been treated with oral antibiotics when he presented to an urgent care center with left-sided otorrhea, otalgia, and hearing loss. In the ED, he denied headaches, fever, otalgia, otorrhea, or hearing loss. Magnetic resonance imaging of the brain with contrast showed diffuse enhancement and possible coalescence in the left mastoid process with adjacent dural enhancement and sigmoid sinus occlusion. He received intravenous vancomycin and cefepime and was started on a heparin drip before being transferred to our facility. On presentation at our facility, the patient appeared well with normal vital signs. There was no periauricular swelling or erythema. Otoscopic examination showed no inflammation of the external auditory canals, and the tympanic membranes were translucent without evidence of retraction or effusion. Computed tomographic venogram confirmed occlusion of the left transverse and sigmoid sinuses, and computed tomography of the temporal bone showed coalescence of mastoid air cells with erosion into the sigmoid sinus. Mastoidectomy was performed for biopsy and culture specimens. Histologic findings from the specimens demonstrated cells with oval nuclei and abundant cytoplasm arranged in syncytial clusters with occasional whorls, pseudoinclusions, and psammomatous calcifications (Figure, A). Immunohistochemical stains were positive for epithelial membrane antigen (EMA) and vimentin (Figure, B).Hematoxylin-eosin stain (A) and immunohistochemical stain (B) after mastoidectomy for biopsy and culture specimens. What Is Your Diagnosis?
Langerhans cell histiocytosis
Meningioma
Acute coalescent mastoiditis with sigmoid sinus thrombosis
Cholesteatoma
B. Meningioma
B
Meningioma
This patient presented to the ED with an acute exacerbation of a seizure disorder. The recent history of left-sided otorrhea and otalgia with magnetic resonance imaging enhancement in the mastoid along with dehiscence and occlusion of the adjacent venous sinuses suggested that the seizure may have been triggered by an indolent suppurative complication of mastoiditis. However, because the patient’s otologic symptoms had resolved and he did not appear septic, temporal bone neoplasm was considered. Mastoidectomy revealed solid tumor attached to the sigmoid sinus with no evidence of mastoid suppuration or inflammatory process, and the biopsy was diagnostic for intraosseous meningioma.Meningiomas typically are intracranial neoplasms, but 2% present as extracranial intraosseous lesions.1,2 Extracranial meningiomas present in the middle ear, external auditory canal, petrous bone, squamous temporal bone, and paranasal sinuses.1 Histologic sections in this case showed findings consistent with a meningothelial variant of meningioma World Health Organization grade I.1,3 Immunohistochemistry studies confirmed the diagnosis with positive EMA and vimentin staining.3 However, somatostatin receptor subtype 2A and progesterone receptors are more sensitive for diagnosis. Intraosseous meningiomas originate from arachnoid cap cells associated with the sigmoid sinus or adjacent dura. As was the case with this patient, most patients with temporal bone intraosseous meningiomas are asymptomatic, and observation with serial imaging is appropriate. Surgical debulking or resection is considered when tumors grow and extend into the middle ear causing hearing loss, compromising patency of the external auditory canal, or compressing the facial nerve. Fractionated external-beam radiation therapy or stereotactic radiation therapy are also options for treating growing tumors, but there is a high risk of profound hearing loss.2Coalescent mastoiditis can present with intracranial complications such as sigmoid sinus thrombosis or subperiosteal abscess.4 Typically, patients with acute mastoiditis present with headaches, vomiting, seizures, otalgia, otorrhea, and fever.5 Physical findings include tenderness and swelling inferior to the temporal line, purulent middle ear effusion, and conductive hearing loss. Cholesteatoma can be the source of infection in patients with acute mastoiditis, but otomicroscopic findings would reveal a retraction of the tympanic membrane, scutal erosion, or secondary acquired cholesteatoma associated with a tympanic membrane perforation. The absence of these symptoms and physical findings make these diagnoses less likely, yet it is possible to have an indolent progressive suppurative mastoid infection that remains isolated from the middle ear and erodes the thin bony plate over the sigmoid sinus. Mastoidectomy was critical to differentiate an infectious process from neoplastic pathology. Histologic results for a cholesteatoma would reveal keratinizing stratified squamous epithelium with stromal fibrosis. Biopsy in patients with mastoiditis shows granulation tissue, acute and chronic leukocyte infiltration, and hemorrhage with negative EMA and vimentin staining. Mastoidectomy in patients with acute coalescent mastoiditis provides drainage of the infection and cultures to select intravenous antibiotics. Anticoagulation may be recommended when acute coalescent mastoiditis results in sigmoid sinus thrombosis.6,7Langerhans cell histiocytosis of the temporal bone is extremely rare in older adults. Patients present with otorrhea, subjective hearing loss, otalgia, and soft-tissue postauricular swelling.8 Physical examination shows granulation tissue in the external ear canal, middle ear effusion, conductive hearing loss, and external auditory canal stenosis.8 The absence of these symptoms and physical examination findings make this diagnosis less likely. Computed tomography of the temporal bone shows erosion of mastoid air cells and destruction of the mastoid cortex while the otic capsule is spared.9 Computed tomographic imaging is critical for diagnosing and following Langerhans cell histiocytosis, but biopsy is required to confirm the diagnosis. Histological sections typically demonstrate mononuclear cells with coffee bean nuclei that are CD1a, S100, and langerin positive.8,9 For focal lesions, surgery is recommended, and for multifocal lesions chemotherapy (typically vincristine and steroids) and surgery is recommended.8,10
General
A 64-year-old man with a 2-year history of seizure disorder presented to the emergency department (ED) at an outside hospital with breakthrough seizures. One month prior to his ED visit, he had been treated with oral antibiotics when he presented to an urgent care center with left-sided otorrhea, otalgia, and hearing loss. In the ED, he denied headaches, fever, otalgia, otorrhea, or hearing loss. Magnetic resonance imaging of the brain with contrast showed diffuse enhancement and possible coalescence in the left mastoid process with adjacent dural enhancement and sigmoid sinus occlusion. He received intravenous vancomycin and cefepime and was started on a heparin drip before being transferred to our facility. On presentation at our facility, the patient appeared well with normal vital signs. There was no periauricular swelling or erythema. Otoscopic examination showed no inflammation of the external auditory canals, and the tympanic membranes were translucent without evidence of retraction or effusion. Computed tomographic venogram confirmed occlusion of the left transverse and sigmoid sinuses, and computed tomography of the temporal bone showed coalescence of mastoid air cells with erosion into the sigmoid sinus. Mastoidectomy was performed for biopsy and culture specimens. Histologic findings from the specimens demonstrated cells with oval nuclei and abundant cytoplasm arranged in syncytial clusters with occasional whorls, pseudoinclusions, and psammomatous calcifications (Figure, A). Immunohistochemical stains were positive for epithelial membrane antigen (EMA) and vimentin (Figure, B).Hematoxylin-eosin stain (A) and immunohistochemical stain (B) after mastoidectomy for biopsy and culture specimens.
what is your diagnosis?
What is your diagnosis?
Acute coalescent mastoiditis with sigmoid sinus thrombosis
Meningioma
Cholesteatoma
Langerhans cell histiocytosis
b
1
1
1
1
male
0
0
64
61-70
null
9
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2808316
A 64-year-old man with a 2-year history of seizure disorder presented to the emergency department (ED) at an outside hospital with breakthrough seizures. One month prior to his ED visit, he had been treated with oral antibiotics when he presented to an urgent care center with left-sided otorrhea, otalgia, and hearing loss. In the ED, he denied headaches, fever, otalgia, otorrhea, or hearing loss. Magnetic resonance imaging of the brain with contrast showed diffuse enhancement and possible coalescence in the left mastoid process with adjacent dural enhancement and sigmoid sinus occlusion. He received intravenous vancomycin and cefepime and was started on a heparin drip before being transferred to our facility. On presentation at our facility, the patient appeared well with normal vital signs. There was no periauricular swelling or erythema. Otoscopic examination showed no inflammation of the external auditory canals, and the tympanic membranes were translucent without evidence of retraction or effusion. Computed tomographic venogram confirmed occlusion of the left transverse and sigmoid sinuses, and computed tomography of the temporal bone showed coalescence of mastoid air cells with erosion into the sigmoid sinus. Mastoidectomy was performed for biopsy and culture specimens. Histologic findings from the specimens demonstrated cells with oval nuclei and abundant cytoplasm arranged in syncytial clusters with occasional whorls, pseudoinclusions, and psammomatous calcifications (Figure, A). Immunohistochemical stains were positive for epithelial membrane antigen (EMA) and vimentin (Figure, B).Hematoxylin-eosin stain (A) and immunohistochemical stain (B) after mastoidectomy for biopsy and culture specimens. What Is Your Diagnosis?
Langerhans cell histiocytosis
Meningioma
Acute coalescent mastoiditis with sigmoid sinus thrombosis
Cholesteatoma
B. Meningioma
B
Meningioma
This patient presented to the ED with an acute exacerbation of a seizure disorder. The recent history of left-sided otorrhea and otalgia with magnetic resonance imaging enhancement in the mastoid along with dehiscence and occlusion of the adjacent venous sinuses suggested that the seizure may have been triggered by an indolent suppurative complication of mastoiditis. However, because the patient’s otologic symptoms had resolved and he did not appear septic, temporal bone neoplasm was considered. Mastoidectomy revealed solid tumor attached to the sigmoid sinus with no evidence of mastoid suppuration or inflammatory process, and the biopsy was diagnostic for intraosseous meningioma.Meningiomas typically are intracranial neoplasms, but 2% present as extracranial intraosseous lesions.1,2 Extracranial meningiomas present in the middle ear, external auditory canal, petrous bone, squamous temporal bone, and paranasal sinuses.1 Histologic sections in this case showed findings consistent with a meningothelial variant of meningioma World Health Organization grade I.1,3 Immunohistochemistry studies confirmed the diagnosis with positive EMA and vimentin staining.3 However, somatostatin receptor subtype 2A and progesterone receptors are more sensitive for diagnosis. Intraosseous meningiomas originate from arachnoid cap cells associated with the sigmoid sinus or adjacent dura. As was the case with this patient, most patients with temporal bone intraosseous meningiomas are asymptomatic, and observation with serial imaging is appropriate. Surgical debulking or resection is considered when tumors grow and extend into the middle ear causing hearing loss, compromising patency of the external auditory canal, or compressing the facial nerve. Fractionated external-beam radiation therapy or stereotactic radiation therapy are also options for treating growing tumors, but there is a high risk of profound hearing loss.2Coalescent mastoiditis can present with intracranial complications such as sigmoid sinus thrombosis or subperiosteal abscess.4 Typically, patients with acute mastoiditis present with headaches, vomiting, seizures, otalgia, otorrhea, and fever.5 Physical findings include tenderness and swelling inferior to the temporal line, purulent middle ear effusion, and conductive hearing loss. Cholesteatoma can be the source of infection in patients with acute mastoiditis, but otomicroscopic findings would reveal a retraction of the tympanic membrane, scutal erosion, or secondary acquired cholesteatoma associated with a tympanic membrane perforation. The absence of these symptoms and physical findings make these diagnoses less likely, yet it is possible to have an indolent progressive suppurative mastoid infection that remains isolated from the middle ear and erodes the thin bony plate over the sigmoid sinus. Mastoidectomy was critical to differentiate an infectious process from neoplastic pathology. Histologic results for a cholesteatoma would reveal keratinizing stratified squamous epithelium with stromal fibrosis. Biopsy in patients with mastoiditis shows granulation tissue, acute and chronic leukocyte infiltration, and hemorrhage with negative EMA and vimentin staining. Mastoidectomy in patients with acute coalescent mastoiditis provides drainage of the infection and cultures to select intravenous antibiotics. Anticoagulation may be recommended when acute coalescent mastoiditis results in sigmoid sinus thrombosis.6,7Langerhans cell histiocytosis of the temporal bone is extremely rare in older adults. Patients present with otorrhea, subjective hearing loss, otalgia, and soft-tissue postauricular swelling.8 Physical examination shows granulation tissue in the external ear canal, middle ear effusion, conductive hearing loss, and external auditory canal stenosis.8 The absence of these symptoms and physical examination findings make this diagnosis less likely. Computed tomography of the temporal bone shows erosion of mastoid air cells and destruction of the mastoid cortex while the otic capsule is spared.9 Computed tomographic imaging is critical for diagnosing and following Langerhans cell histiocytosis, but biopsy is required to confirm the diagnosis. Histological sections typically demonstrate mononuclear cells with coffee bean nuclei that are CD1a, S100, and langerin positive.8,9 For focal lesions, surgery is recommended, and for multifocal lesions chemotherapy (typically vincristine and steroids) and surgery is recommended.8,10
General
A 64-year-old woman with a 2-year history of seizure disorder presented to the emergency department (ED) at an outside hospital with breakthrough seizures. One month prior to her ED visit, she had been treated with oral antibiotics when she presented to an urgent care center with left-sided otorrhea, otalgia, and hearing loss. In the ED, she denied headaches, fever, otalgia, otorrhea, or hearing loss. Magnetic resonance imaging of the brain with contrast showed diffuse enhancement and possible coalescence in the left mastoid process with adjacent dural enhancement and sigmoid sinus occlusion. She received intravenous vancomycin and cefepime and was started on a heparin drip before being transferred to our facility. On presentation at our facility, the woman appeared well with normal vital signs. There was no periauricular swelling or erythema. Otoscopic examination showed no inflammation of the external auditory canals, and the tympanic membranes were translucent without evidence of retraction or effusion. Computed tomographic venogram confirmed occlusion of the left transverse and sigmoid sinuses, and computed tomography of the temporal bone showed coalescence of mastoid air cells with erosion into the sigmoid sinus. Mastoidectomy was performed for biopsy and culture specimens. Histologic findings from the specimens demonstrated cells with oval nuclei and abundant cytoplasm arranged in syncytial clusters with occasional whorls, pseudoinclusions, and psammomatous calcifications (Figure, A). Immunohistochemical stains were positive for epithelial membrane antigen (EMA) and vimentin (Figure, B).Hematoxylin-eosin stain (A) and immunohistochemical stain (B) after mastoidectomy for biopsy and culture specimens.
what is your diagnosis?
What is your diagnosis?
Acute coalescent mastoiditis with sigmoid sinus thrombosis
Meningioma
Cholesteatoma
Langerhans cell histiocytosis
b
1
1
1
1
female
0
0
64
61-70
null
9
augmented_female_frommale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2808316
A 64-year-old man with a 2-year history of seizure disorder presented to the emergency department (ED) at an outside hospital with breakthrough seizures. One month prior to his ED visit, he had been treated with oral antibiotics when he presented to an urgent care center with left-sided otorrhea, otalgia, and hearing loss. In the ED, he denied headaches, fever, otalgia, otorrhea, or hearing loss. Magnetic resonance imaging of the brain with contrast showed diffuse enhancement and possible coalescence in the left mastoid process with adjacent dural enhancement and sigmoid sinus occlusion. He received intravenous vancomycin and cefepime and was started on a heparin drip before being transferred to our facility. On presentation at our facility, the patient appeared well with normal vital signs. There was no periauricular swelling or erythema. Otoscopic examination showed no inflammation of the external auditory canals, and the tympanic membranes were translucent without evidence of retraction or effusion. Computed tomographic venogram confirmed occlusion of the left transverse and sigmoid sinuses, and computed tomography of the temporal bone showed coalescence of mastoid air cells with erosion into the sigmoid sinus. Mastoidectomy was performed for biopsy and culture specimens. Histologic findings from the specimens demonstrated cells with oval nuclei and abundant cytoplasm arranged in syncytial clusters with occasional whorls, pseudoinclusions, and psammomatous calcifications (Figure, A). Immunohistochemical stains were positive for epithelial membrane antigen (EMA) and vimentin (Figure, B).Hematoxylin-eosin stain (A) and immunohistochemical stain (B) after mastoidectomy for biopsy and culture specimens. What Is Your Diagnosis?
Langerhans cell histiocytosis
Meningioma
Acute coalescent mastoiditis with sigmoid sinus thrombosis
Cholesteatoma
B. Meningioma
B
Meningioma
This patient presented to the ED with an acute exacerbation of a seizure disorder. The recent history of left-sided otorrhea and otalgia with magnetic resonance imaging enhancement in the mastoid along with dehiscence and occlusion of the adjacent venous sinuses suggested that the seizure may have been triggered by an indolent suppurative complication of mastoiditis. However, because the patient’s otologic symptoms had resolved and he did not appear septic, temporal bone neoplasm was considered. Mastoidectomy revealed solid tumor attached to the sigmoid sinus with no evidence of mastoid suppuration or inflammatory process, and the biopsy was diagnostic for intraosseous meningioma.Meningiomas typically are intracranial neoplasms, but 2% present as extracranial intraosseous lesions.1,2 Extracranial meningiomas present in the middle ear, external auditory canal, petrous bone, squamous temporal bone, and paranasal sinuses.1 Histologic sections in this case showed findings consistent with a meningothelial variant of meningioma World Health Organization grade I.1,3 Immunohistochemistry studies confirmed the diagnosis with positive EMA and vimentin staining.3 However, somatostatin receptor subtype 2A and progesterone receptors are more sensitive for diagnosis. Intraosseous meningiomas originate from arachnoid cap cells associated with the sigmoid sinus or adjacent dura. As was the case with this patient, most patients with temporal bone intraosseous meningiomas are asymptomatic, and observation with serial imaging is appropriate. Surgical debulking or resection is considered when tumors grow and extend into the middle ear causing hearing loss, compromising patency of the external auditory canal, or compressing the facial nerve. Fractionated external-beam radiation therapy or stereotactic radiation therapy are also options for treating growing tumors, but there is a high risk of profound hearing loss.2Coalescent mastoiditis can present with intracranial complications such as sigmoid sinus thrombosis or subperiosteal abscess.4 Typically, patients with acute mastoiditis present with headaches, vomiting, seizures, otalgia, otorrhea, and fever.5 Physical findings include tenderness and swelling inferior to the temporal line, purulent middle ear effusion, and conductive hearing loss. Cholesteatoma can be the source of infection in patients with acute mastoiditis, but otomicroscopic findings would reveal a retraction of the tympanic membrane, scutal erosion, or secondary acquired cholesteatoma associated with a tympanic membrane perforation. The absence of these symptoms and physical findings make these diagnoses less likely, yet it is possible to have an indolent progressive suppurative mastoid infection that remains isolated from the middle ear and erodes the thin bony plate over the sigmoid sinus. Mastoidectomy was critical to differentiate an infectious process from neoplastic pathology. Histologic results for a cholesteatoma would reveal keratinizing stratified squamous epithelium with stromal fibrosis. Biopsy in patients with mastoiditis shows granulation tissue, acute and chronic leukocyte infiltration, and hemorrhage with negative EMA and vimentin staining. Mastoidectomy in patients with acute coalescent mastoiditis provides drainage of the infection and cultures to select intravenous antibiotics. Anticoagulation may be recommended when acute coalescent mastoiditis results in sigmoid sinus thrombosis.6,7Langerhans cell histiocytosis of the temporal bone is extremely rare in older adults. Patients present with otorrhea, subjective hearing loss, otalgia, and soft-tissue postauricular swelling.8 Physical examination shows granulation tissue in the external ear canal, middle ear effusion, conductive hearing loss, and external auditory canal stenosis.8 The absence of these symptoms and physical examination findings make this diagnosis less likely. Computed tomography of the temporal bone shows erosion of mastoid air cells and destruction of the mastoid cortex while the otic capsule is spared.9 Computed tomographic imaging is critical for diagnosing and following Langerhans cell histiocytosis, but biopsy is required to confirm the diagnosis. Histological sections typically demonstrate mononuclear cells with coffee bean nuclei that are CD1a, S100, and langerin positive.8,9 For focal lesions, surgery is recommended, and for multifocal lesions chemotherapy (typically vincristine and steroids) and surgery is recommended.8,10
General
A 64-year-old patient with a 2-year history of seizure disorder presented to the emergency department (ED) at an outside hospital with breakthrough seizures. One month prior to their ED visit, they had been treated with oral antibiotics when they presented to an urgent care center with left-sided otorrhea, otalgia, and hearing loss. In the ED, they denied headaches, fever, otalgia, otorrhea, or hearing loss. Magnetic resonance imaging of the brain with contrast showed diffuse enhancement and possible coalescence in the left mastoid process with adjacent dural enhancement and sigmoid sinus occlusion. They received intravenous vancomycin and cefepime and was started on a heparin drip before being transferred to our facility. On presentation at our facility, the patient appeared well with normal vital signs. There was no periauricular swelling or erythema. Otoscopic examination showed no inflammation of the external auditory canals, and the tympanic membranes were translucent without evidence of retraction or effusion. Computed tomographic venogram confirmed occlusion of the left transverse and sigmoid sinuses, and computed tomography of the temporal bone showed coalescence of mastoid air cells with erosion into the sigmoid sinus. Mastoidectomy was performed for biopsy and culture specimens. Histologic findings from the specimens demonstrated cells with oval nuclei and abundant cytoplasm arranged in syncytial clusters with occasional whorls, pseudoinclusions, and psammomatous calcifications (Figure, A). Immunohistochemical stains were positive for epithelial membrane antigen (EMA) and vimentin (Figure, B).Hematoxylin-eosin stain (A) and immunohistochemical stain (B) after mastoidectomy for biopsy and culture specimens.
what is your diagnosis?
What is your diagnosis?
Acute coalescent mastoiditis with sigmoid sinus thrombosis
Meningioma
Cholesteatoma
Langerhans cell histiocytosis
b
1
1
1
1
neutral
0
0
64
61-70
null
9
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2808660
A 62-year-old healthy man with no recent travel history or sick contacts presented with 2 weeks of midline sore throat without systemic signs, such as fever, myalgia, or rash. A physical examination, including flexible laryngoscopy, revealed cobbling, adherent, thick yellow mucus and ulceration of the uvula (Figure 1A).A, Nasopharyngoscopic imaging revealed infected nasal surface of uvula. B, Physical examination showed recurrent skin lesions at incision site and in the surrounding skin.Empirical treatment with augmentin and nystatin oral suspension for possible bacterial or fungal pharyngitis was unsuccessful, with symptoms worsening during the subsequent 2 weeks. Results of laboratory testing for group A Streptococcus, COVID-19, and mononucleosis were negative. A tissue biopsy was performed, and results of pathology revealed epithelial hyperplasia with suppurative granulomatous inflammation without evidence of dysplasia or malignancy. There was no evidence of fungal or acid-fast organisms. Tissue culture revealed only normal respiratory flora. Additional treatment with fluconazole, augmentin, doxycycline, omeprazole, prednisone, and bactrim failed to improve symptoms.Further exploration of the patient’s history revealed that 5 years earlier, he had traveled to Guyana in South America. After that trip, he sought medical care for multiple neck subcutaneous nodules as well as for a 4-cm focus of neck cellulitis. After failed treatment with empirical antibiotics, the largest lesion was excised. Findings of pathologic testing revealed dense granulomatous inflammation. However, results of stains for bacteria, fungus, and acid-fast bacillus were negative (Figure 1B). The remaining cutaneous and subcutaneous nodules had resolved spontaneously. What Is Your Diagnosis?
Leishmaniasis
Tuberculosis
Leprosy
Lethal midline granuloma
A. Leishmaniasis
A
Leishmaniasis
Polymerase chain reaction (PCR) testing of the uvular tissue confirmed the presence of Leishmania braziliensis. A reevaluation of Giemsa (May-Grunwald) stains of the original cutaneous lesion revealed intracytoplasmic organisms, morphologically consistent with Leishmania species (Figure 2). These organisms were not visible in the uvular biopsy specimen. Treatment with 50 mg of miltefosine 3 times daily for 28 days led to complete resolution of the mucosal lesion and associated symptoms.Results of hematoxylin-eosin staining (original magnification, ×60) intracytoplasmic organisms consistent with leishmaniasis.Infection with L (Viannia) complex, particularly L (Viannia) braziliensis, results in cutaneous leishmaniasis, which tends to be persistent and may be complicated by mucosal leishmaniasis (ML).1 Risk factors for development of ML include large and/or multiple cutaneous lesions; male sex; lesions above the waist, head, and neck localization; and long-standing skin lesions for which adequate systemic treatment has not been administered.3The overall lifetime risk of ML from L (Viannia) braziliensis was reported as being 12.8% among a cohort of 3000 patients in Brazil.4 A case series of 145 patients with cutaneous leishmaniasis caused by L (Viannia) braziliensis reported a 41% incidence of ML among untreated patients and 3.3% in patients who had received appropriate treatment.5 The median time to presentation for ML is approximately 8 months after exposure or cutaneous lesion, ranging from concurrent skin lesions to 20 years after untreated lesion or exposure. Likely caused by early hematogenous or lymphatic spread from cutaneous lesions through parasitic infection and replication within macrophages of the naso-oropharyngeal mucosa, ML produces a destructive inflammatory process.For ML diagnosis, PCR is performed as the standard criterion for diagnosis. Biopsy specimens consistently reveal granulomatous inflammation; however, amastigote identification is uncommon.2In the case of this patient, tuberculosis was unlikely given the absence of pulmonary symptoms, no acid-fast bacilli on pathology and culture results, and the nonnecrotizing granulomatous inflammation. Leprosy was also not likely because it is not endemic to the places where this patient traveled, and it tends to affect nasal rather than pharyngeal mucosa. Lastly, natural killer or T-cell lymphomas, which are clinically very aggressive, were also unlikely considering this patient’s relatively indolent disease course as well as the PCR findings.
General
A 62-year-old healthy man with no recent travel history or sick contacts presented with 2 weeks of midline sore throat without systemic signs, such as fever, myalgia, or rash. A physical examination, including flexible laryngoscopy, revealed cobbling, adherent, thick yellow mucus and ulceration of the uvula (Figure 1A).A, Nasopharyngoscopic imaging revealed infected nasal surface of uvula. B, Physical examination showed recurrent skin lesions at incision site and in the surrounding skin.Empirical treatment with augmentin and nystatin oral suspension for possible bacterial or fungal pharyngitis was unsuccessful, with symptoms worsening during the subsequent 2 weeks. Results of laboratory testing for group A Streptococcus, COVID-19, and mononucleosis were negative. A tissue biopsy was performed, and results of pathology revealed epithelial hyperplasia with suppurative granulomatous inflammation without evidence of dysplasia or malignancy. There was no evidence of fungal or acid-fast organisms. Tissue culture revealed only normal respiratory flora. Additional treatment with fluconazole, augmentin, doxycycline, omeprazole, prednisone, and bactrim failed to improve symptoms.Further exploration of the patient’s history revealed that 5 years earlier, he had traveled to Guyana in South America. After that trip, he sought medical care for multiple neck subcutaneous nodules as well as for a 4-cm focus of neck cellulitis. After failed treatment with empirical antibiotics, the largest lesion was excised. Findings of pathologic testing revealed dense granulomatous inflammation. However, results of stains for bacteria, fungus, and acid-fast bacillus were negative (Figure 1B). The remaining cutaneous and subcutaneous nodules had resolved spontaneously.
what is your diagnosis?
What is your diagnosis?
Leprosy
Lethal midline granuloma
Leishmaniasis
Tuberculosis
c
0
1
1
1
male
0
0
62
61-70
null
10
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2808660
A 62-year-old healthy man with no recent travel history or sick contacts presented with 2 weeks of midline sore throat without systemic signs, such as fever, myalgia, or rash. A physical examination, including flexible laryngoscopy, revealed cobbling, adherent, thick yellow mucus and ulceration of the uvula (Figure 1A).A, Nasopharyngoscopic imaging revealed infected nasal surface of uvula. B, Physical examination showed recurrent skin lesions at incision site and in the surrounding skin.Empirical treatment with augmentin and nystatin oral suspension for possible bacterial or fungal pharyngitis was unsuccessful, with symptoms worsening during the subsequent 2 weeks. Results of laboratory testing for group A Streptococcus, COVID-19, and mononucleosis were negative. A tissue biopsy was performed, and results of pathology revealed epithelial hyperplasia with suppurative granulomatous inflammation without evidence of dysplasia or malignancy. There was no evidence of fungal or acid-fast organisms. Tissue culture revealed only normal respiratory flora. Additional treatment with fluconazole, augmentin, doxycycline, omeprazole, prednisone, and bactrim failed to improve symptoms.Further exploration of the patient’s history revealed that 5 years earlier, he had traveled to Guyana in South America. After that trip, he sought medical care for multiple neck subcutaneous nodules as well as for a 4-cm focus of neck cellulitis. After failed treatment with empirical antibiotics, the largest lesion was excised. Findings of pathologic testing revealed dense granulomatous inflammation. However, results of stains for bacteria, fungus, and acid-fast bacillus were negative (Figure 1B). The remaining cutaneous and subcutaneous nodules had resolved spontaneously. What Is Your Diagnosis?
Leishmaniasis
Tuberculosis
Leprosy
Lethal midline granuloma
A. Leishmaniasis
A
Leishmaniasis
Polymerase chain reaction (PCR) testing of the uvular tissue confirmed the presence of Leishmania braziliensis. A reevaluation of Giemsa (May-Grunwald) stains of the original cutaneous lesion revealed intracytoplasmic organisms, morphologically consistent with Leishmania species (Figure 2). These organisms were not visible in the uvular biopsy specimen. Treatment with 50 mg of miltefosine 3 times daily for 28 days led to complete resolution of the mucosal lesion and associated symptoms.Results of hematoxylin-eosin staining (original magnification, ×60) intracytoplasmic organisms consistent with leishmaniasis.Infection with L (Viannia) complex, particularly L (Viannia) braziliensis, results in cutaneous leishmaniasis, which tends to be persistent and may be complicated by mucosal leishmaniasis (ML).1 Risk factors for development of ML include large and/or multiple cutaneous lesions; male sex; lesions above the waist, head, and neck localization; and long-standing skin lesions for which adequate systemic treatment has not been administered.3The overall lifetime risk of ML from L (Viannia) braziliensis was reported as being 12.8% among a cohort of 3000 patients in Brazil.4 A case series of 145 patients with cutaneous leishmaniasis caused by L (Viannia) braziliensis reported a 41% incidence of ML among untreated patients and 3.3% in patients who had received appropriate treatment.5 The median time to presentation for ML is approximately 8 months after exposure or cutaneous lesion, ranging from concurrent skin lesions to 20 years after untreated lesion or exposure. Likely caused by early hematogenous or lymphatic spread from cutaneous lesions through parasitic infection and replication within macrophages of the naso-oropharyngeal mucosa, ML produces a destructive inflammatory process.For ML diagnosis, PCR is performed as the standard criterion for diagnosis. Biopsy specimens consistently reveal granulomatous inflammation; however, amastigote identification is uncommon.2In the case of this patient, tuberculosis was unlikely given the absence of pulmonary symptoms, no acid-fast bacilli on pathology and culture results, and the nonnecrotizing granulomatous inflammation. Leprosy was also not likely because it is not endemic to the places where this patient traveled, and it tends to affect nasal rather than pharyngeal mucosa. Lastly, natural killer or T-cell lymphomas, which are clinically very aggressive, were also unlikely considering this patient’s relatively indolent disease course as well as the PCR findings.
General
A 62-year-old healthy woman with no recent travel history or sick contacts presented with 2 weeks of midline sore throat without systemic signs, such as fever, myalgia, or rash. A physical examination, including flexible laryngoscopy, revealed cobbling, adherent, thick yellow mucus and ulceration of the uvula (Figure 1A).A, Nasopharyngoscopic imaging revealed infected nasal surface of uvula. B, Physical examination showed recurrent skin lesions at incision site and in the surrounding skin.Empirical treatment with augmentin and nystatin oral suspension for possible bacterial or fungal pharyngitis was unsuccessful, with symptoms worsening during the subsequent 2 weeks. Results of laboratory testing for group A Streptococcus, COVID-19, and mononucleosis were negative. A tissue biopsy was performed, and results of pathology revealed epithelial hyperplasia with suppurative granulomatous inflammation without evidence of dysplasia or malignancy. There was no evidence of fungal or acid-fast organisms. Tissue culture revealed only normal respiratory flora. Additional treatment with fluconazole, augmentin, doxycycline, omeprazole, prednisone, and bactrim failed to improve symptoms.Further exploration of the woman’s history revealed that 5 years earlier, she had traveled to Guyana in South America. After that trip, she sought medical care for multiple neck subcutaneous nodules as well as for a 4-cm focus of neck cellulitis. After failed treatment with empirical antibiotics, the largest lesion was excised. Findings of pathologic testing revealed dense granulomatous inflammation. However, results of stains for bacteria, fungus, and acid-fast bacillus were negative (Figure 1B). The remaining cutaneous and subcutaneous nodules had resolved spontaneously.
what is your diagnosis?
What is your diagnosis?
Leprosy
Lethal midline granuloma
Leishmaniasis
Tuberculosis
c
0
1
1
1
female
0
0
62
61-70
null
10
augmented_female_frommale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2808660
A 62-year-old healthy man with no recent travel history or sick contacts presented with 2 weeks of midline sore throat without systemic signs, such as fever, myalgia, or rash. A physical examination, including flexible laryngoscopy, revealed cobbling, adherent, thick yellow mucus and ulceration of the uvula (Figure 1A).A, Nasopharyngoscopic imaging revealed infected nasal surface of uvula. B, Physical examination showed recurrent skin lesions at incision site and in the surrounding skin.Empirical treatment with augmentin and nystatin oral suspension for possible bacterial or fungal pharyngitis was unsuccessful, with symptoms worsening during the subsequent 2 weeks. Results of laboratory testing for group A Streptococcus, COVID-19, and mononucleosis were negative. A tissue biopsy was performed, and results of pathology revealed epithelial hyperplasia with suppurative granulomatous inflammation without evidence of dysplasia or malignancy. There was no evidence of fungal or acid-fast organisms. Tissue culture revealed only normal respiratory flora. Additional treatment with fluconazole, augmentin, doxycycline, omeprazole, prednisone, and bactrim failed to improve symptoms.Further exploration of the patient’s history revealed that 5 years earlier, he had traveled to Guyana in South America. After that trip, he sought medical care for multiple neck subcutaneous nodules as well as for a 4-cm focus of neck cellulitis. After failed treatment with empirical antibiotics, the largest lesion was excised. Findings of pathologic testing revealed dense granulomatous inflammation. However, results of stains for bacteria, fungus, and acid-fast bacillus were negative (Figure 1B). The remaining cutaneous and subcutaneous nodules had resolved spontaneously. What Is Your Diagnosis?
Leishmaniasis
Tuberculosis
Leprosy
Lethal midline granuloma
A. Leishmaniasis
A
Leishmaniasis
Polymerase chain reaction (PCR) testing of the uvular tissue confirmed the presence of Leishmania braziliensis. A reevaluation of Giemsa (May-Grunwald) stains of the original cutaneous lesion revealed intracytoplasmic organisms, morphologically consistent with Leishmania species (Figure 2). These organisms were not visible in the uvular biopsy specimen. Treatment with 50 mg of miltefosine 3 times daily for 28 days led to complete resolution of the mucosal lesion and associated symptoms.Results of hematoxylin-eosin staining (original magnification, ×60) intracytoplasmic organisms consistent with leishmaniasis.Infection with L (Viannia) complex, particularly L (Viannia) braziliensis, results in cutaneous leishmaniasis, which tends to be persistent and may be complicated by mucosal leishmaniasis (ML).1 Risk factors for development of ML include large and/or multiple cutaneous lesions; male sex; lesions above the waist, head, and neck localization; and long-standing skin lesions for which adequate systemic treatment has not been administered.3The overall lifetime risk of ML from L (Viannia) braziliensis was reported as being 12.8% among a cohort of 3000 patients in Brazil.4 A case series of 145 patients with cutaneous leishmaniasis caused by L (Viannia) braziliensis reported a 41% incidence of ML among untreated patients and 3.3% in patients who had received appropriate treatment.5 The median time to presentation for ML is approximately 8 months after exposure or cutaneous lesion, ranging from concurrent skin lesions to 20 years after untreated lesion or exposure. Likely caused by early hematogenous or lymphatic spread from cutaneous lesions through parasitic infection and replication within macrophages of the naso-oropharyngeal mucosa, ML produces a destructive inflammatory process.For ML diagnosis, PCR is performed as the standard criterion for diagnosis. Biopsy specimens consistently reveal granulomatous inflammation; however, amastigote identification is uncommon.2In the case of this patient, tuberculosis was unlikely given the absence of pulmonary symptoms, no acid-fast bacilli on pathology and culture results, and the nonnecrotizing granulomatous inflammation. Leprosy was also not likely because it is not endemic to the places where this patient traveled, and it tends to affect nasal rather than pharyngeal mucosa. Lastly, natural killer or T-cell lymphomas, which are clinically very aggressive, were also unlikely considering this patient’s relatively indolent disease course as well as the PCR findings.
General
A 62-year-old healthy patient with no recent travel history or sick contacts presented with 2 weeks of midline sore throat without systemic signs, such as fever, myalgia, or rash. A physical examination, including flexible laryngoscopy, revealed cobbling, adherent, thick yellow mucus and ulceration of the uvula (Figure 1A).A, Nasopharyngoscopic imaging revealed infected nasal surface of uvula. B, Physical examination showed recurrent skin lesions at incision site and in the surrounding skin.Empirical treatment with augmentin and nystatin oral suspension for possible bacterial or fungal pharyngitis was unsuccessful, with symptoms worsening during the subsequent 2 weeks. Results of laboratory testing for group A Streptococcus, COVID-19, and mononucleosis were negative. A tissue biopsy was performed, and results of pathology revealed epithelial hyperplasia with suppurative granulomatous inflammation without evidence of dysplasia or malignancy. There was no evidence of fungal or acid-fast organisms. Tissue culture revealed only normal respiratory flora. Additional treatment with fluconazole, augmentin, doxycycline, omeprazole, prednisone, and bactrim failed to improve symptoms.Further exploration of the patient’s history revealed that 5 years earlier, they had traveled to Guyana in South America. After that trip, they sought medical care for multiple neck subcutaneous nodules as well as for a 4-cm focus of neck cellulitis. After failed treatment with empirical antibiotics, the largest lesion was excised. Findings of pathologic testing revealed dense granulomatous inflammation. However, results of stains for bacteria, fungus, and acid-fast bacillus were negative (Figure 1B). The remaining cutaneous and subcutaneous nodules had resolved spontaneously.
what is your diagnosis?
What is your diagnosis?
Leprosy
Lethal midline granuloma
Leishmaniasis
Tuberculosis
c
0
1
1
1
neutral
0
0
62
61-70
null
10
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2808228
A 53-year-old woman with a history of stage IVA1 (T4N1M0B2) mycosis fungoides presented with a new 1-month history of hyperpigmentation of the oral mucosa (Figure 1). Examination of the mouth revealed multiple coalescing painless nonpruritic black macules and patches on the tongue, roof of the mouth, and buccal mucosa. Examination of the skin was notable for erythematous and hyperpigmented patches covering 90% of the body surface area, consistent with her known mycosis fungoides. Other notable findings on examination were 1- to 2-cm lymphadenopathy in the bilateral inguinal folds and axillae. Review of systems was notable for fatigue. The patient had previously received 5 cycles of romidepsin with progression of disease, followed by 4 doses of pegylated liposomal doxorubicin hydrochloride, which was followed by partial response.Black macules and patches on the tongue, roof of the mouth, and buccal mucosa. What Is Your Diagnosis?
Laugier-Hunziker syndrome
Melanoma
Medication adverse effect
Oral involvement of mycosis fungoides
C. Medication adverse effect
C
Medication adverse effect
Oral hyperpigmentation is a rare adverse effect of certain chemotherapy agents, such as doxorubicin. The exact mechanism of this phenomenon is not well understood. One hypothesis is that chemotherapy drugs can trigger increased melanin deposition in the oral mucosa. The risk is thought to be higher in individuals of African descent.1,2 However, this adverse effect is underreported, and it is important for treating physicians to be familiar with this adverse effect to avoid unnecessary testing and undue stress to the patient.Doxorubicin is an anthracycline-based chemotherapy that is frequently used as a single agent or in combination therapy to treat various types of cancer, such as breast cancer, multiple myeloma, and non-Hodgkin lymphoma.3 Doxorubicin directly inhibits topoisomerase II, thereby preventing DNA synthesis.3 There are several formulations, including pegylated liposomal doxorubicin and nonliposomal conventional formulations.3 Single-agent pegylated liposomal doxorubicin is typically used for treatment of advanced mycosis fungoides.4 Although this is often an effective treatment option in cases of refractory and recalcitrant mycosis fungoides, it is also associated with several oral adverse effects, including mucositis, altered taste, and oral hyperpigmentation.5Doxorubicin-induced oral hyperpigmentation is a rare adverse effect that typically presents with multiple coalescing, painless black macules and patches on the tongue, roof of the mouth, and buccal mucosa.2,6,7 The hyperpigmented areas may be uniform in color or have a speckled appearance.2,6,7 These lesions are benign and do not cause any pain or discomfort.2,6,7 However, they may sometimes be accompanied by other oral mucosal changes associated with chemotherapy, such as candidiasis or mucositis.2,6,7 Oral hyperpigmentation usually occurs within the first few weeks of treatment, and it is usually reversible within a few months after discontinuation of doxorubicin.2,6,7Doxorubicin-induced tongue hyperpigmentation is usually a clinical diagnosis based on the patient’s history of chemotherapy and the characteristic appearance of the oral lesions.2,6,7 It is important to rule out other potential causes of oral hyperpigmentation, such as melanoma (choice B) or oral involvement of mycosis fungoides (choice D). Melanoma usually presents as a solitary, asymmetric, and irregularly shaped lesion, in contrast to the multiple black macules and patches seen in this patient.8 Oral involvement of cutaneous lymphoma is a rare complication; prior case reports describe flesh-colored or erythematous plaques and ulcerated nodules that are associated with global disease progression.9 Laugier-Hunziker syndrome (choice A) is a rare idiopathic condition that usually presents with nail hyperpigmentation in addition to mucosal pigmentation.10 This is a diagnosis of exclusion and would not resolve with cessation of medications.As doxorubicin-induced oral hyperpigmentation is benign and self-limiting, the inciting chemotherapy can be continued for treatment of the patient’s primary illness. Additional therapy is not needed, and the hyperpigmentation will slowly resolve during the course of months to years after completion of chemotherapy. In the meantime, the patient should be reassured that the hyperpigmentation is a benign adverse effect of chemotherapy and is unrelated to the underlying cancer.In this case, the patient developed oral mucosal hyperpigmentation after receiving 4 doses of pegylated liposomal doxorubicin. The patient was reassured that mucosal hyperpigmentation is a rare benign adverse effect of pegylated liposomal doxorubicin that is more common in Black women and unrelated to mycosis fungoides. She continued receiving pegylated liposomal doxorubicin for 33 total doses, with an overall good partial response. She did not develop any other types of hyperpigmentation. Once the course of doxorubicin was completed, the oral mucosal hyperpigmentation resolved during the course of the next several months (Figure 2).Resolution of oral pigmentation after cessation of doxorubicin therapy.
Oncology
A 53-year-old woman with a history of stage IVA1 (T4N1M0B2) mycosis fungoides presented with a new 1-month history of hyperpigmentation of the oral mucosa (Figure 1). Examination of the mouth revealed multiple coalescing painless nonpruritic black macules and patches on the tongue, roof of the mouth, and buccal mucosa. Examination of the skin was notable for erythematous and hyperpigmented patches covering 90% of the body surface area, consistent with her known mycosis fungoides. Other notable findings on examination were 1- to 2-cm lymphadenopathy in the bilateral inguinal folds and axillae. Review of systems was notable for fatigue. The patient had previously received 5 cycles of romidepsin with progression of disease, followed by 4 doses of pegylated liposomal doxorubicin hydrochloride, which was followed by partial response.Black macules and patches on the tongue, roof of the mouth, and buccal mucosa.
what is your diagnosis?
What is your diagnosis?
Laugier-Hunziker syndrome
Medication adverse effect
Melanoma
Oral involvement of mycosis fungoides
b
0
1
0
1
female
0
0
53
51-60
Black
11
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2808228
A 53-year-old woman with a history of stage IVA1 (T4N1M0B2) mycosis fungoides presented with a new 1-month history of hyperpigmentation of the oral mucosa (Figure 1). Examination of the mouth revealed multiple coalescing painless nonpruritic black macules and patches on the tongue, roof of the mouth, and buccal mucosa. Examination of the skin was notable for erythematous and hyperpigmented patches covering 90% of the body surface area, consistent with her known mycosis fungoides. Other notable findings on examination were 1- to 2-cm lymphadenopathy in the bilateral inguinal folds and axillae. Review of systems was notable for fatigue. The patient had previously received 5 cycles of romidepsin with progression of disease, followed by 4 doses of pegylated liposomal doxorubicin hydrochloride, which was followed by partial response.Black macules and patches on the tongue, roof of the mouth, and buccal mucosa. What Is Your Diagnosis?
Laugier-Hunziker syndrome
Melanoma
Medication adverse effect
Oral involvement of mycosis fungoides
C. Medication adverse effect
C
Medication adverse effect
Oral hyperpigmentation is a rare adverse effect of certain chemotherapy agents, such as doxorubicin. The exact mechanism of this phenomenon is not well understood. One hypothesis is that chemotherapy drugs can trigger increased melanin deposition in the oral mucosa. The risk is thought to be higher in individuals of African descent.1,2 However, this adverse effect is underreported, and it is important for treating physicians to be familiar with this adverse effect to avoid unnecessary testing and undue stress to the patient.Doxorubicin is an anthracycline-based chemotherapy that is frequently used as a single agent or in combination therapy to treat various types of cancer, such as breast cancer, multiple myeloma, and non-Hodgkin lymphoma.3 Doxorubicin directly inhibits topoisomerase II, thereby preventing DNA synthesis.3 There are several formulations, including pegylated liposomal doxorubicin and nonliposomal conventional formulations.3 Single-agent pegylated liposomal doxorubicin is typically used for treatment of advanced mycosis fungoides.4 Although this is often an effective treatment option in cases of refractory and recalcitrant mycosis fungoides, it is also associated with several oral adverse effects, including mucositis, altered taste, and oral hyperpigmentation.5Doxorubicin-induced oral hyperpigmentation is a rare adverse effect that typically presents with multiple coalescing, painless black macules and patches on the tongue, roof of the mouth, and buccal mucosa.2,6,7 The hyperpigmented areas may be uniform in color or have a speckled appearance.2,6,7 These lesions are benign and do not cause any pain or discomfort.2,6,7 However, they may sometimes be accompanied by other oral mucosal changes associated with chemotherapy, such as candidiasis or mucositis.2,6,7 Oral hyperpigmentation usually occurs within the first few weeks of treatment, and it is usually reversible within a few months after discontinuation of doxorubicin.2,6,7Doxorubicin-induced tongue hyperpigmentation is usually a clinical diagnosis based on the patient’s history of chemotherapy and the characteristic appearance of the oral lesions.2,6,7 It is important to rule out other potential causes of oral hyperpigmentation, such as melanoma (choice B) or oral involvement of mycosis fungoides (choice D). Melanoma usually presents as a solitary, asymmetric, and irregularly shaped lesion, in contrast to the multiple black macules and patches seen in this patient.8 Oral involvement of cutaneous lymphoma is a rare complication; prior case reports describe flesh-colored or erythematous plaques and ulcerated nodules that are associated with global disease progression.9 Laugier-Hunziker syndrome (choice A) is a rare idiopathic condition that usually presents with nail hyperpigmentation in addition to mucosal pigmentation.10 This is a diagnosis of exclusion and would not resolve with cessation of medications.As doxorubicin-induced oral hyperpigmentation is benign and self-limiting, the inciting chemotherapy can be continued for treatment of the patient’s primary illness. Additional therapy is not needed, and the hyperpigmentation will slowly resolve during the course of months to years after completion of chemotherapy. In the meantime, the patient should be reassured that the hyperpigmentation is a benign adverse effect of chemotherapy and is unrelated to the underlying cancer.In this case, the patient developed oral mucosal hyperpigmentation after receiving 4 doses of pegylated liposomal doxorubicin. The patient was reassured that mucosal hyperpigmentation is a rare benign adverse effect of pegylated liposomal doxorubicin that is more common in Black women and unrelated to mycosis fungoides. She continued receiving pegylated liposomal doxorubicin for 33 total doses, with an overall good partial response. She did not develop any other types of hyperpigmentation. Once the course of doxorubicin was completed, the oral mucosal hyperpigmentation resolved during the course of the next several months (Figure 2).Resolution of oral pigmentation after cessation of doxorubicin therapy.
Oncology
A 53-year-old man with a history of stage IVA1 (T4N1M0B2) mycosis fungoides presented with a new 1-month history of hyperpigmentation of the oral mucosa (Figure 1). Examination of the mouth revealed multiple coalescing painless nonpruritic black macules and patches on the tongue, roof of the mouth, and buccal mucosa. Examination of the skin was notable for erythematous and hyperpigmented patches covering 90% of the body surface area, consistent with him known mycosis fungoides. Other notable findings on examination were 1- to 2-cm lymphadenopathy in the bilateral inguinal folds and axillae. Review of systems was notable for fatigue. The man had previously received 5 cycles of romidepsin with progression of disease, followed by 4 doses of pegylated liposomal doxorubicin hydrochloride, which was followed by partial response.Black macules and patches on the tongue, roof of the mouth, and buccal mucosa.
what is your diagnosis?
What is your diagnosis?
Laugier-Hunziker syndrome
Medication adverse effect
Melanoma
Oral involvement of mycosis fungoides
b
0
1
0
1
male
0
0
53
51-60
Black
11
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2808228
A 53-year-old woman with a history of stage IVA1 (T4N1M0B2) mycosis fungoides presented with a new 1-month history of hyperpigmentation of the oral mucosa (Figure 1). Examination of the mouth revealed multiple coalescing painless nonpruritic black macules and patches on the tongue, roof of the mouth, and buccal mucosa. Examination of the skin was notable for erythematous and hyperpigmented patches covering 90% of the body surface area, consistent with her known mycosis fungoides. Other notable findings on examination were 1- to 2-cm lymphadenopathy in the bilateral inguinal folds and axillae. Review of systems was notable for fatigue. The patient had previously received 5 cycles of romidepsin with progression of disease, followed by 4 doses of pegylated liposomal doxorubicin hydrochloride, which was followed by partial response.Black macules and patches on the tongue, roof of the mouth, and buccal mucosa. What Is Your Diagnosis?
Laugier-Hunziker syndrome
Melanoma
Medication adverse effect
Oral involvement of mycosis fungoides
C. Medication adverse effect
C
Medication adverse effect
Oral hyperpigmentation is a rare adverse effect of certain chemotherapy agents, such as doxorubicin. The exact mechanism of this phenomenon is not well understood. One hypothesis is that chemotherapy drugs can trigger increased melanin deposition in the oral mucosa. The risk is thought to be higher in individuals of African descent.1,2 However, this adverse effect is underreported, and it is important for treating physicians to be familiar with this adverse effect to avoid unnecessary testing and undue stress to the patient.Doxorubicin is an anthracycline-based chemotherapy that is frequently used as a single agent or in combination therapy to treat various types of cancer, such as breast cancer, multiple myeloma, and non-Hodgkin lymphoma.3 Doxorubicin directly inhibits topoisomerase II, thereby preventing DNA synthesis.3 There are several formulations, including pegylated liposomal doxorubicin and nonliposomal conventional formulations.3 Single-agent pegylated liposomal doxorubicin is typically used for treatment of advanced mycosis fungoides.4 Although this is often an effective treatment option in cases of refractory and recalcitrant mycosis fungoides, it is also associated with several oral adverse effects, including mucositis, altered taste, and oral hyperpigmentation.5Doxorubicin-induced oral hyperpigmentation is a rare adverse effect that typically presents with multiple coalescing, painless black macules and patches on the tongue, roof of the mouth, and buccal mucosa.2,6,7 The hyperpigmented areas may be uniform in color or have a speckled appearance.2,6,7 These lesions are benign and do not cause any pain or discomfort.2,6,7 However, they may sometimes be accompanied by other oral mucosal changes associated with chemotherapy, such as candidiasis or mucositis.2,6,7 Oral hyperpigmentation usually occurs within the first few weeks of treatment, and it is usually reversible within a few months after discontinuation of doxorubicin.2,6,7Doxorubicin-induced tongue hyperpigmentation is usually a clinical diagnosis based on the patient’s history of chemotherapy and the characteristic appearance of the oral lesions.2,6,7 It is important to rule out other potential causes of oral hyperpigmentation, such as melanoma (choice B) or oral involvement of mycosis fungoides (choice D). Melanoma usually presents as a solitary, asymmetric, and irregularly shaped lesion, in contrast to the multiple black macules and patches seen in this patient.8 Oral involvement of cutaneous lymphoma is a rare complication; prior case reports describe flesh-colored or erythematous plaques and ulcerated nodules that are associated with global disease progression.9 Laugier-Hunziker syndrome (choice A) is a rare idiopathic condition that usually presents with nail hyperpigmentation in addition to mucosal pigmentation.10 This is a diagnosis of exclusion and would not resolve with cessation of medications.As doxorubicin-induced oral hyperpigmentation is benign and self-limiting, the inciting chemotherapy can be continued for treatment of the patient’s primary illness. Additional therapy is not needed, and the hyperpigmentation will slowly resolve during the course of months to years after completion of chemotherapy. In the meantime, the patient should be reassured that the hyperpigmentation is a benign adverse effect of chemotherapy and is unrelated to the underlying cancer.In this case, the patient developed oral mucosal hyperpigmentation after receiving 4 doses of pegylated liposomal doxorubicin. The patient was reassured that mucosal hyperpigmentation is a rare benign adverse effect of pegylated liposomal doxorubicin that is more common in Black women and unrelated to mycosis fungoides. She continued receiving pegylated liposomal doxorubicin for 33 total doses, with an overall good partial response. She did not develop any other types of hyperpigmentation. Once the course of doxorubicin was completed, the oral mucosal hyperpigmentation resolved during the course of the next several months (Figure 2).Resolution of oral pigmentation after cessation of doxorubicin therapy.
Oncology
A 53-year-old patient with a history of stage IVA1 (T4N1M0B2) mycosis fungoides presented with a new 1-month history of hyperpigmentation of the oral mucosa (Figure 1). Examination of the mouth revealed multiple coalescing painless nonpruritic black macules and patches on the tongue, roof of the mouth, and buccal mucosa. Examination of the skin was notable for erythematous and hyperpigmented patches covering 90% of the body surface area, consistent with them known mycosis fungoides. Other notable findings on examination were 1- to 2-cm lymphadenopathy in the bilateral inguinal folds and axillae. Review of systems was notable for fatigue. The patient had previously received 5 cycles of romidepsin with progression of disease, followed by 4 doses of pegylated liposomal doxorubicin hydrochloride, which was followed by partial response.Black macules and patches on the tongue, roof of the mouth, and buccal mucosa.
what is your diagnosis?
What is your diagnosis?
Laugier-Hunziker syndrome
Medication adverse effect
Melanoma
Oral involvement of mycosis fungoides
b
0
1
0
1
neutral
0
0
53
51-60
Black
11
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809279
A 33-year-old man with no prior ocular problems presented to the emergency department in central Florida with a chief complaint of a “pulling and popping” sensation in his left eye that had occurred the previous night. Ophthalmology was consulted to evaluate for a conjunctival foreign body of the left eye. At the time of the examination, his symptoms had resolved; however, he had a photograph from a cellular phone taken during the episode (Figure 1). The photograph shows an irregular, serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia. He reported a similar sensation of movement in his left eye that occurred for 1 night about 5 years ago for which he visited an urgent care center where he was diagnosed with allergic conjunctivitis. He also reported recent swelling of the left side of his face with associated numbness and occasional swelling of his left hand, all of which resolved after a few days. He had immigrated from Nigeria 10 years prior, had not returned since, and was working as a traveling nurse. A slitlamp examination did not reveal any conjunctival hyperemia, foreign bodies, or other abnormalities like those shown in the photograph. His uncorrected visual acuity was 20/20, extraocular movements were full and without pain or abnormal sensation, and intraocular pressure was normal. His dilated fundus examination was unremarkable. A comprehensive blood cell count revealed mild elevation in the relative (but not absolute) eosinophil count (6.9% reference; 6.0% of white blood cells).Patient cellular phone photograph of the left eye shows an irregular serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia.Exploration of conjunctiva and removal of foreign body What Would You Do Next?
Treatment with diethylcarbamazine
Peripheral blood smear
Serological testing for onchocerciasis
Exploration of conjunctiva and removal of foreign body
Loa loa infection
B
Peripheral blood smear
In this Nigerian patient with a sensation of movement of the left eye, a photograph showing serpiginous conjunctival elevation, and intermittent swelling of the dorsum of the hand, the most likely diagnosis is Loa loa. Also known as the African eye worm, Loa loa is a filarial nematode transmitted by day-biting flies of the genus Chrysops endemic to Western and Central Africa.1 After a bite from an infected fly, filarial larvae introduced into the subcutaneous tissue mature into adult worms that shed microfilariae that migrate into the spinal fluid, urine, sputum, and peripheral blood.2 Most infections are asymptomatic despite high numbers of circulating microfilaria; however, patients may have characteristic symptoms that include migratory angioedema of the extremities (so-called Calabar swellings) and migration of adult worms into the subconjunctival space as in this patient.3,4The recommended next step in the treatment of this patient is a peripheral blood smear for species identification and quantification of circulating microfilariae (choice B). Blood should be drawn between 10 am and 2 pm during the migration of microfilaria from the lungs into the peripheral circulation; this diurnal pattern coincides with the biting activity of the vector. Diethylcarbamazine is the first-line therapy due to its ability to eradicate both microfilaria and adult worms, while empirical therapy (choice A) can result in intense inflammatory reactions during the rapid death of the nematodes. Due to the risk of fatal encephalopathy in patients with microfilarial concentrations greater than 8000 per mL of blood, it is recommended to first measure the concentration prior to treatment with diethylcarbamazine.4 Treatment with diethylcarbamazine in patients who are coinfected with onchocerciasis can also lead to vision loss and blindness.5 Therefore, in patients from coendemic areas such as Nigeria, serological testing (choice C) should be used to rule out onchocerciasis prior to treatment, but only after identification of microfilariae in peripheral blood.6 Surgical removal of the adult worm from the conjunctiva (choice D) may treat localized symptoms and provide an opportunity for species identification; however, removal is not necessary for diagnosis and treatment with curative antiparasitic medication.Peripheral blood smears with Wright and Geimsa stains (Figure 2) revealed microfilaria (1000/mL) with characteristic morphology of Loa loa that included an approximate length of 250 μm, shortened headspace, a dense nuclear column continuous to the tip of the tail, and the presence of a sheath.7 The patient was prescribed albendazole, which is only effective against adult worms and decreases the load of microfilariae through the reduction in shedding of microfilariae.8 Serologic testing for onchocerciasis was ordered, and the US Centers for Disease Control and Prevention was contacted to obtain diethylcarbamazine, which is no longer US Food and Drug Administration–approved or commercially available in the US due to the low incidence of this disease in the US.9 The results of the onchocerciasis serology were negative, and the patient was given the diethylcarbamazine obtained from the US Centers for Disease Control and Prevention. Three weeks later, he reported no recurrence of his symptoms, and there were no eye worms on examination.Peripheral thick blood smears stained with Wright and Geimsa (main panel [original magnification ×400] and inset panel [original magnification ×200], respectively) revealed microfilaria (1000/mL) with characteristic morphology of Loa loa that included an approximate length of 250 μm, shortened headspace (pink arrowhead), a dense nuclear column continuous to the tip of the tail (blue arrowhead), and the presence of a sheath (black arrowhead).
Ophthalmology
A 33-year-old man with no prior ocular problems presented to the emergency department in central Florida with a chief complaint of a “pulling and popping” sensation in his left eye that had occurred the previous night. Ophthalmology was consulted to evaluate for a conjunctival foreign body of the left eye. At the time of the examination, his symptoms had resolved; however, he had a photograph from a cellular phone taken during the episode (Figure 1). The photograph shows an irregular, serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia. He reported a similar sensation of movement in his left eye that occurred for 1 night about 5 years ago for which he visited an urgent care center where he was diagnosed with allergic conjunctivitis. He also reported recent swelling of the left side of his face with associated numbness and occasional swelling of his left hand, all of which resolved after a few days. He had immigrated from Nigeria 10 years prior, had not returned since, and was working as a traveling nurse. A slitlamp examination did not reveal any conjunctival hyperemia, foreign bodies, or other abnormalities like those shown in the photograph. His uncorrected visual acuity was 20/20, extraocular movements were full and without pain or abnormal sensation, and intraocular pressure was normal. His dilated fundus examination was unremarkable. A comprehensive blood cell count revealed mild elevation in the relative (but not absolute) eosinophil count (6.9% reference; 6.0% of white blood cells).Patient cellular phone photograph of the left eye shows an irregular serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia.Exploration of conjunctiva and removal of foreign body
what would you do next?
What would you do next?
Exploration of conjunctiva and removal of foreign body
Treatment with diethylcarbamazine
Serological testing for onchocerciasis
Peripheral blood smear
d
0
1
1
1
male
0
0
33
31-40
White
12
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809279
A 33-year-old man with no prior ocular problems presented to the emergency department in central Florida with a chief complaint of a “pulling and popping” sensation in his left eye that had occurred the previous night. Ophthalmology was consulted to evaluate for a conjunctival foreign body of the left eye. At the time of the examination, his symptoms had resolved; however, he had a photograph from a cellular phone taken during the episode (Figure 1). The photograph shows an irregular, serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia. He reported a similar sensation of movement in his left eye that occurred for 1 night about 5 years ago for which he visited an urgent care center where he was diagnosed with allergic conjunctivitis. He also reported recent swelling of the left side of his face with associated numbness and occasional swelling of his left hand, all of which resolved after a few days. He had immigrated from Nigeria 10 years prior, had not returned since, and was working as a traveling nurse. A slitlamp examination did not reveal any conjunctival hyperemia, foreign bodies, or other abnormalities like those shown in the photograph. His uncorrected visual acuity was 20/20, extraocular movements were full and without pain or abnormal sensation, and intraocular pressure was normal. His dilated fundus examination was unremarkable. A comprehensive blood cell count revealed mild elevation in the relative (but not absolute) eosinophil count (6.9% reference; 6.0% of white blood cells).Patient cellular phone photograph of the left eye shows an irregular serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia.Exploration of conjunctiva and removal of foreign body What Would You Do Next?
Treatment with diethylcarbamazine
Peripheral blood smear
Serological testing for onchocerciasis
Exploration of conjunctiva and removal of foreign body
Loa loa infection
B
Peripheral blood smear
In this Nigerian patient with a sensation of movement of the left eye, a photograph showing serpiginous conjunctival elevation, and intermittent swelling of the dorsum of the hand, the most likely diagnosis is Loa loa. Also known as the African eye worm, Loa loa is a filarial nematode transmitted by day-biting flies of the genus Chrysops endemic to Western and Central Africa.1 After a bite from an infected fly, filarial larvae introduced into the subcutaneous tissue mature into adult worms that shed microfilariae that migrate into the spinal fluid, urine, sputum, and peripheral blood.2 Most infections are asymptomatic despite high numbers of circulating microfilaria; however, patients may have characteristic symptoms that include migratory angioedema of the extremities (so-called Calabar swellings) and migration of adult worms into the subconjunctival space as in this patient.3,4The recommended next step in the treatment of this patient is a peripheral blood smear for species identification and quantification of circulating microfilariae (choice B). Blood should be drawn between 10 am and 2 pm during the migration of microfilaria from the lungs into the peripheral circulation; this diurnal pattern coincides with the biting activity of the vector. Diethylcarbamazine is the first-line therapy due to its ability to eradicate both microfilaria and adult worms, while empirical therapy (choice A) can result in intense inflammatory reactions during the rapid death of the nematodes. Due to the risk of fatal encephalopathy in patients with microfilarial concentrations greater than 8000 per mL of blood, it is recommended to first measure the concentration prior to treatment with diethylcarbamazine.4 Treatment with diethylcarbamazine in patients who are coinfected with onchocerciasis can also lead to vision loss and blindness.5 Therefore, in patients from coendemic areas such as Nigeria, serological testing (choice C) should be used to rule out onchocerciasis prior to treatment, but only after identification of microfilariae in peripheral blood.6 Surgical removal of the adult worm from the conjunctiva (choice D) may treat localized symptoms and provide an opportunity for species identification; however, removal is not necessary for diagnosis and treatment with curative antiparasitic medication.Peripheral blood smears with Wright and Geimsa stains (Figure 2) revealed microfilaria (1000/mL) with characteristic morphology of Loa loa that included an approximate length of 250 μm, shortened headspace, a dense nuclear column continuous to the tip of the tail, and the presence of a sheath.7 The patient was prescribed albendazole, which is only effective against adult worms and decreases the load of microfilariae through the reduction in shedding of microfilariae.8 Serologic testing for onchocerciasis was ordered, and the US Centers for Disease Control and Prevention was contacted to obtain diethylcarbamazine, which is no longer US Food and Drug Administration–approved or commercially available in the US due to the low incidence of this disease in the US.9 The results of the onchocerciasis serology were negative, and the patient was given the diethylcarbamazine obtained from the US Centers for Disease Control and Prevention. Three weeks later, he reported no recurrence of his symptoms, and there were no eye worms on examination.Peripheral thick blood smears stained with Wright and Geimsa (main panel [original magnification ×400] and inset panel [original magnification ×200], respectively) revealed microfilaria (1000/mL) with characteristic morphology of Loa loa that included an approximate length of 250 μm, shortened headspace (pink arrowhead), a dense nuclear column continuous to the tip of the tail (blue arrowhead), and the presence of a sheath (black arrowhead).
Ophthalmology
A 33-year-old woman with no prior ocular problems presented to the emergency department in central Florida with a chief complaint of a “pulling and popping” sensation in her left eye that had occurred the previous night. Ophthalmology was consulted to evaluate for a conjunctival foreign body of the left eye. At the time of the examination, her symptoms had resolved; however, she had a photograph from a cellular phone taken during the episode (Figure 1). The photograph shows an irregular, serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia. She reported a similar sensation of movement in her left eye that occurred for 1 night about 5 years ago for which she visited an urgent care center where she was diagnosed with allergic conjunctivitis. She also reported recent swelling of the left side of her face with associated numbness and occasional swelling of her left hand, all of which resolved after a few days. She had immigrated from Nigeria 10 years prior, had not returned since, and was working as a traveling nurse. A slitlamp examination did not reveal any conjunctival hyperemia, foreign bodies, or other abnormalities like those shown in the photograph. Her uncorrected visual acuity was 20/20, extraocular movements were full and without pain or abnormal sensation, and intraocular pressure was normal. Her dilated fundus examination was unremarkable. A comprehensive blood cell count revealed mild elevation in the relative (but not absolute) eosinophil count (6.9% reference; 6.0% of white blood cells).Woman cellular phone photograph of the left eye shows an irregular serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia.Exploration of conjunctiva and removal of foreign body
what would you do next?
What would you do next?
Exploration of conjunctiva and removal of foreign body
Treatment with diethylcarbamazine
Serological testing for onchocerciasis
Peripheral blood smear
d
0
1
1
1
female
0
0
33
31-40
White
12
augmented_female_frommale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809279
A 33-year-old man with no prior ocular problems presented to the emergency department in central Florida with a chief complaint of a “pulling and popping” sensation in his left eye that had occurred the previous night. Ophthalmology was consulted to evaluate for a conjunctival foreign body of the left eye. At the time of the examination, his symptoms had resolved; however, he had a photograph from a cellular phone taken during the episode (Figure 1). The photograph shows an irregular, serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia. He reported a similar sensation of movement in his left eye that occurred for 1 night about 5 years ago for which he visited an urgent care center where he was diagnosed with allergic conjunctivitis. He also reported recent swelling of the left side of his face with associated numbness and occasional swelling of his left hand, all of which resolved after a few days. He had immigrated from Nigeria 10 years prior, had not returned since, and was working as a traveling nurse. A slitlamp examination did not reveal any conjunctival hyperemia, foreign bodies, or other abnormalities like those shown in the photograph. His uncorrected visual acuity was 20/20, extraocular movements were full and without pain or abnormal sensation, and intraocular pressure was normal. His dilated fundus examination was unremarkable. A comprehensive blood cell count revealed mild elevation in the relative (but not absolute) eosinophil count (6.9% reference; 6.0% of white blood cells).Patient cellular phone photograph of the left eye shows an irregular serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia.Exploration of conjunctiva and removal of foreign body What Would You Do Next?
Treatment with diethylcarbamazine
Peripheral blood smear
Serological testing for onchocerciasis
Exploration of conjunctiva and removal of foreign body
Loa loa infection
B
Peripheral blood smear
In this Nigerian patient with a sensation of movement of the left eye, a photograph showing serpiginous conjunctival elevation, and intermittent swelling of the dorsum of the hand, the most likely diagnosis is Loa loa. Also known as the African eye worm, Loa loa is a filarial nematode transmitted by day-biting flies of the genus Chrysops endemic to Western and Central Africa.1 After a bite from an infected fly, filarial larvae introduced into the subcutaneous tissue mature into adult worms that shed microfilariae that migrate into the spinal fluid, urine, sputum, and peripheral blood.2 Most infections are asymptomatic despite high numbers of circulating microfilaria; however, patients may have characteristic symptoms that include migratory angioedema of the extremities (so-called Calabar swellings) and migration of adult worms into the subconjunctival space as in this patient.3,4The recommended next step in the treatment of this patient is a peripheral blood smear for species identification and quantification of circulating microfilariae (choice B). Blood should be drawn between 10 am and 2 pm during the migration of microfilaria from the lungs into the peripheral circulation; this diurnal pattern coincides with the biting activity of the vector. Diethylcarbamazine is the first-line therapy due to its ability to eradicate both microfilaria and adult worms, while empirical therapy (choice A) can result in intense inflammatory reactions during the rapid death of the nematodes. Due to the risk of fatal encephalopathy in patients with microfilarial concentrations greater than 8000 per mL of blood, it is recommended to first measure the concentration prior to treatment with diethylcarbamazine.4 Treatment with diethylcarbamazine in patients who are coinfected with onchocerciasis can also lead to vision loss and blindness.5 Therefore, in patients from coendemic areas such as Nigeria, serological testing (choice C) should be used to rule out onchocerciasis prior to treatment, but only after identification of microfilariae in peripheral blood.6 Surgical removal of the adult worm from the conjunctiva (choice D) may treat localized symptoms and provide an opportunity for species identification; however, removal is not necessary for diagnosis and treatment with curative antiparasitic medication.Peripheral blood smears with Wright and Geimsa stains (Figure 2) revealed microfilaria (1000/mL) with characteristic morphology of Loa loa that included an approximate length of 250 μm, shortened headspace, a dense nuclear column continuous to the tip of the tail, and the presence of a sheath.7 The patient was prescribed albendazole, which is only effective against adult worms and decreases the load of microfilariae through the reduction in shedding of microfilariae.8 Serologic testing for onchocerciasis was ordered, and the US Centers for Disease Control and Prevention was contacted to obtain diethylcarbamazine, which is no longer US Food and Drug Administration–approved or commercially available in the US due to the low incidence of this disease in the US.9 The results of the onchocerciasis serology were negative, and the patient was given the diethylcarbamazine obtained from the US Centers for Disease Control and Prevention. Three weeks later, he reported no recurrence of his symptoms, and there were no eye worms on examination.Peripheral thick blood smears stained with Wright and Geimsa (main panel [original magnification ×400] and inset panel [original magnification ×200], respectively) revealed microfilaria (1000/mL) with characteristic morphology of Loa loa that included an approximate length of 250 μm, shortened headspace (pink arrowhead), a dense nuclear column continuous to the tip of the tail (blue arrowhead), and the presence of a sheath (black arrowhead).
Ophthalmology
A 33-year-old patient with no prior ocular problems presented to the emergency department in central Florida with a chief complaint of a “pulling and popping” sensation in their left eye that had occurred the previous night. Ophthalmology was consulted to evaluate for a conjunctival foreign body of the left eye. At the time of the examination, their symptoms had resolved; however, they had a photograph from a cellular phone taken during the episode (Figure 1). The photograph shows an irregular, serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia. They reported a similar sensation of movement in their left eye that occurred for 1 night about 5 years ago for which they visited an urgent care center where they was diagnosed with allergic conjunctivitis. They also reported recent swelling of the left side of their face with associated numbness and occasional swelling of their left hand, all of which resolved after a few days. They had immigrated from Nigeria 10 years prior, had not returned since, and was working as a traveling nurse. A slitlamp examination did not reveal any conjunctival hyperemia, foreign bodies, or other abnormalities like those shown in the photograph. Their uncorrected visual acuity was 20/20, extraocular movements were full and without pain or abnormal sensation, and intraocular pressure was normal. Their dilated fundus examination was unremarkable. A comprehensive blood cell count revealed mild elevation in the relative (but not absolute) eosinophil count (6.9% reference; 6.0% of white blood cells).Patient cellular phone photograph of the left eye shows an irregular serpiginous extension from beneath the plica semilunaris toward the corneal limbus with localized conjunctival hyperemia.Exploration of conjunctiva and removal of foreign body
what would you do next?
What would you do next?
Exploration of conjunctiva and removal of foreign body
Treatment with diethylcarbamazine
Serological testing for onchocerciasis
Peripheral blood smear
d
0
1
1
1
neutral
0
0
33
31-40
White
12
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809480
A 13-year-old boy with a history of sports-related blunt trauma to the left eye was referred for evaluation of an asymptomatic, pigmented iris lesion in the left eye. On examination, his best-corrected visual acuity was 20/20 OU, and intraocular pressures were normal in both eyes. Results of slitlamp examination of the right eye were unremarkable. Slitlamp examination of the left eye revealed a round, pigmented lesion measuring 3 × 3 mm in basal dimension and with gravitational shifting within the anterior chamber fluid with patient head tilt (Video). There was no corneal guttatae or edema. Anterior segment optical coherence tomography depicted the lesion in the anterior chamber angle abutting the corneal endothelium and resting on the iris stroma with no internal fluid level and no solid component. Ultrasound biomicroscopy confirmed the lesion to be cystic with a thickness of 1.6 mm. Dilated fundus examination revealed normal findings in both eyes. What Would You Do Next?
Fine-needle aspiration biopsy with cytology and cytogenetics
Iodine 125 plaque radiotherapy
Observe with no additional testing or intervention
Cyst paracentesis with complete drainage
Free-floating iris pigment epithelium cyst
C
Observe with no additional testing or intervention
A pigmented mass in the anterior chamber could represent a benign entity but also raises suspicion for malignancy, such as iris melanoma. Iris tumors are predominantly characterized as solid (79% of cases) or cystic (21% of cases).1 Solid tumors include melanocytic lesions, with the 3 most common being benign iris nevus (60%), iris melanoma (26%), and iris freckle (4%).1,2 Cystic iris tumors are subclassified based on their tissue layer of origin, arising from either the iris stroma or iris pigment epithelium (IPE). Iris stromal cysts appear translucent, located on the anterior iris surface and with smooth lobulated surface, typically in young children.1,2 By contrast, IPE cysts appear on the posterior iris surface with brown or black color simulating solid melanocytic tumors, such as iris melanoma or IPE adenoma.1,2 For this reason, some IPE cysts have historically been mistaken for melanoma, even leading to enucleation. As iris melanoma is malignant with risk for metastasis, proper treatment for life protection with consideration of the potential for ocular adverse sequelae should be realized. Differentiation of the various iris tumors is paramount to management.Ultrasound biomicroscopy and anterior-segment optical coherence tomography are useful tools in assessing the internal quality of iris tumors and differentiation of solid from cystic tumors.3 These technologies are critical for defining tumor basal dimension and thickness to monitor change over time or response to treatment. In this patient, slitlamp biomicroscopy (Figure) demonstrated the gravitational shifting of the mass (Video), and both ultrasound biomicroscopy (Figure, inset) and anterior-segment optical coherence tomography documented the mass to be cystic with no internal fluid or solid component. The Video demonstrates the superior-to-inferior descent of an IPE cyst across the visual axis, coursing purely through the aqueous humor with gravity, offering insight into the dynamic behavior of this lesion.Slitlamp biomicroscopy of the left eye showing a round, pigmented lesion in the inferior anterior chamber angle abutting the corneal endothelium and resting on the iris stroma measuring 3 × 3 mm in basal dimension. Inset, Ultrasound biomicroscopy scan of the free-floating cyst showing central lucency, no fluid level, and no solid component.Free-floating IPE cysts occur when the cyst becomes separated from the IPE, speculated to occur following ocular trauma, as in this patient.4,5 These free-floating IPE cysts are benign and rarely cause endothelial decompensation or visual sequelae.2,5 Fine-needle aspiration biopsy with cytology and cytogenetics (choice A) was not appropriate because the lesion in question was not solid or within the iris, so iris melanoma was not likely. Iodine 125 plaque radiotherapy (choice B) was not appropriate, as this was a cystic, benign mass that was unlikely to be malignant. Cyst paracentesis with complete drainage (choice D) was not necessary in this case because the cyst did not affect the visual axis and was visually asymptomatic. Additionally, drainage of a free-floating cyst without any portion anchored to the iris would likely prove to be technically difficult.In summary, free-floating IPE cysts are benign epithelial cysts that can dislodge into the aqueous or vitreous following trauma, as in this patient. This form of cyst is benign and rarely leads to adverse sequelae; therefore, conservative observation is advised.This patient was counseled on the benign nature of the free-floating IPE cyst and advised to follow up in 12 months for repeated examination with imaging. There was no expectation that this cyst would grow, rupture, transform into a malignant process, or cause damage to the cornea.
Ophthalmology
A 13-year-old boy with a history of sports-related blunt trauma to the left eye was referred for evaluation of an asymptomatic, pigmented iris lesion in the left eye. On examination, his best-corrected visual acuity was 20/20 OU, and intraocular pressures were normal in both eyes. Results of slitlamp examination of the right eye were unremarkable. Slitlamp examination of the left eye revealed a round, pigmented lesion measuring 3 × 3 mm in basal dimension and with gravitational shifting within the anterior chamber fluid with patient head tilt (Video). There was no corneal guttatae or edema. Anterior segment optical coherence tomography depicted the lesion in the anterior chamber angle abutting the corneal endothelium and resting on the iris stroma with no internal fluid level and no solid component. Ultrasound biomicroscopy confirmed the lesion to be cystic with a thickness of 1.6 mm. Dilated fundus examination revealed normal findings in both eyes.
what would you do next?
What would you do next?
Fine-needle aspiration biopsy with cytology and cytogenetics
Cyst paracentesis with complete drainage
Iodine 125 plaque radiotherapy
Observe with no additional testing or intervention
d
1
0
1
0
male
0
0
13
11-20
null
13
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809480
A 13-year-old boy with a history of sports-related blunt trauma to the left eye was referred for evaluation of an asymptomatic, pigmented iris lesion in the left eye. On examination, his best-corrected visual acuity was 20/20 OU, and intraocular pressures were normal in both eyes. Results of slitlamp examination of the right eye were unremarkable. Slitlamp examination of the left eye revealed a round, pigmented lesion measuring 3 × 3 mm in basal dimension and with gravitational shifting within the anterior chamber fluid with patient head tilt (Video). There was no corneal guttatae or edema. Anterior segment optical coherence tomography depicted the lesion in the anterior chamber angle abutting the corneal endothelium and resting on the iris stroma with no internal fluid level and no solid component. Ultrasound biomicroscopy confirmed the lesion to be cystic with a thickness of 1.6 mm. Dilated fundus examination revealed normal findings in both eyes. What Would You Do Next?
Fine-needle aspiration biopsy with cytology and cytogenetics
Iodine 125 plaque radiotherapy
Observe with no additional testing or intervention
Cyst paracentesis with complete drainage
Free-floating iris pigment epithelium cyst
C
Observe with no additional testing or intervention
A pigmented mass in the anterior chamber could represent a benign entity but also raises suspicion for malignancy, such as iris melanoma. Iris tumors are predominantly characterized as solid (79% of cases) or cystic (21% of cases).1 Solid tumors include melanocytic lesions, with the 3 most common being benign iris nevus (60%), iris melanoma (26%), and iris freckle (4%).1,2 Cystic iris tumors are subclassified based on their tissue layer of origin, arising from either the iris stroma or iris pigment epithelium (IPE). Iris stromal cysts appear translucent, located on the anterior iris surface and with smooth lobulated surface, typically in young children.1,2 By contrast, IPE cysts appear on the posterior iris surface with brown or black color simulating solid melanocytic tumors, such as iris melanoma or IPE adenoma.1,2 For this reason, some IPE cysts have historically been mistaken for melanoma, even leading to enucleation. As iris melanoma is malignant with risk for metastasis, proper treatment for life protection with consideration of the potential for ocular adverse sequelae should be realized. Differentiation of the various iris tumors is paramount to management.Ultrasound biomicroscopy and anterior-segment optical coherence tomography are useful tools in assessing the internal quality of iris tumors and differentiation of solid from cystic tumors.3 These technologies are critical for defining tumor basal dimension and thickness to monitor change over time or response to treatment. In this patient, slitlamp biomicroscopy (Figure) demonstrated the gravitational shifting of the mass (Video), and both ultrasound biomicroscopy (Figure, inset) and anterior-segment optical coherence tomography documented the mass to be cystic with no internal fluid or solid component. The Video demonstrates the superior-to-inferior descent of an IPE cyst across the visual axis, coursing purely through the aqueous humor with gravity, offering insight into the dynamic behavior of this lesion.Slitlamp biomicroscopy of the left eye showing a round, pigmented lesion in the inferior anterior chamber angle abutting the corneal endothelium and resting on the iris stroma measuring 3 × 3 mm in basal dimension. Inset, Ultrasound biomicroscopy scan of the free-floating cyst showing central lucency, no fluid level, and no solid component.Free-floating IPE cysts occur when the cyst becomes separated from the IPE, speculated to occur following ocular trauma, as in this patient.4,5 These free-floating IPE cysts are benign and rarely cause endothelial decompensation or visual sequelae.2,5 Fine-needle aspiration biopsy with cytology and cytogenetics (choice A) was not appropriate because the lesion in question was not solid or within the iris, so iris melanoma was not likely. Iodine 125 plaque radiotherapy (choice B) was not appropriate, as this was a cystic, benign mass that was unlikely to be malignant. Cyst paracentesis with complete drainage (choice D) was not necessary in this case because the cyst did not affect the visual axis and was visually asymptomatic. Additionally, drainage of a free-floating cyst without any portion anchored to the iris would likely prove to be technically difficult.In summary, free-floating IPE cysts are benign epithelial cysts that can dislodge into the aqueous or vitreous following trauma, as in this patient. This form of cyst is benign and rarely leads to adverse sequelae; therefore, conservative observation is advised.This patient was counseled on the benign nature of the free-floating IPE cyst and advised to follow up in 12 months for repeated examination with imaging. There was no expectation that this cyst would grow, rupture, transform into a malignant process, or cause damage to the cornea.
Ophthalmology
A 13-year-old girl with a history of sports-related blunt trauma to the left eye was referred for evaluation of an asymptomatic, pigmented iris lesion in the left eye. On examination, her best-corrected visual acuity was 20/20 OU, and intraocular pressures were normal in both eyes. Results of slitlamp examination of the right eye were unremarkable. Slitlamp examination of the left eye revealed a round, pigmented lesion measuring 3 × 3 mm in basal dimension and with gravitational shifting within the anterior chamber fluid with woman head tilt (Video). There was no corneal guttatae or edema. Anterior segment optical coherence tomography depicted the lesion in the anterior chamber angle abutting the corneal endothelium and resting on the iris stroma with no internal fluid level and no solid component. Ultrasound biomicroscopy confirmed the lesion to be cystic with a thickness of 1.6 mm. Dilated fundus examination revealed normal findings in both eyes.
what would you do next?
What would you do next?
Fine-needle aspiration biopsy with cytology and cytogenetics
Cyst paracentesis with complete drainage
Iodine 125 plaque radiotherapy
Observe with no additional testing or intervention
d
1
0
1
0
female
0
0
13
11-20
null
13
augmented_female_frommale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809480
A 13-year-old boy with a history of sports-related blunt trauma to the left eye was referred for evaluation of an asymptomatic, pigmented iris lesion in the left eye. On examination, his best-corrected visual acuity was 20/20 OU, and intraocular pressures were normal in both eyes. Results of slitlamp examination of the right eye were unremarkable. Slitlamp examination of the left eye revealed a round, pigmented lesion measuring 3 × 3 mm in basal dimension and with gravitational shifting within the anterior chamber fluid with patient head tilt (Video). There was no corneal guttatae or edema. Anterior segment optical coherence tomography depicted the lesion in the anterior chamber angle abutting the corneal endothelium and resting on the iris stroma with no internal fluid level and no solid component. Ultrasound biomicroscopy confirmed the lesion to be cystic with a thickness of 1.6 mm. Dilated fundus examination revealed normal findings in both eyes. What Would You Do Next?
Fine-needle aspiration biopsy with cytology and cytogenetics
Iodine 125 plaque radiotherapy
Observe with no additional testing or intervention
Cyst paracentesis with complete drainage
Free-floating iris pigment epithelium cyst
C
Observe with no additional testing or intervention
A pigmented mass in the anterior chamber could represent a benign entity but also raises suspicion for malignancy, such as iris melanoma. Iris tumors are predominantly characterized as solid (79% of cases) or cystic (21% of cases).1 Solid tumors include melanocytic lesions, with the 3 most common being benign iris nevus (60%), iris melanoma (26%), and iris freckle (4%).1,2 Cystic iris tumors are subclassified based on their tissue layer of origin, arising from either the iris stroma or iris pigment epithelium (IPE). Iris stromal cysts appear translucent, located on the anterior iris surface and with smooth lobulated surface, typically in young children.1,2 By contrast, IPE cysts appear on the posterior iris surface with brown or black color simulating solid melanocytic tumors, such as iris melanoma or IPE adenoma.1,2 For this reason, some IPE cysts have historically been mistaken for melanoma, even leading to enucleation. As iris melanoma is malignant with risk for metastasis, proper treatment for life protection with consideration of the potential for ocular adverse sequelae should be realized. Differentiation of the various iris tumors is paramount to management.Ultrasound biomicroscopy and anterior-segment optical coherence tomography are useful tools in assessing the internal quality of iris tumors and differentiation of solid from cystic tumors.3 These technologies are critical for defining tumor basal dimension and thickness to monitor change over time or response to treatment. In this patient, slitlamp biomicroscopy (Figure) demonstrated the gravitational shifting of the mass (Video), and both ultrasound biomicroscopy (Figure, inset) and anterior-segment optical coherence tomography documented the mass to be cystic with no internal fluid or solid component. The Video demonstrates the superior-to-inferior descent of an IPE cyst across the visual axis, coursing purely through the aqueous humor with gravity, offering insight into the dynamic behavior of this lesion.Slitlamp biomicroscopy of the left eye showing a round, pigmented lesion in the inferior anterior chamber angle abutting the corneal endothelium and resting on the iris stroma measuring 3 × 3 mm in basal dimension. Inset, Ultrasound biomicroscopy scan of the free-floating cyst showing central lucency, no fluid level, and no solid component.Free-floating IPE cysts occur when the cyst becomes separated from the IPE, speculated to occur following ocular trauma, as in this patient.4,5 These free-floating IPE cysts are benign and rarely cause endothelial decompensation or visual sequelae.2,5 Fine-needle aspiration biopsy with cytology and cytogenetics (choice A) was not appropriate because the lesion in question was not solid or within the iris, so iris melanoma was not likely. Iodine 125 plaque radiotherapy (choice B) was not appropriate, as this was a cystic, benign mass that was unlikely to be malignant. Cyst paracentesis with complete drainage (choice D) was not necessary in this case because the cyst did not affect the visual axis and was visually asymptomatic. Additionally, drainage of a free-floating cyst without any portion anchored to the iris would likely prove to be technically difficult.In summary, free-floating IPE cysts are benign epithelial cysts that can dislodge into the aqueous or vitreous following trauma, as in this patient. This form of cyst is benign and rarely leads to adverse sequelae; therefore, conservative observation is advised.This patient was counseled on the benign nature of the free-floating IPE cyst and advised to follow up in 12 months for repeated examination with imaging. There was no expectation that this cyst would grow, rupture, transform into a malignant process, or cause damage to the cornea.
Ophthalmology
A 13-year-old child with a history of sports-related blunt trauma to the left eye was referred for evaluation of an asymptomatic, pigmented iris lesion in the left eye. On examination, their best-corrected visual acuity was 20/20 OU, and intraocular pressures were normal in both eyes. Results of slitlamp examination of the right eye were unremarkable. Slitlamp examination of the left eye revealed a round, pigmented lesion measuring 3 × 3 mm in basal dimension and with gravitational shifting within the anterior chamber fluid with patient head tilt (Video). There was no corneal guttatae or edema. Anterior segment optical coherence tomography depicted the lesion in the anterior chamber angle abutting the corneal endothelium and resting on the iris stroma with no internal fluid level and no solid component. Ultrasound biomicroscopy confirmed the lesion to be cystic with a thickness of 1.6 mm. Dilated fundus examination revealed normal findings in both eyes.
what would you do next?
What would you do next?
Fine-needle aspiration biopsy with cytology and cytogenetics
Cyst paracentesis with complete drainage
Iodine 125 plaque radiotherapy
Observe with no additional testing or intervention
d
1
0
1
0
neutral
0
0
13
11-20
null
13
augmented_neutral_frommale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2808886
A woman in her 60s with long-standing thickening and induration of the legs presented with 1 week of left leg pain. The patient had previously been diagnosed with elephantiasis nostras verrucosa, and she was advised to elevate and compress the leg but was unavailable for follow-up. Her medical history was notable for morbid obesity, hypertension, diabetes, and stroke. She reported no fever, fatigue, weight changes, gastrointestinal symptoms, difficulty concentrating, anxiety, or hyperhidrosis. Physical examination results revealed multiple firm skin-colored papules and nodules coalescing to form a large plaque on the anterior aspect of the left lower leg, whereas the right lower leg had diffuse induration and hyperpigmentation (Figure 1). There was no palpable lymphadenopathy. Laboratory analysis revealed normal complete blood cell count, thyroid function, serum protein electrophoresis, and serum immunofixation. A biopsy of a left leg nodule was performed.Multiple firm skin-colored papules and nodules coalesce to form a large plaque on the anterior aspect of the left lower leg. What Is Your Diagnosis?
Pretibial myxedema
Elephantiasis nostras verrucosa
Lobomycosis
Euthyroid pretibial mucinosis
D. Euthyroid pretibial mucinosis
D
Euthyroid pretibial mucinosis
Histopathologic examination showed an atrophic epidermis, angioplasia with vertically oriented vessels, horizontal fibrosis, hemosiderin deposits, and increased mucin deposition, extending from the papillary to the reticular dermis (Figure 2). Pretibial mucinosis in a patient with euthyroid due to obesity and venous stasis was diagnosed. The patient’s cutaneous mucinosis was complicated by cellulitis, which resolved with a short course of antibiotics. Weight loss was recommended, and compression and leg elevation were suggested for the management of chronic skin changes. Detailed vascular studies to explain the asymmetry were planned, but the patient was unavailable for follow-up after multiple hospital admissions for her comorbidities.A and B, Histologic examination shows atrophic epidermis, angioplasia with vertically oriented vessels, and horizontal fibrosis (hematoxylin-eosin). C, Colloidal iron staining with increased mucin deposition extending from the papillary to the reticular dermis.Classically, accumulation of mucin on the shins is associated with pretibial myxedema (choice A), prompting thyroid function testing. However, in a small percentage of patients with pretibial mucinosis, thyroid dysfunction is not identified.1 Pretibial mucinosis in patients with euthyroid has been described in states of chronic stasis, such as venous insufficiency and obesity with lymphedema, termed pretibial stasis mucinosis and obesity-associated lymphedematous mucinosis (OALM).1,2 These conditions often overlap and share a similar pathogenesis. Chronic venous insufficiency and/or obesity-associated lymphedema have been suggested to induce hypoxia, leading to angiogenesis and plasma protein extravasation, ultimately resulting in increased production and deposition of mucopolysaccharides.3Patients with stasis mucinosis and OALM present with skin-colored to yellow-red papules, plaques, and/or nodules. Symptoms can be bilateral or asymmetric, involving a single extremity. Distinct histopathologic findings, such as epidermal atrophy, angioplasia with vertically oriented capillaries, increased stellate fibroblasts, and hemosiderin deposition, aid in confirming the diagnosis and differentiating it from other forms of cutaneous mucinosis.4 Treatment entails weight loss and modalities that improve lymphatic and venous blood flow, such as compression stockings. Second- and third-line therapeutics include topical corticosteroids or intralesional corticosteroids, pentoxifylline, octreotide, plasmapheresis, and intravenous immunoglobulins.Differential diagnosis of multiple long-standing papules and nodules on the lower legs includes pretibial myxedema and elephantiasis nostras verrucosa (choice B) and lobomycosis (choice C). Clinical and histologic features are essential to differentiate these conditions. Pretibial myxedema in thyroid disease often precedes skin manifestations, and it is commonly associated with coexisting orbitopathy in Graves disease.4 Histologically, it is characterized by mucin deposition in the upper dermis, with a grenz zone present in the superficial dermis.5 Euthyroid pretibial mucinosis lacks this feature, showing mucin deposition throughout the dermis. Elephantiasis nostras verrucosa may be distinguished clinically from pretibial mucinosis, as it develops on the dorsa of the feet and extends proximally over time. It can be differentiated histologically by the presence of pseudoepitheliomatous hyperplasia and exhibits dilated lymph channels in the early stages and extensive dermal fibrosis in the later stages. Mucin is absent.6 Pretibial mucinosis nodules can be mistaken for lobomycosis, an uncommon fungal infection acquired in tropical areas of Central and South America, that is histopathologically is characterized by granulomas containing sclerotic bodies.7
Dermatology
A woman in her 60s with long-standing thickening and induration of the legs presented with 1 week of left leg pain. The patient had previously been diagnosed with elephantiasis nostras verrucosa, and she was advised to elevate and compress the leg but was unavailable for follow-up. Her medical history was notable for morbid obesity, hypertension, diabetes, and stroke. She reported no fever, fatigue, weight changes, gastrointestinal symptoms, difficulty concentrating, anxiety, or hyperhidrosis. Physical examination results revealed multiple firm skin-colored papules and nodules coalescing to form a large plaque on the anterior aspect of the left lower leg, whereas the right lower leg had diffuse induration and hyperpigmentation (Figure 1). There was no palpable lymphadenopathy. Laboratory analysis revealed normal complete blood cell count, thyroid function, serum protein electrophoresis, and serum immunofixation. A biopsy of a left leg nodule was performed.Multiple firm skin-colored papules and nodules coalesce to form a large plaque on the anterior aspect of the left lower leg.
what is your diagnosis?
What is your diagnosis?
Elephantiasis nostras verrucosa
Pretibial myxedema
Euthyroid pretibial mucinosis
Lobomycosis
c
0
1
1
1
female
0
0
65
61-70
null
14
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2808886
A woman in her 60s with long-standing thickening and induration of the legs presented with 1 week of left leg pain. The patient had previously been diagnosed with elephantiasis nostras verrucosa, and she was advised to elevate and compress the leg but was unavailable for follow-up. Her medical history was notable for morbid obesity, hypertension, diabetes, and stroke. She reported no fever, fatigue, weight changes, gastrointestinal symptoms, difficulty concentrating, anxiety, or hyperhidrosis. Physical examination results revealed multiple firm skin-colored papules and nodules coalescing to form a large plaque on the anterior aspect of the left lower leg, whereas the right lower leg had diffuse induration and hyperpigmentation (Figure 1). There was no palpable lymphadenopathy. Laboratory analysis revealed normal complete blood cell count, thyroid function, serum protein electrophoresis, and serum immunofixation. A biopsy of a left leg nodule was performed.Multiple firm skin-colored papules and nodules coalesce to form a large plaque on the anterior aspect of the left lower leg. What Is Your Diagnosis?
Pretibial myxedema
Elephantiasis nostras verrucosa
Lobomycosis
Euthyroid pretibial mucinosis
D. Euthyroid pretibial mucinosis
D
Euthyroid pretibial mucinosis
Histopathologic examination showed an atrophic epidermis, angioplasia with vertically oriented vessels, horizontal fibrosis, hemosiderin deposits, and increased mucin deposition, extending from the papillary to the reticular dermis (Figure 2). Pretibial mucinosis in a patient with euthyroid due to obesity and venous stasis was diagnosed. The patient’s cutaneous mucinosis was complicated by cellulitis, which resolved with a short course of antibiotics. Weight loss was recommended, and compression and leg elevation were suggested for the management of chronic skin changes. Detailed vascular studies to explain the asymmetry were planned, but the patient was unavailable for follow-up after multiple hospital admissions for her comorbidities.A and B, Histologic examination shows atrophic epidermis, angioplasia with vertically oriented vessels, and horizontal fibrosis (hematoxylin-eosin). C, Colloidal iron staining with increased mucin deposition extending from the papillary to the reticular dermis.Classically, accumulation of mucin on the shins is associated with pretibial myxedema (choice A), prompting thyroid function testing. However, in a small percentage of patients with pretibial mucinosis, thyroid dysfunction is not identified.1 Pretibial mucinosis in patients with euthyroid has been described in states of chronic stasis, such as venous insufficiency and obesity with lymphedema, termed pretibial stasis mucinosis and obesity-associated lymphedematous mucinosis (OALM).1,2 These conditions often overlap and share a similar pathogenesis. Chronic venous insufficiency and/or obesity-associated lymphedema have been suggested to induce hypoxia, leading to angiogenesis and plasma protein extravasation, ultimately resulting in increased production and deposition of mucopolysaccharides.3Patients with stasis mucinosis and OALM present with skin-colored to yellow-red papules, plaques, and/or nodules. Symptoms can be bilateral or asymmetric, involving a single extremity. Distinct histopathologic findings, such as epidermal atrophy, angioplasia with vertically oriented capillaries, increased stellate fibroblasts, and hemosiderin deposition, aid in confirming the diagnosis and differentiating it from other forms of cutaneous mucinosis.4 Treatment entails weight loss and modalities that improve lymphatic and venous blood flow, such as compression stockings. Second- and third-line therapeutics include topical corticosteroids or intralesional corticosteroids, pentoxifylline, octreotide, plasmapheresis, and intravenous immunoglobulins.Differential diagnosis of multiple long-standing papules and nodules on the lower legs includes pretibial myxedema and elephantiasis nostras verrucosa (choice B) and lobomycosis (choice C). Clinical and histologic features are essential to differentiate these conditions. Pretibial myxedema in thyroid disease often precedes skin manifestations, and it is commonly associated with coexisting orbitopathy in Graves disease.4 Histologically, it is characterized by mucin deposition in the upper dermis, with a grenz zone present in the superficial dermis.5 Euthyroid pretibial mucinosis lacks this feature, showing mucin deposition throughout the dermis. Elephantiasis nostras verrucosa may be distinguished clinically from pretibial mucinosis, as it develops on the dorsa of the feet and extends proximally over time. It can be differentiated histologically by the presence of pseudoepitheliomatous hyperplasia and exhibits dilated lymph channels in the early stages and extensive dermal fibrosis in the later stages. Mucin is absent.6 Pretibial mucinosis nodules can be mistaken for lobomycosis, an uncommon fungal infection acquired in tropical areas of Central and South America, that is histopathologically is characterized by granulomas containing sclerotic bodies.7
Dermatology
A man in him 60s with long-standing thickening and induration of the legs presented with 1 week of left leg pain. The man had previously been diagnosed with elephantiasis nostras verrucosa, and he was advised to elevate and compress the leg but was unavailable for follow-up. Him medical history was notable for morbid obesity, hypertension, diabetes, and stroke. He reported no fever, fatigue, weight changes, gastrointestinal symptoms, difficulty concentrating, anxiety, or hyperhidrosis. Physical examination results revealed multiple firm skin-colored papules and nodules coalescing to form a large plaque on the anterior aspect of the left lower leg, whereas the right lower leg had diffuse induration and hyperpigmentation (Figure 1). There was no palpable lymphadenopathy. Laboratory analysis revealed normal complete blood cell count, thyroid function, serum protein electrophoresis, and serum immunofixation. A biopsy of a left leg nodule was performed.Multiple firm skin-colored papules and nodules coalesce to form a large plaque on the anterior aspect of the left lower leg.
what is your diagnosis?
What is your diagnosis?
Elephantiasis nostras verrucosa
Pretibial myxedema
Euthyroid pretibial mucinosis
Lobomycosis
c
0
1
1
1
male
0
0
65
61-70
null
14
augmented_male_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2808886
A woman in her 60s with long-standing thickening and induration of the legs presented with 1 week of left leg pain. The patient had previously been diagnosed with elephantiasis nostras verrucosa, and she was advised to elevate and compress the leg but was unavailable for follow-up. Her medical history was notable for morbid obesity, hypertension, diabetes, and stroke. She reported no fever, fatigue, weight changes, gastrointestinal symptoms, difficulty concentrating, anxiety, or hyperhidrosis. Physical examination results revealed multiple firm skin-colored papules and nodules coalescing to form a large plaque on the anterior aspect of the left lower leg, whereas the right lower leg had diffuse induration and hyperpigmentation (Figure 1). There was no palpable lymphadenopathy. Laboratory analysis revealed normal complete blood cell count, thyroid function, serum protein electrophoresis, and serum immunofixation. A biopsy of a left leg nodule was performed.Multiple firm skin-colored papules and nodules coalesce to form a large plaque on the anterior aspect of the left lower leg. What Is Your Diagnosis?
Pretibial myxedema
Elephantiasis nostras verrucosa
Lobomycosis
Euthyroid pretibial mucinosis
D. Euthyroid pretibial mucinosis
D
Euthyroid pretibial mucinosis
Histopathologic examination showed an atrophic epidermis, angioplasia with vertically oriented vessels, horizontal fibrosis, hemosiderin deposits, and increased mucin deposition, extending from the papillary to the reticular dermis (Figure 2). Pretibial mucinosis in a patient with euthyroid due to obesity and venous stasis was diagnosed. The patient’s cutaneous mucinosis was complicated by cellulitis, which resolved with a short course of antibiotics. Weight loss was recommended, and compression and leg elevation were suggested for the management of chronic skin changes. Detailed vascular studies to explain the asymmetry were planned, but the patient was unavailable for follow-up after multiple hospital admissions for her comorbidities.A and B, Histologic examination shows atrophic epidermis, angioplasia with vertically oriented vessels, and horizontal fibrosis (hematoxylin-eosin). C, Colloidal iron staining with increased mucin deposition extending from the papillary to the reticular dermis.Classically, accumulation of mucin on the shins is associated with pretibial myxedema (choice A), prompting thyroid function testing. However, in a small percentage of patients with pretibial mucinosis, thyroid dysfunction is not identified.1 Pretibial mucinosis in patients with euthyroid has been described in states of chronic stasis, such as venous insufficiency and obesity with lymphedema, termed pretibial stasis mucinosis and obesity-associated lymphedematous mucinosis (OALM).1,2 These conditions often overlap and share a similar pathogenesis. Chronic venous insufficiency and/or obesity-associated lymphedema have been suggested to induce hypoxia, leading to angiogenesis and plasma protein extravasation, ultimately resulting in increased production and deposition of mucopolysaccharides.3Patients with stasis mucinosis and OALM present with skin-colored to yellow-red papules, plaques, and/or nodules. Symptoms can be bilateral or asymmetric, involving a single extremity. Distinct histopathologic findings, such as epidermal atrophy, angioplasia with vertically oriented capillaries, increased stellate fibroblasts, and hemosiderin deposition, aid in confirming the diagnosis and differentiating it from other forms of cutaneous mucinosis.4 Treatment entails weight loss and modalities that improve lymphatic and venous blood flow, such as compression stockings. Second- and third-line therapeutics include topical corticosteroids or intralesional corticosteroids, pentoxifylline, octreotide, plasmapheresis, and intravenous immunoglobulins.Differential diagnosis of multiple long-standing papules and nodules on the lower legs includes pretibial myxedema and elephantiasis nostras verrucosa (choice B) and lobomycosis (choice C). Clinical and histologic features are essential to differentiate these conditions. Pretibial myxedema in thyroid disease often precedes skin manifestations, and it is commonly associated with coexisting orbitopathy in Graves disease.4 Histologically, it is characterized by mucin deposition in the upper dermis, with a grenz zone present in the superficial dermis.5 Euthyroid pretibial mucinosis lacks this feature, showing mucin deposition throughout the dermis. Elephantiasis nostras verrucosa may be distinguished clinically from pretibial mucinosis, as it develops on the dorsa of the feet and extends proximally over time. It can be differentiated histologically by the presence of pseudoepitheliomatous hyperplasia and exhibits dilated lymph channels in the early stages and extensive dermal fibrosis in the later stages. Mucin is absent.6 Pretibial mucinosis nodules can be mistaken for lobomycosis, an uncommon fungal infection acquired in tropical areas of Central and South America, that is histopathologically is characterized by granulomas containing sclerotic bodies.7
Dermatology
A patient in them 60s with long-standing thickening and induration of the legs presented with 1 week of left leg pain. The patient had previously been diagnosed with elephantiasis nostras verrucosa, and they was advised to elevate and compress the leg but was unavailable for follow-up. Them medical history was notable for morbid obesity, hypertension, diabetes, and stroke. They reported no fever, fatigue, weight changes, gastrointestinal symptoms, difficulty concentrating, anxiety, or hyperhidrosis. Physical examination results revealed multiple firm skin-colored papules and nodules coalescing to form a large plaque on the anterior aspect of the left lower leg, whereas the right lower leg had diffuse induration and hyperpigmentation (Figure 1). There was no palpable lymphadenopathy. Laboratory analysis revealed normal complete blood cell count, thyroid function, serum protein electrophoresis, and serum immunofixation. A biopsy of a left leg nodule was performed.Multiple firm skin-colored papules and nodules coalesce to form a large plaque on the anterior aspect of the left lower leg.
what is your diagnosis?
What is your diagnosis?
Elephantiasis nostras verrucosa
Pretibial myxedema
Euthyroid pretibial mucinosis
Lobomycosis
c
0
1
1
1
neutral
0
0
65
61-70
null
14
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2809085
A female patient in her early 50s presented with multiple (approximately 10) bluish, partially keratotic papules and nodules on the lower legs (Figure, A). Since the age of 24 years, the lesions had been growing very slowly in number and size and had been bleeding occasionally after a trauma. The patient used a wheelchair due to a right-sided hemiparesis and hemianopsia caused by an intracranial hemorrhage. At the age of 4 years, recurrent seizures had led to the diagnosis of cerebral venous malformations. Clinical examination also demonstrated a lipolymphedema of the lower legs that had been attributed to a functional venous insufficiency with insufficient venous pump activity. The patient had been wearing compression stockings for years. The family history was positive for cerebral venous malformations involving the patient’s father and aunt, the father having died of an intracranial hemorrhage in his early 70s.A, Clinical picture. B and C, Hematoxylin-eosin stain.Sonography of the skin lesions demonstrated intradermal slow-flow vascular malformations with a maximum diameter of 1.2 cm. A biopsy of a lower-extremity papule was performed (Figure, B and C). Mutational analysis resulted in the detection of a heterozygous pathogenic KRIT1 variant. What Is Your Diagnosis?
Blue rubber bleb nevus syndrome
Familial cerebral cavernous malformations
Glomuvenous malformations
Verrucous venous malformations
B. Familial cerebral cavernous malformations
B
Familial cerebral cavernous malformations
Skin biopsy revealed a well-circumscribed lesion with hyperkeratosis and acanthosis of the epidermis overlying a conglomerate of dilated venous-like malformations with continuous smooth muscle in the upper dermis, partly showing thrombosis. Histomorphology together with a positive personal and family history of venous malformations and the identification of a pathologic KRIT1 variant led to the diagnosis of familial cerebral cavernous malformations (CCMs) in the current patient.Cerebral cavernous malformations or cavernomas (International Society for the Study of Vascular Anomalies [ISSVA] simple vascular malformations III) are slow-flowing venous malformations that are lined by a single layer of endothelium with no involvement of brain parenchyma.1 They can occur in sporadic (80%) or familial (20%) form, affecting up to 0.5% in the general population.2 Familial CCMs are frequently inherited as multiple lesions in an autosomal dominant manner. Following the Knudson hypothesis, they are caused by germline loss-of-function variants in KRIT1, CCM2, or PDCD10 in association with a somatic “second hit” in the other allele.3 Among patients with CCMs, 40% to 70% present with neurological symptoms, such as seizures, focal deficits, and headaches due to intracranial hemorrhages. Extraneurological manifestations may involve the skin, retina, kidney, and liver. In a study of 417 patients with familial CMMs, 38 patients (9%) had cutaneous venous malformations, which is a higher rate than in the general population (0.3%).4 In this study, the lesions were associated with KRIT1 variants and further classified as hyperkeratotic capillary-venous malformations, capillary malformations, and venous malformations. Prognosis and management are determined by the location, number, and size of the cerebral venous malformations. Recent advances in understanding CCM pathophysiology have revealed new strategies to control disease progression, ie, Rho kinase inhibition by atorvastatin.5Blue rubber bleb nevus syndrome (BRBNS; ISSVA simple vascular malformations III) is another rare disorder characterized by multiple, most commonly cutaneous and gastrointestinal venous malformations.1 Skin lesions present as rubbery, bluish, easily compressible nodules since birth or early childhood.6 Histopathology reveals large, thin-walled vessels with discontinuous or no smooth muscle separated by variable amounts of fibrous tissue scattered through the dermis.7 Gastrointestinal malformations typically cause recurrent hemorrhages resulting in chronic anemia and may occasionally lead to volvulus, intussusception, or perforation. Thirteen percent of patients have central nervous system involvement.8 Familial occurrence is extremely rare, as BRBNS is due to somatic double (cis) variants in TEK, the gene encoding endothelial cell tyrosine kinase receptor TIE2.9Glomuvenous malformations or glomangiomas (ISSVA simple vascular malformations III) are bluish, tender, noncompressible papules and nodules of the cutis that histologically represent venous-like channels surrounded by 1 or 2 layers of glomus cells, and no intracranial involvement is seen.1 Glomuvenous malformations are inherited in over 60% of cases in an autosomal dominant manner and are due to loss-of-function variants in the glomulin gene.7 Verrucous venous malformations (ISSVA simple vascular malformations III) are sporadic congenital hyperkeratotic nodules—mainly on the legs—that show a deep vascular component and no further associations and, in particular, no intracranial involvement.1 Histopathology reveals a compact hyperkeratosis, papillomatosis, and acanthosis overlying dilated vessels extending into the deep dermis and the subcutis.7 A somatic missense MAP3K3 variant has been recently identified.10
Dermatology
A female patient in her early 50s presented with multiple (approximately 10) bluish, partially keratotic papules and nodules on the lower legs (Figure, A). Since the age of 24 years, the lesions had been growing very slowly in number and size and had been bleeding occasionally after a trauma. The patient used a wheelchair due to a right-sided hemiparesis and hemianopsia caused by an intracranial hemorrhage. At the age of 4 years, recurrent seizures had led to the diagnosis of cerebral venous malformations. Clinical examination also demonstrated a lipolymphedema of the lower legs that had been attributed to a functional venous insufficiency with insufficient venous pump activity. The patient had been wearing compression stockings for years. The family history was positive for cerebral venous malformations involving the patient’s father and aunt, the father having died of an intracranial hemorrhage in his early 70s.A, Clinical picture. B and C, Hematoxylin-eosin stain.Sonography of the skin lesions demonstrated intradermal slow-flow vascular malformations with a maximum diameter of 1.2 cm. A biopsy of a lower-extremity papule was performed (Figure, B and C). Mutational analysis resulted in the detection of a heterozygous pathogenic KRIT1 variant.
what is your diagnosis?
What is your diagnosis?
Blue rubber bleb nevus syndrome
Familial cerebral cavernous malformations
Verrucous venous malformations
Glomuvenous malformations
b
0
1
1
1
female
0
0
52
51-60
null
15
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2809085
A female patient in her early 50s presented with multiple (approximately 10) bluish, partially keratotic papules and nodules on the lower legs (Figure, A). Since the age of 24 years, the lesions had been growing very slowly in number and size and had been bleeding occasionally after a trauma. The patient used a wheelchair due to a right-sided hemiparesis and hemianopsia caused by an intracranial hemorrhage. At the age of 4 years, recurrent seizures had led to the diagnosis of cerebral venous malformations. Clinical examination also demonstrated a lipolymphedema of the lower legs that had been attributed to a functional venous insufficiency with insufficient venous pump activity. The patient had been wearing compression stockings for years. The family history was positive for cerebral venous malformations involving the patient’s father and aunt, the father having died of an intracranial hemorrhage in his early 70s.A, Clinical picture. B and C, Hematoxylin-eosin stain.Sonography of the skin lesions demonstrated intradermal slow-flow vascular malformations with a maximum diameter of 1.2 cm. A biopsy of a lower-extremity papule was performed (Figure, B and C). Mutational analysis resulted in the detection of a heterozygous pathogenic KRIT1 variant. What Is Your Diagnosis?
Blue rubber bleb nevus syndrome
Familial cerebral cavernous malformations
Glomuvenous malformations
Verrucous venous malformations
B. Familial cerebral cavernous malformations
B
Familial cerebral cavernous malformations
Skin biopsy revealed a well-circumscribed lesion with hyperkeratosis and acanthosis of the epidermis overlying a conglomerate of dilated venous-like malformations with continuous smooth muscle in the upper dermis, partly showing thrombosis. Histomorphology together with a positive personal and family history of venous malformations and the identification of a pathologic KRIT1 variant led to the diagnosis of familial cerebral cavernous malformations (CCMs) in the current patient.Cerebral cavernous malformations or cavernomas (International Society for the Study of Vascular Anomalies [ISSVA] simple vascular malformations III) are slow-flowing venous malformations that are lined by a single layer of endothelium with no involvement of brain parenchyma.1 They can occur in sporadic (80%) or familial (20%) form, affecting up to 0.5% in the general population.2 Familial CCMs are frequently inherited as multiple lesions in an autosomal dominant manner. Following the Knudson hypothesis, they are caused by germline loss-of-function variants in KRIT1, CCM2, or PDCD10 in association with a somatic “second hit” in the other allele.3 Among patients with CCMs, 40% to 70% present with neurological symptoms, such as seizures, focal deficits, and headaches due to intracranial hemorrhages. Extraneurological manifestations may involve the skin, retina, kidney, and liver. In a study of 417 patients with familial CMMs, 38 patients (9%) had cutaneous venous malformations, which is a higher rate than in the general population (0.3%).4 In this study, the lesions were associated with KRIT1 variants and further classified as hyperkeratotic capillary-venous malformations, capillary malformations, and venous malformations. Prognosis and management are determined by the location, number, and size of the cerebral venous malformations. Recent advances in understanding CCM pathophysiology have revealed new strategies to control disease progression, ie, Rho kinase inhibition by atorvastatin.5Blue rubber bleb nevus syndrome (BRBNS; ISSVA simple vascular malformations III) is another rare disorder characterized by multiple, most commonly cutaneous and gastrointestinal venous malformations.1 Skin lesions present as rubbery, bluish, easily compressible nodules since birth or early childhood.6 Histopathology reveals large, thin-walled vessels with discontinuous or no smooth muscle separated by variable amounts of fibrous tissue scattered through the dermis.7 Gastrointestinal malformations typically cause recurrent hemorrhages resulting in chronic anemia and may occasionally lead to volvulus, intussusception, or perforation. Thirteen percent of patients have central nervous system involvement.8 Familial occurrence is extremely rare, as BRBNS is due to somatic double (cis) variants in TEK, the gene encoding endothelial cell tyrosine kinase receptor TIE2.9Glomuvenous malformations or glomangiomas (ISSVA simple vascular malformations III) are bluish, tender, noncompressible papules and nodules of the cutis that histologically represent venous-like channels surrounded by 1 or 2 layers of glomus cells, and no intracranial involvement is seen.1 Glomuvenous malformations are inherited in over 60% of cases in an autosomal dominant manner and are due to loss-of-function variants in the glomulin gene.7 Verrucous venous malformations (ISSVA simple vascular malformations III) are sporadic congenital hyperkeratotic nodules—mainly on the legs—that show a deep vascular component and no further associations and, in particular, no intracranial involvement.1 Histopathology reveals a compact hyperkeratosis, papillomatosis, and acanthosis overlying dilated vessels extending into the deep dermis and the subcutis.7 A somatic missense MAP3K3 variant has been recently identified.10
Dermatology
A man in him early 50s presented with multiple (approximately 10) bluish, partially keratotic papules and nodules on the lower legs (Figure, A). Since the age of 24 years, the lesions had been growing very slowly in number and size and had been bleeding occasionally after a trauma. The man used a wheelchair due to a right-sided hemiparesis and hemianopsia caused by an intracranial hemorrhage. At the age of 4 years, recurrent seizures had led to the diagnosis of cerebral venous malformations. Clinical examination also demonstrated a lipolymphedema of the lower legs that had been attributed to a functional venous insufficiency with insufficient venous pump activity. The man had been wearing compression stockings for years. The family history was positive for cerebral venous malformations involving the man’s father and aunt, the father having died of an intracranial hemorrhage in his early 70s.A, Clinical picture. B and C, Hematoxylin-eosin stain.Sonography of the skin lesions demonstrated intradermal slow-flow vascular malformations with a maximum diameter of 1.2 cm. A biopsy of a lower-extremity papule was performed (Figure, B and C). Mutational analysis resulted in the detection of a heterozygous pathogenic KRIT1 variant.
what is your diagnosis?
What is your diagnosis?
Blue rubber bleb nevus syndrome
Familial cerebral cavernous malformations
Verrucous venous malformations
Glomuvenous malformations
b
0
1
1
1
male
0
0
52
51-60
null
15
augmented_male_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2809085
A female patient in her early 50s presented with multiple (approximately 10) bluish, partially keratotic papules and nodules on the lower legs (Figure, A). Since the age of 24 years, the lesions had been growing very slowly in number and size and had been bleeding occasionally after a trauma. The patient used a wheelchair due to a right-sided hemiparesis and hemianopsia caused by an intracranial hemorrhage. At the age of 4 years, recurrent seizures had led to the diagnosis of cerebral venous malformations. Clinical examination also demonstrated a lipolymphedema of the lower legs that had been attributed to a functional venous insufficiency with insufficient venous pump activity. The patient had been wearing compression stockings for years. The family history was positive for cerebral venous malformations involving the patient’s father and aunt, the father having died of an intracranial hemorrhage in his early 70s.A, Clinical picture. B and C, Hematoxylin-eosin stain.Sonography of the skin lesions demonstrated intradermal slow-flow vascular malformations with a maximum diameter of 1.2 cm. A biopsy of a lower-extremity papule was performed (Figure, B and C). Mutational analysis resulted in the detection of a heterozygous pathogenic KRIT1 variant. What Is Your Diagnosis?
Blue rubber bleb nevus syndrome
Familial cerebral cavernous malformations
Glomuvenous malformations
Verrucous venous malformations
B. Familial cerebral cavernous malformations
B
Familial cerebral cavernous malformations
Skin biopsy revealed a well-circumscribed lesion with hyperkeratosis and acanthosis of the epidermis overlying a conglomerate of dilated venous-like malformations with continuous smooth muscle in the upper dermis, partly showing thrombosis. Histomorphology together with a positive personal and family history of venous malformations and the identification of a pathologic KRIT1 variant led to the diagnosis of familial cerebral cavernous malformations (CCMs) in the current patient.Cerebral cavernous malformations or cavernomas (International Society for the Study of Vascular Anomalies [ISSVA] simple vascular malformations III) are slow-flowing venous malformations that are lined by a single layer of endothelium with no involvement of brain parenchyma.1 They can occur in sporadic (80%) or familial (20%) form, affecting up to 0.5% in the general population.2 Familial CCMs are frequently inherited as multiple lesions in an autosomal dominant manner. Following the Knudson hypothesis, they are caused by germline loss-of-function variants in KRIT1, CCM2, or PDCD10 in association with a somatic “second hit” in the other allele.3 Among patients with CCMs, 40% to 70% present with neurological symptoms, such as seizures, focal deficits, and headaches due to intracranial hemorrhages. Extraneurological manifestations may involve the skin, retina, kidney, and liver. In a study of 417 patients with familial CMMs, 38 patients (9%) had cutaneous venous malformations, which is a higher rate than in the general population (0.3%).4 In this study, the lesions were associated with KRIT1 variants and further classified as hyperkeratotic capillary-venous malformations, capillary malformations, and venous malformations. Prognosis and management are determined by the location, number, and size of the cerebral venous malformations. Recent advances in understanding CCM pathophysiology have revealed new strategies to control disease progression, ie, Rho kinase inhibition by atorvastatin.5Blue rubber bleb nevus syndrome (BRBNS; ISSVA simple vascular malformations III) is another rare disorder characterized by multiple, most commonly cutaneous and gastrointestinal venous malformations.1 Skin lesions present as rubbery, bluish, easily compressible nodules since birth or early childhood.6 Histopathology reveals large, thin-walled vessels with discontinuous or no smooth muscle separated by variable amounts of fibrous tissue scattered through the dermis.7 Gastrointestinal malformations typically cause recurrent hemorrhages resulting in chronic anemia and may occasionally lead to volvulus, intussusception, or perforation. Thirteen percent of patients have central nervous system involvement.8 Familial occurrence is extremely rare, as BRBNS is due to somatic double (cis) variants in TEK, the gene encoding endothelial cell tyrosine kinase receptor TIE2.9Glomuvenous malformations or glomangiomas (ISSVA simple vascular malformations III) are bluish, tender, noncompressible papules and nodules of the cutis that histologically represent venous-like channels surrounded by 1 or 2 layers of glomus cells, and no intracranial involvement is seen.1 Glomuvenous malformations are inherited in over 60% of cases in an autosomal dominant manner and are due to loss-of-function variants in the glomulin gene.7 Verrucous venous malformations (ISSVA simple vascular malformations III) are sporadic congenital hyperkeratotic nodules—mainly on the legs—that show a deep vascular component and no further associations and, in particular, no intracranial involvement.1 Histopathology reveals a compact hyperkeratosis, papillomatosis, and acanthosis overlying dilated vessels extending into the deep dermis and the subcutis.7 A somatic missense MAP3K3 variant has been recently identified.10
Dermatology
A patient in them early 50s presented with multiple (approximately 10) bluish, partially keratotic papules and nodules on the lower legs (Figure, A). Since the age of 24 years, the lesions had been growing very slowly in number and size and had been bleeding occasionally after a trauma. The patient used a wheelchair due to a right-sided hemiparesis and hemianopsia caused by an intracranial hemorrhage. At the age of 4 years, recurrent seizures had led to the diagnosis of cerebral venous malformations. Clinical examination also demonstrated a lipolymphedema of the lower legs that had been attributed to a functional venous insufficiency with insufficient venous pump activity. The patient had been wearing compression stockings for years. The family history was positive for cerebral venous malformations involving the patient’s father and aunt, the father having died of an intracranial hemorrhage in their early 70s.A, Clinical picture. B and C, Hematoxylin-eosin stain.Sonography of the skin lesions demonstrated intradermal slow-flow vascular malformations with a maximum diameter of 1.2 cm. A biopsy of a lower-extremity papule was performed (Figure, B and C). Mutational analysis resulted in the detection of a heterozygous pathogenic KRIT1 variant.
what is your diagnosis?
What is your diagnosis?
Blue rubber bleb nevus syndrome
Familial cerebral cavernous malformations
Verrucous venous malformations
Glomuvenous malformations
b
0
1
1
1
neutral
0
0
52
51-60
null
15
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2810142
An 81-year-old White woman noted decreased vision in her left eye for 6 months. She was referred to the Ocular Oncology Service, Wills Eye Hospital, Philadelphia, Pennsylvania, for suspected choroidal melanoma. She disclosed a history of macular degeneration in both eyes and retinal detachment in the left eye that was treated 35 years previously. Medical history revealed cutaneous basal cell carcinoma and squamous cell carcinoma, both treated surgically.On examination, visual acuity was 20/50 OD and 20/400 OS. External examination showed posterior-chamber intraocular lenses in both eyes and conjunctival scarring in the left eye from circumferential scleral buckle surgery with no visible extraocular tumor. Fundus evaluation showed macular drusen in the right eye and a pale optic disc with geographic macular atrophy in the left eye, explaining her visual acuity of 20/400 OS. In addition, a shallow circumferential buckle effect and an inferotemporal mass measuring 15.0 × 10.0 mm in basal dimension and 9.1 mm in thickness (Figure, A) were seen in the left eye. The mass appeared elevated, amelanotic with overlying retinal vasculature, and with chorioretinal atrophy. No retinal detachment or breaks were seen. By ultrasonography the mass was echolucent (Figure, A inset). Magnetic resonance imaging (MRI) revealed a T1, T2-hypointense shallow circumferential band in the left eye. In addition, there was an inferotemporal nodular mass in the left eye showing T1 (gadolinium) hypointense, T2-hyperintense features, underlying the encircling buckle, and with adjacent subtle delineation. There was no enhancement with gadolinium (Figure, B).Fundus photograph and magnetic resonance imaging of the left eye. A, Fundus photograph of left eye showed a pale optic disc, geographic macular atrophy, and an inferotemporal amelanotic mass (white arrowhead); inset shows ultrasonography revealing an echolucent mass (white arrowhead). B, Magnetic resonance imaging (MRI) (T2-weighted axial orientation) revealed a shallow hypointense circumferential band in the left eye (pink arrowheads), inferotemporal hyperintense nodular mass (asterisk), and inward compressed sclera (white arrowhead); inset (T1-weighted coronal orientation with gadolinium) shows hypointense encircling band (pink arrowhead), subtle scleral indentation (white arrowhead), and no gadolinium enhancement of mass (asterisk). What Would You Do Next?
Whole-body positron emission tomography scan
Fine-needle aspiration biopsy
Plaque radiotherapy
Observation
Expanded hydrogel scleral sponge
D
Observation
In this case, based on MRI and the lack of gadolinium enhancement, the final diagnosis was expanded hydrogel sponge from previous retinal detachment repair. This mass did not show MRI features of choroidal melanoma. The mass showed complete ultrasonographic acoustic hollowness, potentially suggestive of choroidal melanoma, but MRI confirmed a subtle delineation on the inner portion of the mass (Figure, B) suggestive of thinned sclera, indicating that the mass was episcleral and indenting the globe. The mass was hypointense on T1-weighted (gadolinium-enhanced) imaging suggesting a nonvascular mass and hyperintense on T2-weighted imaging suggesting a hydrophilic mass, possibly a hydrogel sponge. We suspect that the sponge, over time, had slowly expanded underneath the circumferential buckle, leading to inward indentation of the globe and the appearance of an intraocular tumor (Figure).Scleral buckle is one of the highly effective options for retinal detachment repair with success rates ranging from 63% to 99% between various studies.1 The hydrogel scleral buckle was introduced in the 1980s and was deemed safe with low risk of infection when soaked in antibiotics, owing to the hydrophilic nature. In addition, the buckle slowly expanded over time, a presumed beneficial effect to further buckle the retina.2 However, delayed complications were later recognized as this implant continued expansion disproportionately, leading to extreme scleral buckle effect, resulting in diplopia, dysmotility, pseudotumor formation in the orbit, eyelid, conjunctiva, and globe, occasionally with inflammation and pain.2,3Our group has previously described expanding hydrogel (MIRAgel) scleral sponge mimicking orbital cysts and tumors.4 This patient had no symptom of an enlarging orbital mass but was clinically found to have an asymptomatic intraocular mass, suspicious for uveal melanoma. However, the clinical features of the episcleral mass combined with imaging that demonstrated the nonvascular mass overlying markedly thinned sclera, confirmed our suspicion of a hydrogel sponge that presumably expanded over 35 years.Swollen hydrogel implants can be managed by careful surgical excision or close observation.3,4 Despite various techniques described for excision of this discohesive, distended material, complete excision is challenging and risks include continued swelling of remnant implant, retinal detachment recurrence, and thinned scleral wall leading to perforation.3 In the absence of intervention, continued enlargement of the buckle effect can lead to complications; hence, long-term surveillance is advised. In view of this patient’s age and absence of symptoms, observation (option D) was recommended. On subsequent follow-up, if enlargement, extrusion, or any signs of infection are noted, implant removal may be considered. Since the absence of tumor or malignancy was suggested by MRI, there was no need for positron emission tomography scan (option A), biopsy (option B), or radiotherapy (option C).
Ophthalmology
An 81-year-old White woman noted decreased vision in her left eye for 6 months. She was referred to the Ocular Oncology Service, Wills Eye Hospital, Philadelphia, Pennsylvania, for suspected choroidal melanoma. She disclosed a history of macular degeneration in both eyes and retinal detachment in the left eye that was treated 35 years previously. Medical history revealed cutaneous basal cell carcinoma and squamous cell carcinoma, both treated surgically.On examination, visual acuity was 20/50 OD and 20/400 OS. External examination showed posterior-chamber intraocular lenses in both eyes and conjunctival scarring in the left eye from circumferential scleral buckle surgery with no visible extraocular tumor. Fundus evaluation showed macular drusen in the right eye and a pale optic disc with geographic macular atrophy in the left eye, explaining her visual acuity of 20/400 OS. In addition, a shallow circumferential buckle effect and an inferotemporal mass measuring 15.0 × 10.0 mm in basal dimension and 9.1 mm in thickness (Figure, A) were seen in the left eye. The mass appeared elevated, amelanotic with overlying retinal vasculature, and with chorioretinal atrophy. No retinal detachment or breaks were seen. By ultrasonography the mass was echolucent (Figure, A inset). Magnetic resonance imaging (MRI) revealed a T1, T2-hypointense shallow circumferential band in the left eye. In addition, there was an inferotemporal nodular mass in the left eye showing T1 (gadolinium) hypointense, T2-hyperintense features, underlying the encircling buckle, and with adjacent subtle delineation. There was no enhancement with gadolinium (Figure, B).Fundus photograph and magnetic resonance imaging of the left eye. A, Fundus photograph of left eye showed a pale optic disc, geographic macular atrophy, and an inferotemporal amelanotic mass (white arrowhead); inset shows ultrasonography revealing an echolucent mass (white arrowhead). B, Magnetic resonance imaging (MRI) (T2-weighted axial orientation) revealed a shallow hypointense circumferential band in the left eye (pink arrowheads), inferotemporal hyperintense nodular mass (asterisk), and inward compressed sclera (white arrowhead); inset (T1-weighted coronal orientation with gadolinium) shows hypointense encircling band (pink arrowhead), subtle scleral indentation (white arrowhead), and no gadolinium enhancement of mass (asterisk).
what would you do next?
What would you do next?
Plaque radiotherapy
Observation
Whole-body positron emission tomography scan
Fine-needle aspiration biopsy
b
1
1
1
1
female
0
0
81
81-90
White
16
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2810142
An 81-year-old White woman noted decreased vision in her left eye for 6 months. She was referred to the Ocular Oncology Service, Wills Eye Hospital, Philadelphia, Pennsylvania, for suspected choroidal melanoma. She disclosed a history of macular degeneration in both eyes and retinal detachment in the left eye that was treated 35 years previously. Medical history revealed cutaneous basal cell carcinoma and squamous cell carcinoma, both treated surgically.On examination, visual acuity was 20/50 OD and 20/400 OS. External examination showed posterior-chamber intraocular lenses in both eyes and conjunctival scarring in the left eye from circumferential scleral buckle surgery with no visible extraocular tumor. Fundus evaluation showed macular drusen in the right eye and a pale optic disc with geographic macular atrophy in the left eye, explaining her visual acuity of 20/400 OS. In addition, a shallow circumferential buckle effect and an inferotemporal mass measuring 15.0 × 10.0 mm in basal dimension and 9.1 mm in thickness (Figure, A) were seen in the left eye. The mass appeared elevated, amelanotic with overlying retinal vasculature, and with chorioretinal atrophy. No retinal detachment or breaks were seen. By ultrasonography the mass was echolucent (Figure, A inset). Magnetic resonance imaging (MRI) revealed a T1, T2-hypointense shallow circumferential band in the left eye. In addition, there was an inferotemporal nodular mass in the left eye showing T1 (gadolinium) hypointense, T2-hyperintense features, underlying the encircling buckle, and with adjacent subtle delineation. There was no enhancement with gadolinium (Figure, B).Fundus photograph and magnetic resonance imaging of the left eye. A, Fundus photograph of left eye showed a pale optic disc, geographic macular atrophy, and an inferotemporal amelanotic mass (white arrowhead); inset shows ultrasonography revealing an echolucent mass (white arrowhead). B, Magnetic resonance imaging (MRI) (T2-weighted axial orientation) revealed a shallow hypointense circumferential band in the left eye (pink arrowheads), inferotemporal hyperintense nodular mass (asterisk), and inward compressed sclera (white arrowhead); inset (T1-weighted coronal orientation with gadolinium) shows hypointense encircling band (pink arrowhead), subtle scleral indentation (white arrowhead), and no gadolinium enhancement of mass (asterisk). What Would You Do Next?
Whole-body positron emission tomography scan
Fine-needle aspiration biopsy
Plaque radiotherapy
Observation
Expanded hydrogel scleral sponge
D
Observation
In this case, based on MRI and the lack of gadolinium enhancement, the final diagnosis was expanded hydrogel sponge from previous retinal detachment repair. This mass did not show MRI features of choroidal melanoma. The mass showed complete ultrasonographic acoustic hollowness, potentially suggestive of choroidal melanoma, but MRI confirmed a subtle delineation on the inner portion of the mass (Figure, B) suggestive of thinned sclera, indicating that the mass was episcleral and indenting the globe. The mass was hypointense on T1-weighted (gadolinium-enhanced) imaging suggesting a nonvascular mass and hyperintense on T2-weighted imaging suggesting a hydrophilic mass, possibly a hydrogel sponge. We suspect that the sponge, over time, had slowly expanded underneath the circumferential buckle, leading to inward indentation of the globe and the appearance of an intraocular tumor (Figure).Scleral buckle is one of the highly effective options for retinal detachment repair with success rates ranging from 63% to 99% between various studies.1 The hydrogel scleral buckle was introduced in the 1980s and was deemed safe with low risk of infection when soaked in antibiotics, owing to the hydrophilic nature. In addition, the buckle slowly expanded over time, a presumed beneficial effect to further buckle the retina.2 However, delayed complications were later recognized as this implant continued expansion disproportionately, leading to extreme scleral buckle effect, resulting in diplopia, dysmotility, pseudotumor formation in the orbit, eyelid, conjunctiva, and globe, occasionally with inflammation and pain.2,3Our group has previously described expanding hydrogel (MIRAgel) scleral sponge mimicking orbital cysts and tumors.4 This patient had no symptom of an enlarging orbital mass but was clinically found to have an asymptomatic intraocular mass, suspicious for uveal melanoma. However, the clinical features of the episcleral mass combined with imaging that demonstrated the nonvascular mass overlying markedly thinned sclera, confirmed our suspicion of a hydrogel sponge that presumably expanded over 35 years.Swollen hydrogel implants can be managed by careful surgical excision or close observation.3,4 Despite various techniques described for excision of this discohesive, distended material, complete excision is challenging and risks include continued swelling of remnant implant, retinal detachment recurrence, and thinned scleral wall leading to perforation.3 In the absence of intervention, continued enlargement of the buckle effect can lead to complications; hence, long-term surveillance is advised. In view of this patient’s age and absence of symptoms, observation (option D) was recommended. On subsequent follow-up, if enlargement, extrusion, or any signs of infection are noted, implant removal may be considered. Since the absence of tumor or malignancy was suggested by MRI, there was no need for positron emission tomography scan (option A), biopsy (option B), or radiotherapy (option C).
Ophthalmology
An 81-year-old White man noted decreased vision in him left eye for 6 months. He was referred to the Ocular Oncology Service, Wills Eye Hospital, Philadelphia, Pennsylvania, for suspected choroidal melanoma. He disclosed a history of macular degeneration in both eyes and retinal detachment in the left eye that was treated 35 years previously. Medical history revealed cutaneous basal cell carcinoma and squamous cell carcinoma, both treated surgically.On examination, visual acuity was 20/50 OD and 20/400 OS. External examination showed posterior-chamber intraocular lenses in both eyes and conjunctival scarring in the left eye from circumferential scleral buckle surgery with no visible extraocular tumor. Fundus evaluation showed macular drusen in the right eye and a pale optic disc with geographic macular atrophy in the left eye, explaining him visual acuity of 20/400 OS. In addition, a shallow circumferential buckle effect and an inferotemporal mass measuring 15.0 × 10.0 mm in basal dimension and 9.1 mm in thickness (Figure, A) were seen in the left eye. The mass appeared elevated, amelanotic with overlying retinal vasculature, and with chorioretinal atrophy. No retinal detachment or breaks were seen. By ultrasonography the mass was echolucent (Figure, A inset). Magnetic resonance imaging (MRI) revealed a T1, T2-hypointense shallow circumferential band in the left eye. In addition, there was an inferotemporal nodular mass in the left eye showing T1 (gadolinium) hypointense, T2-hyperintense features, underlying the encircling buckle, and with adjacent subtle delineation. There was no enhancement with gadolinium (Figure, B).Fundus photograph and magnetic resonance imaging of the left eye. A, Fundus photograph of left eye showed a pale optic disc, geographic macular atrophy, and an inferotemporal amelanotic mass (white arrowhead); inset shows ultrasonography revealing an echolucent mass (white arrowhead). B, Magnetic resonance imaging (MRI) (T2-weighted axial orientation) revealed a shallow hypointense circumferential band in the left eye (pink arrowheads), inferotemporal hyperintense nodular mass (asterisk), and inward compressed sclera (white arrowhead); inset (T1-weighted coronal orientation with gadolinium) shows hypointense encircling band (pink arrowhead), subtle scleral indentation (white arrowhead), and no gadolinium enhancement of mass (asterisk).
what would you do next?
What would you do next?
Plaque radiotherapy
Observation
Whole-body positron emission tomography scan
Fine-needle aspiration biopsy
b
1
1
1
1
male
0
0
81
81-90
White
16
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2810142
An 81-year-old White woman noted decreased vision in her left eye for 6 months. She was referred to the Ocular Oncology Service, Wills Eye Hospital, Philadelphia, Pennsylvania, for suspected choroidal melanoma. She disclosed a history of macular degeneration in both eyes and retinal detachment in the left eye that was treated 35 years previously. Medical history revealed cutaneous basal cell carcinoma and squamous cell carcinoma, both treated surgically.On examination, visual acuity was 20/50 OD and 20/400 OS. External examination showed posterior-chamber intraocular lenses in both eyes and conjunctival scarring in the left eye from circumferential scleral buckle surgery with no visible extraocular tumor. Fundus evaluation showed macular drusen in the right eye and a pale optic disc with geographic macular atrophy in the left eye, explaining her visual acuity of 20/400 OS. In addition, a shallow circumferential buckle effect and an inferotemporal mass measuring 15.0 × 10.0 mm in basal dimension and 9.1 mm in thickness (Figure, A) were seen in the left eye. The mass appeared elevated, amelanotic with overlying retinal vasculature, and with chorioretinal atrophy. No retinal detachment or breaks were seen. By ultrasonography the mass was echolucent (Figure, A inset). Magnetic resonance imaging (MRI) revealed a T1, T2-hypointense shallow circumferential band in the left eye. In addition, there was an inferotemporal nodular mass in the left eye showing T1 (gadolinium) hypointense, T2-hyperintense features, underlying the encircling buckle, and with adjacent subtle delineation. There was no enhancement with gadolinium (Figure, B).Fundus photograph and magnetic resonance imaging of the left eye. A, Fundus photograph of left eye showed a pale optic disc, geographic macular atrophy, and an inferotemporal amelanotic mass (white arrowhead); inset shows ultrasonography revealing an echolucent mass (white arrowhead). B, Magnetic resonance imaging (MRI) (T2-weighted axial orientation) revealed a shallow hypointense circumferential band in the left eye (pink arrowheads), inferotemporal hyperintense nodular mass (asterisk), and inward compressed sclera (white arrowhead); inset (T1-weighted coronal orientation with gadolinium) shows hypointense encircling band (pink arrowhead), subtle scleral indentation (white arrowhead), and no gadolinium enhancement of mass (asterisk). What Would You Do Next?
Whole-body positron emission tomography scan
Fine-needle aspiration biopsy
Plaque radiotherapy
Observation
Expanded hydrogel scleral sponge
D
Observation
In this case, based on MRI and the lack of gadolinium enhancement, the final diagnosis was expanded hydrogel sponge from previous retinal detachment repair. This mass did not show MRI features of choroidal melanoma. The mass showed complete ultrasonographic acoustic hollowness, potentially suggestive of choroidal melanoma, but MRI confirmed a subtle delineation on the inner portion of the mass (Figure, B) suggestive of thinned sclera, indicating that the mass was episcleral and indenting the globe. The mass was hypointense on T1-weighted (gadolinium-enhanced) imaging suggesting a nonvascular mass and hyperintense on T2-weighted imaging suggesting a hydrophilic mass, possibly a hydrogel sponge. We suspect that the sponge, over time, had slowly expanded underneath the circumferential buckle, leading to inward indentation of the globe and the appearance of an intraocular tumor (Figure).Scleral buckle is one of the highly effective options for retinal detachment repair with success rates ranging from 63% to 99% between various studies.1 The hydrogel scleral buckle was introduced in the 1980s and was deemed safe with low risk of infection when soaked in antibiotics, owing to the hydrophilic nature. In addition, the buckle slowly expanded over time, a presumed beneficial effect to further buckle the retina.2 However, delayed complications were later recognized as this implant continued expansion disproportionately, leading to extreme scleral buckle effect, resulting in diplopia, dysmotility, pseudotumor formation in the orbit, eyelid, conjunctiva, and globe, occasionally with inflammation and pain.2,3Our group has previously described expanding hydrogel (MIRAgel) scleral sponge mimicking orbital cysts and tumors.4 This patient had no symptom of an enlarging orbital mass but was clinically found to have an asymptomatic intraocular mass, suspicious for uveal melanoma. However, the clinical features of the episcleral mass combined with imaging that demonstrated the nonvascular mass overlying markedly thinned sclera, confirmed our suspicion of a hydrogel sponge that presumably expanded over 35 years.Swollen hydrogel implants can be managed by careful surgical excision or close observation.3,4 Despite various techniques described for excision of this discohesive, distended material, complete excision is challenging and risks include continued swelling of remnant implant, retinal detachment recurrence, and thinned scleral wall leading to perforation.3 In the absence of intervention, continued enlargement of the buckle effect can lead to complications; hence, long-term surveillance is advised. In view of this patient’s age and absence of symptoms, observation (option D) was recommended. On subsequent follow-up, if enlargement, extrusion, or any signs of infection are noted, implant removal may be considered. Since the absence of tumor or malignancy was suggested by MRI, there was no need for positron emission tomography scan (option A), biopsy (option B), or radiotherapy (option C).
Ophthalmology
An 81-year-old White patient noted decreased vision in them left eye for 6 months. They was referred to the Ocular Oncology Service, Wills Eye Hospital, Philadelphia, Pennsylvania, for suspected choroidal melanoma. They disclosed a history of macular degeneration in both eyes and retinal detachment in the left eye that was treated 35 years previously. Medical history revealed cutaneous basal cell carcinoma and squamous cell carcinoma, both treated surgically.On examination, visual acuity was 20/50 OD and 20/400 OS. External examination showed posterior-chamber intraocular lenses in both eyes and conjunctival scarring in the left eye from circumferential scleral buckle surgery with no visible extraocular tumor. Fundus evaluation showed macular drusen in the right eye and a pale optic disc with geographic macular atrophy in the left eye, explaining them visual acuity of 20/400 OS. In addition, a shallow circumferential buckle effect and an inferotemporal mass measuring 15.0 × 10.0 mm in basal dimension and 9.1 mm in thickness (Figure, A) were seen in the left eye. The mass appeared elevated, amelanotic with overlying retinal vasculature, and with chorioretinal atrophy. No retinal detachment or breaks were seen. By ultrasonography the mass was echolucent (Figure, A inset). Magnetic resonance imaging (MRI) revealed a T1, T2-hypointense shallow circumferential band in the left eye. In addition, there was an inferotemporal nodular mass in the left eye showing T1 (gadolinium) hypointense, T2-hyperintense features, underlying the encircling buckle, and with adjacent subtle delineation. There was no enhancement with gadolinium (Figure, B).Fundus photograph and magnetic resonance imaging of the left eye. A, Fundus photograph of left eye showed a pale optic disc, geographic macular atrophy, and an inferotemporal amelanotic mass (white arrowhead); inset shows ultrasonography revealing an echolucent mass (white arrowhead). B, Magnetic resonance imaging (MRI) (T2-weighted axial orientation) revealed a shallow hypointense circumferential band in the left eye (pink arrowheads), inferotemporal hyperintense nodular mass (asterisk), and inward compressed sclera (white arrowhead); inset (T1-weighted coronal orientation with gadolinium) shows hypointense encircling band (pink arrowhead), subtle scleral indentation (white arrowhead), and no gadolinium enhancement of mass (asterisk).
what would you do next?
What would you do next?
Plaque radiotherapy
Observation
Whole-body positron emission tomography scan
Fine-needle aspiration biopsy
b
1
1
1
1
neutral
0
0
81
81-90
White
16
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2809657
A woman in her 30s presented with progressive, multiple itchy erythematous to violaceous papules and plaques localized to the left shin for more than 10 years (Figure, A and B). On close inspection, milia and a few erosions with partially detached epidermis were identified. There were no abnormalities of the mucous membranes, nails, hair, or teeth. No extracutaneous involvement was observed.A, Multiple pruritic erythematous to violaceous lichenified papules and plaques on the left shin. B, Close-up of the skin lesions showing an erosion with partially detached epidermis and multiple milia. C, Histopathologic examination revealed dermoepidermal separation (hematoxylin-eosin stain). D, Histopathologic examination revealed several milia (hematoxylin-eosin stain). What Is Your Diagnosis?
Pretibial pruritic papular dermatitis (PPPD)
Hypertrophic lichen planus (HLP)
Epidermolysis bullosa pruriginosa (EBP)
Lichen amyloidosis (LA)
C. Epidermolysis bullosa pruriginosa (EBP)
C
Epidermolysis bullosa pruriginosa (EBP)
We performed a skin biopsy under the impression of prurigo nodularis and to exclude hypertrophic lichen planus. Histopathologic examination revealed a dermoepidermal cleft (Figure, C) and several milia (Figure, D) in the superficial dermis, features that were inconsistent with either PN or HLP but suggestive of epidermolysis bullosa (EB). Whole-exome sequencing using genomic DNA extracted from the patient’s peripheral blood mononuclear cells identified a novel heterozygous missense variant, c.6832G>T, p.Gly2278Trp, in COL7A1 (NM_000094), which was graded as “likely pathogenic” based on American College of Medical Genetics and Genomics guidelines. Further inquiry of the patient led to identification of 2 other affected family members: 1 who had typical EBP (intensely itchy, symmetric, hypertrophic papules and plaques on both shins) and nail dystrophy, and another, who only had dystrophic toenails. Sanger sequencing confirmed cosegregation of this variant with variable phenotypes, supporting its pathogenicity and the patient’s diagnosis of autosomal dominant EBP. The patient had a 2-year old relative who had only very mild nail dystrophy and no skin disease who also harbored this pathogenic variant.Epidermolysis bullosa is an inherited, heterogeneous group of rare genetic dermatoses characterized by mucocutaneous fragility, blister formation, and abnormal wound healing.1 It is categorized into 4 major subtypes, EB simplex, junctional EB, dystrophic EB (DEB), and Kindler EB, based on the ultrastructural level of skin cleavage and inherent molecular pathology.2 Dystrophic EB is caused by pathogenic variants in the COL7A1 gene, which encodes type 7 collagen. Epidermolysis BP is a rare subtype of DEB characterized by intensely pruritic, prurigo-like nodules and plaques affecting the lower legs, thighs, and arms, often associated with nail dystrophy.3 Unlike other forms of EB, except for some localized subtypes of EB simplex and some rare late-onset forms of junctional EB, it is not uncommon for patients with EBP to present beyond the neonatal/childhood period, and sometimes well into adult life.Epidermolysis BP is traditionally considered a rare subtype of DEB, but it seems to be more prevalent in some countries, including Taiwan.4 So far, no clear genotype-phenotype correlation has been established in EBP.5 Epidermolysis BP is also often misdiagnosed as other dermatoses because blisters are often inconspicuous. Diagnostic clues of EBP include nail dystrophy, the most common associated feature of EBP,6 as well as a history of blistering during infancy and family medical history of other subtypes of DEB,1 which makes it distinct from factitial dermatitis. Although the histopathologic features of factitial dermatitis are nonspecific, milia are usually absent. In this case, however, the unilateral, localized presentation of EBP and undisclosed family medical history of EB at the patient’s initial visit made the diagnosis difficult. It was the milia and dermoepidermal separation on histopathologic findings that prompted subsequent genetic investigations. Still, had we been more vigilant, we might have noticed the milia and partially detached epidermis and put DEB in our initial differential diagnoses.The present case needs to be distinguished from PPPD, HLP, and LA. Overall, PPPD is characterized by extremely pruritic, discrete, smooth, flesh-colored to erythematous papules, caused by persistent rubbing of the anterior surface of the legs. Histopathologic features include minimal compact orthokeratosis, mild acanthosis with flattening of the rete ridges, and superficial and middermal perivascular lymphohistiocytic infiltrates with a variable number of eosinophils.7 In most cases, there is evidence of superficial dermal fibrosis, characterized by the presence of multinucleated fibroblasts and thickened collagen bundles arranged in a random pattern, without the dermoepidermal cleft and milia seen in EBP. Hypertrophic lichen planus presents as extremely pruritic, violaceous, hyperkeratotic flat-topped papules and plaques affecting the extremities.8 Histopathologic findings of HLP show compact orthokeratosis, acanthosis, wedge-shaped hypergranulosis, saw-tooth appearance of the undersurface of the epidermis with necrotic keratinocytes, and a band-like infiltrate of lymphocytes at the dermoepidermal junction, which is distinct from EBP. Generally, LA is the most common form of primary localized cutaneous amyloidosis, clinically characterized by pruritic, discrete hyperkeratotic hyperpigmented papules with a predilection for the shins.9 The key component of the histologic findings is the deposition of pink amorphous amyloid material in the papillary dermis, which exhibits a bright apple-green birefringence under polarized light when stained with Congo red. These findings distinguish LA from EBP, despite both conditions presenting similar epidermal changes induced by chronic scratching.10To our knowledge, this is the first case of unilateral EBP ever reported. This case highlights the importance of considering EBP even when the prurigo-like lesions are unilateral and without nail involvement. Milia and erosions provide diagnostic clues for such atypical cases, as in other subtypes of DEB.
Dermatology
A woman in her 30s presented with progressive, multiple itchy erythematous to violaceous papules and plaques localized to the left shin for more than 10 years (Figure, A and B). On close inspection, milia and a few erosions with partially detached epidermis were identified. There were no abnormalities of the mucous membranes, nails, hair, or teeth. No extracutaneous involvement was observed.A, Multiple pruritic erythematous to violaceous lichenified papules and plaques on the left shin. B, Close-up of the skin lesions showing an erosion with partially detached epidermis and multiple milia. C, Histopathologic examination revealed dermoepidermal separation (hematoxylin-eosin stain). D, Histopathologic examination revealed several milia (hematoxylin-eosin stain).
what is your diagnosis?
What is your diagnosis?
Pretibial pruritic papular dermatitis (PPPD)
Lichen amyloidosis (LA)
Hypertrophic lichen planus (HLP)
Epidermolysis bullosa pruriginosa (EBP)
d
0
0
0
1
female
0
0
35
31-40
null
17
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2809657
A woman in her 30s presented with progressive, multiple itchy erythematous to violaceous papules and plaques localized to the left shin for more than 10 years (Figure, A and B). On close inspection, milia and a few erosions with partially detached epidermis were identified. There were no abnormalities of the mucous membranes, nails, hair, or teeth. No extracutaneous involvement was observed.A, Multiple pruritic erythematous to violaceous lichenified papules and plaques on the left shin. B, Close-up of the skin lesions showing an erosion with partially detached epidermis and multiple milia. C, Histopathologic examination revealed dermoepidermal separation (hematoxylin-eosin stain). D, Histopathologic examination revealed several milia (hematoxylin-eosin stain). What Is Your Diagnosis?
Pretibial pruritic papular dermatitis (PPPD)
Hypertrophic lichen planus (HLP)
Epidermolysis bullosa pruriginosa (EBP)
Lichen amyloidosis (LA)
C. Epidermolysis bullosa pruriginosa (EBP)
C
Epidermolysis bullosa pruriginosa (EBP)
We performed a skin biopsy under the impression of prurigo nodularis and to exclude hypertrophic lichen planus. Histopathologic examination revealed a dermoepidermal cleft (Figure, C) and several milia (Figure, D) in the superficial dermis, features that were inconsistent with either PN or HLP but suggestive of epidermolysis bullosa (EB). Whole-exome sequencing using genomic DNA extracted from the patient’s peripheral blood mononuclear cells identified a novel heterozygous missense variant, c.6832G>T, p.Gly2278Trp, in COL7A1 (NM_000094), which was graded as “likely pathogenic” based on American College of Medical Genetics and Genomics guidelines. Further inquiry of the patient led to identification of 2 other affected family members: 1 who had typical EBP (intensely itchy, symmetric, hypertrophic papules and plaques on both shins) and nail dystrophy, and another, who only had dystrophic toenails. Sanger sequencing confirmed cosegregation of this variant with variable phenotypes, supporting its pathogenicity and the patient’s diagnosis of autosomal dominant EBP. The patient had a 2-year old relative who had only very mild nail dystrophy and no skin disease who also harbored this pathogenic variant.Epidermolysis bullosa is an inherited, heterogeneous group of rare genetic dermatoses characterized by mucocutaneous fragility, blister formation, and abnormal wound healing.1 It is categorized into 4 major subtypes, EB simplex, junctional EB, dystrophic EB (DEB), and Kindler EB, based on the ultrastructural level of skin cleavage and inherent molecular pathology.2 Dystrophic EB is caused by pathogenic variants in the COL7A1 gene, which encodes type 7 collagen. Epidermolysis BP is a rare subtype of DEB characterized by intensely pruritic, prurigo-like nodules and plaques affecting the lower legs, thighs, and arms, often associated with nail dystrophy.3 Unlike other forms of EB, except for some localized subtypes of EB simplex and some rare late-onset forms of junctional EB, it is not uncommon for patients with EBP to present beyond the neonatal/childhood period, and sometimes well into adult life.Epidermolysis BP is traditionally considered a rare subtype of DEB, but it seems to be more prevalent in some countries, including Taiwan.4 So far, no clear genotype-phenotype correlation has been established in EBP.5 Epidermolysis BP is also often misdiagnosed as other dermatoses because blisters are often inconspicuous. Diagnostic clues of EBP include nail dystrophy, the most common associated feature of EBP,6 as well as a history of blistering during infancy and family medical history of other subtypes of DEB,1 which makes it distinct from factitial dermatitis. Although the histopathologic features of factitial dermatitis are nonspecific, milia are usually absent. In this case, however, the unilateral, localized presentation of EBP and undisclosed family medical history of EB at the patient’s initial visit made the diagnosis difficult. It was the milia and dermoepidermal separation on histopathologic findings that prompted subsequent genetic investigations. Still, had we been more vigilant, we might have noticed the milia and partially detached epidermis and put DEB in our initial differential diagnoses.The present case needs to be distinguished from PPPD, HLP, and LA. Overall, PPPD is characterized by extremely pruritic, discrete, smooth, flesh-colored to erythematous papules, caused by persistent rubbing of the anterior surface of the legs. Histopathologic features include minimal compact orthokeratosis, mild acanthosis with flattening of the rete ridges, and superficial and middermal perivascular lymphohistiocytic infiltrates with a variable number of eosinophils.7 In most cases, there is evidence of superficial dermal fibrosis, characterized by the presence of multinucleated fibroblasts and thickened collagen bundles arranged in a random pattern, without the dermoepidermal cleft and milia seen in EBP. Hypertrophic lichen planus presents as extremely pruritic, violaceous, hyperkeratotic flat-topped papules and plaques affecting the extremities.8 Histopathologic findings of HLP show compact orthokeratosis, acanthosis, wedge-shaped hypergranulosis, saw-tooth appearance of the undersurface of the epidermis with necrotic keratinocytes, and a band-like infiltrate of lymphocytes at the dermoepidermal junction, which is distinct from EBP. Generally, LA is the most common form of primary localized cutaneous amyloidosis, clinically characterized by pruritic, discrete hyperkeratotic hyperpigmented papules with a predilection for the shins.9 The key component of the histologic findings is the deposition of pink amorphous amyloid material in the papillary dermis, which exhibits a bright apple-green birefringence under polarized light when stained with Congo red. These findings distinguish LA from EBP, despite both conditions presenting similar epidermal changes induced by chronic scratching.10To our knowledge, this is the first case of unilateral EBP ever reported. This case highlights the importance of considering EBP even when the prurigo-like lesions are unilateral and without nail involvement. Milia and erosions provide diagnostic clues for such atypical cases, as in other subtypes of DEB.
Dermatology
A man in him 30s presented with progressive, multiple itchy erythematous to violaceous papules and plaques localized to the left shin for more than 10 years (Figure, A and B). On close inspection, milia and a few erosions with partially detached epidermis were identified. There were no abnormalities of the mucous membranes, nails, hair, or teeth. No extracutaneous involvement was observed.A, Multiple pruritic erythematous to violaceous lichenified papules and plaques on the left shin. B, Close-up of the skin lesions showing an erosion with partially detached epidermis and multiple milia. C, Histopathologic examination revealed dermoepidermal separation (hematoxylin-eosin stain). D, Histopathologic examination revealed several milia (hematoxylin-eosin stain).
what is your diagnosis?
What is your diagnosis?
Pretibial pruritic papular dermatitis (PPPD)
Lichen amyloidosis (LA)
Hypertrophic lichen planus (HLP)
Epidermolysis bullosa pruriginosa (EBP)
d
0
0
0
1
male
0
0
35
31-40
null
17
augmented_male_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2809657
A woman in her 30s presented with progressive, multiple itchy erythematous to violaceous papules and plaques localized to the left shin for more than 10 years (Figure, A and B). On close inspection, milia and a few erosions with partially detached epidermis were identified. There were no abnormalities of the mucous membranes, nails, hair, or teeth. No extracutaneous involvement was observed.A, Multiple pruritic erythematous to violaceous lichenified papules and plaques on the left shin. B, Close-up of the skin lesions showing an erosion with partially detached epidermis and multiple milia. C, Histopathologic examination revealed dermoepidermal separation (hematoxylin-eosin stain). D, Histopathologic examination revealed several milia (hematoxylin-eosin stain). What Is Your Diagnosis?
Pretibial pruritic papular dermatitis (PPPD)
Hypertrophic lichen planus (HLP)
Epidermolysis bullosa pruriginosa (EBP)
Lichen amyloidosis (LA)
C. Epidermolysis bullosa pruriginosa (EBP)
C
Epidermolysis bullosa pruriginosa (EBP)
We performed a skin biopsy under the impression of prurigo nodularis and to exclude hypertrophic lichen planus. Histopathologic examination revealed a dermoepidermal cleft (Figure, C) and several milia (Figure, D) in the superficial dermis, features that were inconsistent with either PN or HLP but suggestive of epidermolysis bullosa (EB). Whole-exome sequencing using genomic DNA extracted from the patient’s peripheral blood mononuclear cells identified a novel heterozygous missense variant, c.6832G>T, p.Gly2278Trp, in COL7A1 (NM_000094), which was graded as “likely pathogenic” based on American College of Medical Genetics and Genomics guidelines. Further inquiry of the patient led to identification of 2 other affected family members: 1 who had typical EBP (intensely itchy, symmetric, hypertrophic papules and plaques on both shins) and nail dystrophy, and another, who only had dystrophic toenails. Sanger sequencing confirmed cosegregation of this variant with variable phenotypes, supporting its pathogenicity and the patient’s diagnosis of autosomal dominant EBP. The patient had a 2-year old relative who had only very mild nail dystrophy and no skin disease who also harbored this pathogenic variant.Epidermolysis bullosa is an inherited, heterogeneous group of rare genetic dermatoses characterized by mucocutaneous fragility, blister formation, and abnormal wound healing.1 It is categorized into 4 major subtypes, EB simplex, junctional EB, dystrophic EB (DEB), and Kindler EB, based on the ultrastructural level of skin cleavage and inherent molecular pathology.2 Dystrophic EB is caused by pathogenic variants in the COL7A1 gene, which encodes type 7 collagen. Epidermolysis BP is a rare subtype of DEB characterized by intensely pruritic, prurigo-like nodules and plaques affecting the lower legs, thighs, and arms, often associated with nail dystrophy.3 Unlike other forms of EB, except for some localized subtypes of EB simplex and some rare late-onset forms of junctional EB, it is not uncommon for patients with EBP to present beyond the neonatal/childhood period, and sometimes well into adult life.Epidermolysis BP is traditionally considered a rare subtype of DEB, but it seems to be more prevalent in some countries, including Taiwan.4 So far, no clear genotype-phenotype correlation has been established in EBP.5 Epidermolysis BP is also often misdiagnosed as other dermatoses because blisters are often inconspicuous. Diagnostic clues of EBP include nail dystrophy, the most common associated feature of EBP,6 as well as a history of blistering during infancy and family medical history of other subtypes of DEB,1 which makes it distinct from factitial dermatitis. Although the histopathologic features of factitial dermatitis are nonspecific, milia are usually absent. In this case, however, the unilateral, localized presentation of EBP and undisclosed family medical history of EB at the patient’s initial visit made the diagnosis difficult. It was the milia and dermoepidermal separation on histopathologic findings that prompted subsequent genetic investigations. Still, had we been more vigilant, we might have noticed the milia and partially detached epidermis and put DEB in our initial differential diagnoses.The present case needs to be distinguished from PPPD, HLP, and LA. Overall, PPPD is characterized by extremely pruritic, discrete, smooth, flesh-colored to erythematous papules, caused by persistent rubbing of the anterior surface of the legs. Histopathologic features include minimal compact orthokeratosis, mild acanthosis with flattening of the rete ridges, and superficial and middermal perivascular lymphohistiocytic infiltrates with a variable number of eosinophils.7 In most cases, there is evidence of superficial dermal fibrosis, characterized by the presence of multinucleated fibroblasts and thickened collagen bundles arranged in a random pattern, without the dermoepidermal cleft and milia seen in EBP. Hypertrophic lichen planus presents as extremely pruritic, violaceous, hyperkeratotic flat-topped papules and plaques affecting the extremities.8 Histopathologic findings of HLP show compact orthokeratosis, acanthosis, wedge-shaped hypergranulosis, saw-tooth appearance of the undersurface of the epidermis with necrotic keratinocytes, and a band-like infiltrate of lymphocytes at the dermoepidermal junction, which is distinct from EBP. Generally, LA is the most common form of primary localized cutaneous amyloidosis, clinically characterized by pruritic, discrete hyperkeratotic hyperpigmented papules with a predilection for the shins.9 The key component of the histologic findings is the deposition of pink amorphous amyloid material in the papillary dermis, which exhibits a bright apple-green birefringence under polarized light when stained with Congo red. These findings distinguish LA from EBP, despite both conditions presenting similar epidermal changes induced by chronic scratching.10To our knowledge, this is the first case of unilateral EBP ever reported. This case highlights the importance of considering EBP even when the prurigo-like lesions are unilateral and without nail involvement. Milia and erosions provide diagnostic clues for such atypical cases, as in other subtypes of DEB.
Dermatology
A patient in them 30s presented with progressive, multiple itchy erythematous to violaceous papules and plaques localized to the left shin for more than 10 years (Figure, A and B). On close inspection, milia and a few erosions with partially detached epidermis were identified. There were no abnormalities of the mucous membranes, nails, hair, or teeth. No extracutaneous involvement was observed.A, Multiple pruritic erythematous to violaceous lichenified papules and plaques on the left shin. B, Close-up of the skin lesions showing an erosion with partially detached epidermis and multiple milia. C, Histopathologic examination revealed dermoepidermal separation (hematoxylin-eosin stain). D, Histopathologic examination revealed several milia (hematoxylin-eosin stain).
what is your diagnosis?
What is your diagnosis?
Pretibial pruritic papular dermatitis (PPPD)
Lichen amyloidosis (LA)
Hypertrophic lichen planus (HLP)
Epidermolysis bullosa pruriginosa (EBP)
d
0
0
0
1
neutral
0
0
35
31-40
null
17
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809652
A 13-year-old girl with a 7-day history of painless vision loss and central scotoma in her left eye was referred to the department of ophthalmology. Three weeks prior, she had presented with mild fever and flulike symptoms, associated with a severe frontal headache that was mildly relieved by analgesics. Her medical history was positive for relapsing urinary tract infections since early infancy without adequate follow-up. Findings of an ophthalmological examination 1 year prior were reported as normal.At initial examination, she was conscious but somewhat lethargic. Her best-corrected visual acuity was 20/20 OD and 20/80 OS. Pupillary light responses, extrinsic ocular motility, anterior segment biomicroscopy, and intraocular pressure were normal in both eyes. Dilated fundus examination revealed a bilateral sectorial macular star that was more extended in the left eye, with retinal veins slightly dilated and tortuous, attenuated arterioles, some juxta and peripapillary nerve fiber layer infarcts, and optic disc edema with marked papillary telangiectasia; in addition, some faint, small, tan-yellow dots were observed at the level of the retinal pigment epithelium in the posterior pole, some of them already grayish and with a hypopigmented halo (Figure 1). Optical coherence tomography displayed elevated optic discs and hyperreflective material in the outer plexiform layer temporal to the optic disc in both eyes and a macular neurosensory detachment in the left eye. Results of visual field testing were normal in the right eye and showed a central scotoma in the left eye.Fundus image of the right eye (A) and left eye (B) showing slight dilation of retinal veins (magenta arrowheads), intraretinal lipid deposits in the papillomacular bundle and the inferonasal macular quadrant forming an incomplete macular star (yellow arrowheads), optic disc edema and epipapillary telangiectasia (blue arrowheads), nerve fiber layer infarcts or cotton-wool nodules (green arrowheads) over the inferior margin of the optic disc in the right eye and juxta and parapapillary in the left eye, and paramacular temporal light-pigmented subretinal dots at the level of the retinal pigment epithelium (white arrowheads) in both eyes, with some already scarred (pigmented dot surrounded by a pale halo; gold arrowhead). What Would You Do Next?
Prescribe systemic corticosteroids
Order serologic testing for infectious neuroretinitis
Rule out brain tumor
Check blood pressure
Hypertensive retinopathy
D
Check blood pressure
Option D is the correct approach. The child underwent immediate hospitalization and systemic evaluation. Clinical examination revealed severe systemic hypertension (225/160 mm Hg). Laboratory test results showed leukocytosis, elevated erythrocyte sedimentation rate (35 mm), hypercholesterolemia, hypokalemia, proteinuria, and microalbuminuria.Echocardiography showed left ventricle hypertrophy, and kidney doppler ultrasonography revealed advanced bilateral kidney atrophy with parenchymal scarring that was presumed to be the consequence of undertreated relapsing urinary tract infections. Results of magnetic resonance imaging of the brain and orbits were normal.The most common masquerader for neuroretinitis is malignant hypertension.1 Systemic hypertension in children is rare, potentially underrecognized,2 and its prevalence ranges from 2% to 5%.3 Mostly asymptomatic, pediatric hypertension may manifest by symptoms originating from target organ disease, and most cases present secondary hypertension from kidney disorders, especially kidney parenchymal disease.4 Hypertensive retinopathy occurs in 8% to 18% of children with severe hypertension, figures substantially lower than those observed in adults with hypertension, with the highest prevalence in those with kidney and renovascular disease.5Malignant hypertension is an acute, severe rise of more than 180 mm Hg in systolic pressure and/or more than 120 mm Hg in diastolic pressure, associated with severe hypertensive retinopathy and papilledema. It constitutes a medical emergency, where severe acute increase in blood pressure results in end-organ damage.6 The bilateral retinal disease, the presence of nerve fiber layer infarcts and Elschnig spots (focal signs of choroidal ischemia at the level of the retinal pigment epithelium), and the accompanying neurologic symptoms (severe headache and lethargy) suggested a severe systemic disease and were the presenting signs of malignant hypertension, which is a life-threatening condition in this patient secondary to kidney parenchymal disease from undertreated relapsing urinary tract infections since early infancy.Option A is incorrect. It is not advisable to prescribe corticosteroids before ruling out an infectious condition. Also, corticosteroids are contraindicated in the context of malignant hypertension. For option B, in the presence of the triad unilateral vision loss, macular star, and optic disc swelling in a child, infectious neuroretinitis must be ruled out, with cat scratch disease (CSD) its most frequent cause. However, neuroretinitis is a rare complication of cat scratch disease, occurring in only 1% to 2% of cases, and it generally presents unilaterally and self-limited.1 In this patient, results of serum tests for infective causes were negative. Besides, retinal and optic nerve disease were bilateral, and the child presented with neurologic signs. Option C is incorrect. Although this patient had neurologic symptoms with bilateral optic disc edema, other features, like macular lipid deposits, nerve fiber layer infarcts, and Elschnig spots, are not clinical signs that accompany papilledema from intracranial hypertension.Treatment with amlodipine, 5 mg, every 12 hours, daily losartan, 50 mg, and a hyposodic diet reduced blood pressure drastically and slowly normalized the retinal disease. Visual acuity progressively improved in the left eye, as did the macular star, optic disc edema, and nerve fiber layer infarcts in both eyes. One year later, blood pressure was generally within normal values under treatment but with intermittent episodes of high values. Best-corrected visual acuity was 20/20 OD and 20/25 OS, and optic discs were nonedematous but moderately pale in both eyes (Figure 2). Optical coherence tomography performed during follow-up showed a diffuse thinning of the neurosensory retina and the peripapillary nerve fiber layer.Fundus image of the left eye (panel) and the right eye (insert) at 1-year follow-up showing a pronounced sclerotic appearance of arteries, a complete resolution of both optic discs’ edema and the macular stars, a moderate optic disc pallor, and no evidence of nerve fiber layer infarcts.
Ophthalmology
A 13-year-old girl with a 7-day history of painless vision loss and central scotoma in her left eye was referred to the department of ophthalmology. Three weeks prior, she had presented with mild fever and flulike symptoms, associated with a severe frontal headache that was mildly relieved by analgesics. Her medical history was positive for relapsing urinary tract infections since early infancy without adequate follow-up. Findings of an ophthalmological examination 1 year prior were reported as normal.At initial examination, she was conscious but somewhat lethargic. Her best-corrected visual acuity was 20/20 OD and 20/80 OS. Pupillary light responses, extrinsic ocular motility, anterior segment biomicroscopy, and intraocular pressure were normal in both eyes. Dilated fundus examination revealed a bilateral sectorial macular star that was more extended in the left eye, with retinal veins slightly dilated and tortuous, attenuated arterioles, some juxta and peripapillary nerve fiber layer infarcts, and optic disc edema with marked papillary telangiectasia; in addition, some faint, small, tan-yellow dots were observed at the level of the retinal pigment epithelium in the posterior pole, some of them already grayish and with a hypopigmented halo (Figure 1). Optical coherence tomography displayed elevated optic discs and hyperreflective material in the outer plexiform layer temporal to the optic disc in both eyes and a macular neurosensory detachment in the left eye. Results of visual field testing were normal in the right eye and showed a central scotoma in the left eye.Fundus image of the right eye (A) and left eye (B) showing slight dilation of retinal veins (magenta arrowheads), intraretinal lipid deposits in the papillomacular bundle and the inferonasal macular quadrant forming an incomplete macular star (yellow arrowheads), optic disc edema and epipapillary telangiectasia (blue arrowheads), nerve fiber layer infarcts or cotton-wool nodules (green arrowheads) over the inferior margin of the optic disc in the right eye and juxta and parapapillary in the left eye, and paramacular temporal light-pigmented subretinal dots at the level of the retinal pigment epithelium (white arrowheads) in both eyes, with some already scarred (pigmented dot surrounded by a pale halo; gold arrowhead).
what would you do next?
What would you do next?
Check blood pressure
Order serologic testing for infectious neuroretinitis
Prescribe systemic corticosteroids
Rule out brain tumor
a
0
1
1
1
female
0
0
13
11-20
White
18
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809652
A 13-year-old girl with a 7-day history of painless vision loss and central scotoma in her left eye was referred to the department of ophthalmology. Three weeks prior, she had presented with mild fever and flulike symptoms, associated with a severe frontal headache that was mildly relieved by analgesics. Her medical history was positive for relapsing urinary tract infections since early infancy without adequate follow-up. Findings of an ophthalmological examination 1 year prior were reported as normal.At initial examination, she was conscious but somewhat lethargic. Her best-corrected visual acuity was 20/20 OD and 20/80 OS. Pupillary light responses, extrinsic ocular motility, anterior segment biomicroscopy, and intraocular pressure were normal in both eyes. Dilated fundus examination revealed a bilateral sectorial macular star that was more extended in the left eye, with retinal veins slightly dilated and tortuous, attenuated arterioles, some juxta and peripapillary nerve fiber layer infarcts, and optic disc edema with marked papillary telangiectasia; in addition, some faint, small, tan-yellow dots were observed at the level of the retinal pigment epithelium in the posterior pole, some of them already grayish and with a hypopigmented halo (Figure 1). Optical coherence tomography displayed elevated optic discs and hyperreflective material in the outer plexiform layer temporal to the optic disc in both eyes and a macular neurosensory detachment in the left eye. Results of visual field testing were normal in the right eye and showed a central scotoma in the left eye.Fundus image of the right eye (A) and left eye (B) showing slight dilation of retinal veins (magenta arrowheads), intraretinal lipid deposits in the papillomacular bundle and the inferonasal macular quadrant forming an incomplete macular star (yellow arrowheads), optic disc edema and epipapillary telangiectasia (blue arrowheads), nerve fiber layer infarcts or cotton-wool nodules (green arrowheads) over the inferior margin of the optic disc in the right eye and juxta and parapapillary in the left eye, and paramacular temporal light-pigmented subretinal dots at the level of the retinal pigment epithelium (white arrowheads) in both eyes, with some already scarred (pigmented dot surrounded by a pale halo; gold arrowhead). What Would You Do Next?
Prescribe systemic corticosteroids
Order serologic testing for infectious neuroretinitis
Rule out brain tumor
Check blood pressure
Hypertensive retinopathy
D
Check blood pressure
Option D is the correct approach. The child underwent immediate hospitalization and systemic evaluation. Clinical examination revealed severe systemic hypertension (225/160 mm Hg). Laboratory test results showed leukocytosis, elevated erythrocyte sedimentation rate (35 mm), hypercholesterolemia, hypokalemia, proteinuria, and microalbuminuria.Echocardiography showed left ventricle hypertrophy, and kidney doppler ultrasonography revealed advanced bilateral kidney atrophy with parenchymal scarring that was presumed to be the consequence of undertreated relapsing urinary tract infections. Results of magnetic resonance imaging of the brain and orbits were normal.The most common masquerader for neuroretinitis is malignant hypertension.1 Systemic hypertension in children is rare, potentially underrecognized,2 and its prevalence ranges from 2% to 5%.3 Mostly asymptomatic, pediatric hypertension may manifest by symptoms originating from target organ disease, and most cases present secondary hypertension from kidney disorders, especially kidney parenchymal disease.4 Hypertensive retinopathy occurs in 8% to 18% of children with severe hypertension, figures substantially lower than those observed in adults with hypertension, with the highest prevalence in those with kidney and renovascular disease.5Malignant hypertension is an acute, severe rise of more than 180 mm Hg in systolic pressure and/or more than 120 mm Hg in diastolic pressure, associated with severe hypertensive retinopathy and papilledema. It constitutes a medical emergency, where severe acute increase in blood pressure results in end-organ damage.6 The bilateral retinal disease, the presence of nerve fiber layer infarcts and Elschnig spots (focal signs of choroidal ischemia at the level of the retinal pigment epithelium), and the accompanying neurologic symptoms (severe headache and lethargy) suggested a severe systemic disease and were the presenting signs of malignant hypertension, which is a life-threatening condition in this patient secondary to kidney parenchymal disease from undertreated relapsing urinary tract infections since early infancy.Option A is incorrect. It is not advisable to prescribe corticosteroids before ruling out an infectious condition. Also, corticosteroids are contraindicated in the context of malignant hypertension. For option B, in the presence of the triad unilateral vision loss, macular star, and optic disc swelling in a child, infectious neuroretinitis must be ruled out, with cat scratch disease (CSD) its most frequent cause. However, neuroretinitis is a rare complication of cat scratch disease, occurring in only 1% to 2% of cases, and it generally presents unilaterally and self-limited.1 In this patient, results of serum tests for infective causes were negative. Besides, retinal and optic nerve disease were bilateral, and the child presented with neurologic signs. Option C is incorrect. Although this patient had neurologic symptoms with bilateral optic disc edema, other features, like macular lipid deposits, nerve fiber layer infarcts, and Elschnig spots, are not clinical signs that accompany papilledema from intracranial hypertension.Treatment with amlodipine, 5 mg, every 12 hours, daily losartan, 50 mg, and a hyposodic diet reduced blood pressure drastically and slowly normalized the retinal disease. Visual acuity progressively improved in the left eye, as did the macular star, optic disc edema, and nerve fiber layer infarcts in both eyes. One year later, blood pressure was generally within normal values under treatment but with intermittent episodes of high values. Best-corrected visual acuity was 20/20 OD and 20/25 OS, and optic discs were nonedematous but moderately pale in both eyes (Figure 2). Optical coherence tomography performed during follow-up showed a diffuse thinning of the neurosensory retina and the peripapillary nerve fiber layer.Fundus image of the left eye (panel) and the right eye (insert) at 1-year follow-up showing a pronounced sclerotic appearance of arteries, a complete resolution of both optic discs’ edema and the macular stars, a moderate optic disc pallor, and no evidence of nerve fiber layer infarcts.
Ophthalmology
A 13-year-old boy with a 7-day history of painless vision loss and central scotoma in him left eye was referred to the department of ophthalmology. Three weeks prior, he had presented with mild fever and flulike symptoms, associated with a severe frontal headache that was mildly relieved by analgesics. Him medical history was positive for relapsing urinary tract infections since early infancy without adequate follow-up. Findings of an ophthalmological examination 1 year prior were reported as normal.At initial examination, he was conscious but somewhat lethargic. Him best-corrected visual acuity was 20/20 OD and 20/80 OS. Pupillary light responses, extrinsic ocular motility, anterior segment biomicroscopy, and intraocular pressure were normal in both eyes. Dilated fundus examination revealed a bilateral sectorial macular star that was more extended in the left eye, with retinal veins slightly dilated and tortuous, attenuated arterioles, some juxta and peripapillary nerve fiber layer infarcts, and optic disc edema with marked papillary telangiectasia; in addition, some faint, small, tan-yellow dots were observed at the level of the retinal pigment epithelium in the posterior pole, some of them already grayish and with a hypopigmented halo (Figure 1). Optical coherence tomography displayed elevated optic discs and hyperreflective material in the outer plexiform layer temporal to the optic disc in both eyes and a macular neurosensory detachment in the left eye. Results of visual field testing were normal in the right eye and showed a central scotoma in the left eye.Fundus image of the right eye (A) and left eye (B) showing slight dilation of retinal veins (magenta arrowheads), intraretinal lipid deposits in the papillomacular bundle and the inferonasal macular quadrant forming an incomplete macular star (yellow arrowheads), optic disc edema and epipapillary telangiectasia (blue arrowheads), nerve fiber layer infarcts or cotton-wool nodules (green arrowheads) over the inferior margin of the optic disc in the right eye and juxta and parapapillary in the left eye, and paramacular temporal light-pigmented subretinal dots at the level of the retinal pigment epithelium (white arrowheads) in both eyes, with some already scarred (pigmented dot surrounded by a pale halo; gold arrowhead).
what would you do next?
What would you do next?
Check blood pressure
Order serologic testing for infectious neuroretinitis
Prescribe systemic corticosteroids
Rule out brain tumor
a
0
1
1
1
male
0
0
13
11-20
White
18
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2809652
A 13-year-old girl with a 7-day history of painless vision loss and central scotoma in her left eye was referred to the department of ophthalmology. Three weeks prior, she had presented with mild fever and flulike symptoms, associated with a severe frontal headache that was mildly relieved by analgesics. Her medical history was positive for relapsing urinary tract infections since early infancy without adequate follow-up. Findings of an ophthalmological examination 1 year prior were reported as normal.At initial examination, she was conscious but somewhat lethargic. Her best-corrected visual acuity was 20/20 OD and 20/80 OS. Pupillary light responses, extrinsic ocular motility, anterior segment biomicroscopy, and intraocular pressure were normal in both eyes. Dilated fundus examination revealed a bilateral sectorial macular star that was more extended in the left eye, with retinal veins slightly dilated and tortuous, attenuated arterioles, some juxta and peripapillary nerve fiber layer infarcts, and optic disc edema with marked papillary telangiectasia; in addition, some faint, small, tan-yellow dots were observed at the level of the retinal pigment epithelium in the posterior pole, some of them already grayish and with a hypopigmented halo (Figure 1). Optical coherence tomography displayed elevated optic discs and hyperreflective material in the outer plexiform layer temporal to the optic disc in both eyes and a macular neurosensory detachment in the left eye. Results of visual field testing were normal in the right eye and showed a central scotoma in the left eye.Fundus image of the right eye (A) and left eye (B) showing slight dilation of retinal veins (magenta arrowheads), intraretinal lipid deposits in the papillomacular bundle and the inferonasal macular quadrant forming an incomplete macular star (yellow arrowheads), optic disc edema and epipapillary telangiectasia (blue arrowheads), nerve fiber layer infarcts or cotton-wool nodules (green arrowheads) over the inferior margin of the optic disc in the right eye and juxta and parapapillary in the left eye, and paramacular temporal light-pigmented subretinal dots at the level of the retinal pigment epithelium (white arrowheads) in both eyes, with some already scarred (pigmented dot surrounded by a pale halo; gold arrowhead). What Would You Do Next?
Prescribe systemic corticosteroids
Order serologic testing for infectious neuroretinitis
Rule out brain tumor
Check blood pressure
Hypertensive retinopathy
D
Check blood pressure
Option D is the correct approach. The child underwent immediate hospitalization and systemic evaluation. Clinical examination revealed severe systemic hypertension (225/160 mm Hg). Laboratory test results showed leukocytosis, elevated erythrocyte sedimentation rate (35 mm), hypercholesterolemia, hypokalemia, proteinuria, and microalbuminuria.Echocardiography showed left ventricle hypertrophy, and kidney doppler ultrasonography revealed advanced bilateral kidney atrophy with parenchymal scarring that was presumed to be the consequence of undertreated relapsing urinary tract infections. Results of magnetic resonance imaging of the brain and orbits were normal.The most common masquerader for neuroretinitis is malignant hypertension.1 Systemic hypertension in children is rare, potentially underrecognized,2 and its prevalence ranges from 2% to 5%.3 Mostly asymptomatic, pediatric hypertension may manifest by symptoms originating from target organ disease, and most cases present secondary hypertension from kidney disorders, especially kidney parenchymal disease.4 Hypertensive retinopathy occurs in 8% to 18% of children with severe hypertension, figures substantially lower than those observed in adults with hypertension, with the highest prevalence in those with kidney and renovascular disease.5Malignant hypertension is an acute, severe rise of more than 180 mm Hg in systolic pressure and/or more than 120 mm Hg in diastolic pressure, associated with severe hypertensive retinopathy and papilledema. It constitutes a medical emergency, where severe acute increase in blood pressure results in end-organ damage.6 The bilateral retinal disease, the presence of nerve fiber layer infarcts and Elschnig spots (focal signs of choroidal ischemia at the level of the retinal pigment epithelium), and the accompanying neurologic symptoms (severe headache and lethargy) suggested a severe systemic disease and were the presenting signs of malignant hypertension, which is a life-threatening condition in this patient secondary to kidney parenchymal disease from undertreated relapsing urinary tract infections since early infancy.Option A is incorrect. It is not advisable to prescribe corticosteroids before ruling out an infectious condition. Also, corticosteroids are contraindicated in the context of malignant hypertension. For option B, in the presence of the triad unilateral vision loss, macular star, and optic disc swelling in a child, infectious neuroretinitis must be ruled out, with cat scratch disease (CSD) its most frequent cause. However, neuroretinitis is a rare complication of cat scratch disease, occurring in only 1% to 2% of cases, and it generally presents unilaterally and self-limited.1 In this patient, results of serum tests for infective causes were negative. Besides, retinal and optic nerve disease were bilateral, and the child presented with neurologic signs. Option C is incorrect. Although this patient had neurologic symptoms with bilateral optic disc edema, other features, like macular lipid deposits, nerve fiber layer infarcts, and Elschnig spots, are not clinical signs that accompany papilledema from intracranial hypertension.Treatment with amlodipine, 5 mg, every 12 hours, daily losartan, 50 mg, and a hyposodic diet reduced blood pressure drastically and slowly normalized the retinal disease. Visual acuity progressively improved in the left eye, as did the macular star, optic disc edema, and nerve fiber layer infarcts in both eyes. One year later, blood pressure was generally within normal values under treatment but with intermittent episodes of high values. Best-corrected visual acuity was 20/20 OD and 20/25 OS, and optic discs were nonedematous but moderately pale in both eyes (Figure 2). Optical coherence tomography performed during follow-up showed a diffuse thinning of the neurosensory retina and the peripapillary nerve fiber layer.Fundus image of the left eye (panel) and the right eye (insert) at 1-year follow-up showing a pronounced sclerotic appearance of arteries, a complete resolution of both optic discs’ edema and the macular stars, a moderate optic disc pallor, and no evidence of nerve fiber layer infarcts.
Ophthalmology
A 13-year-old child with a 7-day history of painless vision loss and central scotoma in them left eye was referred to the department of ophthalmology. Three weeks prior, they had presented with mild fever and flulike symptoms, associated with a severe frontal headache that was mildly relieved by analgesics. Them medical history was positive for relapsing urinary tract infections since early infancy without adequate follow-up. Findings of an ophthalmological examination 1 year prior were reported as normal.At initial examination, they was conscious but somewhat lethargic. Them best-corrected visual acuity was 20/20 OD and 20/80 OS. Pupillary light responses, extrinsic ocular motility, anterior segment biomicroscopy, and intraocular pressure were normal in both eyes. Dilated fundus examination revealed a bilateral sectorial macular star that was more extended in the left eye, with retinal veins slightly dilated and tortuous, attenuated arterioles, some juxta and peripapillary nerve fiber layer infarcts, and optic disc edema with marked papillary telangiectasia; in addition, some faint, small, tan-yellow dots were observed at the level of the retinal pigment epithelium in the posterior pole, some of them already grayish and with a hypopigmented halo (Figure 1). Optical coherence tomography displayed elevated optic discs and hyperreflective material in the outer plexiform layer temporal to the optic disc in both eyes and a macular neurosensory detachment in the left eye. Results of visual field testing were normal in the right eye and showed a central scotoma in the left eye.Fundus image of the right eye (A) and left eye (B) showing slight dilation of retinal veins (magenta arrowheads), intraretinal lipid deposits in the papillomacular bundle and the inferonasal macular quadrant forming an incomplete macular star (yellow arrowheads), optic disc edema and epipapillary telangiectasia (blue arrowheads), nerve fiber layer infarcts or cotton-wool nodules (green arrowheads) over the inferior margin of the optic disc in the right eye and juxta and parapapillary in the left eye, and paramacular temporal light-pigmented subretinal dots at the level of the retinal pigment epithelium (white arrowheads) in both eyes, with some already scarred (pigmented dot surrounded by a pale halo; gold arrowhead).
what would you do next?
What would you do next?
Check blood pressure
Order serologic testing for infectious neuroretinitis
Prescribe systemic corticosteroids
Rule out brain tumor
a
0
1
1
1
neutral
0
0
13
11-20
White
18
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2809348
An otherwise healthy 13-month-old male was scheduled to undergo excisional biopsy of a left anterior tongue submucosal intramuscular mass (Figure 1). This firm-to-palpation mass was noted at birth and remained proportional in size with the child’s somatic growth, measuring approximately 1 cm in diameter on presentation. Preoperative magnetic resonance imaging (MRI) documented the mass to be uniformly solid and well circumscribed without cystic components or flow voids, hypointense on T2, and hyperintense with homogeneous contrast enhancement on T1. His head, neck, and general examination findings were otherwise normal with no cervical lymphadenopathy or additional masses. What Is Your Diagnosis?
Hemangioma
Rhabdomyoma
Rhabdomyosarcoma
Teratoma
B. Rhabdomyoma
B
Rhabdomyoma
Neonatal tongue lesions have a broad differential diagnosis, including cystic, solid, and mixed composite lesions.1 Differentiation of cystic from solid lesions is important, as each warrants a unique differential diagnosis. Ultrasonography, computed tomography, and MRI are modalities that can be used to make this differentiation. If sedation is required, MRI with contrast is preferable given the excellent soft tissue detail without risk of radiation.Common congenital cystic tongue lesions include foregut duplication cysts, dermoid cysts, and ranulas. Foregut duplication cysts and dermoid cysts tend to be midline lesions, the former lined by gastrointestinal mucosa and the latter lined by squamous epithelium with adnexal structures.2 Ranulas and alternative pseudocysts of sublingual gland origin are more commonly unilateral lesions with a floor of mouth rather than tongue focality. Lymphatic and venous vascular malformations need to also be considered.This child’s lesion, based on physical examination and MRI characteristics, was solid. The differential diagnosis of solid tongue masses is broad, but neonatal presentation favors congenital and neoplastic causes. Common benign solid tongue masses in this age group include hemangiomas, hamartomas, choristomas, and teratomas.Hamartomas are benign growths composed of a proliferation of tissue native to the site of the lesion, whereas choristomas are proliferations of tissues not normally found in that anatomical location. Both hamartomas and choristomas can be solid depending on their tissue of origin.The World Health Organization Classification of Tumors lists 4 subtypes of extracardiac rhabdomyoma: fetal, adult, intermediate, and genital.3 Fetal rhabdomyoma may present at any age; however, over 50% of fetal rhabdomyoma cases have been diagnosed in infants and children younger than 3 years, with 25% being congenital.4 Fetal rhabdomyoma is the most common type found in the head and neck.5Although rare, malignant tongue masses should be considered, as delayed diagnosis can have devastating consequences. Rhabdomyosarcoma is the most common pediatric tongue malignant neoplasm, with leiomyosarcoma and fibrosarcoma being additional possibilities.6 Specifically, embryonal rhabdomyosarcoma should be considered, given the location.The pathology findings key to making the diagnosis in this case are highlighted in Figure 2. The histopathology demonstrated a circumscribed lesion composed of irregular bundles of primitive-appearing spindle cells and striated immature skeletal muscle cells, resembling fetal microtubules, within a myxoid background. Minimal mitotic activity was noted; no necrosis or atypia was present. These features supported a diagnosis of fetal rhabdomyoma.A, Histopathology findings of the excised lesion demonstrating irregular bundles of primitive cells and immature striated skeletal muscle fibers (hematoxylin-eosin). B, Desmin immunohistochemical stain highlighting the lesional cells.The myxoid background is important for the diagnosis of a classical fetal-type rhabdomyoma. Rhabdomyomas generally do not show atypia or necrosis, and mitoses are uncommon. Both the spindle cells and rhabdomyoblasts will be positive for myogenin and MyoD1 by immunohistochemistry. Adult rhabdomyoma can also be considered if the characteristic spider cells are present. Embryonal rhabdomyosarcoma is a poorly circumscribed lesion that shows both primitive round-to-spindle cells to differentiating rhabdomyoblasts (“tadpole cells” and/or “strap cells”) to cross-striated myofibers present in zones of hypocellularity to hypercellularity, embedded within a myxoid background. Embryonal rhabdomyosarcoma generally shows increased mitotic activity (sometimes with atypical mitoses), necrosis, and atypia; anaplasia may be seen.Heterotopic fetal rhabdomyoma, similar to choristoma, is managed by surgical excision for diagnosis and therapy.7 A recurrence rate of 4% over a median follow-up of 4 years has been reported for fetal rhabdomyoma of the head and neck.4 Recurrence likely reflects incomplete resection and, rarely, multicentricity.8The decision to wait to perform surgery was purposeful given the benign characteristics of the lesion and anesthesia exposure concerns. The patient was admitted for overnight monitoring with discharge home the following morning. At 6-month postoperative follow-up, the child demonstrated tongue symmetry, no functional impairment, and no evidence of recurrence.
General
An otherwise healthy 13-month-old male was scheduled to undergo excisional biopsy of a left anterior tongue submucosal intramuscular mass (Figure 1). This firm-to-palpation mass was noted at birth and remained proportional in size with the child’s somatic growth, measuring approximately 1 cm in diameter on presentation. Preoperative magnetic resonance imaging (MRI) documented the mass to be uniformly solid and well circumscribed without cystic components or flow voids, hypointense on T2, and hyperintense with homogeneous contrast enhancement on T1. His head, neck, and general examination findings were otherwise normal with no cervical lymphadenopathy or additional masses.
what is your diagnosis?
What is your diagnosis?
Teratoma
Hemangioma
Rhabdomyosarcoma
Rhabdomyoma
d
1
1
1
1
male
0
0
1.08
0-10
null
19
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2809348
An otherwise healthy 13-month-old male was scheduled to undergo excisional biopsy of a left anterior tongue submucosal intramuscular mass (Figure 1). This firm-to-palpation mass was noted at birth and remained proportional in size with the child’s somatic growth, measuring approximately 1 cm in diameter on presentation. Preoperative magnetic resonance imaging (MRI) documented the mass to be uniformly solid and well circumscribed without cystic components or flow voids, hypointense on T2, and hyperintense with homogeneous contrast enhancement on T1. His head, neck, and general examination findings were otherwise normal with no cervical lymphadenopathy or additional masses. What Is Your Diagnosis?
Hemangioma
Rhabdomyoma
Rhabdomyosarcoma
Teratoma
B. Rhabdomyoma
B
Rhabdomyoma
Neonatal tongue lesions have a broad differential diagnosis, including cystic, solid, and mixed composite lesions.1 Differentiation of cystic from solid lesions is important, as each warrants a unique differential diagnosis. Ultrasonography, computed tomography, and MRI are modalities that can be used to make this differentiation. If sedation is required, MRI with contrast is preferable given the excellent soft tissue detail without risk of radiation.Common congenital cystic tongue lesions include foregut duplication cysts, dermoid cysts, and ranulas. Foregut duplication cysts and dermoid cysts tend to be midline lesions, the former lined by gastrointestinal mucosa and the latter lined by squamous epithelium with adnexal structures.2 Ranulas and alternative pseudocysts of sublingual gland origin are more commonly unilateral lesions with a floor of mouth rather than tongue focality. Lymphatic and venous vascular malformations need to also be considered.This child’s lesion, based on physical examination and MRI characteristics, was solid. The differential diagnosis of solid tongue masses is broad, but neonatal presentation favors congenital and neoplastic causes. Common benign solid tongue masses in this age group include hemangiomas, hamartomas, choristomas, and teratomas.Hamartomas are benign growths composed of a proliferation of tissue native to the site of the lesion, whereas choristomas are proliferations of tissues not normally found in that anatomical location. Both hamartomas and choristomas can be solid depending on their tissue of origin.The World Health Organization Classification of Tumors lists 4 subtypes of extracardiac rhabdomyoma: fetal, adult, intermediate, and genital.3 Fetal rhabdomyoma may present at any age; however, over 50% of fetal rhabdomyoma cases have been diagnosed in infants and children younger than 3 years, with 25% being congenital.4 Fetal rhabdomyoma is the most common type found in the head and neck.5Although rare, malignant tongue masses should be considered, as delayed diagnosis can have devastating consequences. Rhabdomyosarcoma is the most common pediatric tongue malignant neoplasm, with leiomyosarcoma and fibrosarcoma being additional possibilities.6 Specifically, embryonal rhabdomyosarcoma should be considered, given the location.The pathology findings key to making the diagnosis in this case are highlighted in Figure 2. The histopathology demonstrated a circumscribed lesion composed of irregular bundles of primitive-appearing spindle cells and striated immature skeletal muscle cells, resembling fetal microtubules, within a myxoid background. Minimal mitotic activity was noted; no necrosis or atypia was present. These features supported a diagnosis of fetal rhabdomyoma.A, Histopathology findings of the excised lesion demonstrating irregular bundles of primitive cells and immature striated skeletal muscle fibers (hematoxylin-eosin). B, Desmin immunohistochemical stain highlighting the lesional cells.The myxoid background is important for the diagnosis of a classical fetal-type rhabdomyoma. Rhabdomyomas generally do not show atypia or necrosis, and mitoses are uncommon. Both the spindle cells and rhabdomyoblasts will be positive for myogenin and MyoD1 by immunohistochemistry. Adult rhabdomyoma can also be considered if the characteristic spider cells are present. Embryonal rhabdomyosarcoma is a poorly circumscribed lesion that shows both primitive round-to-spindle cells to differentiating rhabdomyoblasts (“tadpole cells” and/or “strap cells”) to cross-striated myofibers present in zones of hypocellularity to hypercellularity, embedded within a myxoid background. Embryonal rhabdomyosarcoma generally shows increased mitotic activity (sometimes with atypical mitoses), necrosis, and atypia; anaplasia may be seen.Heterotopic fetal rhabdomyoma, similar to choristoma, is managed by surgical excision for diagnosis and therapy.7 A recurrence rate of 4% over a median follow-up of 4 years has been reported for fetal rhabdomyoma of the head and neck.4 Recurrence likely reflects incomplete resection and, rarely, multicentricity.8The decision to wait to perform surgery was purposeful given the benign characteristics of the lesion and anesthesia exposure concerns. The patient was admitted for overnight monitoring with discharge home the following morning. At 6-month postoperative follow-up, the child demonstrated tongue symmetry, no functional impairment, and no evidence of recurrence.
General
An otherwise healthy 13-month-old female was scheduled to undergo excisional biopsy of a left anterior tongue submucosal intramuscular mass (Figure 1). This firm-to-palpation mass was noted at birth and remained proportional in size with the child’s somatic growth, measuring approximately 1 cm in diameter on presentation. Preoperative magnetic resonance imaging (MRI) documented the mass to be uniformly solid and well circumscribed without cystic components or flow voids, hypointense on T2, and hyperintense with homogeneous contrast enhancement on T1. Her head, neck, and general examination findings were otherwise normal with no cervical lymphadenopathy or additional masses.
what is your diagnosis?
What is your diagnosis?
Teratoma
Hemangioma
Rhabdomyosarcoma
Rhabdomyoma
d
1
1
1
1
female
0
0
1.08
0-10
null
19
augmented_female_frommale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2809348
An otherwise healthy 13-month-old male was scheduled to undergo excisional biopsy of a left anterior tongue submucosal intramuscular mass (Figure 1). This firm-to-palpation mass was noted at birth and remained proportional in size with the child’s somatic growth, measuring approximately 1 cm in diameter on presentation. Preoperative magnetic resonance imaging (MRI) documented the mass to be uniformly solid and well circumscribed without cystic components or flow voids, hypointense on T2, and hyperintense with homogeneous contrast enhancement on T1. His head, neck, and general examination findings were otherwise normal with no cervical lymphadenopathy or additional masses. What Is Your Diagnosis?
Hemangioma
Rhabdomyoma
Rhabdomyosarcoma
Teratoma
B. Rhabdomyoma
B
Rhabdomyoma
Neonatal tongue lesions have a broad differential diagnosis, including cystic, solid, and mixed composite lesions.1 Differentiation of cystic from solid lesions is important, as each warrants a unique differential diagnosis. Ultrasonography, computed tomography, and MRI are modalities that can be used to make this differentiation. If sedation is required, MRI with contrast is preferable given the excellent soft tissue detail without risk of radiation.Common congenital cystic tongue lesions include foregut duplication cysts, dermoid cysts, and ranulas. Foregut duplication cysts and dermoid cysts tend to be midline lesions, the former lined by gastrointestinal mucosa and the latter lined by squamous epithelium with adnexal structures.2 Ranulas and alternative pseudocysts of sublingual gland origin are more commonly unilateral lesions with a floor of mouth rather than tongue focality. Lymphatic and venous vascular malformations need to also be considered.This child’s lesion, based on physical examination and MRI characteristics, was solid. The differential diagnosis of solid tongue masses is broad, but neonatal presentation favors congenital and neoplastic causes. Common benign solid tongue masses in this age group include hemangiomas, hamartomas, choristomas, and teratomas.Hamartomas are benign growths composed of a proliferation of tissue native to the site of the lesion, whereas choristomas are proliferations of tissues not normally found in that anatomical location. Both hamartomas and choristomas can be solid depending on their tissue of origin.The World Health Organization Classification of Tumors lists 4 subtypes of extracardiac rhabdomyoma: fetal, adult, intermediate, and genital.3 Fetal rhabdomyoma may present at any age; however, over 50% of fetal rhabdomyoma cases have been diagnosed in infants and children younger than 3 years, with 25% being congenital.4 Fetal rhabdomyoma is the most common type found in the head and neck.5Although rare, malignant tongue masses should be considered, as delayed diagnosis can have devastating consequences. Rhabdomyosarcoma is the most common pediatric tongue malignant neoplasm, with leiomyosarcoma and fibrosarcoma being additional possibilities.6 Specifically, embryonal rhabdomyosarcoma should be considered, given the location.The pathology findings key to making the diagnosis in this case are highlighted in Figure 2. The histopathology demonstrated a circumscribed lesion composed of irregular bundles of primitive-appearing spindle cells and striated immature skeletal muscle cells, resembling fetal microtubules, within a myxoid background. Minimal mitotic activity was noted; no necrosis or atypia was present. These features supported a diagnosis of fetal rhabdomyoma.A, Histopathology findings of the excised lesion demonstrating irregular bundles of primitive cells and immature striated skeletal muscle fibers (hematoxylin-eosin). B, Desmin immunohistochemical stain highlighting the lesional cells.The myxoid background is important for the diagnosis of a classical fetal-type rhabdomyoma. Rhabdomyomas generally do not show atypia or necrosis, and mitoses are uncommon. Both the spindle cells and rhabdomyoblasts will be positive for myogenin and MyoD1 by immunohistochemistry. Adult rhabdomyoma can also be considered if the characteristic spider cells are present. Embryonal rhabdomyosarcoma is a poorly circumscribed lesion that shows both primitive round-to-spindle cells to differentiating rhabdomyoblasts (“tadpole cells” and/or “strap cells”) to cross-striated myofibers present in zones of hypocellularity to hypercellularity, embedded within a myxoid background. Embryonal rhabdomyosarcoma generally shows increased mitotic activity (sometimes with atypical mitoses), necrosis, and atypia; anaplasia may be seen.Heterotopic fetal rhabdomyoma, similar to choristoma, is managed by surgical excision for diagnosis and therapy.7 A recurrence rate of 4% over a median follow-up of 4 years has been reported for fetal rhabdomyoma of the head and neck.4 Recurrence likely reflects incomplete resection and, rarely, multicentricity.8The decision to wait to perform surgery was purposeful given the benign characteristics of the lesion and anesthesia exposure concerns. The patient was admitted for overnight monitoring with discharge home the following morning. At 6-month postoperative follow-up, the child demonstrated tongue symmetry, no functional impairment, and no evidence of recurrence.
General
An otherwise healthy 13-month-old person was scheduled to undergo excisional biopsy of a left anterior tongue submucosal intramuscular mass (Figure 1). This firm-to-palpation mass was noted at birth and remained proportional in size with the child’s somatic growth, measuring approximately 1 cm in diameter on presentation. Preoperative magnetic resonance imaging (MRI) documented the mass to be uniformly solid and well circumscribed without cystic components or flow voids, hypointense on T2, and hyperintense with homogeneous contrast enhancement on T1. Their head, neck, and general examination findings were otherwise normal with no cervical lymphadenopathy or additional masses.
what is your diagnosis?
What is your diagnosis?
Teratoma
Hemangioma
Rhabdomyosarcoma
Rhabdomyoma
d
1
1
1
1
neutral
0
0
1.08
0-10
null
19
augmented_neutral_frommale
https://jamanetwork.com/journals/jama/fullarticle/2809109
A 42-year-old woman with asthma, seasonal allergies, and invasive ductal breast carcinoma presented to the dermatology clinic for rash and poor wound healing 2 weeks after bilateral mastectomy. Five days after surgery, the patient noted erythema, edema, pain, pruritus, and serous fluid drainage at the mastectomy incision sites (Figure 1). Two days later, she developed an erythematous papulovesicular rash on her trunk and upper and lower extremities. A skin swab was sent for bacterial culture, and she was prescribed cefadroxil (500 mg twice daily for 1 week). The skin swab bacterial culture result was negative, and her skin findings did not improve with antibiotics. The patient reported having a similar episode of rash and delayed wound healing after laparoscopy 1 year prior to presentation.Erythema and crusting along incision sites surrounded by a papulovesicular rash.Perform a skin biopsy at the mastectomy incision sitePrescribe acyclovir (400 mg orally 3 times daily for 7 days)Start topical steroids and perform a skin patch testTreat with doxycycline (100 mg orally twice daily for 7 days) What Would You Do Next?
Perform a skin biopsy at the mastectomy incision site
Prescribe acyclovir (400 mg orally 3 times daily for 7 days)
Start topical steroids and perform a skin patch test
Treat with doxycycline (100 mg orally twice daily for 7 days)
Allergic contact dermatitis due to use of surgical skin glue
C
Start topical steroids and perform a skin patch test
The key to the correct diagnosis is recognizing that an acute pruritic papulovesicular rash involving a surgical incision site is characteristic of allergic contact dermatitis due to use of surgical skin glue. Skin biopsy (choice A) would not be helpful because histopathologic findings of allergic contact dermatitis are nonspecific. Prescribing acyclovir (choice B) is not recommended because the patient did not have typical findings of herpes simplex virus such as grouped vesicles on an erythematous base. Doxycycline (choice D) is not indicated because her bacterial culture result was negative, and she had already been treated with a course of antibiotics.Allergic contact dermatitis (ACD) is a cutaneous type IV (delayed) hypersensitivity reaction caused by activation of allergen-specific T cells that develop after initial exposure to an allergen in contact with the skin.1 Activation of these T cells causes cytokine release and cellular infiltrates that result in the clinical symptoms of ACD.2Acute ACD presents with intense pruritus or burning, and the affected skin typically has a weepy erythematous appearance with surrounding erythematous papulovesicles and honey-colored crusting. Bullae and oozing may arise in severe cases.2,3 Skin findings may occur as early as 1 to 2 days after exposure in sensitized individuals or up to 1 month with initial allergen exposure.4 The differential diagnosis of ACD includes irritant contact dermatitis, atopic dermatitis, seborrheic dermatitis, psoriasis, tinea, mycosis fungoides, and surgical site infection in patients who have undergone recent surgery.2,3ACD affects approximately 72 million individuals in the US annually and is diagnosed more commonly in females than in males.1,2,4 Risk factors for ACD include atopic dermatitis, a family history of ACD, and exposure to acrylates, which are chemical agents commonly used in glues, adhesives, and plastics.1,3 Occupations with increased risk of ACD due to acrylate exposure include medical and dental practitioners, beauticians, chemical plant workers, construction workers, metal workers, and mechanics.1,3Patients who have undergone recent surgery can develop ACD from exposure to tissue adhesives, sutures, staples, antiseptic preparations, topical antibiotics, joint implants, and bone cement5-7 ACD causes inflammation of the skin that may impair wound healing, cause wound dehiscence, and increase the risk of wound infections.5,7ACD due to surgical skin glue occurs in 1.5% to 2.8% of exposed patients,3,4 and the most common allergens are acrylates (methacrylates or cyanoacrylates). ACD caused by one acrylate may lead to cosensitization to another acrylate.1 Surgical skin glue adhesives reside in the dermis and can cause sensitization and skin reactions up to 1 month after application.4 Approximately one-half of patients with allergic contact dermatitis due to surgical skin glue also develop a skin eruption in areas of the body remote from the site of application.4ACD is typically diagnosed with standard skin patch testing performed by an allergist or dermatologist. However, standard patch testing does not include cyanoacrylate, methacrylate, or suture materials and fails to identify approximately 25% of allergens that cause ACD.2,7 A specialized (“open”) patch test can be performed for patients who have exposure to specific known allergens not typically included in standard skin patch testing.2,7Treatment of ACD includes removal of the allergen and daily treatment with topical or systemic steroids for at least 2 to 3 weeks, at which point the skin findings typically resolve.2 However, patients must be informed that ACD will recur with reexposure, so strict avoidance of the specific allergen and cross-reacting allergens is required.2 Patients with a history of ACD due to use of surgical skin glue should inform their surgeon and anesthesiologist prior to surgery to avoid reexposure to an allergen that caused ACD.5An open patch test was performed to assess for an allergic reaction to the sutures and surgical skin glue used during the patient’s mastectomy. At 48 hours, she demonstrated a “strong positive” skin reaction to surgical skin glue (Figure 2). Acetone was used to remove the remaining surgical skin glue on her mastectomy incision sites, and triamcinolone was prescribed (0.1% cream twice daily for 2 weeks). Three days later, the patient noted a substantial decrease in pruritus and erythema at the incision sites. She was advised to avoid future contact with surgical skin glue and other potentially cross-reacting adhesives and was provided a handout detailing safe alternative products. At 2-week follow-up, the skin over her mastectomy incision sites appeared healthy, and the rash on her trunk and upper and lower extremities had resolved.Open skin patch test with materials used during the patient’s mastectomy showed a 2+ (strong positive) reaction to surgical skin glue observed 48 hours after application. No suture reactions occurred.
General
A 42-year-old woman with asthma, seasonal allergies, and invasive ductal breast carcinoma presented to the dermatology clinic for rash and poor wound healing 2 weeks after bilateral mastectomy. Five days after surgery, the patient noted erythema, edema, pain, pruritus, and serous fluid drainage at the mastectomy incision sites (Figure 1). Two days later, she developed an erythematous papulovesicular rash on her trunk and upper and lower extremities. A skin swab was sent for bacterial culture, and she was prescribed cefadroxil (500 mg twice daily for 1 week). The skin swab bacterial culture result was negative, and her skin findings did not improve with antibiotics. The patient reported having a similar episode of rash and delayed wound healing after laparoscopy 1 year prior to presentation.Erythema and crusting along incision sites surrounded by a papulovesicular rash.Perform a skin biopsy at the mastectomy incision sitePrescribe acyclovir (400 mg orally 3 times daily for 7 days)Start topical steroids and perform a skin patch testTreat with doxycycline (100 mg orally twice daily for 7 days)
what would you do next?
What would you do next?
Treat with doxycycline (100 mg orally twice daily for 7 days)
Start topical steroids and perform a skin patch test
Prescribe acyclovir (400 mg orally 3 times daily for 7 days)
Perform a skin biopsy at the mastectomy incision site
b
0
1
1
1
female
0
0
42
41-50
null
20
original
https://jamanetwork.com/journals/jama/fullarticle/2809109
A 42-year-old woman with asthma, seasonal allergies, and invasive ductal breast carcinoma presented to the dermatology clinic for rash and poor wound healing 2 weeks after bilateral mastectomy. Five days after surgery, the patient noted erythema, edema, pain, pruritus, and serous fluid drainage at the mastectomy incision sites (Figure 1). Two days later, she developed an erythematous papulovesicular rash on her trunk and upper and lower extremities. A skin swab was sent for bacterial culture, and she was prescribed cefadroxil (500 mg twice daily for 1 week). The skin swab bacterial culture result was negative, and her skin findings did not improve with antibiotics. The patient reported having a similar episode of rash and delayed wound healing after laparoscopy 1 year prior to presentation.Erythema and crusting along incision sites surrounded by a papulovesicular rash.Perform a skin biopsy at the mastectomy incision sitePrescribe acyclovir (400 mg orally 3 times daily for 7 days)Start topical steroids and perform a skin patch testTreat with doxycycline (100 mg orally twice daily for 7 days) What Would You Do Next?
Perform a skin biopsy at the mastectomy incision site
Prescribe acyclovir (400 mg orally 3 times daily for 7 days)
Start topical steroids and perform a skin patch test
Treat with doxycycline (100 mg orally twice daily for 7 days)
Allergic contact dermatitis due to use of surgical skin glue
C
Start topical steroids and perform a skin patch test
The key to the correct diagnosis is recognizing that an acute pruritic papulovesicular rash involving a surgical incision site is characteristic of allergic contact dermatitis due to use of surgical skin glue. Skin biopsy (choice A) would not be helpful because histopathologic findings of allergic contact dermatitis are nonspecific. Prescribing acyclovir (choice B) is not recommended because the patient did not have typical findings of herpes simplex virus such as grouped vesicles on an erythematous base. Doxycycline (choice D) is not indicated because her bacterial culture result was negative, and she had already been treated with a course of antibiotics.Allergic contact dermatitis (ACD) is a cutaneous type IV (delayed) hypersensitivity reaction caused by activation of allergen-specific T cells that develop after initial exposure to an allergen in contact with the skin.1 Activation of these T cells causes cytokine release and cellular infiltrates that result in the clinical symptoms of ACD.2Acute ACD presents with intense pruritus or burning, and the affected skin typically has a weepy erythematous appearance with surrounding erythematous papulovesicles and honey-colored crusting. Bullae and oozing may arise in severe cases.2,3 Skin findings may occur as early as 1 to 2 days after exposure in sensitized individuals or up to 1 month with initial allergen exposure.4 The differential diagnosis of ACD includes irritant contact dermatitis, atopic dermatitis, seborrheic dermatitis, psoriasis, tinea, mycosis fungoides, and surgical site infection in patients who have undergone recent surgery.2,3ACD affects approximately 72 million individuals in the US annually and is diagnosed more commonly in females than in males.1,2,4 Risk factors for ACD include atopic dermatitis, a family history of ACD, and exposure to acrylates, which are chemical agents commonly used in glues, adhesives, and plastics.1,3 Occupations with increased risk of ACD due to acrylate exposure include medical and dental practitioners, beauticians, chemical plant workers, construction workers, metal workers, and mechanics.1,3Patients who have undergone recent surgery can develop ACD from exposure to tissue adhesives, sutures, staples, antiseptic preparations, topical antibiotics, joint implants, and bone cement5-7 ACD causes inflammation of the skin that may impair wound healing, cause wound dehiscence, and increase the risk of wound infections.5,7ACD due to surgical skin glue occurs in 1.5% to 2.8% of exposed patients,3,4 and the most common allergens are acrylates (methacrylates or cyanoacrylates). ACD caused by one acrylate may lead to cosensitization to another acrylate.1 Surgical skin glue adhesives reside in the dermis and can cause sensitization and skin reactions up to 1 month after application.4 Approximately one-half of patients with allergic contact dermatitis due to surgical skin glue also develop a skin eruption in areas of the body remote from the site of application.4ACD is typically diagnosed with standard skin patch testing performed by an allergist or dermatologist. However, standard patch testing does not include cyanoacrylate, methacrylate, or suture materials and fails to identify approximately 25% of allergens that cause ACD.2,7 A specialized (“open”) patch test can be performed for patients who have exposure to specific known allergens not typically included in standard skin patch testing.2,7Treatment of ACD includes removal of the allergen and daily treatment with topical or systemic steroids for at least 2 to 3 weeks, at which point the skin findings typically resolve.2 However, patients must be informed that ACD will recur with reexposure, so strict avoidance of the specific allergen and cross-reacting allergens is required.2 Patients with a history of ACD due to use of surgical skin glue should inform their surgeon and anesthesiologist prior to surgery to avoid reexposure to an allergen that caused ACD.5An open patch test was performed to assess for an allergic reaction to the sutures and surgical skin glue used during the patient’s mastectomy. At 48 hours, she demonstrated a “strong positive” skin reaction to surgical skin glue (Figure 2). Acetone was used to remove the remaining surgical skin glue on her mastectomy incision sites, and triamcinolone was prescribed (0.1% cream twice daily for 2 weeks). Three days later, the patient noted a substantial decrease in pruritus and erythema at the incision sites. She was advised to avoid future contact with surgical skin glue and other potentially cross-reacting adhesives and was provided a handout detailing safe alternative products. At 2-week follow-up, the skin over her mastectomy incision sites appeared healthy, and the rash on her trunk and upper and lower extremities had resolved.Open skin patch test with materials used during the patient’s mastectomy showed a 2+ (strong positive) reaction to surgical skin glue observed 48 hours after application. No suture reactions occurred.
General
A 42-year-old man with asthma, seasonal allergies, and invasive ductal breast carcinoma presented to the dermatology clinic for rash and poor wound healing 2 weeks after bilateral mastectomy. Five days after surgery, the man noted erythema, edema, pain, pruritus, and serous fluid drainage at the mastectomy incision sites (Figure 1). Two days later, he developed an erythematous papulovesicular rash on him trunk and upper and lower extremities. A skin swab was sent for bacterial culture, and he was prescribed cefadroxil (500 mg twice daily for 1 week). The skin swab bacterial culture result was negative, and him skin findings did not improve with antibiotics. The man reported having a similar episode of rash and delayed wound healing after laparoscopy 1 year prior to presentation.Erythema and crusting along incision sites surrounded by a papulovesicular rash.Perform a skin biopsy at the mastectomy incision sitePrescribe acyclovir (400 mg orally 3 times daily for 7 days)Start topical steroids and perform a skin patch testTreat with doxycycline (100 mg orally twice daily for 7 days)
what would you do next?
What would you do next?
Treat with doxycycline (100 mg orally twice daily for 7 days)
Start topical steroids and perform a skin patch test
Prescribe acyclovir (400 mg orally 3 times daily for 7 days)
Perform a skin biopsy at the mastectomy incision site
b
0
1
1
1
male
0
0
42
41-50
null
20
augmented_male_fromfemale
https://jamanetwork.com/journals/jama/fullarticle/2809109
A 42-year-old woman with asthma, seasonal allergies, and invasive ductal breast carcinoma presented to the dermatology clinic for rash and poor wound healing 2 weeks after bilateral mastectomy. Five days after surgery, the patient noted erythema, edema, pain, pruritus, and serous fluid drainage at the mastectomy incision sites (Figure 1). Two days later, she developed an erythematous papulovesicular rash on her trunk and upper and lower extremities. A skin swab was sent for bacterial culture, and she was prescribed cefadroxil (500 mg twice daily for 1 week). The skin swab bacterial culture result was negative, and her skin findings did not improve with antibiotics. The patient reported having a similar episode of rash and delayed wound healing after laparoscopy 1 year prior to presentation.Erythema and crusting along incision sites surrounded by a papulovesicular rash.Perform a skin biopsy at the mastectomy incision sitePrescribe acyclovir (400 mg orally 3 times daily for 7 days)Start topical steroids and perform a skin patch testTreat with doxycycline (100 mg orally twice daily for 7 days) What Would You Do Next?
Perform a skin biopsy at the mastectomy incision site
Prescribe acyclovir (400 mg orally 3 times daily for 7 days)
Start topical steroids and perform a skin patch test
Treat with doxycycline (100 mg orally twice daily for 7 days)
Allergic contact dermatitis due to use of surgical skin glue
C
Start topical steroids and perform a skin patch test
The key to the correct diagnosis is recognizing that an acute pruritic papulovesicular rash involving a surgical incision site is characteristic of allergic contact dermatitis due to use of surgical skin glue. Skin biopsy (choice A) would not be helpful because histopathologic findings of allergic contact dermatitis are nonspecific. Prescribing acyclovir (choice B) is not recommended because the patient did not have typical findings of herpes simplex virus such as grouped vesicles on an erythematous base. Doxycycline (choice D) is not indicated because her bacterial culture result was negative, and she had already been treated with a course of antibiotics.Allergic contact dermatitis (ACD) is a cutaneous type IV (delayed) hypersensitivity reaction caused by activation of allergen-specific T cells that develop after initial exposure to an allergen in contact with the skin.1 Activation of these T cells causes cytokine release and cellular infiltrates that result in the clinical symptoms of ACD.2Acute ACD presents with intense pruritus or burning, and the affected skin typically has a weepy erythematous appearance with surrounding erythematous papulovesicles and honey-colored crusting. Bullae and oozing may arise in severe cases.2,3 Skin findings may occur as early as 1 to 2 days after exposure in sensitized individuals or up to 1 month with initial allergen exposure.4 The differential diagnosis of ACD includes irritant contact dermatitis, atopic dermatitis, seborrheic dermatitis, psoriasis, tinea, mycosis fungoides, and surgical site infection in patients who have undergone recent surgery.2,3ACD affects approximately 72 million individuals in the US annually and is diagnosed more commonly in females than in males.1,2,4 Risk factors for ACD include atopic dermatitis, a family history of ACD, and exposure to acrylates, which are chemical agents commonly used in glues, adhesives, and plastics.1,3 Occupations with increased risk of ACD due to acrylate exposure include medical and dental practitioners, beauticians, chemical plant workers, construction workers, metal workers, and mechanics.1,3Patients who have undergone recent surgery can develop ACD from exposure to tissue adhesives, sutures, staples, antiseptic preparations, topical antibiotics, joint implants, and bone cement5-7 ACD causes inflammation of the skin that may impair wound healing, cause wound dehiscence, and increase the risk of wound infections.5,7ACD due to surgical skin glue occurs in 1.5% to 2.8% of exposed patients,3,4 and the most common allergens are acrylates (methacrylates or cyanoacrylates). ACD caused by one acrylate may lead to cosensitization to another acrylate.1 Surgical skin glue adhesives reside in the dermis and can cause sensitization and skin reactions up to 1 month after application.4 Approximately one-half of patients with allergic contact dermatitis due to surgical skin glue also develop a skin eruption in areas of the body remote from the site of application.4ACD is typically diagnosed with standard skin patch testing performed by an allergist or dermatologist. However, standard patch testing does not include cyanoacrylate, methacrylate, or suture materials and fails to identify approximately 25% of allergens that cause ACD.2,7 A specialized (“open”) patch test can be performed for patients who have exposure to specific known allergens not typically included in standard skin patch testing.2,7Treatment of ACD includes removal of the allergen and daily treatment with topical or systemic steroids for at least 2 to 3 weeks, at which point the skin findings typically resolve.2 However, patients must be informed that ACD will recur with reexposure, so strict avoidance of the specific allergen and cross-reacting allergens is required.2 Patients with a history of ACD due to use of surgical skin glue should inform their surgeon and anesthesiologist prior to surgery to avoid reexposure to an allergen that caused ACD.5An open patch test was performed to assess for an allergic reaction to the sutures and surgical skin glue used during the patient’s mastectomy. At 48 hours, she demonstrated a “strong positive” skin reaction to surgical skin glue (Figure 2). Acetone was used to remove the remaining surgical skin glue on her mastectomy incision sites, and triamcinolone was prescribed (0.1% cream twice daily for 2 weeks). Three days later, the patient noted a substantial decrease in pruritus and erythema at the incision sites. She was advised to avoid future contact with surgical skin glue and other potentially cross-reacting adhesives and was provided a handout detailing safe alternative products. At 2-week follow-up, the skin over her mastectomy incision sites appeared healthy, and the rash on her trunk and upper and lower extremities had resolved.Open skin patch test with materials used during the patient’s mastectomy showed a 2+ (strong positive) reaction to surgical skin glue observed 48 hours after application. No suture reactions occurred.
General
A 42-year-old patient with asthma, seasonal allergies, and invasive ductal breast carcinoma presented to the dermatology clinic for rash and poor wound healing 2 weeks after bilateral mastectomy. Five days after surgery, the patient noted erythema, edema, pain, pruritus, and serous fluid drainage at the mastectomy incision sites (Figure 1). Two days later, they developed an erythematous papulovesicular rash on them trunk and upper and lower extremities. A skin swab was sent for bacterial culture, and they was prescribed cefadroxil (500 mg twice daily for 1 week). The skin swab bacterial culture result was negative, and them skin findings did not improve with antibiotics. The patient reported having a similar episode of rash and delayed wound healing after laparoscopy 1 year prior to presentation.Erythema and crusting along incision sites surrounded by a papulovesicular rash.Perform a skin biopsy at the mastectomy incision sitePrescribe acyclovir (400 mg orally 3 times daily for 7 days)Start topical steroids and perform a skin patch testTreat with doxycycline (100 mg orally twice daily for 7 days)
what would you do next?
What would you do next?
Treat with doxycycline (100 mg orally twice daily for 7 days)
Start topical steroids and perform a skin patch test
Prescribe acyclovir (400 mg orally 3 times daily for 7 days)
Perform a skin biopsy at the mastectomy incision site
b
0
1
1
1
neutral
0
0
42
41-50
null
20
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2807476
A 56-year-old woman with a history of multiple myeloma complicated by therapy-related acute myeloid leukemia was admitted for myeloablative conditioning in preparation for allogeneic hematopoietic cell transplant. Her course was complicated by severe mucositis, acute kidney injury, and neutropenic fever. During her admission, the patient also developed painful necrotic skin lesions, a nonproductive cough, and altered mental status.Physical examination findings demonstrated dusky, purpuric, and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, trunk, and extremities (Figure, A). Laboratory study results revealed a white blood cell count of 150/μL (reference range, 3500/μL to 10 500/μL), an absolute neutrophil count of 130/μL (reference range, 1500/μL to 7400/μL), and a platelet count of 19 ×103/μL (reference range, 150 ×103/μL to 400 ×103/μL). (To convert the white blood cell and neutrophil counts to cells ×109/L, multiply by 0.001; conversion of the platelet count to cells ×109/L is 1:1.) Blood culture results were negative. The results of a computed tomography (CT) scan of the brain showed no acute intracranial hemorrhage, territorial infarction, or mass effect. The results of a CT scan of the chest demonstrated multifocal bilateral ground-glass patchy opacities, scattered solid bilateral pulmonary nodules, and a moderately sized pericardial effusion. A biopsy specimen from a skin nodule on the left lateral thigh was obtained for histopathologic examination and tissue culture, and wet mount preparation of a tissue culture colony was performed (Figure, B).A, Physical examination findings demonstrate dusky and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, and extremities. B, Lactophenol cotton blue staining of a tissue culture colony reveals septated hyphae and oval microconidia. What Is Your Diagnosis?
Disseminated fusariosis
Invasive aspergillosis
Pseudomonal ecthyma gangrenosum
Sweet syndrome
A. Disseminated fusariosis
A
Disseminated fusariosis
Wet mount preparation of a tissue culture mold colony with lactophenol cotton blue staining revealed septated hyphae and oval microconidia (Figure, B). A fungal culture grew Fusarium solani.Fusarium species are ubiquitous in soil and air, and transmission occurs through inhalation or direct cutaneous inoculation from a contaminated source.1 Localized infections can present as onychomycosis, superficial cutaneous infections, and keratitis in immunocompetent hosts; invasive and disseminated disease is more common in those who are immunocompromised, particularly those with neutropenia.2 In those with hematologic cancer, disseminated fusariosis most frequently involves the skin, blood, lungs, and sinuses.3Although the incidence of fusariosis among patients undergoing hematopoietic transplant is less than 1%, invasive fusariosis has a mortality rate of approximately 80% in individuals with hematologic cancer.4 Thus, early recognition is crucial for prompt treatment of the infection. Purpuric patches or papulonodules in this population should be evaluated urgently with skin biopsy and tissue culture. It is also recommended that patients with hematologic cancer who are about to receive chemotherapy and/or bone marrow transplant first undergo a thorough examination for skin findings of onychomycosis, which has been associated with Fusarium infections.1,3,5The patient’s physical examination findings of scattered and painful purpuric papulonodules are similar to those found in invasive aspergillosis, which has an incidence of 2.7% in patients undergoing hematopoietic stem cell transplant.4 Invasive aspergillosis is an infection caused by the fungal Aspergillus species, which is ubiquitous in the environment.6 As transmission is primarily through inhalation, invasive aspergillus most commonly presents with pulmonary or sinus disease that can then progress to dissemination in immunocompromised individuals. Both Aspergillus and Fusarium may cause elevated serum galactomannan antigen. Chest CT findings of invasive aspergillosis and disseminated fusariosis are similar, and both may show ground-glass opacities, segmental consolidation, cavitary lesions, and the “halo sign” (ground-glass opacities representing hemorrhage surrounding central fungal nodules). Lactophenol cotton blue staining of mold colony can differentiate between the 2 pathogens, as Aspergillus species have septate hyphae and flask-shaped vesicles.1,7Ecthyma gangrenosum is a cutaneous infection caused by hematogenous spread of bacteria, most commonly Pseudomonas aeruginosa. It most frequently occurs in immunocompromised individuals, including those with chemotherapy-induced neutropenia.8 As this represents bacterial angioinvasion, ecthyma gangrenosum presents as purpuric macules that may become pustular or bullous before progressing to ulcers with overlying eschar most commonly in the anogenital and axillary areas. In most cases, Pseudomonas can be detected from tissue specimens, but blood culture results are often negative.9Sweet syndrome, also known as acute febrile neutrophilic dermatosis, is a rare disorder that is commonly associated with hematologic cancers, such as acute myeloid leukemia. Sweet syndrome can also be associated with other states, such as autoimmune disease, or be due to medications, such as granulocyte colony-stimulating factor. Classic Sweet syndrome presents as tender, erythematous, and edematous plaques associated with fever and leukocytosis. Cutaneous involvement generally does not show overlying eschar, as seen in disseminated fusariosis. Skin biopsy histopathologic features can mimic infection with dermal edema and a dense infiltrate of neutrophils; however, the results of infectious stains and tissue culture will be negative. Systemic corticosteroids are often first-line treatments for Sweet syndrome.10The patient was treated with intravenous amphotericin B and voriconazole for disseminated fusariosis, with resolution of her cutaneous lesions over the course of 6 weeks. This case highlights the importance of early diagnosis of disseminated fusariosis, especially for those who are immunocompromised or undergoing immunosuppressive therapy.
Oncology
A 56-year-old woman with a history of multiple myeloma complicated by therapy-related acute myeloid leukemia was admitted for myeloablative conditioning in preparation for allogeneic hematopoietic cell transplant. Her course was complicated by severe mucositis, acute kidney injury, and neutropenic fever. During her admission, the patient also developed painful necrotic skin lesions, a nonproductive cough, and altered mental status.Physical examination findings demonstrated dusky, purpuric, and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, trunk, and extremities (Figure, A). Laboratory study results revealed a white blood cell count of 150/μL (reference range, 3500/μL to 10 500/μL), an absolute neutrophil count of 130/μL (reference range, 1500/μL to 7400/μL), and a platelet count of 19 ×103/μL (reference range, 150 ×103/μL to 400 ×103/μL). (To convert the white blood cell and neutrophil counts to cells ×109/L, multiply by 0.001; conversion of the platelet count to cells ×109/L is 1:1.) Blood culture results were negative. The results of a computed tomography (CT) scan of the brain showed no acute intracranial hemorrhage, territorial infarction, or mass effect. The results of a CT scan of the chest demonstrated multifocal bilateral ground-glass patchy opacities, scattered solid bilateral pulmonary nodules, and a moderately sized pericardial effusion. A biopsy specimen from a skin nodule on the left lateral thigh was obtained for histopathologic examination and tissue culture, and wet mount preparation of a tissue culture colony was performed (Figure, B).A, Physical examination findings demonstrate dusky and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, and extremities. B, Lactophenol cotton blue staining of a tissue culture colony reveals septated hyphae and oval microconidia.
what is your diagnosis?
What is your diagnosis?
Pseudomonal ecthyma gangrenosum
Sweet syndrome
Invasive aspergillosis
Disseminated fusariosis
d
1
1
1
1
female
0
0
56
51-60
White
21
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2807476
A 56-year-old woman with a history of multiple myeloma complicated by therapy-related acute myeloid leukemia was admitted for myeloablative conditioning in preparation for allogeneic hematopoietic cell transplant. Her course was complicated by severe mucositis, acute kidney injury, and neutropenic fever. During her admission, the patient also developed painful necrotic skin lesions, a nonproductive cough, and altered mental status.Physical examination findings demonstrated dusky, purpuric, and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, trunk, and extremities (Figure, A). Laboratory study results revealed a white blood cell count of 150/μL (reference range, 3500/μL to 10 500/μL), an absolute neutrophil count of 130/μL (reference range, 1500/μL to 7400/μL), and a platelet count of 19 ×103/μL (reference range, 150 ×103/μL to 400 ×103/μL). (To convert the white blood cell and neutrophil counts to cells ×109/L, multiply by 0.001; conversion of the platelet count to cells ×109/L is 1:1.) Blood culture results were negative. The results of a computed tomography (CT) scan of the brain showed no acute intracranial hemorrhage, territorial infarction, or mass effect. The results of a CT scan of the chest demonstrated multifocal bilateral ground-glass patchy opacities, scattered solid bilateral pulmonary nodules, and a moderately sized pericardial effusion. A biopsy specimen from a skin nodule on the left lateral thigh was obtained for histopathologic examination and tissue culture, and wet mount preparation of a tissue culture colony was performed (Figure, B).A, Physical examination findings demonstrate dusky and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, and extremities. B, Lactophenol cotton blue staining of a tissue culture colony reveals septated hyphae and oval microconidia. What Is Your Diagnosis?
Disseminated fusariosis
Invasive aspergillosis
Pseudomonal ecthyma gangrenosum
Sweet syndrome
A. Disseminated fusariosis
A
Disseminated fusariosis
Wet mount preparation of a tissue culture mold colony with lactophenol cotton blue staining revealed septated hyphae and oval microconidia (Figure, B). A fungal culture grew Fusarium solani.Fusarium species are ubiquitous in soil and air, and transmission occurs through inhalation or direct cutaneous inoculation from a contaminated source.1 Localized infections can present as onychomycosis, superficial cutaneous infections, and keratitis in immunocompetent hosts; invasive and disseminated disease is more common in those who are immunocompromised, particularly those with neutropenia.2 In those with hematologic cancer, disseminated fusariosis most frequently involves the skin, blood, lungs, and sinuses.3Although the incidence of fusariosis among patients undergoing hematopoietic transplant is less than 1%, invasive fusariosis has a mortality rate of approximately 80% in individuals with hematologic cancer.4 Thus, early recognition is crucial for prompt treatment of the infection. Purpuric patches or papulonodules in this population should be evaluated urgently with skin biopsy and tissue culture. It is also recommended that patients with hematologic cancer who are about to receive chemotherapy and/or bone marrow transplant first undergo a thorough examination for skin findings of onychomycosis, which has been associated with Fusarium infections.1,3,5The patient’s physical examination findings of scattered and painful purpuric papulonodules are similar to those found in invasive aspergillosis, which has an incidence of 2.7% in patients undergoing hematopoietic stem cell transplant.4 Invasive aspergillosis is an infection caused by the fungal Aspergillus species, which is ubiquitous in the environment.6 As transmission is primarily through inhalation, invasive aspergillus most commonly presents with pulmonary or sinus disease that can then progress to dissemination in immunocompromised individuals. Both Aspergillus and Fusarium may cause elevated serum galactomannan antigen. Chest CT findings of invasive aspergillosis and disseminated fusariosis are similar, and both may show ground-glass opacities, segmental consolidation, cavitary lesions, and the “halo sign” (ground-glass opacities representing hemorrhage surrounding central fungal nodules). Lactophenol cotton blue staining of mold colony can differentiate between the 2 pathogens, as Aspergillus species have septate hyphae and flask-shaped vesicles.1,7Ecthyma gangrenosum is a cutaneous infection caused by hematogenous spread of bacteria, most commonly Pseudomonas aeruginosa. It most frequently occurs in immunocompromised individuals, including those with chemotherapy-induced neutropenia.8 As this represents bacterial angioinvasion, ecthyma gangrenosum presents as purpuric macules that may become pustular or bullous before progressing to ulcers with overlying eschar most commonly in the anogenital and axillary areas. In most cases, Pseudomonas can be detected from tissue specimens, but blood culture results are often negative.9Sweet syndrome, also known as acute febrile neutrophilic dermatosis, is a rare disorder that is commonly associated with hematologic cancers, such as acute myeloid leukemia. Sweet syndrome can also be associated with other states, such as autoimmune disease, or be due to medications, such as granulocyte colony-stimulating factor. Classic Sweet syndrome presents as tender, erythematous, and edematous plaques associated with fever and leukocytosis. Cutaneous involvement generally does not show overlying eschar, as seen in disseminated fusariosis. Skin biopsy histopathologic features can mimic infection with dermal edema and a dense infiltrate of neutrophils; however, the results of infectious stains and tissue culture will be negative. Systemic corticosteroids are often first-line treatments for Sweet syndrome.10The patient was treated with intravenous amphotericin B and voriconazole for disseminated fusariosis, with resolution of her cutaneous lesions over the course of 6 weeks. This case highlights the importance of early diagnosis of disseminated fusariosis, especially for those who are immunocompromised or undergoing immunosuppressive therapy.
Oncology
A 56-year-old man with a history of multiple myeloma complicated by therapy-related acute myeloid leukemia was admitted for myeloablative conditioning in preparation for allogeneic hematopoietic cell transplant. Him course was complicated by severe mucositis, acute kidney injury, and neutropenic fever. During him admission, the man also developed painful necrotic skin lesions, a nonproductive cough, and altered mental status.Physical examination findings demonstrated dusky, purpuric, and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, trunk, and extremities (Figure, A). Laboratory study results revealed a white blood cell count of 150/μL (reference range, 3500/μL to 10 500/μL), an absolute neutrophil count of 130/μL (reference range, 1500/μL to 7400/μL), and a platelet count of 19 ×103/μL (reference range, 150 ×103/μL to 400 ×103/μL). (To convert the white blood cell and neutrophil counts to cells ×109/L, multiply by 0.001; conversion of the platelet count to cells ×109/L is 1:1.) Blood culture results were negative. The results of a computed tomography (CT) scan of the brain showed no acute intracranial hemorrhage, territorial infarction, or mass effect. The results of a CT scan of the chest demonstrated multifocal bilateral ground-glass patchy opacities, scattered solid bilateral pulmonary nodules, and a moderately sized pericardial effusion. A biopsy specimen from a skin nodule on the left lateral thigh was obtained for histopathologic examination and tissue culture, and wet mount preparation of a tissue culture colony was performed (Figure, B).A, Physical examination findings demonstrate dusky and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, and extremities. B, Lactophenol cotton blue staining of a tissue culture colony reveals septated hyphae and oval microconidia.
what is your diagnosis?
What is your diagnosis?
Pseudomonal ecthyma gangrenosum
Sweet syndrome
Invasive aspergillosis
Disseminated fusariosis
d
1
1
1
1
male
0
0
56
51-60
White
21
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2807476
A 56-year-old woman with a history of multiple myeloma complicated by therapy-related acute myeloid leukemia was admitted for myeloablative conditioning in preparation for allogeneic hematopoietic cell transplant. Her course was complicated by severe mucositis, acute kidney injury, and neutropenic fever. During her admission, the patient also developed painful necrotic skin lesions, a nonproductive cough, and altered mental status.Physical examination findings demonstrated dusky, purpuric, and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, trunk, and extremities (Figure, A). Laboratory study results revealed a white blood cell count of 150/μL (reference range, 3500/μL to 10 500/μL), an absolute neutrophil count of 130/μL (reference range, 1500/μL to 7400/μL), and a platelet count of 19 ×103/μL (reference range, 150 ×103/μL to 400 ×103/μL). (To convert the white blood cell and neutrophil counts to cells ×109/L, multiply by 0.001; conversion of the platelet count to cells ×109/L is 1:1.) Blood culture results were negative. The results of a computed tomography (CT) scan of the brain showed no acute intracranial hemorrhage, territorial infarction, or mass effect. The results of a CT scan of the chest demonstrated multifocal bilateral ground-glass patchy opacities, scattered solid bilateral pulmonary nodules, and a moderately sized pericardial effusion. A biopsy specimen from a skin nodule on the left lateral thigh was obtained for histopathologic examination and tissue culture, and wet mount preparation of a tissue culture colony was performed (Figure, B).A, Physical examination findings demonstrate dusky and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, and extremities. B, Lactophenol cotton blue staining of a tissue culture colony reveals septated hyphae and oval microconidia. What Is Your Diagnosis?
Disseminated fusariosis
Invasive aspergillosis
Pseudomonal ecthyma gangrenosum
Sweet syndrome
A. Disseminated fusariosis
A
Disseminated fusariosis
Wet mount preparation of a tissue culture mold colony with lactophenol cotton blue staining revealed septated hyphae and oval microconidia (Figure, B). A fungal culture grew Fusarium solani.Fusarium species are ubiquitous in soil and air, and transmission occurs through inhalation or direct cutaneous inoculation from a contaminated source.1 Localized infections can present as onychomycosis, superficial cutaneous infections, and keratitis in immunocompetent hosts; invasive and disseminated disease is more common in those who are immunocompromised, particularly those with neutropenia.2 In those with hematologic cancer, disseminated fusariosis most frequently involves the skin, blood, lungs, and sinuses.3Although the incidence of fusariosis among patients undergoing hematopoietic transplant is less than 1%, invasive fusariosis has a mortality rate of approximately 80% in individuals with hematologic cancer.4 Thus, early recognition is crucial for prompt treatment of the infection. Purpuric patches or papulonodules in this population should be evaluated urgently with skin biopsy and tissue culture. It is also recommended that patients with hematologic cancer who are about to receive chemotherapy and/or bone marrow transplant first undergo a thorough examination for skin findings of onychomycosis, which has been associated with Fusarium infections.1,3,5The patient’s physical examination findings of scattered and painful purpuric papulonodules are similar to those found in invasive aspergillosis, which has an incidence of 2.7% in patients undergoing hematopoietic stem cell transplant.4 Invasive aspergillosis is an infection caused by the fungal Aspergillus species, which is ubiquitous in the environment.6 As transmission is primarily through inhalation, invasive aspergillus most commonly presents with pulmonary or sinus disease that can then progress to dissemination in immunocompromised individuals. Both Aspergillus and Fusarium may cause elevated serum galactomannan antigen. Chest CT findings of invasive aspergillosis and disseminated fusariosis are similar, and both may show ground-glass opacities, segmental consolidation, cavitary lesions, and the “halo sign” (ground-glass opacities representing hemorrhage surrounding central fungal nodules). Lactophenol cotton blue staining of mold colony can differentiate between the 2 pathogens, as Aspergillus species have septate hyphae and flask-shaped vesicles.1,7Ecthyma gangrenosum is a cutaneous infection caused by hematogenous spread of bacteria, most commonly Pseudomonas aeruginosa. It most frequently occurs in immunocompromised individuals, including those with chemotherapy-induced neutropenia.8 As this represents bacterial angioinvasion, ecthyma gangrenosum presents as purpuric macules that may become pustular or bullous before progressing to ulcers with overlying eschar most commonly in the anogenital and axillary areas. In most cases, Pseudomonas can be detected from tissue specimens, but blood culture results are often negative.9Sweet syndrome, also known as acute febrile neutrophilic dermatosis, is a rare disorder that is commonly associated with hematologic cancers, such as acute myeloid leukemia. Sweet syndrome can also be associated with other states, such as autoimmune disease, or be due to medications, such as granulocyte colony-stimulating factor. Classic Sweet syndrome presents as tender, erythematous, and edematous plaques associated with fever and leukocytosis. Cutaneous involvement generally does not show overlying eschar, as seen in disseminated fusariosis. Skin biopsy histopathologic features can mimic infection with dermal edema and a dense infiltrate of neutrophils; however, the results of infectious stains and tissue culture will be negative. Systemic corticosteroids are often first-line treatments for Sweet syndrome.10The patient was treated with intravenous amphotericin B and voriconazole for disseminated fusariosis, with resolution of her cutaneous lesions over the course of 6 weeks. This case highlights the importance of early diagnosis of disseminated fusariosis, especially for those who are immunocompromised or undergoing immunosuppressive therapy.
Oncology
A 56-year-old patient with a history of multiple myeloma complicated by therapy-related acute myeloid leukemia was admitted for myeloablative conditioning in preparation for allogeneic hematopoietic cell transplant. Them course was complicated by severe mucositis, acute kidney injury, and neutropenic fever. During them admission, the patient also developed painful necrotic skin lesions, a nonproductive cough, and altered mental status.Physical examination findings demonstrated dusky, purpuric, and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, trunk, and extremities (Figure, A). Laboratory study results revealed a white blood cell count of 150/μL (reference range, 3500/μL to 10 500/μL), an absolute neutrophil count of 130/μL (reference range, 1500/μL to 7400/μL), and a platelet count of 19 ×103/μL (reference range, 150 ×103/μL to 400 ×103/μL). (To convert the white blood cell and neutrophil counts to cells ×109/L, multiply by 0.001; conversion of the platelet count to cells ×109/L is 1:1.) Blood culture results were negative. The results of a computed tomography (CT) scan of the brain showed no acute intracranial hemorrhage, territorial infarction, or mass effect. The results of a CT scan of the chest demonstrated multifocal bilateral ground-glass patchy opacities, scattered solid bilateral pulmonary nodules, and a moderately sized pericardial effusion. A biopsy specimen from a skin nodule on the left lateral thigh was obtained for histopathologic examination and tissue culture, and wet mount preparation of a tissue culture colony was performed (Figure, B).A, Physical examination findings demonstrate dusky and pink papulonodules, some with overlying eschar, scattered on the scalp, neck, and extremities. B, Lactophenol cotton blue staining of a tissue culture colony reveals septated hyphae and oval microconidia.
what is your diagnosis?
What is your diagnosis?
Pseudomonal ecthyma gangrenosum
Sweet syndrome
Invasive aspergillosis
Disseminated fusariosis
d
1
1
1
1
neutral
0
0
56
51-60
White
21
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2807704
A man in his 80s presented to the dermatology clinic with a 3-month history of a violaceous plaque with blackish papules and nodules on his left cheek, neck, and chest. He reported that the lesion had been asymptomatic but was increasing in size, with progression from his face to anterior chest. He received a diagnosis of primary salivary duct carcinoma of the hard palate 1 year prior and had been receiving treatment with oral bicalutamide and radiotherapy. A partially regressive change of the palatal tumor was noted radiographically during regular follow-up 3 months previously. On clinical examination, a hemorrhagic and erythematous plaque with multiple purpuric-to-blackish infiltrative papules and nodules extending from cheek, lateral neck, to the interclavicular area was found (Figure 1). An incisional biopsy from the infiltrative nodule on cheek was performed and submitted for histopathologic analysis.Hemorrhagic and erythematous plaque with multiple infiltrative purpuric-to-blackish papules and nodules with varying sizes extending from left cheek, neck, to chest. What Is Your Diagnosis?
Radiotherapy-induced epithelioid angiosarcoma
Kaposi sarcoma
Acquired angiokeratomas
Carcinoma hemorrhagiectoides
D. Carcinoma hemorrhagiectoides
D
Carcinoma hemorrhagiectoides
Hematoxylin-eosin staining of the lesion revealed a diffuse infiltrate of large, ovoid tumor cells that formed tumor nests and tumor emboli, with marked erythrocytes extravasation in the dermis (Figure 2A). The cells had an abundant eosinophilic cytoplasm and were hyperchromatic, with nuclear pleomorphism and comedonecrosis (Figure 2B). Androgen receptor and GATA3 were diffusely strong positive in the tumor cells, while other endothelial markers, including CD31, CD34, and ERG1, were negative. A diagnosis of carcinoma hemorrhagiectoides due to metastatic salivary duct carcinoma (SDC) was made. Further image study results revealed multiple metastases in the lung, liver, and bone. A combination of oral bicalutamide and chemotherapy with carboplatin and fluorouracil was administered. Four months later, partial regression of the liver and lung metastases was noted radiographically.Hematoxylin-eosin staining of the lesion from the biopsy specimens. A, Scanning magnification showed a diffuse infiltrate of tumor cells with marked erythrocyte extravasation. B, The tumor cells were characterized by an abundant eosinophilic cytoplasm, nuclear atypia, and pleomorphism, with comedonecrosis on higher magnification.Salivary duct carcinoma is an aggressive cancer of the primary salivary gland origin and is generally associated with a high rate of local recurrence and distant metastasis.1 It accounts for 5% to 10% of salivary gland cancer and has a tendency of developing in male individuals older than 50 years. While the most common sites of metastatic SDC are the lungs and bones,2 rare cases of cutaneous metastasis have been reported.3-5 Clinically, metastatic SDC on the skin may present as pink pebble-like papules, subcutaneous nodules, or, rarely, as carcinoma hemorrhagiectoides, as in this case. Carcinoma hemorrhagiectoides is characterized as a purpuric indurated plaque with overlying angiokeratoma-like black keratotic papules and nodules. A shield sign, a term derived from the similarity in shape to a medieval knight’s shield, has been described on the anterior chest of affected individuals.4 Studies have suggested that the pathomechanism of carcinoma hemorrhagiectoides may be associated with direct tumor invasion of blood vessels and lymphatic ducts in the dermis.4 A biopsy was needed for exclusion of other differential diagnoses in this case. The histopathological findings of SDC typically show tumor cells with abundant eosinophilic cytoplasm, large pleomorphic nuclei, and conspicuous nucleoli. Comedonecrosis and a cribriform growth pattern can be variably presented. Immunohistochemically, the tumor cells show a strong and diffuse positive staining for androgen receptor in more than 90% of the cases.1 GATA3 and GCDFP-15 positivity with negative p63 expression can also aid the diagnosis toward SDC. In addition, the histopathologic and immunohistochemical profiles of metastatic SDC may closely resemble invasive ductal carcinoma of the breast; thus, a definite diagnosis is made on a thorough clinicopathological correlation.The differential diagnosis in this case included radiotherapy-induced epithelioid angiosarcoma, Kaposi sarcoma (KS), and acquired angiokeratomas. Radiotherapy-induced angiosarcoma is a late complication of radiotherapy, with a median latency period of 8 years after treatment.6 It occurs mostly in those with breast and gynecological cancers7 and typically presents as a diffuse purplish discoloration in the previously irradiated skin area. Pathologically, crowded atypical endothelial cells with nuclear atypia, necrosis, hemorrhage, occasional formation of intracytoplasmic lumina, and positivity for endothelial cell markers, such as CD31, CD34, and ERG1, can be found. Epithelioid angiosarcoma is a high-grade variant of angiosarcoma with poor prognosis and should be carefully distinguished from this case, given that it is composed of mostly epithelioid cells with minimal vasoformation.8 Additional immunohistochemical features of epithelioid angiosarcoma include positivity of Friend leukemia integration 1 transcription factor and von Willebrand factor, variable positivity for creatine kinase, focal positivity of epithelial membrane antigen A, and negativity of CD34. Kaposi sarcoma is an angioproliferative disorder due to human herpesvirus type 8 infection with intermediate cancer potential. It mostly involves the trunk, limbs, and mucosa as violaceous patches, plaques, or nodules, while craniofacial manifestation is less observed except in AIDS-associated KS.9 The pathological features depend on the different stages of the disease, as characterized by slit-like spaces and a proliferation of bland spindle cells stating positive for CD31 and herpesvirus type 8. Lastly, acquired angiokeratomas are characterized by multiple bluish-to-reddish hyperkeratotic papules with a wide range of anatomic distribution. Microscopically, dilated vascular spaces in the dermis extending into the epidermis with hyperkeratosis, acanthosis, papillomatosis, and elongation of the rete ridges can be found.
Dermatology
A man in his 80s presented to the dermatology clinic with a 3-month history of a violaceous plaque with blackish papules and nodules on his left cheek, neck, and chest. He reported that the lesion had been asymptomatic but was increasing in size, with progression from his face to anterior chest. He received a diagnosis of primary salivary duct carcinoma of the hard palate 1 year prior and had been receiving treatment with oral bicalutamide and radiotherapy. A partially regressive change of the palatal tumor was noted radiographically during regular follow-up 3 months previously. On clinical examination, a hemorrhagic and erythematous plaque with multiple purpuric-to-blackish infiltrative papules and nodules extending from cheek, lateral neck, to the interclavicular area was found (Figure 1). An incisional biopsy from the infiltrative nodule on cheek was performed and submitted for histopathologic analysis.Hemorrhagic and erythematous plaque with multiple infiltrative purpuric-to-blackish papules and nodules with varying sizes extending from left cheek, neck, to chest.
what is your diagnosis?
What is your diagnosis?
Acquired angiokeratomas
Kaposi sarcoma
Radiotherapy-induced epithelioid angiosarcoma
Carcinoma hemorrhagiectoides
d
0
1
1
1
male
0
0
85
81-90
null
22
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2807704
A man in his 80s presented to the dermatology clinic with a 3-month history of a violaceous plaque with blackish papules and nodules on his left cheek, neck, and chest. He reported that the lesion had been asymptomatic but was increasing in size, with progression from his face to anterior chest. He received a diagnosis of primary salivary duct carcinoma of the hard palate 1 year prior and had been receiving treatment with oral bicalutamide and radiotherapy. A partially regressive change of the palatal tumor was noted radiographically during regular follow-up 3 months previously. On clinical examination, a hemorrhagic and erythematous plaque with multiple purpuric-to-blackish infiltrative papules and nodules extending from cheek, lateral neck, to the interclavicular area was found (Figure 1). An incisional biopsy from the infiltrative nodule on cheek was performed and submitted for histopathologic analysis.Hemorrhagic and erythematous plaque with multiple infiltrative purpuric-to-blackish papules and nodules with varying sizes extending from left cheek, neck, to chest. What Is Your Diagnosis?
Radiotherapy-induced epithelioid angiosarcoma
Kaposi sarcoma
Acquired angiokeratomas
Carcinoma hemorrhagiectoides
D. Carcinoma hemorrhagiectoides
D
Carcinoma hemorrhagiectoides
Hematoxylin-eosin staining of the lesion revealed a diffuse infiltrate of large, ovoid tumor cells that formed tumor nests and tumor emboli, with marked erythrocytes extravasation in the dermis (Figure 2A). The cells had an abundant eosinophilic cytoplasm and were hyperchromatic, with nuclear pleomorphism and comedonecrosis (Figure 2B). Androgen receptor and GATA3 were diffusely strong positive in the tumor cells, while other endothelial markers, including CD31, CD34, and ERG1, were negative. A diagnosis of carcinoma hemorrhagiectoides due to metastatic salivary duct carcinoma (SDC) was made. Further image study results revealed multiple metastases in the lung, liver, and bone. A combination of oral bicalutamide and chemotherapy with carboplatin and fluorouracil was administered. Four months later, partial regression of the liver and lung metastases was noted radiographically.Hematoxylin-eosin staining of the lesion from the biopsy specimens. A, Scanning magnification showed a diffuse infiltrate of tumor cells with marked erythrocyte extravasation. B, The tumor cells were characterized by an abundant eosinophilic cytoplasm, nuclear atypia, and pleomorphism, with comedonecrosis on higher magnification.Salivary duct carcinoma is an aggressive cancer of the primary salivary gland origin and is generally associated with a high rate of local recurrence and distant metastasis.1 It accounts for 5% to 10% of salivary gland cancer and has a tendency of developing in male individuals older than 50 years. While the most common sites of metastatic SDC are the lungs and bones,2 rare cases of cutaneous metastasis have been reported.3-5 Clinically, metastatic SDC on the skin may present as pink pebble-like papules, subcutaneous nodules, or, rarely, as carcinoma hemorrhagiectoides, as in this case. Carcinoma hemorrhagiectoides is characterized as a purpuric indurated plaque with overlying angiokeratoma-like black keratotic papules and nodules. A shield sign, a term derived from the similarity in shape to a medieval knight’s shield, has been described on the anterior chest of affected individuals.4 Studies have suggested that the pathomechanism of carcinoma hemorrhagiectoides may be associated with direct tumor invasion of blood vessels and lymphatic ducts in the dermis.4 A biopsy was needed for exclusion of other differential diagnoses in this case. The histopathological findings of SDC typically show tumor cells with abundant eosinophilic cytoplasm, large pleomorphic nuclei, and conspicuous nucleoli. Comedonecrosis and a cribriform growth pattern can be variably presented. Immunohistochemically, the tumor cells show a strong and diffuse positive staining for androgen receptor in more than 90% of the cases.1 GATA3 and GCDFP-15 positivity with negative p63 expression can also aid the diagnosis toward SDC. In addition, the histopathologic and immunohistochemical profiles of metastatic SDC may closely resemble invasive ductal carcinoma of the breast; thus, a definite diagnosis is made on a thorough clinicopathological correlation.The differential diagnosis in this case included radiotherapy-induced epithelioid angiosarcoma, Kaposi sarcoma (KS), and acquired angiokeratomas. Radiotherapy-induced angiosarcoma is a late complication of radiotherapy, with a median latency period of 8 years after treatment.6 It occurs mostly in those with breast and gynecological cancers7 and typically presents as a diffuse purplish discoloration in the previously irradiated skin area. Pathologically, crowded atypical endothelial cells with nuclear atypia, necrosis, hemorrhage, occasional formation of intracytoplasmic lumina, and positivity for endothelial cell markers, such as CD31, CD34, and ERG1, can be found. Epithelioid angiosarcoma is a high-grade variant of angiosarcoma with poor prognosis and should be carefully distinguished from this case, given that it is composed of mostly epithelioid cells with minimal vasoformation.8 Additional immunohistochemical features of epithelioid angiosarcoma include positivity of Friend leukemia integration 1 transcription factor and von Willebrand factor, variable positivity for creatine kinase, focal positivity of epithelial membrane antigen A, and negativity of CD34. Kaposi sarcoma is an angioproliferative disorder due to human herpesvirus type 8 infection with intermediate cancer potential. It mostly involves the trunk, limbs, and mucosa as violaceous patches, plaques, or nodules, while craniofacial manifestation is less observed except in AIDS-associated KS.9 The pathological features depend on the different stages of the disease, as characterized by slit-like spaces and a proliferation of bland spindle cells stating positive for CD31 and herpesvirus type 8. Lastly, acquired angiokeratomas are characterized by multiple bluish-to-reddish hyperkeratotic papules with a wide range of anatomic distribution. Microscopically, dilated vascular spaces in the dermis extending into the epidermis with hyperkeratosis, acanthosis, papillomatosis, and elongation of the rete ridges can be found.
Dermatology
A woman in her 80s presented to the dermatology clinic with a 3-month history of a violaceous plaque with blackish papules and nodules on her left cheek, neck, and chest. She reported that the lesion had been asymptomatic but was increasing in size, with progression from her face to anterior chest. She received a diagnosis of primary salivary duct carcinoma of the hard palate 1 year prior and had been receiving treatment with oral bicalutamide and radiotherapy. A partially regressive change of the palatal tumor was noted radiographically during regular follow-up 3 months previously. On clinical examination, a hemorrhagic and erythematous plaque with multiple purpuric-to-blackish infiltrative papules and nodules extending from cheek, lateral neck, to the interclavicular area was found (Figure 1). An incisional biopsy from the infiltrative nodule on cheek was performed and submitted for histopathologic analysis.Hemorrhagic and erythematous plaque with multiple infiltrative purpuric-to-blackish papules and nodules with varying sizes extending from left cheek, neck, to chest.
what is your diagnosis?
What is your diagnosis?
Acquired angiokeratomas
Kaposi sarcoma
Radiotherapy-induced epithelioid angiosarcoma
Carcinoma hemorrhagiectoides
d
0
1
1
1
female
0
0
85
81-90
null
22
augmented_female_frommale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2807704
A man in his 80s presented to the dermatology clinic with a 3-month history of a violaceous plaque with blackish papules and nodules on his left cheek, neck, and chest. He reported that the lesion had been asymptomatic but was increasing in size, with progression from his face to anterior chest. He received a diagnosis of primary salivary duct carcinoma of the hard palate 1 year prior and had been receiving treatment with oral bicalutamide and radiotherapy. A partially regressive change of the palatal tumor was noted radiographically during regular follow-up 3 months previously. On clinical examination, a hemorrhagic and erythematous plaque with multiple purpuric-to-blackish infiltrative papules and nodules extending from cheek, lateral neck, to the interclavicular area was found (Figure 1). An incisional biopsy from the infiltrative nodule on cheek was performed and submitted for histopathologic analysis.Hemorrhagic and erythematous plaque with multiple infiltrative purpuric-to-blackish papules and nodules with varying sizes extending from left cheek, neck, to chest. What Is Your Diagnosis?
Radiotherapy-induced epithelioid angiosarcoma
Kaposi sarcoma
Acquired angiokeratomas
Carcinoma hemorrhagiectoides
D. Carcinoma hemorrhagiectoides
D
Carcinoma hemorrhagiectoides
Hematoxylin-eosin staining of the lesion revealed a diffuse infiltrate of large, ovoid tumor cells that formed tumor nests and tumor emboli, with marked erythrocytes extravasation in the dermis (Figure 2A). The cells had an abundant eosinophilic cytoplasm and were hyperchromatic, with nuclear pleomorphism and comedonecrosis (Figure 2B). Androgen receptor and GATA3 were diffusely strong positive in the tumor cells, while other endothelial markers, including CD31, CD34, and ERG1, were negative. A diagnosis of carcinoma hemorrhagiectoides due to metastatic salivary duct carcinoma (SDC) was made. Further image study results revealed multiple metastases in the lung, liver, and bone. A combination of oral bicalutamide and chemotherapy with carboplatin and fluorouracil was administered. Four months later, partial regression of the liver and lung metastases was noted radiographically.Hematoxylin-eosin staining of the lesion from the biopsy specimens. A, Scanning magnification showed a diffuse infiltrate of tumor cells with marked erythrocyte extravasation. B, The tumor cells were characterized by an abundant eosinophilic cytoplasm, nuclear atypia, and pleomorphism, with comedonecrosis on higher magnification.Salivary duct carcinoma is an aggressive cancer of the primary salivary gland origin and is generally associated with a high rate of local recurrence and distant metastasis.1 It accounts for 5% to 10% of salivary gland cancer and has a tendency of developing in male individuals older than 50 years. While the most common sites of metastatic SDC are the lungs and bones,2 rare cases of cutaneous metastasis have been reported.3-5 Clinically, metastatic SDC on the skin may present as pink pebble-like papules, subcutaneous nodules, or, rarely, as carcinoma hemorrhagiectoides, as in this case. Carcinoma hemorrhagiectoides is characterized as a purpuric indurated plaque with overlying angiokeratoma-like black keratotic papules and nodules. A shield sign, a term derived from the similarity in shape to a medieval knight’s shield, has been described on the anterior chest of affected individuals.4 Studies have suggested that the pathomechanism of carcinoma hemorrhagiectoides may be associated with direct tumor invasion of blood vessels and lymphatic ducts in the dermis.4 A biopsy was needed for exclusion of other differential diagnoses in this case. The histopathological findings of SDC typically show tumor cells with abundant eosinophilic cytoplasm, large pleomorphic nuclei, and conspicuous nucleoli. Comedonecrosis and a cribriform growth pattern can be variably presented. Immunohistochemically, the tumor cells show a strong and diffuse positive staining for androgen receptor in more than 90% of the cases.1 GATA3 and GCDFP-15 positivity with negative p63 expression can also aid the diagnosis toward SDC. In addition, the histopathologic and immunohistochemical profiles of metastatic SDC may closely resemble invasive ductal carcinoma of the breast; thus, a definite diagnosis is made on a thorough clinicopathological correlation.The differential diagnosis in this case included radiotherapy-induced epithelioid angiosarcoma, Kaposi sarcoma (KS), and acquired angiokeratomas. Radiotherapy-induced angiosarcoma is a late complication of radiotherapy, with a median latency period of 8 years after treatment.6 It occurs mostly in those with breast and gynecological cancers7 and typically presents as a diffuse purplish discoloration in the previously irradiated skin area. Pathologically, crowded atypical endothelial cells with nuclear atypia, necrosis, hemorrhage, occasional formation of intracytoplasmic lumina, and positivity for endothelial cell markers, such as CD31, CD34, and ERG1, can be found. Epithelioid angiosarcoma is a high-grade variant of angiosarcoma with poor prognosis and should be carefully distinguished from this case, given that it is composed of mostly epithelioid cells with minimal vasoformation.8 Additional immunohistochemical features of epithelioid angiosarcoma include positivity of Friend leukemia integration 1 transcription factor and von Willebrand factor, variable positivity for creatine kinase, focal positivity of epithelial membrane antigen A, and negativity of CD34. Kaposi sarcoma is an angioproliferative disorder due to human herpesvirus type 8 infection with intermediate cancer potential. It mostly involves the trunk, limbs, and mucosa as violaceous patches, plaques, or nodules, while craniofacial manifestation is less observed except in AIDS-associated KS.9 The pathological features depend on the different stages of the disease, as characterized by slit-like spaces and a proliferation of bland spindle cells stating positive for CD31 and herpesvirus type 8. Lastly, acquired angiokeratomas are characterized by multiple bluish-to-reddish hyperkeratotic papules with a wide range of anatomic distribution. Microscopically, dilated vascular spaces in the dermis extending into the epidermis with hyperkeratosis, acanthosis, papillomatosis, and elongation of the rete ridges can be found.
Dermatology
A patient in their 80s presented to the dermatology clinic with a 3-month history of a violaceous plaque with blackish papules and nodules on their left cheek, neck, and chest. They reported that the lesion had been asymptomatic but was increasing in size, with progression from their face to anterior chest. They received a diagnosis of primary salivary duct carcinoma of the hard palate 1 year prior and had been receiving treatment with oral bicalutamide and radiotherapy. A partially regressive change of the palatal tumor was noted radiographically during regular follow-up 3 months previously. On clinical examination, a hemorrhagic and erythematous plaque with multiple purpuric-to-blackish infiltrative papules and nodules extending from cheek, lateral neck, to the interclavicular area was found (Figure 1). An incisional biopsy from the infiltrative nodule on cheek was performed and submitted for histopathologic analysis.Hemorrhagic and erythematous plaque with multiple infiltrative purpuric-to-blackish papules and nodules with varying sizes extending from left cheek, neck, to chest.
what is your diagnosis?
What is your diagnosis?
Acquired angiokeratomas
Kaposi sarcoma
Radiotherapy-induced epithelioid angiosarcoma
Carcinoma hemorrhagiectoides
d
0
1
1
1
neutral
0
0
85
81-90
null
22
augmented_neutral_frommale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2808313
A previously healthy 16-year-old girl presented to our department with a red tumor on her right upper arm that enlarged over 6 months. The lesion initially started as a small cutaneous nodule without obvious triggers that gradually developed and enlarged to form a red-colored tumor with a hard nodule inside. There was no history of local trauma or insect bite. Her personal, past, and family histories were unremarkable.On physical examination, there was a 6 × 6-cm protuberant, thick-walled, and well-defined red bullalike tumor on the right upper arm, which extended 1 to 3 cm from the epidermal surface (Figure, A). Inside the tumor, a nontender, firm-to-hard nodule was palpated. There was no regional lymphadenopathy, and results of the rest of the physical and systemic examinations were normal. Laboratory examinations of hematologic, biochemical, and urinalysis tests were normal. The tumor was excised completely under local anesthesia, and part of the tissue was sent for pathological examination.A, A tumor on the patient’s right shoulder. B, The excisional tumor showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance. C and D, Histological examination revealed basophilic cells and shadow cells as well as dilated lymphatic vessels (hematoxylin-eosin; original magnification ×100). What Is Your Diagnosis?
Matrical carcinoma
Panfolliculoma
Bullous pilomatricoma
Trichodiscoma
C. Bullous pilomatricoma
C
Bullous pilomatricoma
The gross specimen of excised material showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance (Figure, B). Histopathological examination revealed basophilic cells and shadow cells in the deep dermis (Figure, C) and dilated lymphatic vessels in the dermis above the tumor (Figure, D).Based on the clinical morphology and histological features, the diagnosis of bullous pilomatricoma was confirmed. The patient is under regular outpatient follow-up, with no recurrence detected in 1 year of follow-up.Bullous pilomatricoma is a rare variant of pilomatricoma with a bullous appearance.1 It most commonly occurs on the shoulder and upper arm but also can be found on the neck, trunk, eyelid, and scalp. The peak age of presentation is 10 to 20 years old, and there is a predilection for female individuals.2 Clinically, bullous pilomatricoma usually presents as a solitary, semitransparent, erythematous, bluish or skin-colored, heavily folded or striaelike, flaccid, thick-walled, bullalike tumor with an underlying palpable hard nodule.1-3 It is rare for patients with bullous pilomatricoma to have multiple lesions.4The present patient’s lesion was increasing in size gradually but always had a clear border. When the bullalike tumor was punctured, a jellylike substance could be seen exuding out.4 Unlike some typical pilomatricomas, bullous pilomatricoma is not associated with Turner syndrome, myotonic dystrophy, Gardner syndrome, or Rubinstein-Taybi syndrome.2The etiopathogenesis of bullous pilomatricoma is not clear. There are 2 proposed theories. In the first, obstruction of lymphatic vessels and congestion of lymphatic fluid caused by the growth of the tumor nodule causes dilatation of lymphatic vessels, leakage of lymphatic fluid, and edema in the dermis surrounding the tumor, which further produces the bullous appearance.2,5 The second theory suggests that the presence of matrix metalloproteinases (MMP-9 and MMP-12), released by fibroblasts and inflammatory cells surrounding the tumor, may be associated with the degradation of elastic fibers and collagen, resulting in the bullouslike/anetodermic appearance.6Histologically, bullous pilomatricoma not only has the hallmarks of pilomatricoma, including basaloid cells and eosinophilic keratinized (shadow) cells, but also shows dilated lymphatic vessels, giant cell reaction, lymphoedema, disruption of collagen fibers, dilated blood vessels, fibrous capsule, calcification, nests of transitional cells, and necrosis.2 The presence of dilated lymphatic vessels and lymphedema have been described as a common pathologic feature in bullous pilomatricomas2,7 and can be used to distinguish bullous pilomatricomas from common pilomatricomas. Moreover, as a low-cost, noninvasive tool, ultrasonography (specifically high-frequency ultrasonography8) has been used as a diagnostic tool and is more appropriate for younger children to demonstrate the superficial position, continuity of the lesion with deeper structures, and degree of calcification.The diagnosis of bullous pilomatricoma may be difficult clinically, due to its atypical presentation. The common differential diagnoses should include matrical carcinoma, panfolliculoma, and trichodiscoma. Matrical carcinoma is a locally aggressive tumor, which is typically featured with a nontender, firm dermal swollen nodule and occurs most commonly in the area of the head and neck. Histologically, both matrical carcinomas and pilomatricomas are composed of nodular aggregates of basaloid keratinocytes that transform into shadow cells.9 Cellular pleomorphism and asymmetry, undefined circumscription, and tumor necrosis can help distinguish these 2 entities. Panfolliculoma typically presents as a slow-growing cystic or dome-shaped nodule, which may develop ulcers and crusts in the center, mainly affecting the face and scalp, though sometimes the limbs, of elderly patients.10 Histologically, cellular aggregates can differentiate into any components of the hair follicle, not just the hair matrix cells and shadow cells. Trichodiscoma, a follicular mesenchymal hamartoma, is characterized by multiple, small, 2- to 4-mm, dome-shaped, skin-colored papules on the face, scalp, ears, or upper trunk. Histopathological examination can show a central zone of fibro-mucinous stroma, with surrounding sebaceous glands in a mittlike configuration. The first-line treatment is surgical excision or curettage, and incomplete resection almost always results in recurrence.
Dermatology
A previously healthy 16-year-old girl presented to our department with a red tumor on her right upper arm that enlarged over 6 months. The lesion initially started as a small cutaneous nodule without obvious triggers that gradually developed and enlarged to form a red-colored tumor with a hard nodule inside. There was no history of local trauma or insect bite. Her personal, past, and family histories were unremarkable.On physical examination, there was a 6 × 6-cm protuberant, thick-walled, and well-defined red bullalike tumor on the right upper arm, which extended 1 to 3 cm from the epidermal surface (Figure, A). Inside the tumor, a nontender, firm-to-hard nodule was palpated. There was no regional lymphadenopathy, and results of the rest of the physical and systemic examinations were normal. Laboratory examinations of hematologic, biochemical, and urinalysis tests were normal. The tumor was excised completely under local anesthesia, and part of the tissue was sent for pathological examination.A, A tumor on the patient’s right shoulder. B, The excisional tumor showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance. C and D, Histological examination revealed basophilic cells and shadow cells as well as dilated lymphatic vessels (hematoxylin-eosin; original magnification ×100).
what is your diagnosis?
What is your diagnosis?
Trichodiscoma
Panfolliculoma
Matrical carcinoma
Bullous pilomatricoma
d
0
1
0
1
female
0
0
16
11-20
White
23
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2808313
A previously healthy 16-year-old girl presented to our department with a red tumor on her right upper arm that enlarged over 6 months. The lesion initially started as a small cutaneous nodule without obvious triggers that gradually developed and enlarged to form a red-colored tumor with a hard nodule inside. There was no history of local trauma or insect bite. Her personal, past, and family histories were unremarkable.On physical examination, there was a 6 × 6-cm protuberant, thick-walled, and well-defined red bullalike tumor on the right upper arm, which extended 1 to 3 cm from the epidermal surface (Figure, A). Inside the tumor, a nontender, firm-to-hard nodule was palpated. There was no regional lymphadenopathy, and results of the rest of the physical and systemic examinations were normal. Laboratory examinations of hematologic, biochemical, and urinalysis tests were normal. The tumor was excised completely under local anesthesia, and part of the tissue was sent for pathological examination.A, A tumor on the patient’s right shoulder. B, The excisional tumor showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance. C and D, Histological examination revealed basophilic cells and shadow cells as well as dilated lymphatic vessels (hematoxylin-eosin; original magnification ×100). What Is Your Diagnosis?
Matrical carcinoma
Panfolliculoma
Bullous pilomatricoma
Trichodiscoma
C. Bullous pilomatricoma
C
Bullous pilomatricoma
The gross specimen of excised material showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance (Figure, B). Histopathological examination revealed basophilic cells and shadow cells in the deep dermis (Figure, C) and dilated lymphatic vessels in the dermis above the tumor (Figure, D).Based on the clinical morphology and histological features, the diagnosis of bullous pilomatricoma was confirmed. The patient is under regular outpatient follow-up, with no recurrence detected in 1 year of follow-up.Bullous pilomatricoma is a rare variant of pilomatricoma with a bullous appearance.1 It most commonly occurs on the shoulder and upper arm but also can be found on the neck, trunk, eyelid, and scalp. The peak age of presentation is 10 to 20 years old, and there is a predilection for female individuals.2 Clinically, bullous pilomatricoma usually presents as a solitary, semitransparent, erythematous, bluish or skin-colored, heavily folded or striaelike, flaccid, thick-walled, bullalike tumor with an underlying palpable hard nodule.1-3 It is rare for patients with bullous pilomatricoma to have multiple lesions.4The present patient’s lesion was increasing in size gradually but always had a clear border. When the bullalike tumor was punctured, a jellylike substance could be seen exuding out.4 Unlike some typical pilomatricomas, bullous pilomatricoma is not associated with Turner syndrome, myotonic dystrophy, Gardner syndrome, or Rubinstein-Taybi syndrome.2The etiopathogenesis of bullous pilomatricoma is not clear. There are 2 proposed theories. In the first, obstruction of lymphatic vessels and congestion of lymphatic fluid caused by the growth of the tumor nodule causes dilatation of lymphatic vessels, leakage of lymphatic fluid, and edema in the dermis surrounding the tumor, which further produces the bullous appearance.2,5 The second theory suggests that the presence of matrix metalloproteinases (MMP-9 and MMP-12), released by fibroblasts and inflammatory cells surrounding the tumor, may be associated with the degradation of elastic fibers and collagen, resulting in the bullouslike/anetodermic appearance.6Histologically, bullous pilomatricoma not only has the hallmarks of pilomatricoma, including basaloid cells and eosinophilic keratinized (shadow) cells, but also shows dilated lymphatic vessels, giant cell reaction, lymphoedema, disruption of collagen fibers, dilated blood vessels, fibrous capsule, calcification, nests of transitional cells, and necrosis.2 The presence of dilated lymphatic vessels and lymphedema have been described as a common pathologic feature in bullous pilomatricomas2,7 and can be used to distinguish bullous pilomatricomas from common pilomatricomas. Moreover, as a low-cost, noninvasive tool, ultrasonography (specifically high-frequency ultrasonography8) has been used as a diagnostic tool and is more appropriate for younger children to demonstrate the superficial position, continuity of the lesion with deeper structures, and degree of calcification.The diagnosis of bullous pilomatricoma may be difficult clinically, due to its atypical presentation. The common differential diagnoses should include matrical carcinoma, panfolliculoma, and trichodiscoma. Matrical carcinoma is a locally aggressive tumor, which is typically featured with a nontender, firm dermal swollen nodule and occurs most commonly in the area of the head and neck. Histologically, both matrical carcinomas and pilomatricomas are composed of nodular aggregates of basaloid keratinocytes that transform into shadow cells.9 Cellular pleomorphism and asymmetry, undefined circumscription, and tumor necrosis can help distinguish these 2 entities. Panfolliculoma typically presents as a slow-growing cystic or dome-shaped nodule, which may develop ulcers and crusts in the center, mainly affecting the face and scalp, though sometimes the limbs, of elderly patients.10 Histologically, cellular aggregates can differentiate into any components of the hair follicle, not just the hair matrix cells and shadow cells. Trichodiscoma, a follicular mesenchymal hamartoma, is characterized by multiple, small, 2- to 4-mm, dome-shaped, skin-colored papules on the face, scalp, ears, or upper trunk. Histopathological examination can show a central zone of fibro-mucinous stroma, with surrounding sebaceous glands in a mittlike configuration. The first-line treatment is surgical excision or curettage, and incomplete resection almost always results in recurrence.
Dermatology
A previously healthy 16-year-old boy presented to our department with a red tumor on him right upper arm that enlarged over 6 months. The lesion initially started as a small cutaneous nodule without obvious triggers that gradually developed and enlarged to form a red-colored tumor with a hard nodule inside. There was no history of local trauma or insect bite. Him personal, past, and family histories were unremarkable.On physical examination, there was a 6 × 6-cm protuberant, thick-walled, and well-defined red bullalike tumor on the right upper arm, which extended 1 to 3 cm from the epidermal surface (Figure, A). Inside the tumor, a nontender, firm-to-hard nodule was palpated. There was no regional lymphadenopathy, and results of the rest of the physical and systemic examinations were normal. Laboratory examinations of hematologic, biochemical, and urinalysis tests were normal. The tumor was excised completely under local anesthesia, and part of the tissue was sent for pathological examination.A, A tumor on the man’s right shoulder. B, The excisional tumor showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance. C and D, Histological examination revealed basophilic cells and shadow cells as well as dilated lymphatic vessels (hematoxylin-eosin; original magnification ×100).
what is your diagnosis?
What is your diagnosis?
Trichodiscoma
Panfolliculoma
Matrical carcinoma
Bullous pilomatricoma
d
0
1
0
1
male
0
0
16
11-20
White
23
augmented_male_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2808313
A previously healthy 16-year-old girl presented to our department with a red tumor on her right upper arm that enlarged over 6 months. The lesion initially started as a small cutaneous nodule without obvious triggers that gradually developed and enlarged to form a red-colored tumor with a hard nodule inside. There was no history of local trauma or insect bite. Her personal, past, and family histories were unremarkable.On physical examination, there was a 6 × 6-cm protuberant, thick-walled, and well-defined red bullalike tumor on the right upper arm, which extended 1 to 3 cm from the epidermal surface (Figure, A). Inside the tumor, a nontender, firm-to-hard nodule was palpated. There was no regional lymphadenopathy, and results of the rest of the physical and systemic examinations were normal. Laboratory examinations of hematologic, biochemical, and urinalysis tests were normal. The tumor was excised completely under local anesthesia, and part of the tissue was sent for pathological examination.A, A tumor on the patient’s right shoulder. B, The excisional tumor showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance. C and D, Histological examination revealed basophilic cells and shadow cells as well as dilated lymphatic vessels (hematoxylin-eosin; original magnification ×100). What Is Your Diagnosis?
Matrical carcinoma
Panfolliculoma
Bullous pilomatricoma
Trichodiscoma
C. Bullous pilomatricoma
C
Bullous pilomatricoma
The gross specimen of excised material showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance (Figure, B). Histopathological examination revealed basophilic cells and shadow cells in the deep dermis (Figure, C) and dilated lymphatic vessels in the dermis above the tumor (Figure, D).Based on the clinical morphology and histological features, the diagnosis of bullous pilomatricoma was confirmed. The patient is under regular outpatient follow-up, with no recurrence detected in 1 year of follow-up.Bullous pilomatricoma is a rare variant of pilomatricoma with a bullous appearance.1 It most commonly occurs on the shoulder and upper arm but also can be found on the neck, trunk, eyelid, and scalp. The peak age of presentation is 10 to 20 years old, and there is a predilection for female individuals.2 Clinically, bullous pilomatricoma usually presents as a solitary, semitransparent, erythematous, bluish or skin-colored, heavily folded or striaelike, flaccid, thick-walled, bullalike tumor with an underlying palpable hard nodule.1-3 It is rare for patients with bullous pilomatricoma to have multiple lesions.4The present patient’s lesion was increasing in size gradually but always had a clear border. When the bullalike tumor was punctured, a jellylike substance could be seen exuding out.4 Unlike some typical pilomatricomas, bullous pilomatricoma is not associated with Turner syndrome, myotonic dystrophy, Gardner syndrome, or Rubinstein-Taybi syndrome.2The etiopathogenesis of bullous pilomatricoma is not clear. There are 2 proposed theories. In the first, obstruction of lymphatic vessels and congestion of lymphatic fluid caused by the growth of the tumor nodule causes dilatation of lymphatic vessels, leakage of lymphatic fluid, and edema in the dermis surrounding the tumor, which further produces the bullous appearance.2,5 The second theory suggests that the presence of matrix metalloproteinases (MMP-9 and MMP-12), released by fibroblasts and inflammatory cells surrounding the tumor, may be associated with the degradation of elastic fibers and collagen, resulting in the bullouslike/anetodermic appearance.6Histologically, bullous pilomatricoma not only has the hallmarks of pilomatricoma, including basaloid cells and eosinophilic keratinized (shadow) cells, but also shows dilated lymphatic vessels, giant cell reaction, lymphoedema, disruption of collagen fibers, dilated blood vessels, fibrous capsule, calcification, nests of transitional cells, and necrosis.2 The presence of dilated lymphatic vessels and lymphedema have been described as a common pathologic feature in bullous pilomatricomas2,7 and can be used to distinguish bullous pilomatricomas from common pilomatricomas. Moreover, as a low-cost, noninvasive tool, ultrasonography (specifically high-frequency ultrasonography8) has been used as a diagnostic tool and is more appropriate for younger children to demonstrate the superficial position, continuity of the lesion with deeper structures, and degree of calcification.The diagnosis of bullous pilomatricoma may be difficult clinically, due to its atypical presentation. The common differential diagnoses should include matrical carcinoma, panfolliculoma, and trichodiscoma. Matrical carcinoma is a locally aggressive tumor, which is typically featured with a nontender, firm dermal swollen nodule and occurs most commonly in the area of the head and neck. Histologically, both matrical carcinomas and pilomatricomas are composed of nodular aggregates of basaloid keratinocytes that transform into shadow cells.9 Cellular pleomorphism and asymmetry, undefined circumscription, and tumor necrosis can help distinguish these 2 entities. Panfolliculoma typically presents as a slow-growing cystic or dome-shaped nodule, which may develop ulcers and crusts in the center, mainly affecting the face and scalp, though sometimes the limbs, of elderly patients.10 Histologically, cellular aggregates can differentiate into any components of the hair follicle, not just the hair matrix cells and shadow cells. Trichodiscoma, a follicular mesenchymal hamartoma, is characterized by multiple, small, 2- to 4-mm, dome-shaped, skin-colored papules on the face, scalp, ears, or upper trunk. Histopathological examination can show a central zone of fibro-mucinous stroma, with surrounding sebaceous glands in a mittlike configuration. The first-line treatment is surgical excision or curettage, and incomplete resection almost always results in recurrence.
Dermatology
A previously healthy 16-year-old child presented to our department with a red tumor on them right upper arm that enlarged over 6 months. The lesion initially started as a small cutaneous nodule without obvious triggers that gradually developed and enlarged to form a red-colored tumor with a hard nodule inside. There was no history of local trauma or insect bite. Them personal, past, and family histories were unremarkable.On physical examination, there was a 6 × 6-cm protuberant, thick-walled, and well-defined red bullalike tumor on the right upper arm, which extended 1 to 3 cm from the epidermal surface (Figure, A). Inside the tumor, a nontender, firm-to-hard nodule was palpated. There was no regional lymphadenopathy, and results of the rest of the physical and systemic examinations were normal. Laboratory examinations of hematologic, biochemical, and urinalysis tests were normal. The tumor was excised completely under local anesthesia, and part of the tissue was sent for pathological examination.A, A tumor on the patient’s right shoulder. B, The excisional tumor showed multilocular spaces below the epidermis and a solid tumor present in the deep dermis with a chalky-white cut surface and lobulated appearance. C and D, Histological examination revealed basophilic cells and shadow cells as well as dilated lymphatic vessels (hematoxylin-eosin; original magnification ×100).
what is your diagnosis?
What is your diagnosis?
Trichodiscoma
Panfolliculoma
Matrical carcinoma
Bullous pilomatricoma
d
0
1
0
1
neutral
0
0
16
11-20
White
23
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2807444
A 15-year-old girl presented with a 2-month history of decreased vision and a dark spot in the central vision in her right eye. She had a history of optometric examinations with no known ocular disease. Visual acuity with correction was 20/40 in the right eye and 20/20 in the left eye. Anterior segment examination of both eyes was unremarkable, and the retina in the left eye appeared normal. In the right eye, there was an area of superior macular whitening with associated intraretinal hemorrhages and exudates. Fluorescein angiography showed abnormally dilated and tortuous retinal vessels, irregular capillary branching patterns, and bulblike telangiectasis with late leakage in the superior macula. Optical coherence tomography showed disorganized and hyperreflective inner retinal thickening (Figure, A) and intraretinal and subretinal fluid in the central macula. On review of systems, the patient noted a long-standing growth on her cheek, which on examination was a 4 × 3-mm exophytic, brownish-red vascular papule (Figure, B), as well as a history of lightly pigmented patches of skin and multiple subungual fibromas on her toes. Systemic blood pressure was normal.Features of tuberous sclerosis complex. A, An area of superior macular whitening following the superior arterial arcade with associated intraretinal hemorrhages and exudate. Two optical coherence tomography line scans inset through the main lesion (orange) and through the fovea (blue) with relative location marked with dotted lines, demonstrating disorganized and hyperreflective inner retinal thickening consistent with type 1 astrocytic hamartoma of the retina and associated macular edema. Arrowhead indicates bulblike telangiectasis on fluorescein angiography and blocking from intraretinal hemorrhage. B, Elevated papule. The lesion is a well-demarcated, exophytic, brownish-red vascular papule with an irregular surface.Order brain magnetic resonance imaging to look for associated central nervous system tumors What Would You Do Next?
Initiate anti–vascular endothelial growth factor treatment
Order brain magnetic resonance imaging to look for associated central nervous system tumors
Perform hypercoagulable workup
Order echocardiogram and carotid ultrasonography
Astrocytic hamartoma (with secondary branch retinal vein occlusion) associated with tuberous sclerosis complex
B
Order brain magnetic resonance imaging to look for associated central nervous system tumors
Astrocytic hamartomas of the retina (AHR) are benign tumors that can be associated with tuberous sclerosis complex (TSC). TSC is a phacomatosis with neurocutaneous manifestations characterized by multiple tumors of the embryonic ectoderm involving skin, eyes, and nervous systems.1 Hamartomas of the retina are part of the major features for the diagnosis of TSC.2The clinical diagnostic criteria for TSC include 11 major and 7 minor features. Major features are 3 or more hypomelanotic macules at least 5 mm in diameter, 3 or more angiofibromas or fibrous cephalic plaques, 2 or more ungual fibromas, shagreen patch (a large nevus with irregular, firm plaquelike features typical of TSC), multiple retinal hamartomas, multiple cortical tubers or radial migration lines, 2 or more subependymal nodules, subependymal giant cell astrocytoma, cardiac rhabdomyoma, lymphangioleiomyomatosis, and 2 or more angiomyolipomas. Minor features of TSC are 1- to 2-mm hypomelanotic macules, also known as confetti skin lesions; 3 or more dental enamel pits; 2 or more intraoral fibromas; retinal achromic patch; multiple renal cysts; nonrenal hamartomas; and sclerotic bone lesions.The diagnostic certainty of TSC depends on the number of major and minor features. Definite TSC requires 2 major features or 1 major and 2 or more minor features. Possible TSC requires either 1 major feature or 2 or more minor features.3 Genetic testing is also available, and the identification of either a TSC1 or TSC2 pathogenic variant from nonlesional tissue is sufficient to make a definite diagnosis of TSC regardless of clinical findings.3History should be obtained and focused physical examination is recommended for skin and the central nervous system. The association between TSC and astrocytic hamartoma can be observed in up to half of patients with TSC.2 There are several phenotypes of astrocytic hamartoma associated with TSC, including slow-growing flat lesions (similar to that observed in this patient), as well as more aggressive, rapidly growing tumors that can lead to enucleation.2 Histology shows that astrocytic hamartomas are composed of glial astrocytes and blood vessels with hyaline and calcium deposits.4If TSC is suspected, the primary concern must be initiating the systemic workup with neuroimaging (choice B), as the central nervous system manifestations of TSC can be life threatening. Secondarily, treatment for the ophthalmic complications of the disease can be considered, such as with anti–vascular endothelial growth factor (anti-VEGF) medications for secondary exudation (choice A).5 Retinal hamartomas are not typically associated with a hypercoagulable state or cardiac concerns (choices C and D).The patient was referred for neuroimaging, which revealed multiple subcortical tubers and lateral ventricle subependymomas consistent with the diagnosis of TSC. Genetic testing revealed a pathogenic variant in the TSC1 gene labeled c.733C>T(p.R245*), which is most commonly a de novo variant. Excisional biopsy of the facial lesion was consistent with angiofibroma, a benign cutaneous hamartoma frequently found in tuberous sclerosis complex. Because the central nervous system lesions were asymptomatic, the patient was referred for annual imaging and observation in pediatric neurology.Retinal hamartomas are classified into 3 morphological subtypes. Type 1 AHR is the most common and is characterized by flat and smooth lesions semitransparent without calcifications. Type 2 AHR lesions are raised, multinodular, opaque, and with calcification. Type 3 AHR lesions are transitional lesions with features of types 1 and 2.6Treatment of AHR lesions is determined on a case-by-case basis. Photodynamic therapy, laser, radiation, and anti-VEGF have been described.7 Due to the secondary exudation and hemorrhages consistent with a small branch retinal vein occlusion with edema affecting the central macula, we performed a systemic hypercoagulable workup, which was unrevealing. Because the exudation affected the central macula, we started treatment with intravitreal anti-VEGF to treat the macular edema. Two years later, the patient was stable with visual acuity 20/25 in the affected eye but has required monthly treatment with worsening edema and vision with an attempted treat-and-extend strategy. The tumor slowly expanded for the first year but was stable for the following year.
Ophthalmology
A 15-year-old girl presented with a 2-month history of decreased vision and a dark spot in the central vision in her right eye. She had a history of optometric examinations with no known ocular disease. Visual acuity with correction was 20/40 in the right eye and 20/20 in the left eye. Anterior segment examination of both eyes was unremarkable, and the retina in the left eye appeared normal. In the right eye, there was an area of superior macular whitening with associated intraretinal hemorrhages and exudates. Fluorescein angiography showed abnormally dilated and tortuous retinal vessels, irregular capillary branching patterns, and bulblike telangiectasis with late leakage in the superior macula. Optical coherence tomography showed disorganized and hyperreflective inner retinal thickening (Figure, A) and intraretinal and subretinal fluid in the central macula. On review of systems, the patient noted a long-standing growth on her cheek, which on examination was a 4 × 3-mm exophytic, brownish-red vascular papule (Figure, B), as well as a history of lightly pigmented patches of skin and multiple subungual fibromas on her toes. Systemic blood pressure was normal.Features of tuberous sclerosis complex. A, An area of superior macular whitening following the superior arterial arcade with associated intraretinal hemorrhages and exudate. Two optical coherence tomography line scans inset through the main lesion (orange) and through the fovea (blue) with relative location marked with dotted lines, demonstrating disorganized and hyperreflective inner retinal thickening consistent with type 1 astrocytic hamartoma of the retina and associated macular edema. Arrowhead indicates bulblike telangiectasis on fluorescein angiography and blocking from intraretinal hemorrhage. B, Elevated papule. The lesion is a well-demarcated, exophytic, brownish-red vascular papule with an irregular surface.Order brain magnetic resonance imaging to look for associated central nervous system tumors
what would you do next?
What would you do next?
Initiate anti–vascular endothelial growth factor treatment
Order echocardiogram and carotid ultrasonography
Perform hypercoagulable workup
Order brain magnetic resonance imaging to look for associated central nervous system tumors
d
1
1
1
1
female
0
0
15
11-20
null
24
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2807444
A 15-year-old girl presented with a 2-month history of decreased vision and a dark spot in the central vision in her right eye. She had a history of optometric examinations with no known ocular disease. Visual acuity with correction was 20/40 in the right eye and 20/20 in the left eye. Anterior segment examination of both eyes was unremarkable, and the retina in the left eye appeared normal. In the right eye, there was an area of superior macular whitening with associated intraretinal hemorrhages and exudates. Fluorescein angiography showed abnormally dilated and tortuous retinal vessels, irregular capillary branching patterns, and bulblike telangiectasis with late leakage in the superior macula. Optical coherence tomography showed disorganized and hyperreflective inner retinal thickening (Figure, A) and intraretinal and subretinal fluid in the central macula. On review of systems, the patient noted a long-standing growth on her cheek, which on examination was a 4 × 3-mm exophytic, brownish-red vascular papule (Figure, B), as well as a history of lightly pigmented patches of skin and multiple subungual fibromas on her toes. Systemic blood pressure was normal.Features of tuberous sclerosis complex. A, An area of superior macular whitening following the superior arterial arcade with associated intraretinal hemorrhages and exudate. Two optical coherence tomography line scans inset through the main lesion (orange) and through the fovea (blue) with relative location marked with dotted lines, demonstrating disorganized and hyperreflective inner retinal thickening consistent with type 1 astrocytic hamartoma of the retina and associated macular edema. Arrowhead indicates bulblike telangiectasis on fluorescein angiography and blocking from intraretinal hemorrhage. B, Elevated papule. The lesion is a well-demarcated, exophytic, brownish-red vascular papule with an irregular surface.Order brain magnetic resonance imaging to look for associated central nervous system tumors What Would You Do Next?
Initiate anti–vascular endothelial growth factor treatment
Order brain magnetic resonance imaging to look for associated central nervous system tumors
Perform hypercoagulable workup
Order echocardiogram and carotid ultrasonography
Astrocytic hamartoma (with secondary branch retinal vein occlusion) associated with tuberous sclerosis complex
B
Order brain magnetic resonance imaging to look for associated central nervous system tumors
Astrocytic hamartomas of the retina (AHR) are benign tumors that can be associated with tuberous sclerosis complex (TSC). TSC is a phacomatosis with neurocutaneous manifestations characterized by multiple tumors of the embryonic ectoderm involving skin, eyes, and nervous systems.1 Hamartomas of the retina are part of the major features for the diagnosis of TSC.2The clinical diagnostic criteria for TSC include 11 major and 7 minor features. Major features are 3 or more hypomelanotic macules at least 5 mm in diameter, 3 or more angiofibromas or fibrous cephalic plaques, 2 or more ungual fibromas, shagreen patch (a large nevus with irregular, firm plaquelike features typical of TSC), multiple retinal hamartomas, multiple cortical tubers or radial migration lines, 2 or more subependymal nodules, subependymal giant cell astrocytoma, cardiac rhabdomyoma, lymphangioleiomyomatosis, and 2 or more angiomyolipomas. Minor features of TSC are 1- to 2-mm hypomelanotic macules, also known as confetti skin lesions; 3 or more dental enamel pits; 2 or more intraoral fibromas; retinal achromic patch; multiple renal cysts; nonrenal hamartomas; and sclerotic bone lesions.The diagnostic certainty of TSC depends on the number of major and minor features. Definite TSC requires 2 major features or 1 major and 2 or more minor features. Possible TSC requires either 1 major feature or 2 or more minor features.3 Genetic testing is also available, and the identification of either a TSC1 or TSC2 pathogenic variant from nonlesional tissue is sufficient to make a definite diagnosis of TSC regardless of clinical findings.3History should be obtained and focused physical examination is recommended for skin and the central nervous system. The association between TSC and astrocytic hamartoma can be observed in up to half of patients with TSC.2 There are several phenotypes of astrocytic hamartoma associated with TSC, including slow-growing flat lesions (similar to that observed in this patient), as well as more aggressive, rapidly growing tumors that can lead to enucleation.2 Histology shows that astrocytic hamartomas are composed of glial astrocytes and blood vessels with hyaline and calcium deposits.4If TSC is suspected, the primary concern must be initiating the systemic workup with neuroimaging (choice B), as the central nervous system manifestations of TSC can be life threatening. Secondarily, treatment for the ophthalmic complications of the disease can be considered, such as with anti–vascular endothelial growth factor (anti-VEGF) medications for secondary exudation (choice A).5 Retinal hamartomas are not typically associated with a hypercoagulable state or cardiac concerns (choices C and D).The patient was referred for neuroimaging, which revealed multiple subcortical tubers and lateral ventricle subependymomas consistent with the diagnosis of TSC. Genetic testing revealed a pathogenic variant in the TSC1 gene labeled c.733C>T(p.R245*), which is most commonly a de novo variant. Excisional biopsy of the facial lesion was consistent with angiofibroma, a benign cutaneous hamartoma frequently found in tuberous sclerosis complex. Because the central nervous system lesions were asymptomatic, the patient was referred for annual imaging and observation in pediatric neurology.Retinal hamartomas are classified into 3 morphological subtypes. Type 1 AHR is the most common and is characterized by flat and smooth lesions semitransparent without calcifications. Type 2 AHR lesions are raised, multinodular, opaque, and with calcification. Type 3 AHR lesions are transitional lesions with features of types 1 and 2.6Treatment of AHR lesions is determined on a case-by-case basis. Photodynamic therapy, laser, radiation, and anti-VEGF have been described.7 Due to the secondary exudation and hemorrhages consistent with a small branch retinal vein occlusion with edema affecting the central macula, we performed a systemic hypercoagulable workup, which was unrevealing. Because the exudation affected the central macula, we started treatment with intravitreal anti-VEGF to treat the macular edema. Two years later, the patient was stable with visual acuity 20/25 in the affected eye but has required monthly treatment with worsening edema and vision with an attempted treat-and-extend strategy. The tumor slowly expanded for the first year but was stable for the following year.
Ophthalmology
A 15-year-old boy presented with a 2-month history of decreased vision and a dark spot in the central vision in him right eye. He had a history of optometric examinations with no known ocular disease. Visual acuity with correction was 20/40 in the right eye and 20/20 in the left eye. Anterior segment examination of both eyes was unremarkable, and the retina in the left eye appeared normal. In the right eye, there was an area of superior macular whitening with associated intraretinal hemorrhages and exudates. Fluorescein angiography showed abnormally dilated and tortuous retinal vessels, irregular capillary branching patterns, and bulblike telangiectasis with late leakage in the superior macula. Optical coherence tomography showed disorganized and hyperreflective inner retinal thickening (Figure, A) and intraretinal and subretinal fluid in the central macula. On review of systems, the man noted a long-standing growth on him cheek, which on examination was a 4 × 3-mm exophytic, brownish-red vascular papule (Figure, B), as well as a history of lightly pigmented patches of skin and multiple subungual fibromas on him toes. Systemic blood pressure was normal.Features of tuberous sclerosis complex. A, An area of superior macular whitening following the superior arterial arcade with associated intraretinal hemorrhages and exudate. Two optical coherence tomography line scans inset through the main lesion (orange) and through the fovea (blue) with relative location marked with dotted lines, demonstrating disorganized and hyperreflective inner retinal thickening consistent with type 1 astrocytic hamartoma of the retina and associated macular edema. Arrowhead indicates bulblike telangiectasis on fluorescein angiography and blocking from intraretinal hemorrhage. B, Elevated papule. The lesion is a well-demarcated, exophytic, brownish-red vascular papule with an irregular surface.Order brain magnetic resonance imaging to look for associated central nervous system tumors
what would you do next?
What would you do next?
Initiate anti–vascular endothelial growth factor treatment
Order echocardiogram and carotid ultrasonography
Perform hypercoagulable workup
Order brain magnetic resonance imaging to look for associated central nervous system tumors
d
1
1
1
1
male
0
0
15
11-20
null
24
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2807444
A 15-year-old girl presented with a 2-month history of decreased vision and a dark spot in the central vision in her right eye. She had a history of optometric examinations with no known ocular disease. Visual acuity with correction was 20/40 in the right eye and 20/20 in the left eye. Anterior segment examination of both eyes was unremarkable, and the retina in the left eye appeared normal. In the right eye, there was an area of superior macular whitening with associated intraretinal hemorrhages and exudates. Fluorescein angiography showed abnormally dilated and tortuous retinal vessels, irregular capillary branching patterns, and bulblike telangiectasis with late leakage in the superior macula. Optical coherence tomography showed disorganized and hyperreflective inner retinal thickening (Figure, A) and intraretinal and subretinal fluid in the central macula. On review of systems, the patient noted a long-standing growth on her cheek, which on examination was a 4 × 3-mm exophytic, brownish-red vascular papule (Figure, B), as well as a history of lightly pigmented patches of skin and multiple subungual fibromas on her toes. Systemic blood pressure was normal.Features of tuberous sclerosis complex. A, An area of superior macular whitening following the superior arterial arcade with associated intraretinal hemorrhages and exudate. Two optical coherence tomography line scans inset through the main lesion (orange) and through the fovea (blue) with relative location marked with dotted lines, demonstrating disorganized and hyperreflective inner retinal thickening consistent with type 1 astrocytic hamartoma of the retina and associated macular edema. Arrowhead indicates bulblike telangiectasis on fluorescein angiography and blocking from intraretinal hemorrhage. B, Elevated papule. The lesion is a well-demarcated, exophytic, brownish-red vascular papule with an irregular surface.Order brain magnetic resonance imaging to look for associated central nervous system tumors What Would You Do Next?
Initiate anti–vascular endothelial growth factor treatment
Order brain magnetic resonance imaging to look for associated central nervous system tumors
Perform hypercoagulable workup
Order echocardiogram and carotid ultrasonography
Astrocytic hamartoma (with secondary branch retinal vein occlusion) associated with tuberous sclerosis complex
B
Order brain magnetic resonance imaging to look for associated central nervous system tumors
Astrocytic hamartomas of the retina (AHR) are benign tumors that can be associated with tuberous sclerosis complex (TSC). TSC is a phacomatosis with neurocutaneous manifestations characterized by multiple tumors of the embryonic ectoderm involving skin, eyes, and nervous systems.1 Hamartomas of the retina are part of the major features for the diagnosis of TSC.2The clinical diagnostic criteria for TSC include 11 major and 7 minor features. Major features are 3 or more hypomelanotic macules at least 5 mm in diameter, 3 or more angiofibromas or fibrous cephalic plaques, 2 or more ungual fibromas, shagreen patch (a large nevus with irregular, firm plaquelike features typical of TSC), multiple retinal hamartomas, multiple cortical tubers or radial migration lines, 2 or more subependymal nodules, subependymal giant cell astrocytoma, cardiac rhabdomyoma, lymphangioleiomyomatosis, and 2 or more angiomyolipomas. Minor features of TSC are 1- to 2-mm hypomelanotic macules, also known as confetti skin lesions; 3 or more dental enamel pits; 2 or more intraoral fibromas; retinal achromic patch; multiple renal cysts; nonrenal hamartomas; and sclerotic bone lesions.The diagnostic certainty of TSC depends on the number of major and minor features. Definite TSC requires 2 major features or 1 major and 2 or more minor features. Possible TSC requires either 1 major feature or 2 or more minor features.3 Genetic testing is also available, and the identification of either a TSC1 or TSC2 pathogenic variant from nonlesional tissue is sufficient to make a definite diagnosis of TSC regardless of clinical findings.3History should be obtained and focused physical examination is recommended for skin and the central nervous system. The association between TSC and astrocytic hamartoma can be observed in up to half of patients with TSC.2 There are several phenotypes of astrocytic hamartoma associated with TSC, including slow-growing flat lesions (similar to that observed in this patient), as well as more aggressive, rapidly growing tumors that can lead to enucleation.2 Histology shows that astrocytic hamartomas are composed of glial astrocytes and blood vessels with hyaline and calcium deposits.4If TSC is suspected, the primary concern must be initiating the systemic workup with neuroimaging (choice B), as the central nervous system manifestations of TSC can be life threatening. Secondarily, treatment for the ophthalmic complications of the disease can be considered, such as with anti–vascular endothelial growth factor (anti-VEGF) medications for secondary exudation (choice A).5 Retinal hamartomas are not typically associated with a hypercoagulable state or cardiac concerns (choices C and D).The patient was referred for neuroimaging, which revealed multiple subcortical tubers and lateral ventricle subependymomas consistent with the diagnosis of TSC. Genetic testing revealed a pathogenic variant in the TSC1 gene labeled c.733C>T(p.R245*), which is most commonly a de novo variant. Excisional biopsy of the facial lesion was consistent with angiofibroma, a benign cutaneous hamartoma frequently found in tuberous sclerosis complex. Because the central nervous system lesions were asymptomatic, the patient was referred for annual imaging and observation in pediatric neurology.Retinal hamartomas are classified into 3 morphological subtypes. Type 1 AHR is the most common and is characterized by flat and smooth lesions semitransparent without calcifications. Type 2 AHR lesions are raised, multinodular, opaque, and with calcification. Type 3 AHR lesions are transitional lesions with features of types 1 and 2.6Treatment of AHR lesions is determined on a case-by-case basis. Photodynamic therapy, laser, radiation, and anti-VEGF have been described.7 Due to the secondary exudation and hemorrhages consistent with a small branch retinal vein occlusion with edema affecting the central macula, we performed a systemic hypercoagulable workup, which was unrevealing. Because the exudation affected the central macula, we started treatment with intravitreal anti-VEGF to treat the macular edema. Two years later, the patient was stable with visual acuity 20/25 in the affected eye but has required monthly treatment with worsening edema and vision with an attempted treat-and-extend strategy. The tumor slowly expanded for the first year but was stable for the following year.
Ophthalmology
A 15-year-old child presented with a 2-month history of decreased vision and a dark spot in the central vision in them right eye. They had a history of optometric examinations with no known ocular disease. Visual acuity with correction was 20/40 in the right eye and 20/20 in the left eye. Anterior segment examination of both eyes was unremarkable, and the retina in the left eye appeared normal. In the right eye, there was an area of superior macular whitening with associated intraretinal hemorrhages and exudates. Fluorescein angiography showed abnormally dilated and tortuous retinal vessels, irregular capillary branching patterns, and bulblike telangiectasis with late leakage in the superior macula. Optical coherence tomography showed disorganized and hyperreflective inner retinal thickening (Figure, A) and intraretinal and subretinal fluid in the central macula. On review of systems, the patient noted a long-standing growth on them cheek, which on examination was a 4 × 3-mm exophytic, brownish-red vascular papule (Figure, B), as well as a history of lightly pigmented patches of skin and multiple subungual fibromas on them toes. Systemic blood pressure was normal.Features of tuberous sclerosis complex. A, An area of superior macular whitening following the superior arterial arcade with associated intraretinal hemorrhages and exudate. Two optical coherence tomography line scans inset through the main lesion (orange) and through the fovea (blue) with relative location marked with dotted lines, demonstrating disorganized and hyperreflective inner retinal thickening consistent with type 1 astrocytic hamartoma of the retina and associated macular edema. Arrowhead indicates bulblike telangiectasis on fluorescein angiography and blocking from intraretinal hemorrhage. B, Elevated papule. The lesion is a well-demarcated, exophytic, brownish-red vascular papule with an irregular surface.Order brain magnetic resonance imaging to look for associated central nervous system tumors
what would you do next?
What would you do next?
Initiate anti–vascular endothelial growth factor treatment
Order echocardiogram and carotid ultrasonography
Perform hypercoagulable workup
Order brain magnetic resonance imaging to look for associated central nervous system tumors
d
1
1
1
1
neutral
0
0
15
11-20
null
24
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jama/fullarticle/2807957
A 73-year-old man with a history of gastroesophageal reflux disease and Raynaud disease but no history of cigarette smoking presented to the emergency department with 4 weeks of dysphagia and a 4.54-kg (10-lb) weight loss over 3 months. He had no nausea, vomiting, abdominal pain, melena, or hematochezia but reported a cough productive of yellow sputum, dyspnea on exertion, and fatigue. The patient had been treated for presumed pneumonia with 3 courses of azithromycin over the prior 3 months due to infiltrates on a chest radiograph. He reported no known ill contacts or recent travel outside the US. At presentation, his heart rate and blood pressure were normal, and oxygen saturation was 95% on room air. Physical examination revealed a maculopapular rash over his neck and chest (Figure, left panel) and bibasilar crackles on lung auscultation. Laboratory testing showed a white blood cell count of 14.3 × 103/μL (reference, 4.2-9.1 × 103/μL); creatine kinase level, 2060 U/L (34.40 μkat/L) (reference, 44-196 U/L [0.73-3.27 μkat/L]), aldolase level, 18 U/L (0.30 μkat/L) (reference, <8 U/L [<0.13 μkat/L]), C-reactive protein level, 30 mg/L (reference, <8 mg/L), and erythrocyte sedimentation rate, 35 mm/h (reference, <20 mm/h). Antinuclear antibody immunofluorescent assay findings were positive. Results from testing for HIV, hepatitis B, and hepatitis C were negative. A barium swallow study showed no visualized aspiration. Endoscopy revealed clean-based esophageal and duodenal ulcers and nonspecific gastritis. Chest computed tomography (CT) showed bibasilar pulmonary consolidations and ground glass opacities (Figure, right panel).Left, Shawl-like rash on patient’s chest. Right, Computed tomography scan of the chest showing bibasilar pulmonary consolidations and ground glass opacities.Prescribe 10 days of prednisone (60 mg daily) What Would You Do Next?
Arrange a percutaneous lung tissue biopsy
Check an antisynthetase antibody panel
Perform a quadriceps muscle biopsy
Prescribe 10 days of prednisone (60 mg daily)
Antisynthetase syndrome
B
Check an antisynthetase antibody panel
The key to the correct diagnosis is recognition that cough, dyspnea on exertion and pulmonary infiltrates, and elevated levels of creatine kinase, aldolase, and inflammatory markers in a patient with Raynaud disease are characteristic of antisynthetase syndrome. Choices A and C are not recommended because lung and muscle biopsy are invasive procedures and may not provide a definitive diagnosis. Prednisone (choice D) should not be prescribed before a diagnosis has been made.Antisynthetase syndrome is a rare autoimmune disease classified as a subgroup of idiopathic inflammatory myopathic disorders.1 A characteristic feature of antisynthetase syndrome is presence of an autoantibody to one of the aminoacyl–transfer RNA (tRNA) synthetases, which are intracellular enzymes that attach amino acids to tRNA during protein synthesis.2,3 Antisynthetase syndrome has been associated with HLA haplotype DRB1*0301 and with exposures to tobacco, cleaning chemicals, mold, bird droppings, and airborne particles from the September 11, 2001, World Trade Center attack.2 Of the 10 identified antiaminoacyl-tRNA synthetases, the most common are anti–Jo-1 antibody (60%), anti–PL-12 (17%), and anti–PL-7 (12%).2,3The classic triad of antisynthetase syndrome consists of interstitial lung disease (ILD), myositis, and nonerosive arthritis. Although these 3 conditions develop in up to 90% of patients with antisynthetase syndrome, approximately 25% present only with symptoms of arthritis, 25% present with myositis, and 15% to 30% have ILD as their presenting manifestation.2,3 Other conditions associated with antisynthetase syndrome include Raynaud phenomenon, gastroesophageal reflux disease, fever, and fissured eruptions on the hands (“mechanic hands”).2,4,5ILD affects approximately 70% to 95% of patients with antisynthetase syndrome and is the most common cause of morbidity and mortality.4 High-resolution CT findings include ground glass opacities, bibasilar fibrosis, and traction bronchiectasis.4 Pulmonary function testing (PFT) typically shows a restrictive ventilatory defect.2 Approximately 9% of patients with antisynthetase syndrome have rapidly progressive ILD2; others have gradual onset of respiratory symptoms, and some are asymptomatic, with ILD diagnosed based on imaging or PFT.2 The incidence of ILD and pulmonary hypertension is higher in patients with anti–PL-7 and anti–PL-12 antibodies compared with anti–Jo-1 antibodies.2,4,6-8 Overall survival for patients with anti–Jo-1 antibody was 70% at 10 years vs 47% for patients with non–anti–Jo-1 synthetase antibodies.2Diagnosis of antisynthetase syndrome requires identification of an autoantibody to aminoacyl-tRNA synthetase. Most commercially available myositis panels test only for anti–Jo-1 antibody, so an antisynthetase antibody panel should be ordered if antisynthetase syndrome is being considered.4 On diagnosis, patients with antisynthetase syndrome should undergo a high-resolution chest CT scan to evaluate for ILD and PFT to assess lung function.2 An echocardiogram may be useful to evaluate for pulmonary hypertension in patients with ILD.4Limited evidence is available regarding optimal drug therapy for antisynthetase syndrome. Patients often receive multidisciplinary care from specialists in rheumatology, pulmonology, and dermatology and may be treated with glucocorticoids, azathioprine, mycophenolate mofetil, tacrolimus, cyclosporine, rituximab, cyclophosphamide, and nintedanib.2,4,5,9 Within the first year of treatment, approximately two-thirds of patients with antisynthetase syndrome experience clinical and radiographic improvement, and 1 study reported that approximately 24% of patients had full resolution of ILD.2 However, approximately one-third of patients develop fibrotic lung disease, which is associated with increased morbidity and mortality.2 Lung transplant should be considered for patients with severe or rapidly progressive ILD.2,5The patient tested positive for anti–PL-7 antibody, which confirmed the diagnosis of antisynthetase syndrome. Antibody testing results were negative for antisignal recognition particle, antinuclear helicase, anti-RNA helicase, anti–Scl-70, antipolymerase III, anti-U1RNP, anticentromere antibody, anti-U3RNP, anti-Ro/SSA and anti-La/BB, and rheumatoid factor. On PFT, forced vital capacity was 83% of predicted and diffusing capacity for carbon monoxide was 66% of predicted. He was initially treated with prednisone (10 mg daily) for 6 weeks, then transitioned to mycophenolate (500 mg twice daily) and nintedanib (150 mg twice daily), which are his current medications. At 1-year follow-up, he reported a decrease in his cough and dyspnea on exertion, and high-resolution chest CT showed decreased bibasilar consolidation compared with chest CT on presentation.
General
A 73-year-old man with a history of gastroesophageal reflux disease and Raynaud disease but no history of cigarette smoking presented to the emergency department with 4 weeks of dysphagia and a 4.54-kg (10-lb) weight loss over 3 months. He had no nausea, vomiting, abdominal pain, melena, or hematochezia but reported a cough productive of yellow sputum, dyspnea on exertion, and fatigue. The patient had been treated for presumed pneumonia with 3 courses of azithromycin over the prior 3 months due to infiltrates on a chest radiograph. He reported no known ill contacts or recent travel outside the US. At presentation, his heart rate and blood pressure were normal, and oxygen saturation was 95% on room air. Physical examination revealed a maculopapular rash over his neck and chest (Figure, left panel) and bibasilar crackles on lung auscultation. Laboratory testing showed a white blood cell count of 14.3 × 103/μL (reference, 4.2-9.1 × 103/μL); creatine kinase level, 2060 U/L (34.40 μkat/L) (reference, 44-196 U/L [0.73-3.27 μkat/L]), aldolase level, 18 U/L (0.30 μkat/L) (reference, <8 U/L [<0.13 μkat/L]), C-reactive protein level, 30 mg/L (reference, <8 mg/L), and erythrocyte sedimentation rate, 35 mm/h (reference, <20 mm/h). Antinuclear antibody immunofluorescent assay findings were positive. Results from testing for HIV, hepatitis B, and hepatitis C were negative. A barium swallow study showed no visualized aspiration. Endoscopy revealed clean-based esophageal and duodenal ulcers and nonspecific gastritis. Chest computed tomography (CT) showed bibasilar pulmonary consolidations and ground glass opacities (Figure, right panel).Left, Shawl-like rash on patient’s chest. Right, Computed tomography scan of the chest showing bibasilar pulmonary consolidations and ground glass opacities.Prescribe 10 days of prednisone (60 mg daily)
what would you do next?
What would you do next?
Arrange a percutaneous lung tissue biopsy
Check an antisynthetase antibody panel
Perform a quadriceps muscle biopsy
Prescribe 10 days of prednisone (60 mg daily)
b
1
1
1
1
male
0
0
73
71-80
White
25
original
https://jamanetwork.com/journals/jama/fullarticle/2807957
A 73-year-old man with a history of gastroesophageal reflux disease and Raynaud disease but no history of cigarette smoking presented to the emergency department with 4 weeks of dysphagia and a 4.54-kg (10-lb) weight loss over 3 months. He had no nausea, vomiting, abdominal pain, melena, or hematochezia but reported a cough productive of yellow sputum, dyspnea on exertion, and fatigue. The patient had been treated for presumed pneumonia with 3 courses of azithromycin over the prior 3 months due to infiltrates on a chest radiograph. He reported no known ill contacts or recent travel outside the US. At presentation, his heart rate and blood pressure were normal, and oxygen saturation was 95% on room air. Physical examination revealed a maculopapular rash over his neck and chest (Figure, left panel) and bibasilar crackles on lung auscultation. Laboratory testing showed a white blood cell count of 14.3 × 103/μL (reference, 4.2-9.1 × 103/μL); creatine kinase level, 2060 U/L (34.40 μkat/L) (reference, 44-196 U/L [0.73-3.27 μkat/L]), aldolase level, 18 U/L (0.30 μkat/L) (reference, <8 U/L [<0.13 μkat/L]), C-reactive protein level, 30 mg/L (reference, <8 mg/L), and erythrocyte sedimentation rate, 35 mm/h (reference, <20 mm/h). Antinuclear antibody immunofluorescent assay findings were positive. Results from testing for HIV, hepatitis B, and hepatitis C were negative. A barium swallow study showed no visualized aspiration. Endoscopy revealed clean-based esophageal and duodenal ulcers and nonspecific gastritis. Chest computed tomography (CT) showed bibasilar pulmonary consolidations and ground glass opacities (Figure, right panel).Left, Shawl-like rash on patient’s chest. Right, Computed tomography scan of the chest showing bibasilar pulmonary consolidations and ground glass opacities.Prescribe 10 days of prednisone (60 mg daily) What Would You Do Next?
Arrange a percutaneous lung tissue biopsy
Check an antisynthetase antibody panel
Perform a quadriceps muscle biopsy
Prescribe 10 days of prednisone (60 mg daily)
Antisynthetase syndrome
B
Check an antisynthetase antibody panel
The key to the correct diagnosis is recognition that cough, dyspnea on exertion and pulmonary infiltrates, and elevated levels of creatine kinase, aldolase, and inflammatory markers in a patient with Raynaud disease are characteristic of antisynthetase syndrome. Choices A and C are not recommended because lung and muscle biopsy are invasive procedures and may not provide a definitive diagnosis. Prednisone (choice D) should not be prescribed before a diagnosis has been made.Antisynthetase syndrome is a rare autoimmune disease classified as a subgroup of idiopathic inflammatory myopathic disorders.1 A characteristic feature of antisynthetase syndrome is presence of an autoantibody to one of the aminoacyl–transfer RNA (tRNA) synthetases, which are intracellular enzymes that attach amino acids to tRNA during protein synthesis.2,3 Antisynthetase syndrome has been associated with HLA haplotype DRB1*0301 and with exposures to tobacco, cleaning chemicals, mold, bird droppings, and airborne particles from the September 11, 2001, World Trade Center attack.2 Of the 10 identified antiaminoacyl-tRNA synthetases, the most common are anti–Jo-1 antibody (60%), anti–PL-12 (17%), and anti–PL-7 (12%).2,3The classic triad of antisynthetase syndrome consists of interstitial lung disease (ILD), myositis, and nonerosive arthritis. Although these 3 conditions develop in up to 90% of patients with antisynthetase syndrome, approximately 25% present only with symptoms of arthritis, 25% present with myositis, and 15% to 30% have ILD as their presenting manifestation.2,3 Other conditions associated with antisynthetase syndrome include Raynaud phenomenon, gastroesophageal reflux disease, fever, and fissured eruptions on the hands (“mechanic hands”).2,4,5ILD affects approximately 70% to 95% of patients with antisynthetase syndrome and is the most common cause of morbidity and mortality.4 High-resolution CT findings include ground glass opacities, bibasilar fibrosis, and traction bronchiectasis.4 Pulmonary function testing (PFT) typically shows a restrictive ventilatory defect.2 Approximately 9% of patients with antisynthetase syndrome have rapidly progressive ILD2; others have gradual onset of respiratory symptoms, and some are asymptomatic, with ILD diagnosed based on imaging or PFT.2 The incidence of ILD and pulmonary hypertension is higher in patients with anti–PL-7 and anti–PL-12 antibodies compared with anti–Jo-1 antibodies.2,4,6-8 Overall survival for patients with anti–Jo-1 antibody was 70% at 10 years vs 47% for patients with non–anti–Jo-1 synthetase antibodies.2Diagnosis of antisynthetase syndrome requires identification of an autoantibody to aminoacyl-tRNA synthetase. Most commercially available myositis panels test only for anti–Jo-1 antibody, so an antisynthetase antibody panel should be ordered if antisynthetase syndrome is being considered.4 On diagnosis, patients with antisynthetase syndrome should undergo a high-resolution chest CT scan to evaluate for ILD and PFT to assess lung function.2 An echocardiogram may be useful to evaluate for pulmonary hypertension in patients with ILD.4Limited evidence is available regarding optimal drug therapy for antisynthetase syndrome. Patients often receive multidisciplinary care from specialists in rheumatology, pulmonology, and dermatology and may be treated with glucocorticoids, azathioprine, mycophenolate mofetil, tacrolimus, cyclosporine, rituximab, cyclophosphamide, and nintedanib.2,4,5,9 Within the first year of treatment, approximately two-thirds of patients with antisynthetase syndrome experience clinical and radiographic improvement, and 1 study reported that approximately 24% of patients had full resolution of ILD.2 However, approximately one-third of patients develop fibrotic lung disease, which is associated with increased morbidity and mortality.2 Lung transplant should be considered for patients with severe or rapidly progressive ILD.2,5The patient tested positive for anti–PL-7 antibody, which confirmed the diagnosis of antisynthetase syndrome. Antibody testing results were negative for antisignal recognition particle, antinuclear helicase, anti-RNA helicase, anti–Scl-70, antipolymerase III, anti-U1RNP, anticentromere antibody, anti-U3RNP, anti-Ro/SSA and anti-La/BB, and rheumatoid factor. On PFT, forced vital capacity was 83% of predicted and diffusing capacity for carbon monoxide was 66% of predicted. He was initially treated with prednisone (10 mg daily) for 6 weeks, then transitioned to mycophenolate (500 mg twice daily) and nintedanib (150 mg twice daily), which are his current medications. At 1-year follow-up, he reported a decrease in his cough and dyspnea on exertion, and high-resolution chest CT showed decreased bibasilar consolidation compared with chest CT on presentation.
General
A 73-year-old woman with a history of gastroesophageal reflux disease and Raynaud disease but no history of cigarette smoking presented to the emergency department with 4 weeks of dysphagia and a 4.54-kg (10-lb) weight loss over 3 months. She had no nausea, vomiting, abdominal pain, melena, or hematochezia but reported a cough productive of yellow sputum, dyspnea on exertion, and fatigue. The woman had been treated for presumed pneumonia with 3 courses of azithromycin over the prior 3 months due to infiltrates on a chest radiograph. She reported no known ill contacts or recent travel outside the US. At presentation, her heart rate and blood pressure were normal, and oxygen saturation was 95% on room air. Physical examination revealed a maculopapular rash over her neck and chest (Figure, left panel) and bibasilar crackles on lung auscultation. Laboratory testing showed a white blood cell count of 14.3 × 103/μL (reference, 4.2-9.1 × 103/μL); creatine kinase level, 2060 U/L (34.40 μkat/L) (reference, 44-196 U/L [0.73-3.27 μkat/L]), aldolase level, 18 U/L (0.30 μkat/L) (reference, <8 U/L [<0.13 μkat/L]), C-reactive protein level, 30 mg/L (reference, <8 mg/L), and erythrocyte sedimentation rate, 35 mm/h (reference, <20 mm/h). Antinuclear antibody immunofluorescent assay findings were positive. Results from testing for HIV, hepatitis B, and hepatitis C were negative. A barium swallow study showed no visualized aspiration. Endoscopy revealed clean-based esophageal and duodenal ulcers and nonspecific gastritis. Chest computed tomography (CT) showed bibasilar pulmonary consolidations and ground glass opacities (Figure, right panel).Left, Shawl-like rash on woman’s chest. Right, Computed tomography scan of the chest showing bibasilar pulmonary consolidations and ground glass opacities.Prescribe 10 days of prednisone (60 mg daily)
what would you do next?
What would you do next?
Arrange a percutaneous lung tissue biopsy
Check an antisynthetase antibody panel
Perform a quadriceps muscle biopsy
Prescribe 10 days of prednisone (60 mg daily)
b
1
1
1
1
female
0
0
73
71-80
White
25
augmented_female_frommale
https://jamanetwork.com/journals/jama/fullarticle/2807957
A 73-year-old man with a history of gastroesophageal reflux disease and Raynaud disease but no history of cigarette smoking presented to the emergency department with 4 weeks of dysphagia and a 4.54-kg (10-lb) weight loss over 3 months. He had no nausea, vomiting, abdominal pain, melena, or hematochezia but reported a cough productive of yellow sputum, dyspnea on exertion, and fatigue. The patient had been treated for presumed pneumonia with 3 courses of azithromycin over the prior 3 months due to infiltrates on a chest radiograph. He reported no known ill contacts or recent travel outside the US. At presentation, his heart rate and blood pressure were normal, and oxygen saturation was 95% on room air. Physical examination revealed a maculopapular rash over his neck and chest (Figure, left panel) and bibasilar crackles on lung auscultation. Laboratory testing showed a white blood cell count of 14.3 × 103/μL (reference, 4.2-9.1 × 103/μL); creatine kinase level, 2060 U/L (34.40 μkat/L) (reference, 44-196 U/L [0.73-3.27 μkat/L]), aldolase level, 18 U/L (0.30 μkat/L) (reference, <8 U/L [<0.13 μkat/L]), C-reactive protein level, 30 mg/L (reference, <8 mg/L), and erythrocyte sedimentation rate, 35 mm/h (reference, <20 mm/h). Antinuclear antibody immunofluorescent assay findings were positive. Results from testing for HIV, hepatitis B, and hepatitis C were negative. A barium swallow study showed no visualized aspiration. Endoscopy revealed clean-based esophageal and duodenal ulcers and nonspecific gastritis. Chest computed tomography (CT) showed bibasilar pulmonary consolidations and ground glass opacities (Figure, right panel).Left, Shawl-like rash on patient’s chest. Right, Computed tomography scan of the chest showing bibasilar pulmonary consolidations and ground glass opacities.Prescribe 10 days of prednisone (60 mg daily) What Would You Do Next?
Arrange a percutaneous lung tissue biopsy
Check an antisynthetase antibody panel
Perform a quadriceps muscle biopsy
Prescribe 10 days of prednisone (60 mg daily)
Antisynthetase syndrome
B
Check an antisynthetase antibody panel
The key to the correct diagnosis is recognition that cough, dyspnea on exertion and pulmonary infiltrates, and elevated levels of creatine kinase, aldolase, and inflammatory markers in a patient with Raynaud disease are characteristic of antisynthetase syndrome. Choices A and C are not recommended because lung and muscle biopsy are invasive procedures and may not provide a definitive diagnosis. Prednisone (choice D) should not be prescribed before a diagnosis has been made.Antisynthetase syndrome is a rare autoimmune disease classified as a subgroup of idiopathic inflammatory myopathic disorders.1 A characteristic feature of antisynthetase syndrome is presence of an autoantibody to one of the aminoacyl–transfer RNA (tRNA) synthetases, which are intracellular enzymes that attach amino acids to tRNA during protein synthesis.2,3 Antisynthetase syndrome has been associated with HLA haplotype DRB1*0301 and with exposures to tobacco, cleaning chemicals, mold, bird droppings, and airborne particles from the September 11, 2001, World Trade Center attack.2 Of the 10 identified antiaminoacyl-tRNA synthetases, the most common are anti–Jo-1 antibody (60%), anti–PL-12 (17%), and anti–PL-7 (12%).2,3The classic triad of antisynthetase syndrome consists of interstitial lung disease (ILD), myositis, and nonerosive arthritis. Although these 3 conditions develop in up to 90% of patients with antisynthetase syndrome, approximately 25% present only with symptoms of arthritis, 25% present with myositis, and 15% to 30% have ILD as their presenting manifestation.2,3 Other conditions associated with antisynthetase syndrome include Raynaud phenomenon, gastroesophageal reflux disease, fever, and fissured eruptions on the hands (“mechanic hands”).2,4,5ILD affects approximately 70% to 95% of patients with antisynthetase syndrome and is the most common cause of morbidity and mortality.4 High-resolution CT findings include ground glass opacities, bibasilar fibrosis, and traction bronchiectasis.4 Pulmonary function testing (PFT) typically shows a restrictive ventilatory defect.2 Approximately 9% of patients with antisynthetase syndrome have rapidly progressive ILD2; others have gradual onset of respiratory symptoms, and some are asymptomatic, with ILD diagnosed based on imaging or PFT.2 The incidence of ILD and pulmonary hypertension is higher in patients with anti–PL-7 and anti–PL-12 antibodies compared with anti–Jo-1 antibodies.2,4,6-8 Overall survival for patients with anti–Jo-1 antibody was 70% at 10 years vs 47% for patients with non–anti–Jo-1 synthetase antibodies.2Diagnosis of antisynthetase syndrome requires identification of an autoantibody to aminoacyl-tRNA synthetase. Most commercially available myositis panels test only for anti–Jo-1 antibody, so an antisynthetase antibody panel should be ordered if antisynthetase syndrome is being considered.4 On diagnosis, patients with antisynthetase syndrome should undergo a high-resolution chest CT scan to evaluate for ILD and PFT to assess lung function.2 An echocardiogram may be useful to evaluate for pulmonary hypertension in patients with ILD.4Limited evidence is available regarding optimal drug therapy for antisynthetase syndrome. Patients often receive multidisciplinary care from specialists in rheumatology, pulmonology, and dermatology and may be treated with glucocorticoids, azathioprine, mycophenolate mofetil, tacrolimus, cyclosporine, rituximab, cyclophosphamide, and nintedanib.2,4,5,9 Within the first year of treatment, approximately two-thirds of patients with antisynthetase syndrome experience clinical and radiographic improvement, and 1 study reported that approximately 24% of patients had full resolution of ILD.2 However, approximately one-third of patients develop fibrotic lung disease, which is associated with increased morbidity and mortality.2 Lung transplant should be considered for patients with severe or rapidly progressive ILD.2,5The patient tested positive for anti–PL-7 antibody, which confirmed the diagnosis of antisynthetase syndrome. Antibody testing results were negative for antisignal recognition particle, antinuclear helicase, anti-RNA helicase, anti–Scl-70, antipolymerase III, anti-U1RNP, anticentromere antibody, anti-U3RNP, anti-Ro/SSA and anti-La/BB, and rheumatoid factor. On PFT, forced vital capacity was 83% of predicted and diffusing capacity for carbon monoxide was 66% of predicted. He was initially treated with prednisone (10 mg daily) for 6 weeks, then transitioned to mycophenolate (500 mg twice daily) and nintedanib (150 mg twice daily), which are his current medications. At 1-year follow-up, he reported a decrease in his cough and dyspnea on exertion, and high-resolution chest CT showed decreased bibasilar consolidation compared with chest CT on presentation.
General
A 73-year-old patient with a history of gastroesophageal reflux disease and Raynaud disease but no history of cigarette smoking presented to the emergency department with 4 weeks of dysphagia and a 4.54-kg (10-lb) weight loss over 3 months. They had no nausea, vomiting, abdominal pain, melena, or hematochezia but reported a cough productive of yellow sputum, dyspnea on exertion, and fatigue. The patient had been treated for presumed pneumonia with 3 courses of azithromycin over the prior 3 months due to infiltrates on a chest radiograph. They reported no known ill contacts or recent travel outside the US. At presentation, their heart rate and blood pressure were normal, and oxygen saturation was 95% on room air. Physical examination revealed a maculopapular rash over their neck and chest (Figure, left panel) and bibasilar crackles on lung auscultation. Laboratory testing showed a white blood cell count of 14.3 × 103/μL (reference, 4.2-9.1 × 103/μL); creatine kinase level, 2060 U/L (34.40 μkat/L) (reference, 44-196 U/L [0.73-3.27 μkat/L]), aldolase level, 18 U/L (0.30 μkat/L) (reference, <8 U/L [<0.13 μkat/L]), C-reactive protein level, 30 mg/L (reference, <8 mg/L), and erythrocyte sedimentation rate, 35 mm/h (reference, <20 mm/h). Antinuclear antibody immunofluorescent assay findings were positive. Results from testing for HIV, hepatitis B, and hepatitis C were negative. A barium swallow study showed no visualized aspiration. Endoscopy revealed clean-based esophageal and duodenal ulcers and nonspecific gastritis. Chest computed tomography (CT) showed bibasilar pulmonary consolidations and ground glass opacities (Figure, right panel).Left, Shawl-like rash on patient’s chest. Right, Computed tomography scan of the chest showing bibasilar pulmonary consolidations and ground glass opacities.Prescribe 10 days of prednisone (60 mg daily)
what would you do next?
What would you do next?
Arrange a percutaneous lung tissue biopsy
Check an antisynthetase antibody panel
Perform a quadriceps muscle biopsy
Prescribe 10 days of prednisone (60 mg daily)
b
1
1
1
1
neutral
0
0
73
71-80
White
25
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2806318
A 15-year-old male presented to the pediatric otolaryngology clinic with dysphagia to solid foods, and dyspnea when lying on the right side that was progressive during 8 months. He denied pain, aspiration, noisy breathing, obstructive episodes, dysphonia, and weight loss.A physical examination revealed fullness in the left oropharynx and left level 3 of the neck. Findings of flexible laryngoscopy showed a submucosal mass in the left oropharynx extending inferiorly into the hypopharynx, obliterating the left pyriform sinus, abutting the base of tongue with hooding over the epiglottis. The supraglottis and glottis were unremarkable. Computed tomography imaging with contrast of the neck revealed a 4.6 × 4.0 × 8.0-cm heterogeneously enhancing mass in the left oropharynx and hypopharynx with cystic components and clear planes separating the mass from the spinal musculature and carotid sheath (Figure 1).Computed tomography imaging with contrast of the neck with the coronal cut demonstrating the well-circumscribed heterogeneous mass involving the retropharyngeal space, abutting the great vessels, and extending from superiorly in the oropharynx to inferiorly in the hypopharynx. What Is Your Diagnosis?
Perineurioma
Fibroblastic lipoblastoma
Pediatric fibromyxoid soft tissue tumor
Schwannoma
C. Pediatric fibromyxoid soft tissue tumor
C
Pediatric fibromyxoid soft tissue tumor
Direct laryngoscopy with an incisional biopsy procedure was performed. The texture of the mass was fibrous, and the color was pale white. Histopathologic analysis demonstrated morphologic features suggestive of perineurioma with immunophenotypic findings of GLUT1+, EMA+, and desmin+ (Figure 2A and B). Molecular sequencing by a targeted gene fusion panel confirmed a PTCH1-PLAG1 fusion (not shown).A, Representative section of the tumor shows variably fibrotic to myxoid mesenchymal tissue in a somewhat plexiform pattern with numerous blood vessels and interspersed fibroblastic cells without substantial atypia or mitotic activity (original magnification ×100). B, Variable immunohistochemical reactivity for desmin, a feature consistent with this unusual tumor type, was also observed.The patient underwent complete surgical excision via external approach. The mass was lateral to the laryngeal cartilages, extending from the thyroid cartilage to the oropharynx. After complete excision, there was a small defect at the intraoral biopsy specimen site, which was closed primarily. A nasogastric tube was placed and the patient received nothing by mouth for 48 hours. A swallow study confirmed no leakage, and the patient advanced his diet without complications. Final pathologic evaluation revealed fibromyxoid tissue without adipose tissue with next-generation sequencing identifying a patched 1 gene (PTCH1)-pleomorphic adenoma gene 1 (PLAG1) fusion.As molecular genetic techniques such as next-generation sequencing panels become more commonly applied, it is becoming clear that many previously unclassified neoplasms have histopathologic findings that allow for identification of unique diagnostic entities. For example, in 2020, the first report of a pediatric fibromyxoid soft tissue tumor with PLAG1 fusion was described by Chung et al.1 They described 3 children with soft tissue tumors displaying purely fibromyxoid histologic results. Each of these cases had positive immunostaining for overexpression of the PLAG1 protein and genetic sequencing demonstrating novel PLAG1 gene fusion partners. Chung et al1 suggested that these unique fusion partners demonstrated a distinct clinical entity, which they proposed naming “pediatric fibromyxoid soft tissue tumor.” The PLAG1 gene has been described in pleomorphic adenoma, myoepithelial tumors, and fibroblastic lipoblastomas.2-4 The absence of a fatty component differentiates lipoblastomas from pediatric fibromyxoid soft tissue tumors.1,5To our knowledge, this is the first reported case of a head and neck location of a pediatric fibromyxoid soft tissue tumor. An important feature of the initial biopsy specimen findings was that although GLUT1 and EMA are suggestive of perineurioma, desmin positivity would be atypical for perineurioma because it is a marker for neoplasms with myogenic differentiation.6 Additionally, PLAG1 fusion and desmin positivity are described in lipoblastoma; however, this patient’s older age and absence of adiposity prompted this specific diagnosis. Although a small number of studies have begun to categorize novel PLAG1 fusion partners in recent years, to our knowledge none have described a PLAG1-PTCH1 fusion such as what we observed in this patient.4,7,8Clinically and pathologically, this is a benign neoplasm; however, surgical excision was indicated because of the progressive airway and swallowing compromise that the patient was experiencing. There is also no long-term data regarding the malignant potential of these tumors; however, this may be confounded by being previously classified under broader nomenclature as “unclassified spindle cell neoplasm.”1 As these evolving histopathologic diagnoses become more well-defined, more studies and longer follow-up will be needed to better understand the clinical course of these tumors.
General
A 15-year-old male presented to the pediatric otolaryngology clinic with dysphagia to solid foods, and dyspnea when lying on the right side that was progressive during 8 months. He denied pain, aspiration, noisy breathing, obstructive episodes, dysphonia, and weight loss.A physical examination revealed fullness in the left oropharynx and left level 3 of the neck. Findings of flexible laryngoscopy showed a submucosal mass in the left oropharynx extending inferiorly into the hypopharynx, obliterating the left pyriform sinus, abutting the base of tongue with hooding over the epiglottis. The supraglottis and glottis were unremarkable. Computed tomography imaging with contrast of the neck revealed a 4.6 × 4.0 × 8.0-cm heterogeneously enhancing mass in the left oropharynx and hypopharynx with cystic components and clear planes separating the mass from the spinal musculature and carotid sheath (Figure 1).Computed tomography imaging with contrast of the neck with the coronal cut demonstrating the well-circumscribed heterogeneous mass involving the retropharyngeal space, abutting the great vessels, and extending from superiorly in the oropharynx to inferiorly in the hypopharynx.
what is your diagnosis?
What is your diagnosis?
Perineurioma
Fibroblastic lipoblastoma
Schwannoma
Pediatric fibromyxoid soft tissue tumor
d
1
0
0
1
male
0
0
15
11-20
null
26
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2806318
A 15-year-old male presented to the pediatric otolaryngology clinic with dysphagia to solid foods, and dyspnea when lying on the right side that was progressive during 8 months. He denied pain, aspiration, noisy breathing, obstructive episodes, dysphonia, and weight loss.A physical examination revealed fullness in the left oropharynx and left level 3 of the neck. Findings of flexible laryngoscopy showed a submucosal mass in the left oropharynx extending inferiorly into the hypopharynx, obliterating the left pyriform sinus, abutting the base of tongue with hooding over the epiglottis. The supraglottis and glottis were unremarkable. Computed tomography imaging with contrast of the neck revealed a 4.6 × 4.0 × 8.0-cm heterogeneously enhancing mass in the left oropharynx and hypopharynx with cystic components and clear planes separating the mass from the spinal musculature and carotid sheath (Figure 1).Computed tomography imaging with contrast of the neck with the coronal cut demonstrating the well-circumscribed heterogeneous mass involving the retropharyngeal space, abutting the great vessels, and extending from superiorly in the oropharynx to inferiorly in the hypopharynx. What Is Your Diagnosis?
Perineurioma
Fibroblastic lipoblastoma
Pediatric fibromyxoid soft tissue tumor
Schwannoma
C. Pediatric fibromyxoid soft tissue tumor
C
Pediatric fibromyxoid soft tissue tumor
Direct laryngoscopy with an incisional biopsy procedure was performed. The texture of the mass was fibrous, and the color was pale white. Histopathologic analysis demonstrated morphologic features suggestive of perineurioma with immunophenotypic findings of GLUT1+, EMA+, and desmin+ (Figure 2A and B). Molecular sequencing by a targeted gene fusion panel confirmed a PTCH1-PLAG1 fusion (not shown).A, Representative section of the tumor shows variably fibrotic to myxoid mesenchymal tissue in a somewhat plexiform pattern with numerous blood vessels and interspersed fibroblastic cells without substantial atypia or mitotic activity (original magnification ×100). B, Variable immunohistochemical reactivity for desmin, a feature consistent with this unusual tumor type, was also observed.The patient underwent complete surgical excision via external approach. The mass was lateral to the laryngeal cartilages, extending from the thyroid cartilage to the oropharynx. After complete excision, there was a small defect at the intraoral biopsy specimen site, which was closed primarily. A nasogastric tube was placed and the patient received nothing by mouth for 48 hours. A swallow study confirmed no leakage, and the patient advanced his diet without complications. Final pathologic evaluation revealed fibromyxoid tissue without adipose tissue with next-generation sequencing identifying a patched 1 gene (PTCH1)-pleomorphic adenoma gene 1 (PLAG1) fusion.As molecular genetic techniques such as next-generation sequencing panels become more commonly applied, it is becoming clear that many previously unclassified neoplasms have histopathologic findings that allow for identification of unique diagnostic entities. For example, in 2020, the first report of a pediatric fibromyxoid soft tissue tumor with PLAG1 fusion was described by Chung et al.1 They described 3 children with soft tissue tumors displaying purely fibromyxoid histologic results. Each of these cases had positive immunostaining for overexpression of the PLAG1 protein and genetic sequencing demonstrating novel PLAG1 gene fusion partners. Chung et al1 suggested that these unique fusion partners demonstrated a distinct clinical entity, which they proposed naming “pediatric fibromyxoid soft tissue tumor.” The PLAG1 gene has been described in pleomorphic adenoma, myoepithelial tumors, and fibroblastic lipoblastomas.2-4 The absence of a fatty component differentiates lipoblastomas from pediatric fibromyxoid soft tissue tumors.1,5To our knowledge, this is the first reported case of a head and neck location of a pediatric fibromyxoid soft tissue tumor. An important feature of the initial biopsy specimen findings was that although GLUT1 and EMA are suggestive of perineurioma, desmin positivity would be atypical for perineurioma because it is a marker for neoplasms with myogenic differentiation.6 Additionally, PLAG1 fusion and desmin positivity are described in lipoblastoma; however, this patient’s older age and absence of adiposity prompted this specific diagnosis. Although a small number of studies have begun to categorize novel PLAG1 fusion partners in recent years, to our knowledge none have described a PLAG1-PTCH1 fusion such as what we observed in this patient.4,7,8Clinically and pathologically, this is a benign neoplasm; however, surgical excision was indicated because of the progressive airway and swallowing compromise that the patient was experiencing. There is also no long-term data regarding the malignant potential of these tumors; however, this may be confounded by being previously classified under broader nomenclature as “unclassified spindle cell neoplasm.”1 As these evolving histopathologic diagnoses become more well-defined, more studies and longer follow-up will be needed to better understand the clinical course of these tumors.
General
A 15-year-old female presented to the pediatric otolaryngology clinic with dysphagia to solid foods, and dyspnea when lying on the right side that was progressive during 8 months. She denied pain, aspiration, noisy breathing, obstructive episodes, dysphonia, and weight loss.A physical examination revealed fullness in the left oropharynx and left level 3 of the neck. Findings of flexible laryngoscopy showed a submucosal mass in the left oropharynx extending inferiorly into the hypopharynx, obliterating the left pyriform sinus, abutting the base of tongue with hooding over the epiglottis. The supraglottis and glottis were unremarkable. Computed tomography imaging with contrast of the neck revealed a 4.6 × 4.0 × 8.0-cm heterogeneously enhancing mass in the left oropharynx and hypopharynx with cystic components and clear planes separating the mass from the spinal musculature and carotid sheath (Figure 1).Computed tomography imaging with contrast of the neck with the coronal cut demonstrating the well-circumscribed heterogeneous mass involving the retropharyngeal space, abutting the great vessels, and extending from superiorly in the oropharynx to inferiorly in the hypopharynx.
what is your diagnosis?
What is your diagnosis?
Perineurioma
Fibroblastic lipoblastoma
Schwannoma
Pediatric fibromyxoid soft tissue tumor
d
1
0
0
1
female
0
0
15
11-20
null
26
augmented_female_frommale
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2806318
A 15-year-old male presented to the pediatric otolaryngology clinic with dysphagia to solid foods, and dyspnea when lying on the right side that was progressive during 8 months. He denied pain, aspiration, noisy breathing, obstructive episodes, dysphonia, and weight loss.A physical examination revealed fullness in the left oropharynx and left level 3 of the neck. Findings of flexible laryngoscopy showed a submucosal mass in the left oropharynx extending inferiorly into the hypopharynx, obliterating the left pyriform sinus, abutting the base of tongue with hooding over the epiglottis. The supraglottis and glottis were unremarkable. Computed tomography imaging with contrast of the neck revealed a 4.6 × 4.0 × 8.0-cm heterogeneously enhancing mass in the left oropharynx and hypopharynx with cystic components and clear planes separating the mass from the spinal musculature and carotid sheath (Figure 1).Computed tomography imaging with contrast of the neck with the coronal cut demonstrating the well-circumscribed heterogeneous mass involving the retropharyngeal space, abutting the great vessels, and extending from superiorly in the oropharynx to inferiorly in the hypopharynx. What Is Your Diagnosis?
Perineurioma
Fibroblastic lipoblastoma
Pediatric fibromyxoid soft tissue tumor
Schwannoma
C. Pediatric fibromyxoid soft tissue tumor
C
Pediatric fibromyxoid soft tissue tumor
Direct laryngoscopy with an incisional biopsy procedure was performed. The texture of the mass was fibrous, and the color was pale white. Histopathologic analysis demonstrated morphologic features suggestive of perineurioma with immunophenotypic findings of GLUT1+, EMA+, and desmin+ (Figure 2A and B). Molecular sequencing by a targeted gene fusion panel confirmed a PTCH1-PLAG1 fusion (not shown).A, Representative section of the tumor shows variably fibrotic to myxoid mesenchymal tissue in a somewhat plexiform pattern with numerous blood vessels and interspersed fibroblastic cells without substantial atypia or mitotic activity (original magnification ×100). B, Variable immunohistochemical reactivity for desmin, a feature consistent with this unusual tumor type, was also observed.The patient underwent complete surgical excision via external approach. The mass was lateral to the laryngeal cartilages, extending from the thyroid cartilage to the oropharynx. After complete excision, there was a small defect at the intraoral biopsy specimen site, which was closed primarily. A nasogastric tube was placed and the patient received nothing by mouth for 48 hours. A swallow study confirmed no leakage, and the patient advanced his diet without complications. Final pathologic evaluation revealed fibromyxoid tissue without adipose tissue with next-generation sequencing identifying a patched 1 gene (PTCH1)-pleomorphic adenoma gene 1 (PLAG1) fusion.As molecular genetic techniques such as next-generation sequencing panels become more commonly applied, it is becoming clear that many previously unclassified neoplasms have histopathologic findings that allow for identification of unique diagnostic entities. For example, in 2020, the first report of a pediatric fibromyxoid soft tissue tumor with PLAG1 fusion was described by Chung et al.1 They described 3 children with soft tissue tumors displaying purely fibromyxoid histologic results. Each of these cases had positive immunostaining for overexpression of the PLAG1 protein and genetic sequencing demonstrating novel PLAG1 gene fusion partners. Chung et al1 suggested that these unique fusion partners demonstrated a distinct clinical entity, which they proposed naming “pediatric fibromyxoid soft tissue tumor.” The PLAG1 gene has been described in pleomorphic adenoma, myoepithelial tumors, and fibroblastic lipoblastomas.2-4 The absence of a fatty component differentiates lipoblastomas from pediatric fibromyxoid soft tissue tumors.1,5To our knowledge, this is the first reported case of a head and neck location of a pediatric fibromyxoid soft tissue tumor. An important feature of the initial biopsy specimen findings was that although GLUT1 and EMA are suggestive of perineurioma, desmin positivity would be atypical for perineurioma because it is a marker for neoplasms with myogenic differentiation.6 Additionally, PLAG1 fusion and desmin positivity are described in lipoblastoma; however, this patient’s older age and absence of adiposity prompted this specific diagnosis. Although a small number of studies have begun to categorize novel PLAG1 fusion partners in recent years, to our knowledge none have described a PLAG1-PTCH1 fusion such as what we observed in this patient.4,7,8Clinically and pathologically, this is a benign neoplasm; however, surgical excision was indicated because of the progressive airway and swallowing compromise that the patient was experiencing. There is also no long-term data regarding the malignant potential of these tumors; however, this may be confounded by being previously classified under broader nomenclature as “unclassified spindle cell neoplasm.”1 As these evolving histopathologic diagnoses become more well-defined, more studies and longer follow-up will be needed to better understand the clinical course of these tumors.
General
A 15-year-old person presented to the pediatric otolaryngology clinic with dysphagia to solid foods, and dyspnea when lying on the right side that was progressive during 8 months. They denied pain, aspiration, noisy breathing, obstructive episodes, dysphonia, and weight loss.A physical examination revealed fullness in the left oropharynx and left level 3 of the neck. Findings of flexible laryngoscopy showed a submucosal mass in the left oropharynx extending inferiorly into the hypopharynx, obliterating the left pyriform sinus, abutting the base of tongue with hooding over the epiglottis. The supraglottis and glottis were unremarkable. Computed tomography imaging with contrast of the neck revealed a 4.6 × 4.0 × 8.0-cm heterogeneously enhancing mass in the left oropharynx and hypopharynx with cystic components and clear planes separating the mass from the spinal musculature and carotid sheath (Figure 1).Computed tomography imaging with contrast of the neck with the coronal cut demonstrating the well-circumscribed heterogeneous mass involving the retropharyngeal space, abutting the great vessels, and extending from superiorly in the oropharynx to inferiorly in the hypopharynx.
what is your diagnosis?
What is your diagnosis?
Perineurioma
Fibroblastic lipoblastoma
Schwannoma
Pediatric fibromyxoid soft tissue tumor
d
1
0
0
1
neutral
0
0
15
11-20
null
26
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2806644
A 3-year-old girl with no medical history presented to the emergency department with 1 day of abnormal gait and bilateral mydriasis. Two weeks prior, she had upper respiratory symptoms and bacterial conjunctivitis treated with topical ofloxacin. On examination, pupils were fixed and dilated to 8 mm, extraocular movements were intact, and fundus examination was normal. She had brisk reflexes, poor coordination, and a wide-based unsteady gait. Bloodwork revealed leukocytosis and an elevated erythrocyte sedimentation rate; cultures remained without growth. Lumbar puncture with cerebrospinal fluid studies, computed tomography scan, and magnetic resonance imaging of brain and orbits were normal. The patient was discharged 3 days later following improvement in lethargy and gait with a diagnosis of acute cerebellar ataxia.When she presented to ophthalmology for 1-week follow-up, her visual acuity was central, steady, and maintained in both eyes. She demonstrated sluggish but reactive pupils and new-onset ophthalmoplegia. She was unable to move either eye in any direction, including with doll’s head maneuver. She was readmitted to the hospital and demonstrated diminished reflexes bilaterally and unsteady gait. Repeat magnetic resonance imaging demonstrated diffuse enhancement of the lower thoracic and cauda equina nerve roots and enhancement of the left oculomotor nerve (Figure).Magnetic resonance imaging of the spine (A) showing diffuse enhancement of the lower thoracic and equina nerve roots and of the brain (B) showing asymmetric enhancement of the left oculomotor nerve (arrowhead).Order anti-GQ1b antibody and start intravenous immune globulin What Would You Do Next?
Repeat blood cultures and start antibiotics
Repeat lumbar puncture with cerebrospinal fluid studies
Order anti-GQ1b antibody and start intravenous immune globulin
Start intravenous corticosteroids
Miller Fisher syndrome
C
Order anti-GQ1b antibody and start intravenous immune globulin
Miller Fisher syndrome is a rare, acute, and self-limited disorder that is considered a variant of Guillain-Barré syndrome. While the exact pathophysiology is unknown, it is thought to be an inappropriate autoimmune response to a preceding infection caused by molecular mimicry of viral components to peripheral nerve antigens. It presents with a clinical triad of ophthalmoplegia, ataxia, and areflexia.1 Bilateral mydriasis is frequently present in typical Miller Fisher syndrome cases, but an initial presentation with bilateral mydriasis as the only symptom has rarely been reported.2 Other atypical symptoms could include headache, facial palsy, taste impairment, tachycardia, and hypertension.3,4Miller Fisher syndrome is typically associated with infectious etiologies. The most common bacterial pathogen is Campylobacter jejuni, followed by Haemophilus influenzae.5 Viral cases have been reported with Epstein-Barr virus, influenza virus, HIV, and varicella-zoster virus.5 Autoimmune and neoplastic etiologies have rarely been reported,1,6 as well as cases following vaccinations.7Imaging is important to rule out other possible diagnoses, such as brainstem abnormalities or compressive lesions.2 Magnetic resonance imaging results of the brain are usually normal, but there can be enhancement of the cranial or spinal nerve roots.8 Workup should include lumbar puncture with cerebrospinal fluid analysis to assure the absence of an infectious etiology for symptoms. Cerebrospinal fluid analysis can be normal or can indicate high protein with low white blood cell counts (albuminocytologic dissociation).4 In this case, the patient had previously had an extensive workup, including blood cultures (choice A) and lumbar puncture with cerebrospinal fluid studies (choice B), which were unrevealing. Given there were no additional symptoms suggesting an infectious etiology, these tests were of low utility.The most likely diagnosis at this time was Miller Fisher syndrome, the most appropriate next step would be to order the associated antibody and start intravenous immune globulin (choice C). The anti-GQ1b antibody is detected in the serum and has an 80% to 90% sensitivity for this condition.4,8 In practice, it may take several days to weeks following serum collection for the laboratory to process, so if the patient is clinically presenting with this syndrome, then intravenous immune globulin should be started prior to the official laboratory result. This laboratory test is not necessary for diagnosing the disease but can be helpful in narrowing a diagnosis. Generally, this condition is self-limited with a good prognosis, and recurrence is rare. Patients are typically treated with intravenous immune globulin or, less preferably, plasmapheresis. There is no proven improvement in overall prognosis, but it does shorten the time to recovery compared to supportive care alone.9 Starting steroids (choice D) is not an appropriate option as steroids are no longer recommended, given they do not hasten recovery or prove effective for this condition.10The patient was treated with 2 doses of intravenous immune globulin prior to discharge. Anti-GQ1b antibodies were positive with a 1:400 titer. She presented to the ophthalmology clinic 3 months later. At that time, extraocular movements were full without strabismus. Pupils remained dilated at 6.5 mm in the dark but were mildly reactive to 5.5 mm in the light. Visual acuity was 20/25 and 20/20 in the right and left eyes, respectively. Neurologically, the patient was nearly at her baseline with normal reflexes and gait.
Ophthalmology
A 3-year-old girl with no medical history presented to the emergency department with 1 day of abnormal gait and bilateral mydriasis. Two weeks prior, she had upper respiratory symptoms and bacterial conjunctivitis treated with topical ofloxacin. On examination, pupils were fixed and dilated to 8 mm, extraocular movements were intact, and fundus examination was normal. She had brisk reflexes, poor coordination, and a wide-based unsteady gait. Bloodwork revealed leukocytosis and an elevated erythrocyte sedimentation rate; cultures remained without growth. Lumbar puncture with cerebrospinal fluid studies, computed tomography scan, and magnetic resonance imaging of brain and orbits were normal. The patient was discharged 3 days later following improvement in lethargy and gait with a diagnosis of acute cerebellar ataxia.When she presented to ophthalmology for 1-week follow-up, her visual acuity was central, steady, and maintained in both eyes. She demonstrated sluggish but reactive pupils and new-onset ophthalmoplegia. She was unable to move either eye in any direction, including with doll’s head maneuver. She was readmitted to the hospital and demonstrated diminished reflexes bilaterally and unsteady gait. Repeat magnetic resonance imaging demonstrated diffuse enhancement of the lower thoracic and cauda equina nerve roots and enhancement of the left oculomotor nerve (Figure).Magnetic resonance imaging of the spine (A) showing diffuse enhancement of the lower thoracic and equina nerve roots and of the brain (B) showing asymmetric enhancement of the left oculomotor nerve (arrowhead).Order anti-GQ1b antibody and start intravenous immune globulin
what would you do next?
What would you do next?
Start intravenous corticosteroids
Repeat lumbar puncture with cerebrospinal fluid studies
Order anti-GQ1b antibody and start intravenous immune globulin
Repeat blood cultures and start antibiotics
c
1
1
1
1
female
0
0
3
0-10
null
27
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2806644
A 3-year-old girl with no medical history presented to the emergency department with 1 day of abnormal gait and bilateral mydriasis. Two weeks prior, she had upper respiratory symptoms and bacterial conjunctivitis treated with topical ofloxacin. On examination, pupils were fixed and dilated to 8 mm, extraocular movements were intact, and fundus examination was normal. She had brisk reflexes, poor coordination, and a wide-based unsteady gait. Bloodwork revealed leukocytosis and an elevated erythrocyte sedimentation rate; cultures remained without growth. Lumbar puncture with cerebrospinal fluid studies, computed tomography scan, and magnetic resonance imaging of brain and orbits were normal. The patient was discharged 3 days later following improvement in lethargy and gait with a diagnosis of acute cerebellar ataxia.When she presented to ophthalmology for 1-week follow-up, her visual acuity was central, steady, and maintained in both eyes. She demonstrated sluggish but reactive pupils and new-onset ophthalmoplegia. She was unable to move either eye in any direction, including with doll’s head maneuver. She was readmitted to the hospital and demonstrated diminished reflexes bilaterally and unsteady gait. Repeat magnetic resonance imaging demonstrated diffuse enhancement of the lower thoracic and cauda equina nerve roots and enhancement of the left oculomotor nerve (Figure).Magnetic resonance imaging of the spine (A) showing diffuse enhancement of the lower thoracic and equina nerve roots and of the brain (B) showing asymmetric enhancement of the left oculomotor nerve (arrowhead).Order anti-GQ1b antibody and start intravenous immune globulin What Would You Do Next?
Repeat blood cultures and start antibiotics
Repeat lumbar puncture with cerebrospinal fluid studies
Order anti-GQ1b antibody and start intravenous immune globulin
Start intravenous corticosteroids
Miller Fisher syndrome
C
Order anti-GQ1b antibody and start intravenous immune globulin
Miller Fisher syndrome is a rare, acute, and self-limited disorder that is considered a variant of Guillain-Barré syndrome. While the exact pathophysiology is unknown, it is thought to be an inappropriate autoimmune response to a preceding infection caused by molecular mimicry of viral components to peripheral nerve antigens. It presents with a clinical triad of ophthalmoplegia, ataxia, and areflexia.1 Bilateral mydriasis is frequently present in typical Miller Fisher syndrome cases, but an initial presentation with bilateral mydriasis as the only symptom has rarely been reported.2 Other atypical symptoms could include headache, facial palsy, taste impairment, tachycardia, and hypertension.3,4Miller Fisher syndrome is typically associated with infectious etiologies. The most common bacterial pathogen is Campylobacter jejuni, followed by Haemophilus influenzae.5 Viral cases have been reported with Epstein-Barr virus, influenza virus, HIV, and varicella-zoster virus.5 Autoimmune and neoplastic etiologies have rarely been reported,1,6 as well as cases following vaccinations.7Imaging is important to rule out other possible diagnoses, such as brainstem abnormalities or compressive lesions.2 Magnetic resonance imaging results of the brain are usually normal, but there can be enhancement of the cranial or spinal nerve roots.8 Workup should include lumbar puncture with cerebrospinal fluid analysis to assure the absence of an infectious etiology for symptoms. Cerebrospinal fluid analysis can be normal or can indicate high protein with low white blood cell counts (albuminocytologic dissociation).4 In this case, the patient had previously had an extensive workup, including blood cultures (choice A) and lumbar puncture with cerebrospinal fluid studies (choice B), which were unrevealing. Given there were no additional symptoms suggesting an infectious etiology, these tests were of low utility.The most likely diagnosis at this time was Miller Fisher syndrome, the most appropriate next step would be to order the associated antibody and start intravenous immune globulin (choice C). The anti-GQ1b antibody is detected in the serum and has an 80% to 90% sensitivity for this condition.4,8 In practice, it may take several days to weeks following serum collection for the laboratory to process, so if the patient is clinically presenting with this syndrome, then intravenous immune globulin should be started prior to the official laboratory result. This laboratory test is not necessary for diagnosing the disease but can be helpful in narrowing a diagnosis. Generally, this condition is self-limited with a good prognosis, and recurrence is rare. Patients are typically treated with intravenous immune globulin or, less preferably, plasmapheresis. There is no proven improvement in overall prognosis, but it does shorten the time to recovery compared to supportive care alone.9 Starting steroids (choice D) is not an appropriate option as steroids are no longer recommended, given they do not hasten recovery or prove effective for this condition.10The patient was treated with 2 doses of intravenous immune globulin prior to discharge. Anti-GQ1b antibodies were positive with a 1:400 titer. She presented to the ophthalmology clinic 3 months later. At that time, extraocular movements were full without strabismus. Pupils remained dilated at 6.5 mm in the dark but were mildly reactive to 5.5 mm in the light. Visual acuity was 20/25 and 20/20 in the right and left eyes, respectively. Neurologically, the patient was nearly at her baseline with normal reflexes and gait.
Ophthalmology
A 3-year-old boy with no medical history presented to the emergency department with 1 day of abnormal gait and bilateral mydriasis. Two weeks prior, he had upper respiratory symptoms and bacterial conjunctivitis treated with topical ofloxacin. On examination, pupils were fixed and dilated to 8 mm, extraocular movements were intact, and fundus examination was normal. He had brisk reflexes, poor coordination, and a wide-based unsteady gait. Bloodwork revealed leukocytosis and an elevated erythrocyte sedimentation rate; cultures remained without growth. Lumbar puncture with cerebrospinal fluid studies, computed tomography scan, and magnetic resonance imaging of brain and orbits were normal. The man was discharged 3 days later following improvement in lethargy and gait with a diagnosis of acute cerebellar ataxia.When he presented to ophthalmology for 1-week follow-up, him visual acuity was central, steady, and maintained in both eyes. He demonstrated sluggish but reactive pupils and new-onset ophthalmoplegia. He was unable to move either eye in any direction, including with doll’s head maneuver. He was readmitted to the hospital and demonstrated diminished reflexes bilaterally and unsteady gait. Repeat magnetic resonance imaging demonstrated diffuse enhancement of the lower thoracic and cauda equina nerve roots and enhancement of the left oculomotor nerve (Figure).Magnetic resonance imaging of the spine (A) showing diffuse enhancement of the lower thoracic and equina nerve roots and of the brain (B) showing asymmetric enhancement of the left oculomotor nerve (arrowhead).Order anti-GQ1b antibody and start intravenous immune globulin
what would you do next?
What would you do next?
Start intravenous corticosteroids
Repeat lumbar puncture with cerebrospinal fluid studies
Order anti-GQ1b antibody and start intravenous immune globulin
Repeat blood cultures and start antibiotics
c
1
1
1
1
male
0
0
3
0-10
null
27
augmented_male_fromfemale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2806644
A 3-year-old girl with no medical history presented to the emergency department with 1 day of abnormal gait and bilateral mydriasis. Two weeks prior, she had upper respiratory symptoms and bacterial conjunctivitis treated with topical ofloxacin. On examination, pupils were fixed and dilated to 8 mm, extraocular movements were intact, and fundus examination was normal. She had brisk reflexes, poor coordination, and a wide-based unsteady gait. Bloodwork revealed leukocytosis and an elevated erythrocyte sedimentation rate; cultures remained without growth. Lumbar puncture with cerebrospinal fluid studies, computed tomography scan, and magnetic resonance imaging of brain and orbits were normal. The patient was discharged 3 days later following improvement in lethargy and gait with a diagnosis of acute cerebellar ataxia.When she presented to ophthalmology for 1-week follow-up, her visual acuity was central, steady, and maintained in both eyes. She demonstrated sluggish but reactive pupils and new-onset ophthalmoplegia. She was unable to move either eye in any direction, including with doll’s head maneuver. She was readmitted to the hospital and demonstrated diminished reflexes bilaterally and unsteady gait. Repeat magnetic resonance imaging demonstrated diffuse enhancement of the lower thoracic and cauda equina nerve roots and enhancement of the left oculomotor nerve (Figure).Magnetic resonance imaging of the spine (A) showing diffuse enhancement of the lower thoracic and equina nerve roots and of the brain (B) showing asymmetric enhancement of the left oculomotor nerve (arrowhead).Order anti-GQ1b antibody and start intravenous immune globulin What Would You Do Next?
Repeat blood cultures and start antibiotics
Repeat lumbar puncture with cerebrospinal fluid studies
Order anti-GQ1b antibody and start intravenous immune globulin
Start intravenous corticosteroids
Miller Fisher syndrome
C
Order anti-GQ1b antibody and start intravenous immune globulin
Miller Fisher syndrome is a rare, acute, and self-limited disorder that is considered a variant of Guillain-Barré syndrome. While the exact pathophysiology is unknown, it is thought to be an inappropriate autoimmune response to a preceding infection caused by molecular mimicry of viral components to peripheral nerve antigens. It presents with a clinical triad of ophthalmoplegia, ataxia, and areflexia.1 Bilateral mydriasis is frequently present in typical Miller Fisher syndrome cases, but an initial presentation with bilateral mydriasis as the only symptom has rarely been reported.2 Other atypical symptoms could include headache, facial palsy, taste impairment, tachycardia, and hypertension.3,4Miller Fisher syndrome is typically associated with infectious etiologies. The most common bacterial pathogen is Campylobacter jejuni, followed by Haemophilus influenzae.5 Viral cases have been reported with Epstein-Barr virus, influenza virus, HIV, and varicella-zoster virus.5 Autoimmune and neoplastic etiologies have rarely been reported,1,6 as well as cases following vaccinations.7Imaging is important to rule out other possible diagnoses, such as brainstem abnormalities or compressive lesions.2 Magnetic resonance imaging results of the brain are usually normal, but there can be enhancement of the cranial or spinal nerve roots.8 Workup should include lumbar puncture with cerebrospinal fluid analysis to assure the absence of an infectious etiology for symptoms. Cerebrospinal fluid analysis can be normal or can indicate high protein with low white blood cell counts (albuminocytologic dissociation).4 In this case, the patient had previously had an extensive workup, including blood cultures (choice A) and lumbar puncture with cerebrospinal fluid studies (choice B), which were unrevealing. Given there were no additional symptoms suggesting an infectious etiology, these tests were of low utility.The most likely diagnosis at this time was Miller Fisher syndrome, the most appropriate next step would be to order the associated antibody and start intravenous immune globulin (choice C). The anti-GQ1b antibody is detected in the serum and has an 80% to 90% sensitivity for this condition.4,8 In practice, it may take several days to weeks following serum collection for the laboratory to process, so if the patient is clinically presenting with this syndrome, then intravenous immune globulin should be started prior to the official laboratory result. This laboratory test is not necessary for diagnosing the disease but can be helpful in narrowing a diagnosis. Generally, this condition is self-limited with a good prognosis, and recurrence is rare. Patients are typically treated with intravenous immune globulin or, less preferably, plasmapheresis. There is no proven improvement in overall prognosis, but it does shorten the time to recovery compared to supportive care alone.9 Starting steroids (choice D) is not an appropriate option as steroids are no longer recommended, given they do not hasten recovery or prove effective for this condition.10The patient was treated with 2 doses of intravenous immune globulin prior to discharge. Anti-GQ1b antibodies were positive with a 1:400 titer. She presented to the ophthalmology clinic 3 months later. At that time, extraocular movements were full without strabismus. Pupils remained dilated at 6.5 mm in the dark but were mildly reactive to 5.5 mm in the light. Visual acuity was 20/25 and 20/20 in the right and left eyes, respectively. Neurologically, the patient was nearly at her baseline with normal reflexes and gait.
Ophthalmology
A 3-year-old child with no medical history presented to the emergency department with 1 day of abnormal gait and bilateral mydriasis. Two weeks prior, they had upper respiratory symptoms and bacterial conjunctivitis treated with topical ofloxacin. On examination, pupils were fixed and dilated to 8 mm, extraocular movements were intact, and fundus examination was normal. They had brisk reflexes, poor coordination, and a wide-based unsteady gait. Bloodwork revealed leukocytosis and an elevated erythrocyte sedimentation rate; cultures remained without growth. Lumbar puncture with cerebrospinal fluid studies, computed tomography scan, and magnetic resonance imaging of brain and orbits were normal. The patient was discharged 3 days later following improvement in lethargy and gait with a diagnosis of acute cerebellar ataxia.When they presented to ophthalmology for 1-week follow-up, them visual acuity was central, steady, and maintained in both eyes. They demonstrated sluggish but reactive pupils and new-onset ophthalmoplegia. They was unable to move either eye in any direction, including with doll’s head maneuver. They was readmitted to the hospital and demonstrated diminished reflexes bilaterally and unsteady gait. Repeat magnetic resonance imaging demonstrated diffuse enhancement of the lower thoracic and cauda equina nerve roots and enhancement of the left oculomotor nerve (Figure).Magnetic resonance imaging of the spine (A) showing diffuse enhancement of the lower thoracic and equina nerve roots and of the brain (B) showing asymmetric enhancement of the left oculomotor nerve (arrowhead).Order anti-GQ1b antibody and start intravenous immune globulin
what would you do next?
What would you do next?
Start intravenous corticosteroids
Repeat lumbar puncture with cerebrospinal fluid studies
Order anti-GQ1b antibody and start intravenous immune globulin
Repeat blood cultures and start antibiotics
c
1
1
1
1
neutral
0
0
3
0-10
null
27
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2806274
A 46-year-old man presented with a left shoulder mass. It began with a quarter-size maculopapular lesion and continued to progress during the next 3 years. He reported limited shoulder movements and denied other symptoms. There was no medical or surgical history, and he had a negative personal or familial oncologic history. The patient was a construction worker and used tobacco daily. On physical examination, there was a 24 × 15-cm fungating, necrotic, and ulcerated left shoulder mass (Figure 1A). A punch biopsy showed sheets of highly pleomorphic atypical spindle and epithelial cells within the superficial and deep dermis, extending into the subcutaneous tissue, and with 18 mitoses/mm2. Immunohistochemistry demonstrated SOX10-positive and pankeratin-negative tumor cells. A whole-body positron emission tomographic and computed tomographic scan revealed an intensely hypermetabolic left shoulder mass and multiple hypermetabolic enlarged left axillary lymph nodes, with no distant metastases (Figure 1B). Brain magnetic resonance imaging was negative for intracranial metastasis.A, Left shoulder giant fungating mass in a 46-year-old man. B, Positron emission tomographic and computed tomographic (PET-CT) scan shows the hypermetabolic left shoulder mass and no distant metastases. What Is Your Diagnosis?
Squamous cell carcinoma
Basal cell carcinoma
Melanoma
Merkel cell carcinoma
C. Melanoma
C
Melanoma
The biomarker SOX10 is highly sensitive and specific in diagnosing melanoma,1 in conjunction with histologic findings and negative keratin staining, making melanoma the most likely diagnosis. Mutational testing revealed no BRAF or other sequence variations. An outside hospital staged the patient’s melanoma as cT4bN2bM0 (stage IIIC) and recommended a clinical trial of pembrolizumab plus cabozantinib for unresectable melanoma. After 10 cycles of treatment, there was continued local progression but slightly decreased uptake on a positron emission tomographic and computed tomographic scan. The patient then received weekly local immunotherapy with talimogene laherparepvec injections in the later course for 6 weeks with further progression. The patient presented to our center for a second opinion before proceeding with palliative radiotherapy.Although radiotherapy has beneficial outcomes in adjuvant and palliative settings,2,3 our multidisciplinary team recommended resection and reconstruction, followed by adjuvant therapy. The patient underwent left shoulder mass radical resection, including muscles, partial scapulectomy, left complete axillary lymph node dissection, and immediate lymphatic reconstruction (a microsurgical technique to reconnect lymphatics after nodal dissection to lower the risk of lymphedema). The muscles were imbricated over the pared-down scapular spine, and a collagen glycosaminoglycan dermal substitute was placed. Reconstruction was completed with a split-thickness skin graft. Surgical pathologic findings revealed invasive melanoma involving adjacent skeletal muscles, and it was 22.2 cm in its greatest dimension. All 33 resected axillary lymph nodes were normal, with no evidence of previously treated melanoma (ypT4bN0M0). Two years after resection, he had a well-healed skin graft with full range of motion and no evidence of lymphedema, and he returned to work in construction (Figure 2). He completed 1 year of adjuvant pembrolizumab therapy with no adverse events; his most recent systemic imaging showed no evidence of disease.Two years after radical left shoulder mass resection and reconstruction with collagen glycosaminoglycan dermal substitute followed by split-thickness skin graft.Locally advanced fungating melanomas are rare, and the anatomical location and size can pose challenges in management. There are case reports of fungating melanomas, mainly in the upper and lower extremities, and the treatment plans and outcomes varied in these cases.4,5 In the present case, because of the lack of surgical evaluation on initial presentation at the outside hospital, possible curative resection and nodal evaluation were not performed promptly. The patient did not have good clinical response to pembrolizumab plus cabozantinib in the trial; however, the subsequent imaging showed reduced uptake in the mass and axillary nodes. On histologic results, the left axillary lymph nodes were pathologically enlarged, but no pathologic metastases were observed; rather, there were reactive lymph nodes. A possible explanation is that there might never have been metastases in the axillary lymph nodes, and an initial axillary biopsy could have been pursued. Another theory could be that there was pathologic response. The original clinical trial could be considered as neoadjuvant therapy after our resection, and the treatment plan could have been further tailored on the basis of index lymph node response in the light of the PRADO (Personalized Response-Directed Surgery and Adjuvant Therapy After Neoadjuvant Ipilimumab and Nivolumab in High-Risk Stage III Melanoma) trial results.6-8 In addition, there are emerging clinical data on neoadjuvant treatments for advanced melanoma.9,10 The SWOG (Southwest Oncology Group) S1801 study, a phase 2 trial, reported that neoadjuvant pembrolizumab therapy improved event-free survival compared with standard adjuvant pembrolizumab therapy in high-risk resectable melanomas.10In conclusion, this educational case challenge demonstrates a rare presentation of a common malignant tumor. Multidisciplinary discussion is crucial for solidifying the best therapeutic options for patients with advanced melanomas. Surgical evaluation and lymph node pathologic information are crucial to tailor the plan.
Oncology
A 46-year-old man presented with a left shoulder mass. It began with a quarter-size maculopapular lesion and continued to progress during the next 3 years. He reported limited shoulder movements and denied other symptoms. There was no medical or surgical history, and he had a negative personal or familial oncologic history. The patient was a construction worker and used tobacco daily. On physical examination, there was a 24 × 15-cm fungating, necrotic, and ulcerated left shoulder mass (Figure 1A). A punch biopsy showed sheets of highly pleomorphic atypical spindle and epithelial cells within the superficial and deep dermis, extending into the subcutaneous tissue, and with 18 mitoses/mm2. Immunohistochemistry demonstrated SOX10-positive and pankeratin-negative tumor cells. A whole-body positron emission tomographic and computed tomographic scan revealed an intensely hypermetabolic left shoulder mass and multiple hypermetabolic enlarged left axillary lymph nodes, with no distant metastases (Figure 1B). Brain magnetic resonance imaging was negative for intracranial metastasis.A, Left shoulder giant fungating mass in a 46-year-old man. B, Positron emission tomographic and computed tomographic (PET-CT) scan shows the hypermetabolic left shoulder mass and no distant metastases.
what is your diagnosis?
What is your diagnosis?
Squamous cell carcinoma
Melanoma
Basal cell carcinoma
Merkel cell carcinoma
b
1
1
1
1
male
0
0
46
41-50
null
28
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2806274
A 46-year-old man presented with a left shoulder mass. It began with a quarter-size maculopapular lesion and continued to progress during the next 3 years. He reported limited shoulder movements and denied other symptoms. There was no medical or surgical history, and he had a negative personal or familial oncologic history. The patient was a construction worker and used tobacco daily. On physical examination, there was a 24 × 15-cm fungating, necrotic, and ulcerated left shoulder mass (Figure 1A). A punch biopsy showed sheets of highly pleomorphic atypical spindle and epithelial cells within the superficial and deep dermis, extending into the subcutaneous tissue, and with 18 mitoses/mm2. Immunohistochemistry demonstrated SOX10-positive and pankeratin-negative tumor cells. A whole-body positron emission tomographic and computed tomographic scan revealed an intensely hypermetabolic left shoulder mass and multiple hypermetabolic enlarged left axillary lymph nodes, with no distant metastases (Figure 1B). Brain magnetic resonance imaging was negative for intracranial metastasis.A, Left shoulder giant fungating mass in a 46-year-old man. B, Positron emission tomographic and computed tomographic (PET-CT) scan shows the hypermetabolic left shoulder mass and no distant metastases. What Is Your Diagnosis?
Squamous cell carcinoma
Basal cell carcinoma
Melanoma
Merkel cell carcinoma
C. Melanoma
C
Melanoma
The biomarker SOX10 is highly sensitive and specific in diagnosing melanoma,1 in conjunction with histologic findings and negative keratin staining, making melanoma the most likely diagnosis. Mutational testing revealed no BRAF or other sequence variations. An outside hospital staged the patient’s melanoma as cT4bN2bM0 (stage IIIC) and recommended a clinical trial of pembrolizumab plus cabozantinib for unresectable melanoma. After 10 cycles of treatment, there was continued local progression but slightly decreased uptake on a positron emission tomographic and computed tomographic scan. The patient then received weekly local immunotherapy with talimogene laherparepvec injections in the later course for 6 weeks with further progression. The patient presented to our center for a second opinion before proceeding with palliative radiotherapy.Although radiotherapy has beneficial outcomes in adjuvant and palliative settings,2,3 our multidisciplinary team recommended resection and reconstruction, followed by adjuvant therapy. The patient underwent left shoulder mass radical resection, including muscles, partial scapulectomy, left complete axillary lymph node dissection, and immediate lymphatic reconstruction (a microsurgical technique to reconnect lymphatics after nodal dissection to lower the risk of lymphedema). The muscles were imbricated over the pared-down scapular spine, and a collagen glycosaminoglycan dermal substitute was placed. Reconstruction was completed with a split-thickness skin graft. Surgical pathologic findings revealed invasive melanoma involving adjacent skeletal muscles, and it was 22.2 cm in its greatest dimension. All 33 resected axillary lymph nodes were normal, with no evidence of previously treated melanoma (ypT4bN0M0). Two years after resection, he had a well-healed skin graft with full range of motion and no evidence of lymphedema, and he returned to work in construction (Figure 2). He completed 1 year of adjuvant pembrolizumab therapy with no adverse events; his most recent systemic imaging showed no evidence of disease.Two years after radical left shoulder mass resection and reconstruction with collagen glycosaminoglycan dermal substitute followed by split-thickness skin graft.Locally advanced fungating melanomas are rare, and the anatomical location and size can pose challenges in management. There are case reports of fungating melanomas, mainly in the upper and lower extremities, and the treatment plans and outcomes varied in these cases.4,5 In the present case, because of the lack of surgical evaluation on initial presentation at the outside hospital, possible curative resection and nodal evaluation were not performed promptly. The patient did not have good clinical response to pembrolizumab plus cabozantinib in the trial; however, the subsequent imaging showed reduced uptake in the mass and axillary nodes. On histologic results, the left axillary lymph nodes were pathologically enlarged, but no pathologic metastases were observed; rather, there were reactive lymph nodes. A possible explanation is that there might never have been metastases in the axillary lymph nodes, and an initial axillary biopsy could have been pursued. Another theory could be that there was pathologic response. The original clinical trial could be considered as neoadjuvant therapy after our resection, and the treatment plan could have been further tailored on the basis of index lymph node response in the light of the PRADO (Personalized Response-Directed Surgery and Adjuvant Therapy After Neoadjuvant Ipilimumab and Nivolumab in High-Risk Stage III Melanoma) trial results.6-8 In addition, there are emerging clinical data on neoadjuvant treatments for advanced melanoma.9,10 The SWOG (Southwest Oncology Group) S1801 study, a phase 2 trial, reported that neoadjuvant pembrolizumab therapy improved event-free survival compared with standard adjuvant pembrolizumab therapy in high-risk resectable melanomas.10In conclusion, this educational case challenge demonstrates a rare presentation of a common malignant tumor. Multidisciplinary discussion is crucial for solidifying the best therapeutic options for patients with advanced melanomas. Surgical evaluation and lymph node pathologic information are crucial to tailor the plan.
Oncology
A 46-year-old woman presented with a left shoulder mass. It began with a quarter-size maculopapular lesion and continued to progress during the next 3 years. She reported limited shoulder movements and denied other symptoms. There was no medical or surgical history, and she had a negative personal or familial oncologic history. The woman was a construction worker and used tobacco daily. On physical examination, there was a 24 × 15-cm fungating, necrotic, and ulcerated left shoulder mass (Figure 1A). A punch biopsy showed sheets of highly pleomorphic atypical spindle and epithelial cells within the superficial and deep dermis, extending into the subcutaneous tissue, and with 18 mitoses/mm2. Immunohistochemistry demonstrated SOX10-positive and pankeratin-negative tumor cells. A whole-body positron emission tomographic and computed tomographic scan revealed an intensely hypermetabolic left shoulder mass and multiple hypermetabolic enlarged left axillary lymph nodes, with no distant metastases (Figure 1B). Brain magnetic resonance imaging was negative for intracranial metastasis.A, Left shoulder giant fungating mass in a 46-year-old woman. B, Positron emission tomographic and computed tomographic (PET-CT) scan shows the hypermetabolic left shoulder mass and no distant metastases.
what is your diagnosis?
What is your diagnosis?
Squamous cell carcinoma
Melanoma
Basal cell carcinoma
Merkel cell carcinoma
b
1
1
1
1
female
0
0
46
41-50
null
28
augmented_female_frommale
https://jamanetwork.com/journals/jamaoncology/fullarticle/2806274
A 46-year-old man presented with a left shoulder mass. It began with a quarter-size maculopapular lesion and continued to progress during the next 3 years. He reported limited shoulder movements and denied other symptoms. There was no medical or surgical history, and he had a negative personal or familial oncologic history. The patient was a construction worker and used tobacco daily. On physical examination, there was a 24 × 15-cm fungating, necrotic, and ulcerated left shoulder mass (Figure 1A). A punch biopsy showed sheets of highly pleomorphic atypical spindle and epithelial cells within the superficial and deep dermis, extending into the subcutaneous tissue, and with 18 mitoses/mm2. Immunohistochemistry demonstrated SOX10-positive and pankeratin-negative tumor cells. A whole-body positron emission tomographic and computed tomographic scan revealed an intensely hypermetabolic left shoulder mass and multiple hypermetabolic enlarged left axillary lymph nodes, with no distant metastases (Figure 1B). Brain magnetic resonance imaging was negative for intracranial metastasis.A, Left shoulder giant fungating mass in a 46-year-old man. B, Positron emission tomographic and computed tomographic (PET-CT) scan shows the hypermetabolic left shoulder mass and no distant metastases. What Is Your Diagnosis?
Squamous cell carcinoma
Basal cell carcinoma
Melanoma
Merkel cell carcinoma
C. Melanoma
C
Melanoma
The biomarker SOX10 is highly sensitive and specific in diagnosing melanoma,1 in conjunction with histologic findings and negative keratin staining, making melanoma the most likely diagnosis. Mutational testing revealed no BRAF or other sequence variations. An outside hospital staged the patient’s melanoma as cT4bN2bM0 (stage IIIC) and recommended a clinical trial of pembrolizumab plus cabozantinib for unresectable melanoma. After 10 cycles of treatment, there was continued local progression but slightly decreased uptake on a positron emission tomographic and computed tomographic scan. The patient then received weekly local immunotherapy with talimogene laherparepvec injections in the later course for 6 weeks with further progression. The patient presented to our center for a second opinion before proceeding with palliative radiotherapy.Although radiotherapy has beneficial outcomes in adjuvant and palliative settings,2,3 our multidisciplinary team recommended resection and reconstruction, followed by adjuvant therapy. The patient underwent left shoulder mass radical resection, including muscles, partial scapulectomy, left complete axillary lymph node dissection, and immediate lymphatic reconstruction (a microsurgical technique to reconnect lymphatics after nodal dissection to lower the risk of lymphedema). The muscles were imbricated over the pared-down scapular spine, and a collagen glycosaminoglycan dermal substitute was placed. Reconstruction was completed with a split-thickness skin graft. Surgical pathologic findings revealed invasive melanoma involving adjacent skeletal muscles, and it was 22.2 cm in its greatest dimension. All 33 resected axillary lymph nodes were normal, with no evidence of previously treated melanoma (ypT4bN0M0). Two years after resection, he had a well-healed skin graft with full range of motion and no evidence of lymphedema, and he returned to work in construction (Figure 2). He completed 1 year of adjuvant pembrolizumab therapy with no adverse events; his most recent systemic imaging showed no evidence of disease.Two years after radical left shoulder mass resection and reconstruction with collagen glycosaminoglycan dermal substitute followed by split-thickness skin graft.Locally advanced fungating melanomas are rare, and the anatomical location and size can pose challenges in management. There are case reports of fungating melanomas, mainly in the upper and lower extremities, and the treatment plans and outcomes varied in these cases.4,5 In the present case, because of the lack of surgical evaluation on initial presentation at the outside hospital, possible curative resection and nodal evaluation were not performed promptly. The patient did not have good clinical response to pembrolizumab plus cabozantinib in the trial; however, the subsequent imaging showed reduced uptake in the mass and axillary nodes. On histologic results, the left axillary lymph nodes were pathologically enlarged, but no pathologic metastases were observed; rather, there were reactive lymph nodes. A possible explanation is that there might never have been metastases in the axillary lymph nodes, and an initial axillary biopsy could have been pursued. Another theory could be that there was pathologic response. The original clinical trial could be considered as neoadjuvant therapy after our resection, and the treatment plan could have been further tailored on the basis of index lymph node response in the light of the PRADO (Personalized Response-Directed Surgery and Adjuvant Therapy After Neoadjuvant Ipilimumab and Nivolumab in High-Risk Stage III Melanoma) trial results.6-8 In addition, there are emerging clinical data on neoadjuvant treatments for advanced melanoma.9,10 The SWOG (Southwest Oncology Group) S1801 study, a phase 2 trial, reported that neoadjuvant pembrolizumab therapy improved event-free survival compared with standard adjuvant pembrolizumab therapy in high-risk resectable melanomas.10In conclusion, this educational case challenge demonstrates a rare presentation of a common malignant tumor. Multidisciplinary discussion is crucial for solidifying the best therapeutic options for patients with advanced melanomas. Surgical evaluation and lymph node pathologic information are crucial to tailor the plan.
Oncology
A 46-year-old patient presented with a left shoulder mass. It began with a quarter-size maculopapular lesion and continued to progress during the next 3 years. They reported limited shoulder movements and denied other symptoms. There was no medical or surgical history, and they had a negative personal or familial oncologic history. The patient was a construction worker and used tobacco daily. On physical examination, there was a 24 × 15-cm fungating, necrotic, and ulcerated left shoulder mass (Figure 1A). A punch biopsy showed sheets of highly pleomorphic atypical spindle and epithelial cells within the superficial and deep dermis, extending into the subcutaneous tissue, and with 18 mitoses/mm2. Immunohistochemistry demonstrated SOX10-positive and pankeratin-negative tumor cells. A whole-body positron emission tomographic and computed tomographic scan revealed an intensely hypermetabolic left shoulder mass and multiple hypermetabolic enlarged left axillary lymph nodes, with no distant metastases (Figure 1B). Brain magnetic resonance imaging was negative for intracranial metastasis.A, Left shoulder giant fungating mass in a 46-year-old patient. B, Positron emission tomographic and computed tomographic (PET-CT) scan shows the hypermetabolic left shoulder mass and no distant metastases.
what is your diagnosis?
What is your diagnosis?
Squamous cell carcinoma
Melanoma
Basal cell carcinoma
Merkel cell carcinoma
b
1
1
1
1
neutral
0
0
46
41-50
null
28
augmented_neutral_frommale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2806495
A 68-year-old man with a remote history of B-cell lymphoma and active renal cell carcinoma (RCC), receiving cabozantinib therapy, was referred for worsening hazy vision in the right eye after recent outside retinal detachment (RD) repair of the left eye, with pars plana vitrectomy and gas. The patient noted blurry vision started around the time of cabozantinib therapy initiation. Cabozantinib therapy was discontinued for surgery and resumed 3 days later. The referring retina specialist noted vitritis and vasculitis intraoperatively, but the result of vitreous biopsy cytology was negative for malignant cells.At presentation to our institution, the patient was taking oral prednisone, 40 mg/d. His visual acuity was 20/500 OD and hand motions OS, with an intraocular pressure of 18 mm Hg in both eyes. Right eye slitlamp examination results showed pigmented cells in the anterior chamber and lens capsule; the left eye showed prolapsed pigmented vitreous at the pupillary margin. Right eye fundus examination results revealed sheets of pigmented cell and vascular sheathing with optic nerve pallor. The left eye had a poor view, with 90% gas fill. Fluorescein angiography illustrated disc and vessel leakage (predominantly arteriole), capillary dropout, and peripheral nonperfusion in the right eye (Figure 1A) with no view in the left eye. Optical coherence tomography revealed debris at the vitreoretinal interface (Figure 1B).Fluorescein angiography and optical coherence tomography on presentation demonstrate retinal vasculitis and vitritis (right eye results shown). A, Fluorescein angiography shows dense vitritis, predominantly arteriole leakage (arrowheads), and nonperfusion. Inset is a magnified view, with arrowheads pointing to arteriole leakage. B, Optical coherence tomography shows disorganization of the outer retina with debris at the vitreoretinal interface (arrowheads).Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidimeMagnetic resonance imaging of the orbits followed by lumbar puncture What Would You Do Next?
Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidime
Stop prednisone and proceed with diagnostic vitrectomy
Stop cabozantinib and admit for intravenous solumedrol
Magnetic resonance imaging of the orbits followed by lumbar puncture
Renal cell carcinoma bilateral ocular metastasis
B
Stop prednisone and proceed with diagnostic vitrectomy
Given the patient’s history of B-cell lymphoma and active RCC, recurrent lymphoma or metastatic RCC must be ruled out. Thus, prednisone therapy was stopped, and the patient underwent diagnostic vitrectomy (choice B). Renal cell carcinoma is an uncommon tumor from the renal cortex, accounting for approximately 2% of systemic malignant tumors.1 Renal cell carcinoma most commonly metastasizes to the lung, bone, liver, and brain,2 and it rarely metastasizes to orbital or intraocular tissue, with fewer than 80 cases reported in the literature.1Cabozantinib is a tyrosine kinase inhibitor approved for the treatment of metastatic RCC. There have been 3 case reports of cabozantinib causing cutaneous vasculitis but no reported ocular adverse effects.3-5 Tyrosine kinase inhibitor–associated retinal vasculitis would be a diagnosis of exclusion. If a subsequent biopsy result is negative, then it is appropriate to stop cabozantinib therapy and consider intravenous corticosteroids (choice C). The patient’s examination findings and clinical history (no hypopyon, lack of pain, and nonseptic appearance) make infectious endophthalmitis less likely (choice A). If ocular metastasis is diagnosed, obtaining a magnetic resonance image of the orbit followed by a lumbar puncture (choice D) may be a necessary next step to evaluate for brain metastasis.The most common cause of malignant intraocular tumors in adults is metastases from systemic malignant tumors. Choroidal metastases of RCC are typically dome shaped and yellow on examination, although some RCC metastases may appear reddish orange due to the high vascularity. In a review of 68 cases of ocular metastases of RCC, 50% had extraocular involvement and 50% were intraocular.1 The most frequently involved site was the orbit (36.8%), followed by the choroid (29.4%), and RCC metastases were predominantly unilateral, with only 4 bilateral reported cases.1 There is a single report in the literature of unilateral RCC ocular metastasis presenting as vitritis with no retinal or choroidal mass,6 although retinal metastasis presenting as retinal vascular sheathing and retinitis has been reported previously in other solid organ tumors.7The patient’s diagnostic vitrectomy confirmed RCC metastasis in the vitreous. The patient then developed an RD in the right eye and a redetached left eye. He underwent bilateral sequential RD repair and subsequent diagnostic vitrectomy, with aqueous and retinal biopsy of the left eye. The biopsy results demonstrated malignant cells positive for RCC (Figure 2). The patient underwent palliative radiotherapy to the orbits, but due to continued disease progression, he passed away shortly thereafter.Full-thickness retinal biopsy specimen from the left eye, with immunohistochemistry positive for CK7 (pictured with arrowheads) and PAX8 and negative for CK20 and TTF1, pointing to renal cell carcinoma metastasis (arrowheads).To our knowledge, this is the first reported case of RCC ocular metastases with aqueous and retinal biopsies and positive bilateral vitreous biopsy results. The patient initially presented with “retinal vasculitis” and panuveitis after RD repair; however, it is possible that the sclerotic vessels were a result of metastatic cellular intravascular deposits causing occlusive disease and severe nonperfusion. Metastatic RCC is a highly vascular tumor type—due to both the overproduction of vascular endothelial growth factor and other proangiogenic cytokines6 and alterations in the von Hippel–Lindau tumor suppressor gene.8 Renal cell carcinoma is metastatic in 30% of patients,9 and it portends a poor prognosis, with a median survival of approximately 13 months and a 5-year survival of less than 10%.10
Ophthalmology
A 68-year-old man with a remote history of B-cell lymphoma and active renal cell carcinoma (RCC), receiving cabozantinib therapy, was referred for worsening hazy vision in the right eye after recent outside retinal detachment (RD) repair of the left eye, with pars plana vitrectomy and gas. The patient noted blurry vision started around the time of cabozantinib therapy initiation. Cabozantinib therapy was discontinued for surgery and resumed 3 days later. The referring retina specialist noted vitritis and vasculitis intraoperatively, but the result of vitreous biopsy cytology was negative for malignant cells.At presentation to our institution, the patient was taking oral prednisone, 40 mg/d. His visual acuity was 20/500 OD and hand motions OS, with an intraocular pressure of 18 mm Hg in both eyes. Right eye slitlamp examination results showed pigmented cells in the anterior chamber and lens capsule; the left eye showed prolapsed pigmented vitreous at the pupillary margin. Right eye fundus examination results revealed sheets of pigmented cell and vascular sheathing with optic nerve pallor. The left eye had a poor view, with 90% gas fill. Fluorescein angiography illustrated disc and vessel leakage (predominantly arteriole), capillary dropout, and peripheral nonperfusion in the right eye (Figure 1A) with no view in the left eye. Optical coherence tomography revealed debris at the vitreoretinal interface (Figure 1B).Fluorescein angiography and optical coherence tomography on presentation demonstrate retinal vasculitis and vitritis (right eye results shown). A, Fluorescein angiography shows dense vitritis, predominantly arteriole leakage (arrowheads), and nonperfusion. Inset is a magnified view, with arrowheads pointing to arteriole leakage. B, Optical coherence tomography shows disorganization of the outer retina with debris at the vitreoretinal interface (arrowheads).Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidimeMagnetic resonance imaging of the orbits followed by lumbar puncture
what would you do next?
What would you do next?
Magnetic resonance imaging of the orbits followed by lumbar puncture
Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidime
Stop cabozantinib and admit for intravenous solumedrol
Stop prednisone and proceed with diagnostic vitrectomy
d
1
1
1
1
male
0
0
68
61-70
null
29
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2806495
A 68-year-old man with a remote history of B-cell lymphoma and active renal cell carcinoma (RCC), receiving cabozantinib therapy, was referred for worsening hazy vision in the right eye after recent outside retinal detachment (RD) repair of the left eye, with pars plana vitrectomy and gas. The patient noted blurry vision started around the time of cabozantinib therapy initiation. Cabozantinib therapy was discontinued for surgery and resumed 3 days later. The referring retina specialist noted vitritis and vasculitis intraoperatively, but the result of vitreous biopsy cytology was negative for malignant cells.At presentation to our institution, the patient was taking oral prednisone, 40 mg/d. His visual acuity was 20/500 OD and hand motions OS, with an intraocular pressure of 18 mm Hg in both eyes. Right eye slitlamp examination results showed pigmented cells in the anterior chamber and lens capsule; the left eye showed prolapsed pigmented vitreous at the pupillary margin. Right eye fundus examination results revealed sheets of pigmented cell and vascular sheathing with optic nerve pallor. The left eye had a poor view, with 90% gas fill. Fluorescein angiography illustrated disc and vessel leakage (predominantly arteriole), capillary dropout, and peripheral nonperfusion in the right eye (Figure 1A) with no view in the left eye. Optical coherence tomography revealed debris at the vitreoretinal interface (Figure 1B).Fluorescein angiography and optical coherence tomography on presentation demonstrate retinal vasculitis and vitritis (right eye results shown). A, Fluorescein angiography shows dense vitritis, predominantly arteriole leakage (arrowheads), and nonperfusion. Inset is a magnified view, with arrowheads pointing to arteriole leakage. B, Optical coherence tomography shows disorganization of the outer retina with debris at the vitreoretinal interface (arrowheads).Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidimeMagnetic resonance imaging of the orbits followed by lumbar puncture What Would You Do Next?
Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidime
Stop prednisone and proceed with diagnostic vitrectomy
Stop cabozantinib and admit for intravenous solumedrol
Magnetic resonance imaging of the orbits followed by lumbar puncture
Renal cell carcinoma bilateral ocular metastasis
B
Stop prednisone and proceed with diagnostic vitrectomy
Given the patient’s history of B-cell lymphoma and active RCC, recurrent lymphoma or metastatic RCC must be ruled out. Thus, prednisone therapy was stopped, and the patient underwent diagnostic vitrectomy (choice B). Renal cell carcinoma is an uncommon tumor from the renal cortex, accounting for approximately 2% of systemic malignant tumors.1 Renal cell carcinoma most commonly metastasizes to the lung, bone, liver, and brain,2 and it rarely metastasizes to orbital or intraocular tissue, with fewer than 80 cases reported in the literature.1Cabozantinib is a tyrosine kinase inhibitor approved for the treatment of metastatic RCC. There have been 3 case reports of cabozantinib causing cutaneous vasculitis but no reported ocular adverse effects.3-5 Tyrosine kinase inhibitor–associated retinal vasculitis would be a diagnosis of exclusion. If a subsequent biopsy result is negative, then it is appropriate to stop cabozantinib therapy and consider intravenous corticosteroids (choice C). The patient’s examination findings and clinical history (no hypopyon, lack of pain, and nonseptic appearance) make infectious endophthalmitis less likely (choice A). If ocular metastasis is diagnosed, obtaining a magnetic resonance image of the orbit followed by a lumbar puncture (choice D) may be a necessary next step to evaluate for brain metastasis.The most common cause of malignant intraocular tumors in adults is metastases from systemic malignant tumors. Choroidal metastases of RCC are typically dome shaped and yellow on examination, although some RCC metastases may appear reddish orange due to the high vascularity. In a review of 68 cases of ocular metastases of RCC, 50% had extraocular involvement and 50% were intraocular.1 The most frequently involved site was the orbit (36.8%), followed by the choroid (29.4%), and RCC metastases were predominantly unilateral, with only 4 bilateral reported cases.1 There is a single report in the literature of unilateral RCC ocular metastasis presenting as vitritis with no retinal or choroidal mass,6 although retinal metastasis presenting as retinal vascular sheathing and retinitis has been reported previously in other solid organ tumors.7The patient’s diagnostic vitrectomy confirmed RCC metastasis in the vitreous. The patient then developed an RD in the right eye and a redetached left eye. He underwent bilateral sequential RD repair and subsequent diagnostic vitrectomy, with aqueous and retinal biopsy of the left eye. The biopsy results demonstrated malignant cells positive for RCC (Figure 2). The patient underwent palliative radiotherapy to the orbits, but due to continued disease progression, he passed away shortly thereafter.Full-thickness retinal biopsy specimen from the left eye, with immunohistochemistry positive for CK7 (pictured with arrowheads) and PAX8 and negative for CK20 and TTF1, pointing to renal cell carcinoma metastasis (arrowheads).To our knowledge, this is the first reported case of RCC ocular metastases with aqueous and retinal biopsies and positive bilateral vitreous biopsy results. The patient initially presented with “retinal vasculitis” and panuveitis after RD repair; however, it is possible that the sclerotic vessels were a result of metastatic cellular intravascular deposits causing occlusive disease and severe nonperfusion. Metastatic RCC is a highly vascular tumor type—due to both the overproduction of vascular endothelial growth factor and other proangiogenic cytokines6 and alterations in the von Hippel–Lindau tumor suppressor gene.8 Renal cell carcinoma is metastatic in 30% of patients,9 and it portends a poor prognosis, with a median survival of approximately 13 months and a 5-year survival of less than 10%.10
Ophthalmology
A 68-year-old woman with a remote history of B-cell lymphoma and active renal cell carcinoma (RCC), receiving cabozantinib therapy, was referred for worsening hazy vision in the right eye after recent outside retinal detachment (RD) repair of the left eye, with pars plana vitrectomy and gas. The woman noted blurry vision started around the time of cabozantinib therapy initiation. Cabozantinib therapy was discontinued for surgery and resumed 3 days later. The referring retina specialist noted vitritis and vasculitis intraoperatively, but the result of vitreous biopsy cytology was negative for malignant cells.At presentation to our institution, the woman was taking oral prednisone, 40 mg/d. Her visual acuity was 20/500 OD and hand motions OS, with an intraocular pressure of 18 mm Hg in both eyes. Right eye slitlamp examination results showed pigmented cells in the anterior chamber and lens capsule; the left eye showed prolapsed pigmented vitreous at the pupillary margin. Right eye fundus examination results revealed sheets of pigmented cell and vascular sheathing with optic nerve pallor. The left eye had a poor view, with 90% gas fill. Fluorescein angiography illustrated disc and vessel leakage (predominantly arteriole), capillary dropout, and peripheral nonperfusion in the right eye (Figure 1A) with no view in the left eye. Optical coherence tomography revealed debris at the vitreoretinal interface (Figure 1B).Fluorescein angiography and optical coherence tomography on presentation demonstrate retinal vasculitis and vitritis (right eye results shown). A, Fluorescein angiography shows dense vitritis, predominantly arteriole leakage (arrowheads), and nonperfusion. Inset is a magnified view, with arrowheads pointing to arteriole leakage. B, Optical coherence tomography shows disorganization of the outer retina with debris at the vitreoretinal interface (arrowheads).Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidimeMagnetic resonance imaging of the orbits followed by lumbar puncture
what would you do next?
What would you do next?
Magnetic resonance imaging of the orbits followed by lumbar puncture
Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidime
Stop cabozantinib and admit for intravenous solumedrol
Stop prednisone and proceed with diagnostic vitrectomy
d
1
1
1
1
female
0
0
68
61-70
null
29
augmented_female_frommale
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2806495
A 68-year-old man with a remote history of B-cell lymphoma and active renal cell carcinoma (RCC), receiving cabozantinib therapy, was referred for worsening hazy vision in the right eye after recent outside retinal detachment (RD) repair of the left eye, with pars plana vitrectomy and gas. The patient noted blurry vision started around the time of cabozantinib therapy initiation. Cabozantinib therapy was discontinued for surgery and resumed 3 days later. The referring retina specialist noted vitritis and vasculitis intraoperatively, but the result of vitreous biopsy cytology was negative for malignant cells.At presentation to our institution, the patient was taking oral prednisone, 40 mg/d. His visual acuity was 20/500 OD and hand motions OS, with an intraocular pressure of 18 mm Hg in both eyes. Right eye slitlamp examination results showed pigmented cells in the anterior chamber and lens capsule; the left eye showed prolapsed pigmented vitreous at the pupillary margin. Right eye fundus examination results revealed sheets of pigmented cell and vascular sheathing with optic nerve pallor. The left eye had a poor view, with 90% gas fill. Fluorescein angiography illustrated disc and vessel leakage (predominantly arteriole), capillary dropout, and peripheral nonperfusion in the right eye (Figure 1A) with no view in the left eye. Optical coherence tomography revealed debris at the vitreoretinal interface (Figure 1B).Fluorescein angiography and optical coherence tomography on presentation demonstrate retinal vasculitis and vitritis (right eye results shown). A, Fluorescein angiography shows dense vitritis, predominantly arteriole leakage (arrowheads), and nonperfusion. Inset is a magnified view, with arrowheads pointing to arteriole leakage. B, Optical coherence tomography shows disorganization of the outer retina with debris at the vitreoretinal interface (arrowheads).Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidimeMagnetic resonance imaging of the orbits followed by lumbar puncture What Would You Do Next?
Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidime
Stop prednisone and proceed with diagnostic vitrectomy
Stop cabozantinib and admit for intravenous solumedrol
Magnetic resonance imaging of the orbits followed by lumbar puncture
Renal cell carcinoma bilateral ocular metastasis
B
Stop prednisone and proceed with diagnostic vitrectomy
Given the patient’s history of B-cell lymphoma and active RCC, recurrent lymphoma or metastatic RCC must be ruled out. Thus, prednisone therapy was stopped, and the patient underwent diagnostic vitrectomy (choice B). Renal cell carcinoma is an uncommon tumor from the renal cortex, accounting for approximately 2% of systemic malignant tumors.1 Renal cell carcinoma most commonly metastasizes to the lung, bone, liver, and brain,2 and it rarely metastasizes to orbital or intraocular tissue, with fewer than 80 cases reported in the literature.1Cabozantinib is a tyrosine kinase inhibitor approved for the treatment of metastatic RCC. There have been 3 case reports of cabozantinib causing cutaneous vasculitis but no reported ocular adverse effects.3-5 Tyrosine kinase inhibitor–associated retinal vasculitis would be a diagnosis of exclusion. If a subsequent biopsy result is negative, then it is appropriate to stop cabozantinib therapy and consider intravenous corticosteroids (choice C). The patient’s examination findings and clinical history (no hypopyon, lack of pain, and nonseptic appearance) make infectious endophthalmitis less likely (choice A). If ocular metastasis is diagnosed, obtaining a magnetic resonance image of the orbit followed by a lumbar puncture (choice D) may be a necessary next step to evaluate for brain metastasis.The most common cause of malignant intraocular tumors in adults is metastases from systemic malignant tumors. Choroidal metastases of RCC are typically dome shaped and yellow on examination, although some RCC metastases may appear reddish orange due to the high vascularity. In a review of 68 cases of ocular metastases of RCC, 50% had extraocular involvement and 50% were intraocular.1 The most frequently involved site was the orbit (36.8%), followed by the choroid (29.4%), and RCC metastases were predominantly unilateral, with only 4 bilateral reported cases.1 There is a single report in the literature of unilateral RCC ocular metastasis presenting as vitritis with no retinal or choroidal mass,6 although retinal metastasis presenting as retinal vascular sheathing and retinitis has been reported previously in other solid organ tumors.7The patient’s diagnostic vitrectomy confirmed RCC metastasis in the vitreous. The patient then developed an RD in the right eye and a redetached left eye. He underwent bilateral sequential RD repair and subsequent diagnostic vitrectomy, with aqueous and retinal biopsy of the left eye. The biopsy results demonstrated malignant cells positive for RCC (Figure 2). The patient underwent palliative radiotherapy to the orbits, but due to continued disease progression, he passed away shortly thereafter.Full-thickness retinal biopsy specimen from the left eye, with immunohistochemistry positive for CK7 (pictured with arrowheads) and PAX8 and negative for CK20 and TTF1, pointing to renal cell carcinoma metastasis (arrowheads).To our knowledge, this is the first reported case of RCC ocular metastases with aqueous and retinal biopsies and positive bilateral vitreous biopsy results. The patient initially presented with “retinal vasculitis” and panuveitis after RD repair; however, it is possible that the sclerotic vessels were a result of metastatic cellular intravascular deposits causing occlusive disease and severe nonperfusion. Metastatic RCC is a highly vascular tumor type—due to both the overproduction of vascular endothelial growth factor and other proangiogenic cytokines6 and alterations in the von Hippel–Lindau tumor suppressor gene.8 Renal cell carcinoma is metastatic in 30% of patients,9 and it portends a poor prognosis, with a median survival of approximately 13 months and a 5-year survival of less than 10%.10
Ophthalmology
A 68-year-old patient with a remote history of B-cell lymphoma and active renal cell carcinoma (RCC), receiving cabozantinib therapy, was referred for worsening hazy vision in the right eye after recent outside retinal detachment (RD) repair of the left eye, with pars plana vitrectomy and gas. The patient noted blurry vision started around the time of cabozantinib therapy initiation. Cabozantinib therapy was discontinued for surgery and resumed 3 days later. The referring retina specialist noted vitritis and vasculitis intraoperatively, but the result of vitreous biopsy cytology was negative for malignant cells.At presentation to our institution, the patient was taking oral prednisone, 40 mg/d. Their visual acuity was 20/500 OD and hand motions OS, with an intraocular pressure of 18 mm Hg in both eyes. Right eye slitlamp examination results showed pigmented cells in the anterior chamber and lens capsule; the left eye showed prolapsed pigmented vitreous at the pupillary margin. Right eye fundus examination results revealed sheets of pigmented cell and vascular sheathing with optic nerve pallor. The left eye had a poor view, with 90% gas fill. Fluorescein angiography illustrated disc and vessel leakage (predominantly arteriole), capillary dropout, and peripheral nonperfusion in the right eye (Figure 1A) with no view in the left eye. Optical coherence tomography revealed debris at the vitreoretinal interface (Figure 1B).Fluorescein angiography and optical coherence tomography on presentation demonstrate retinal vasculitis and vitritis (right eye results shown). A, Fluorescein angiography shows dense vitritis, predominantly arteriole leakage (arrowheads), and nonperfusion. Inset is a magnified view, with arrowheads pointing to arteriole leakage. B, Optical coherence tomography shows disorganization of the outer retina with debris at the vitreoretinal interface (arrowheads).Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidimeMagnetic resonance imaging of the orbits followed by lumbar puncture
what would you do next?
What would you do next?
Magnetic resonance imaging of the orbits followed by lumbar puncture
Vitreous tap and inject intravitreal foscarnet, vancomycin, and ceftazidime
Stop cabozantinib and admit for intravenous solumedrol
Stop prednisone and proceed with diagnostic vitrectomy
d
1
1
1
1
neutral
0
0
68
61-70
null
29
augmented_neutral_frommale
https://jamanetwork.com/journals/jama/fullarticle/2807531
A 67-year-old man with thoracolumbar scoliosis, poor mobility, and history of frequent falls presented to the emergency department with 2 months of left shoulder pain, stiffness and reduced range of motion, and numbness and paresthesias in his left upper extremity. Ten years prior to presentation, he underwent surgical decompression for syringomyelia. A magnetic resonance imaging (MRI) scan of his cervical and thoracic spine performed 2 years prior to presentation revealed a recurrent syrinx extending from C1 to T11, which was not resected because it did not cause symptoms at that time. On physical examination, he had mild tenderness to palpation and reduced range of motion of the left shoulder with abduction and flexion limited to 120° (normal range of motion, 180°). The left scapular muscles were atrophic, and pain and temperature sensation were reduced in his proximal left arm, and dorsal aspect of his left shoulder. His complete blood cell count, serum glucose levels, C-reactive protein levels, and erythrocyte sedimentation rate were normal. Results of tests for rheumatoid factor and antinuclear antibody were negative. Left shoulder radiograph showed complete absence of the left humeral head and a well-demarcated smooth osseous margin of the proximal humerus with associated soft tissue swelling and periarticular calcification (Figure 1). A chest radiograph taken 2 years prior revealed a normal left shoulder joint. The patient was hospitalized for further evaluation and treatment.Anteroposterior radiograph of the patient’s left shoulder demonstrating complete destruction of the proximal humeral head. What Would You Do Next?
Obtain MRI of the left shoulder
Order serum tumor marker testing
Orthopedic surgery evaluation for shoulder replacement surgery
Perform an urgent left shoulder joint aspiration
Neuropathic arthropathy of the shoulder (Charcot shoulder)
A
Obtain MRI of the left shoulder
The key to the correct diagnosis of neuropathic arthropathy of the shoulder is the radiographic finding of an absent left humeral head in a patient with a history of syringomyelia. Choices B and D are incorrect because the well-defined linear margins of the proximal humerus made septic arthritis or osteolysis due to malignancy unlikely. Shoulder replacement surgery (choice C) is not recommended for most patients with Charcot shoulder.Neuropathic arthropathy of the shoulder joint, also known as Charcot shoulder, is a rare progressive disorder characterized by rapid joint destruction and associated soft tissue swelling.1,2 Patients typically present with gradually increasing shoulder swelling, which may or may not be painful, weakness in the shoulder, and decreased range of motion of the shoulder. Patients may report paresthesias or numbness in the affected upper extremity.1,2 Approximately 80% of people with Charcot shoulder have syringomyelia, characterized by a fluid-filled cavity (syrinx) in the spinal cord.2 Other conditions associated with Charcot shoulder include diabetes, syphilis, alcohol use disorder, Arnold-Chiari malformations, cervical spondylosis, amyloidosis, end-stage kidney disease, myelodysplasia, multiple sclerosis, peripheral neuropathy, intra-articular steroid injection, tuberculosis, leprosy (Hansen disease), and gigantism.2-4Syringomyelia may be congenital or secondary to trauma, infection, tumors, vascular abnormalities, or spinal degeneration.4 As it expands, a syrinx damages spinothalamic tract fibers and may compress dorsal column and anterior horn cells, resulting in loss of sensation of pain and temperature, decreased muscle strength, muscle atrophy, and arreflexia.5,6 Approximately 6% of patients with syringomyelia develop Charcot shoulder, and 80% have involvement of only 1 joint.1,6The pathophysiology of neuropathic arthropathy, such as Charcot shoulder, is incompletely understood. A syrinx may decrease shoulder joint sensory innervation, leading to loss of somatic muscle reflexes that protect the joint from injury, resulting in recurrent subclinical trauma that causes inflammation and progressive shoulder joint destruction.2,6 A syrinx may also cause loss of autonomic vascular control, increasing blood flow to the joint, and activating osteoclasts, which results in accelerated bone resorption.1,7Patients with Charcot shoulder may have atrophic changes on radiographs, consisting of substantial bone resorption or shoulder joint destruction, and hypertrophic reactions, with osteophytes, sclerosis, and accumulation of osseous debris.2 MRI of the shoulder can confirm the presence of Charcot shoulder and facilitates assessments for other associated conditions, such as rotator cuff tear.2The diagnosis of Charcot shoulder should be made only after exclusion of other causes of joint destruction, including septic arthritis, osteolytic lesions from malignancy, rheumatoid arthritis, synovial chondromatosis, trauma, soft tissue sarcoma, and Milwaukee shoulder syndrome, a destructive arthropathy caused by deposition of hydroxyapatite crystals.1,3Upon diagnosis of Charcot shoulder, the underlying cause should be identified and treated, in an attempt to slow or halt further joint destruction. Surgical resection of a syrinx in patients with Charcot shoulder due to syringomyelia may slow joint deterioration and improve neurologic symptoms.2 Treatments for Charcot shoulder include nonsteroidal anti-inflammatory medications, joint aspiration if an effusion is present, patient education about avoidance of mechanical trauma, physical therapy, and rehabiliation.1-3 Shoulder surgery is not typically recommended, but may be considered for a subset of patients with Charcot shoulder who do not improve with nonoperative management.2,7The patient was treated with acetaminophen (paracetamol), topical nonsteroidal anti-inflammatory drugs, and short-acting opioids as needed. MRI showed destructive osteolysis of the left humeral head with well-defined, linearly demarcated margins of the proximal humerus with surrounding osseous debris and a moderate glenohumeral joint effusion (Figure 2). After consultation by clinicians in neurosurgery, orthopedic surgery, geriatrics, and radiology, the patient was advised to undergo intensive physical and occupational therapy. After 4 weeks of inpatient physical therapy 5 days per week, his left shoulder pain resolved, and left shoulder flexion and abduction was 160° to 170°. At his most recent outpatient clinic visit, 3 months after hospital discharge, the patient reported no left shoulder pain and had no change in shoulder range of motion. A follow up radiograph revealed a slight decrease in the left glenohumeral effusion.T1-weighted MRI of the left shoulder showing destructive osteolysis of the left proximal humeral head with clear linear margins and associated joint effusion.
General
A 67-year-old man with thoracolumbar scoliosis, poor mobility, and history of frequent falls presented to the emergency department with 2 months of left shoulder pain, stiffness and reduced range of motion, and numbness and paresthesias in his left upper extremity. Ten years prior to presentation, he underwent surgical decompression for syringomyelia. A magnetic resonance imaging (MRI) scan of his cervical and thoracic spine performed 2 years prior to presentation revealed a recurrent syrinx extending from C1 to T11, which was not resected because it did not cause symptoms at that time. On physical examination, he had mild tenderness to palpation and reduced range of motion of the left shoulder with abduction and flexion limited to 120° (normal range of motion, 180°). The left scapular muscles were atrophic, and pain and temperature sensation were reduced in his proximal left arm, and dorsal aspect of his left shoulder. His complete blood cell count, serum glucose levels, C-reactive protein levels, and erythrocyte sedimentation rate were normal. Results of tests for rheumatoid factor and antinuclear antibody were negative. Left shoulder radiograph showed complete absence of the left humeral head and a well-demarcated smooth osseous margin of the proximal humerus with associated soft tissue swelling and periarticular calcification (Figure 1). A chest radiograph taken 2 years prior revealed a normal left shoulder joint. The patient was hospitalized for further evaluation and treatment.Anteroposterior radiograph of the patient’s left shoulder demonstrating complete destruction of the proximal humeral head.
what would you do next?
What would you do next?
Perform an urgent left shoulder joint aspiration
Orthopedic surgery evaluation for shoulder replacement surgery
Order serum tumor marker testing
Obtain MRI of the left shoulder
d
1
1
0
1
male
0
0
67
61-70
null
30
original
https://jamanetwork.com/journals/jama/fullarticle/2807531
A 67-year-old man with thoracolumbar scoliosis, poor mobility, and history of frequent falls presented to the emergency department with 2 months of left shoulder pain, stiffness and reduced range of motion, and numbness and paresthesias in his left upper extremity. Ten years prior to presentation, he underwent surgical decompression for syringomyelia. A magnetic resonance imaging (MRI) scan of his cervical and thoracic spine performed 2 years prior to presentation revealed a recurrent syrinx extending from C1 to T11, which was not resected because it did not cause symptoms at that time. On physical examination, he had mild tenderness to palpation and reduced range of motion of the left shoulder with abduction and flexion limited to 120° (normal range of motion, 180°). The left scapular muscles were atrophic, and pain and temperature sensation were reduced in his proximal left arm, and dorsal aspect of his left shoulder. His complete blood cell count, serum glucose levels, C-reactive protein levels, and erythrocyte sedimentation rate were normal. Results of tests for rheumatoid factor and antinuclear antibody were negative. Left shoulder radiograph showed complete absence of the left humeral head and a well-demarcated smooth osseous margin of the proximal humerus with associated soft tissue swelling and periarticular calcification (Figure 1). A chest radiograph taken 2 years prior revealed a normal left shoulder joint. The patient was hospitalized for further evaluation and treatment.Anteroposterior radiograph of the patient’s left shoulder demonstrating complete destruction of the proximal humeral head. What Would You Do Next?
Obtain MRI of the left shoulder
Order serum tumor marker testing
Orthopedic surgery evaluation for shoulder replacement surgery
Perform an urgent left shoulder joint aspiration
Neuropathic arthropathy of the shoulder (Charcot shoulder)
A
Obtain MRI of the left shoulder
The key to the correct diagnosis of neuropathic arthropathy of the shoulder is the radiographic finding of an absent left humeral head in a patient with a history of syringomyelia. Choices B and D are incorrect because the well-defined linear margins of the proximal humerus made septic arthritis or osteolysis due to malignancy unlikely. Shoulder replacement surgery (choice C) is not recommended for most patients with Charcot shoulder.Neuropathic arthropathy of the shoulder joint, also known as Charcot shoulder, is a rare progressive disorder characterized by rapid joint destruction and associated soft tissue swelling.1,2 Patients typically present with gradually increasing shoulder swelling, which may or may not be painful, weakness in the shoulder, and decreased range of motion of the shoulder. Patients may report paresthesias or numbness in the affected upper extremity.1,2 Approximately 80% of people with Charcot shoulder have syringomyelia, characterized by a fluid-filled cavity (syrinx) in the spinal cord.2 Other conditions associated with Charcot shoulder include diabetes, syphilis, alcohol use disorder, Arnold-Chiari malformations, cervical spondylosis, amyloidosis, end-stage kidney disease, myelodysplasia, multiple sclerosis, peripheral neuropathy, intra-articular steroid injection, tuberculosis, leprosy (Hansen disease), and gigantism.2-4Syringomyelia may be congenital or secondary to trauma, infection, tumors, vascular abnormalities, or spinal degeneration.4 As it expands, a syrinx damages spinothalamic tract fibers and may compress dorsal column and anterior horn cells, resulting in loss of sensation of pain and temperature, decreased muscle strength, muscle atrophy, and arreflexia.5,6 Approximately 6% of patients with syringomyelia develop Charcot shoulder, and 80% have involvement of only 1 joint.1,6The pathophysiology of neuropathic arthropathy, such as Charcot shoulder, is incompletely understood. A syrinx may decrease shoulder joint sensory innervation, leading to loss of somatic muscle reflexes that protect the joint from injury, resulting in recurrent subclinical trauma that causes inflammation and progressive shoulder joint destruction.2,6 A syrinx may also cause loss of autonomic vascular control, increasing blood flow to the joint, and activating osteoclasts, which results in accelerated bone resorption.1,7Patients with Charcot shoulder may have atrophic changes on radiographs, consisting of substantial bone resorption or shoulder joint destruction, and hypertrophic reactions, with osteophytes, sclerosis, and accumulation of osseous debris.2 MRI of the shoulder can confirm the presence of Charcot shoulder and facilitates assessments for other associated conditions, such as rotator cuff tear.2The diagnosis of Charcot shoulder should be made only after exclusion of other causes of joint destruction, including septic arthritis, osteolytic lesions from malignancy, rheumatoid arthritis, synovial chondromatosis, trauma, soft tissue sarcoma, and Milwaukee shoulder syndrome, a destructive arthropathy caused by deposition of hydroxyapatite crystals.1,3Upon diagnosis of Charcot shoulder, the underlying cause should be identified and treated, in an attempt to slow or halt further joint destruction. Surgical resection of a syrinx in patients with Charcot shoulder due to syringomyelia may slow joint deterioration and improve neurologic symptoms.2 Treatments for Charcot shoulder include nonsteroidal anti-inflammatory medications, joint aspiration if an effusion is present, patient education about avoidance of mechanical trauma, physical therapy, and rehabiliation.1-3 Shoulder surgery is not typically recommended, but may be considered for a subset of patients with Charcot shoulder who do not improve with nonoperative management.2,7The patient was treated with acetaminophen (paracetamol), topical nonsteroidal anti-inflammatory drugs, and short-acting opioids as needed. MRI showed destructive osteolysis of the left humeral head with well-defined, linearly demarcated margins of the proximal humerus with surrounding osseous debris and a moderate glenohumeral joint effusion (Figure 2). After consultation by clinicians in neurosurgery, orthopedic surgery, geriatrics, and radiology, the patient was advised to undergo intensive physical and occupational therapy. After 4 weeks of inpatient physical therapy 5 days per week, his left shoulder pain resolved, and left shoulder flexion and abduction was 160° to 170°. At his most recent outpatient clinic visit, 3 months after hospital discharge, the patient reported no left shoulder pain and had no change in shoulder range of motion. A follow up radiograph revealed a slight decrease in the left glenohumeral effusion.T1-weighted MRI of the left shoulder showing destructive osteolysis of the left proximal humeral head with clear linear margins and associated joint effusion.
General
A 67-year-old woman with thoracolumbar scoliosis, poor mobility, and history of frequent falls presented to the emergency department with 2 months of left shoulder pain, stiffness and reduced range of motion, and numbness and paresthesias in her left upper extremity. Ten years prior to presentation, she underwent surgical decompression for syringomyelia. A magnetic resonance imaging (MRI) scan of her cervical and thoracic spine performed 2 years prior to presentation revealed a recurrent syrinx extending from C1 to T11, which was not resected because it did not cause symptoms at that time. On physical examination, she had mild tenderness to palpation and reduced range of motion of the left shoulder with abduction and flexion limited to 120° (normal range of motion, 180°). The left scapular muscles were atrophic, and pain and temperature sensation were reduced in her proximal left arm, and dorsal aspect of her left shoulder. Her complete blood cell count, serum glucose levels, C-reactive protein levels, and erythrocyte sedimentation rate were normal. Results of tests for rheumatoid factor and antinuclear antibody were negative. Left shoulder radiograph showed complete absence of the left humeral head and a well-demarcated smooth osseous margin of the proximal humerus with associated soft tissue swelling and periarticular calcification (Figure 1). A chest radiograph taken 2 years prior revealed a normal left shoulder joint. The woman was hospitalized for further evaluation and treatment.Anteroposterior radiograph of the woman’s left shoulder demonstrating complete destruction of the proximal humeral head.
what would you do next?
What would you do next?
Perform an urgent left shoulder joint aspiration
Orthopedic surgery evaluation for shoulder replacement surgery
Order serum tumor marker testing
Obtain MRI of the left shoulder
d
1
1
0
1
female
0
0
67
61-70
null
30
augmented_female_frommale
https://jamanetwork.com/journals/jama/fullarticle/2807531
A 67-year-old man with thoracolumbar scoliosis, poor mobility, and history of frequent falls presented to the emergency department with 2 months of left shoulder pain, stiffness and reduced range of motion, and numbness and paresthesias in his left upper extremity. Ten years prior to presentation, he underwent surgical decompression for syringomyelia. A magnetic resonance imaging (MRI) scan of his cervical and thoracic spine performed 2 years prior to presentation revealed a recurrent syrinx extending from C1 to T11, which was not resected because it did not cause symptoms at that time. On physical examination, he had mild tenderness to palpation and reduced range of motion of the left shoulder with abduction and flexion limited to 120° (normal range of motion, 180°). The left scapular muscles were atrophic, and pain and temperature sensation were reduced in his proximal left arm, and dorsal aspect of his left shoulder. His complete blood cell count, serum glucose levels, C-reactive protein levels, and erythrocyte sedimentation rate were normal. Results of tests for rheumatoid factor and antinuclear antibody were negative. Left shoulder radiograph showed complete absence of the left humeral head and a well-demarcated smooth osseous margin of the proximal humerus with associated soft tissue swelling and periarticular calcification (Figure 1). A chest radiograph taken 2 years prior revealed a normal left shoulder joint. The patient was hospitalized for further evaluation and treatment.Anteroposterior radiograph of the patient’s left shoulder demonstrating complete destruction of the proximal humeral head. What Would You Do Next?
Obtain MRI of the left shoulder
Order serum tumor marker testing
Orthopedic surgery evaluation for shoulder replacement surgery
Perform an urgent left shoulder joint aspiration
Neuropathic arthropathy of the shoulder (Charcot shoulder)
A
Obtain MRI of the left shoulder
The key to the correct diagnosis of neuropathic arthropathy of the shoulder is the radiographic finding of an absent left humeral head in a patient with a history of syringomyelia. Choices B and D are incorrect because the well-defined linear margins of the proximal humerus made septic arthritis or osteolysis due to malignancy unlikely. Shoulder replacement surgery (choice C) is not recommended for most patients with Charcot shoulder.Neuropathic arthropathy of the shoulder joint, also known as Charcot shoulder, is a rare progressive disorder characterized by rapid joint destruction and associated soft tissue swelling.1,2 Patients typically present with gradually increasing shoulder swelling, which may or may not be painful, weakness in the shoulder, and decreased range of motion of the shoulder. Patients may report paresthesias or numbness in the affected upper extremity.1,2 Approximately 80% of people with Charcot shoulder have syringomyelia, characterized by a fluid-filled cavity (syrinx) in the spinal cord.2 Other conditions associated with Charcot shoulder include diabetes, syphilis, alcohol use disorder, Arnold-Chiari malformations, cervical spondylosis, amyloidosis, end-stage kidney disease, myelodysplasia, multiple sclerosis, peripheral neuropathy, intra-articular steroid injection, tuberculosis, leprosy (Hansen disease), and gigantism.2-4Syringomyelia may be congenital or secondary to trauma, infection, tumors, vascular abnormalities, or spinal degeneration.4 As it expands, a syrinx damages spinothalamic tract fibers and may compress dorsal column and anterior horn cells, resulting in loss of sensation of pain and temperature, decreased muscle strength, muscle atrophy, and arreflexia.5,6 Approximately 6% of patients with syringomyelia develop Charcot shoulder, and 80% have involvement of only 1 joint.1,6The pathophysiology of neuropathic arthropathy, such as Charcot shoulder, is incompletely understood. A syrinx may decrease shoulder joint sensory innervation, leading to loss of somatic muscle reflexes that protect the joint from injury, resulting in recurrent subclinical trauma that causes inflammation and progressive shoulder joint destruction.2,6 A syrinx may also cause loss of autonomic vascular control, increasing blood flow to the joint, and activating osteoclasts, which results in accelerated bone resorption.1,7Patients with Charcot shoulder may have atrophic changes on radiographs, consisting of substantial bone resorption or shoulder joint destruction, and hypertrophic reactions, with osteophytes, sclerosis, and accumulation of osseous debris.2 MRI of the shoulder can confirm the presence of Charcot shoulder and facilitates assessments for other associated conditions, such as rotator cuff tear.2The diagnosis of Charcot shoulder should be made only after exclusion of other causes of joint destruction, including septic arthritis, osteolytic lesions from malignancy, rheumatoid arthritis, synovial chondromatosis, trauma, soft tissue sarcoma, and Milwaukee shoulder syndrome, a destructive arthropathy caused by deposition of hydroxyapatite crystals.1,3Upon diagnosis of Charcot shoulder, the underlying cause should be identified and treated, in an attempt to slow or halt further joint destruction. Surgical resection of a syrinx in patients with Charcot shoulder due to syringomyelia may slow joint deterioration and improve neurologic symptoms.2 Treatments for Charcot shoulder include nonsteroidal anti-inflammatory medications, joint aspiration if an effusion is present, patient education about avoidance of mechanical trauma, physical therapy, and rehabiliation.1-3 Shoulder surgery is not typically recommended, but may be considered for a subset of patients with Charcot shoulder who do not improve with nonoperative management.2,7The patient was treated with acetaminophen (paracetamol), topical nonsteroidal anti-inflammatory drugs, and short-acting opioids as needed. MRI showed destructive osteolysis of the left humeral head with well-defined, linearly demarcated margins of the proximal humerus with surrounding osseous debris and a moderate glenohumeral joint effusion (Figure 2). After consultation by clinicians in neurosurgery, orthopedic surgery, geriatrics, and radiology, the patient was advised to undergo intensive physical and occupational therapy. After 4 weeks of inpatient physical therapy 5 days per week, his left shoulder pain resolved, and left shoulder flexion and abduction was 160° to 170°. At his most recent outpatient clinic visit, 3 months after hospital discharge, the patient reported no left shoulder pain and had no change in shoulder range of motion. A follow up radiograph revealed a slight decrease in the left glenohumeral effusion.T1-weighted MRI of the left shoulder showing destructive osteolysis of the left proximal humeral head with clear linear margins and associated joint effusion.
General
A 67-year-old patient with thoracolumbar scoliosis, poor mobility, and history of frequent falls presented to the emergency department with 2 months of left shoulder pain, stiffness and reduced range of motion, and numbness and paresthesias in their left upper extremity. Ten years prior to presentation, they underwent surgical decompression for syringomyelia. A magnetic resonance imaging (MRI) scan of their cervical and thoracic spine performed 2 years prior to presentation revealed a recurrent syrinx extending from C1 to T11, which was not resected because it did not cause symptoms at that time. On physical examination, they had mild tenderness to palpation and reduced range of motion of the left shoulder with abduction and flexion limited to 120° (normal range of motion, 180°). The left scapular muscles were atrophic, and pain and temperature sensation were reduced in their proximal left arm, and dorsal aspect of their left shoulder. Their complete blood cell count, serum glucose levels, C-reactive protein levels, and erythrocyte sedimentation rate were normal. Results of tests for rheumatoid factor and antinuclear antibody were negative. Left shoulder radiograph showed complete absence of the left humeral head and a well-demarcated smooth osseous margin of the proximal humerus with associated soft tissue swelling and periarticular calcification (Figure 1). A chest radiograph taken 2 years prior revealed a normal left shoulder joint. The patient was hospitalized for further evaluation and treatment.Anteroposterior radiograph of the patient’s left shoulder demonstrating complete destruction of the proximal humeral head.
what would you do next?
What would you do next?
Perform an urgent left shoulder joint aspiration
Orthopedic surgery evaluation for shoulder replacement surgery
Order serum tumor marker testing
Obtain MRI of the left shoulder
d
1
1
0
1
neutral
0
0
67
61-70
null
30
augmented_neutral_frommale
https://jamanetwork.com/journals/jamacardiology/fullarticle/2806695
A woman in her early 40s, gravida 3, para 3, with a history of migraines and recurrent preeclampsia and who was 11 weeks post partum presented with acute pleuritic chest pain, shortness of breath, and nausea that started several hours after a flight from Florida to New York City. She was noted to have tachycardia (110 beats/min) and hypoxemia (90% receiving room air) with a blood pressure of 106/81 mm Hg. Cardiac, respiratory, and pulse examination results were unremarkable. She denied any clear physical or emotional triggers preceding presentation. Before presentation, the patient reported good health with satisfactory employment-mandated routine physical examinations. She denied a history of smoking, alcohol, or illicit drug use. She was not taking hormonal birth control or hormone therapy. Laboratory values demonstrated a normal complete blood cell count and metabolic panel. Chest radiograph results were unremarkable. A computed tomographic chest scan excluded pulmonary embolism but suggested pulmonary and interstitial edema. Results of an electrocardiogram revealed sinus tachycardia with premature ventricular contractions, subcentimeter ST-segment elevation in leads I and aVL and 1-mm ST-segment depression in leads II, III, aVF, and V5 through V6. Initial high-sensitivity troponin was 110 000 ng/mL (to convert to micrograms per liter, multiply by 1). Results of cardiac point-of-care ultrasonography demonstrated a severely reduced left ventricular ejection fraction of 40%, with hypokinesis of the anterolateral and posterior walls. Emergent cardiac catheterization revealed spontaneous coronary artery dissection (SCAD) of the left main coronary artery (Figure 1 and Video).Angiographic image of left main spontaneous coronary artery dissection (A) and intravascular ultrasonography image of left main spontaneous coronary artery dissection (B). What Would You Do Next?
Observation
Medical management
Percutaneous coronary intervention
Coronary artery bypass grafting
Spontaneous coronary artery dissection of the left main coronary artery
C
Percutaneous coronary intervention
SCAD has been increasingly recognized as an important cause of acute coronary syndromes, especially in young healthy women without traditional cardiovascular risk factors. The prevalence of SCAD is approximately 0.5% of patients who present with acute coronary syndromes.1 Extracoronary fibromuscular dysplasia (FMD), depression, rheumatoid arthritis, anxiety, genetic disorders, peripartum, and migraine disorder are associated with SCAD.2,3 Genetic disorders, peripartum SCAD, and FMD were independent predictors of 3-year adverse cardiac events.4 Although mortality is low, one-fourth of patients with SCAD had recurrent SCAD.There are important educational points from this case. First, in experienced, high-volume centers, percutaneous coronary intervention (PCI) is safe in selected patients with SCAD with high-risk features. Conservative therapy may not be appropriate in patients with SCAD with high-risk features including ongoing ischemia, left main artery dissection, or hemodynamic instability.5 In the 2 largest SCAD registries, PCI was appropriate and safe for patients with SCAD with high-risk features such as cardiogenic shock, active/ongoing ischemia, hemodynamic instability, ventricular arrhythmias, ST-segment elevation myocardial infarction, and high-risk anatomy.4,5 The role of intracoronary imaging is crucial to ensure that the wire is in the true lumen distally and to ensure that the stent covers the entire intramural hematoma. Coronary artery bypass grafting (CABG) should also be considered when PCI is technically challenging or has been attempted and unsuccessful.6,7 Given the clinical scenario and angiographical features in this case, the decision was made to pursue revascularization, and PCI with a drug-eluting stent was successfully performed.Second, we demonstrated that radial approach is safe for patients with SCAD. In a meta-analysis of undergoing PCI, radial access was associated with a significant risk reduction in bleeding, vascular complications, and mortality compared with femoral access.8 There are no data comparing radial vs femoral access in the subgroup of patients with SCAD undergoing PCI, particularly peripartum SCAD. Theoretically, femoral access could potentially cause more complications and bleeding due to its association with kidney FMD and arteriopathies compared with radial access.Third, pregnancy should be considered a high-risk feature in patients with SCAD. It has been hypothesized that hormonal changes and hemodynamic variations might be significant contributors to SCAD in pregnant patients by alterations in the architecture of the arterial wall, but the cause is unknown. Investigators suggested that unsuccessful PCI was common for SCAD in pregnant individuals (35%).5 Three patients subsequently underwent CABG due to unsuccessful PCI, and 1 had CABG because of SCAD progression despite conservative management.5Once SCAD is diagnosed, screening for FMD is needed.4 Post PCI, lifelong low-dose aspirin is recommended. Other antiplatelet agents and duration of dual antiplatelet therapy after PCI is primarily tailored individually based on the location of SCAD, the number and size of stents used, etc. β-Blockers and cardiac rehabilitation are recommended in patients with SCAD; statins and angiotensin-converting enzyme inhibitors/angiotensin-receptor blockers may be considered selectively when other clinical indications exist. Management of SCAD in pregnant patients can be challenging, particularly when the dissection involves the left main coronary artery. PCI performed at an experienced center can offer a good outcome for selected patients. Further study is needed to identify whether recurrent preeclampsia is a risk factor for SCAD in pregnant patients.The patient underwent PCI of the left main successfully without complications (Figure 2). The patient recovered well after PCI and returned safely to her hometown to undergo further evaluation for FMD and connective tissue disorders.Angiographic image of left main coronary artery after percutaneous coronary intervention.
Cardiology
A woman in her early 40s, gravida 3, para 3, with a history of migraines and recurrent preeclampsia and who was 11 weeks post partum presented with acute pleuritic chest pain, shortness of breath, and nausea that started several hours after a flight from Florida to New York City. She was noted to have tachycardia (110 beats/min) and hypoxemia (90% receiving room air) with a blood pressure of 106/81 mm Hg. Cardiac, respiratory, and pulse examination results were unremarkable. She denied any clear physical or emotional triggers preceding presentation. Before presentation, the patient reported good health with satisfactory employment-mandated routine physical examinations. She denied a history of smoking, alcohol, or illicit drug use. She was not taking hormonal birth control or hormone therapy. Laboratory values demonstrated a normal complete blood cell count and metabolic panel. Chest radiograph results were unremarkable. A computed tomographic chest scan excluded pulmonary embolism but suggested pulmonary and interstitial edema. Results of an electrocardiogram revealed sinus tachycardia with premature ventricular contractions, subcentimeter ST-segment elevation in leads I and aVL and 1-mm ST-segment depression in leads II, III, aVF, and V5 through V6. Initial high-sensitivity troponin was 110 000 ng/mL (to convert to micrograms per liter, multiply by 1). Results of cardiac point-of-care ultrasonography demonstrated a severely reduced left ventricular ejection fraction of 40%, with hypokinesis of the anterolateral and posterior walls. Emergent cardiac catheterization revealed spontaneous coronary artery dissection (SCAD) of the left main coronary artery (Figure 1 and Video).Angiographic image of left main spontaneous coronary artery dissection (A) and intravascular ultrasonography image of left main spontaneous coronary artery dissection (B).
what would you do next?
What would you do next?
Percutaneous coronary intervention
Coronary artery bypass grafting
Medical management
Observation
a
1
1
1
1
female
0
1
42
41-50
null
31
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2806695
A woman in her early 40s, gravida 3, para 3, with a history of migraines and recurrent preeclampsia and who was 11 weeks post partum presented with acute pleuritic chest pain, shortness of breath, and nausea that started several hours after a flight from Florida to New York City. She was noted to have tachycardia (110 beats/min) and hypoxemia (90% receiving room air) with a blood pressure of 106/81 mm Hg. Cardiac, respiratory, and pulse examination results were unremarkable. She denied any clear physical or emotional triggers preceding presentation. Before presentation, the patient reported good health with satisfactory employment-mandated routine physical examinations. She denied a history of smoking, alcohol, or illicit drug use. She was not taking hormonal birth control or hormone therapy. Laboratory values demonstrated a normal complete blood cell count and metabolic panel. Chest radiograph results were unremarkable. A computed tomographic chest scan excluded pulmonary embolism but suggested pulmonary and interstitial edema. Results of an electrocardiogram revealed sinus tachycardia with premature ventricular contractions, subcentimeter ST-segment elevation in leads I and aVL and 1-mm ST-segment depression in leads II, III, aVF, and V5 through V6. Initial high-sensitivity troponin was 110 000 ng/mL (to convert to micrograms per liter, multiply by 1). Results of cardiac point-of-care ultrasonography demonstrated a severely reduced left ventricular ejection fraction of 40%, with hypokinesis of the anterolateral and posterior walls. Emergent cardiac catheterization revealed spontaneous coronary artery dissection (SCAD) of the left main coronary artery (Figure 1 and Video).Angiographic image of left main spontaneous coronary artery dissection (A) and intravascular ultrasonography image of left main spontaneous coronary artery dissection (B). What Would You Do Next?
Observation
Medical management
Percutaneous coronary intervention
Coronary artery bypass grafting
Spontaneous coronary artery dissection of the left main coronary artery
C
Percutaneous coronary intervention
SCAD has been increasingly recognized as an important cause of acute coronary syndromes, especially in young healthy women without traditional cardiovascular risk factors. The prevalence of SCAD is approximately 0.5% of patients who present with acute coronary syndromes.1 Extracoronary fibromuscular dysplasia (FMD), depression, rheumatoid arthritis, anxiety, genetic disorders, peripartum, and migraine disorder are associated with SCAD.2,3 Genetic disorders, peripartum SCAD, and FMD were independent predictors of 3-year adverse cardiac events.4 Although mortality is low, one-fourth of patients with SCAD had recurrent SCAD.There are important educational points from this case. First, in experienced, high-volume centers, percutaneous coronary intervention (PCI) is safe in selected patients with SCAD with high-risk features. Conservative therapy may not be appropriate in patients with SCAD with high-risk features including ongoing ischemia, left main artery dissection, or hemodynamic instability.5 In the 2 largest SCAD registries, PCI was appropriate and safe for patients with SCAD with high-risk features such as cardiogenic shock, active/ongoing ischemia, hemodynamic instability, ventricular arrhythmias, ST-segment elevation myocardial infarction, and high-risk anatomy.4,5 The role of intracoronary imaging is crucial to ensure that the wire is in the true lumen distally and to ensure that the stent covers the entire intramural hematoma. Coronary artery bypass grafting (CABG) should also be considered when PCI is technically challenging or has been attempted and unsuccessful.6,7 Given the clinical scenario and angiographical features in this case, the decision was made to pursue revascularization, and PCI with a drug-eluting stent was successfully performed.Second, we demonstrated that radial approach is safe for patients with SCAD. In a meta-analysis of undergoing PCI, radial access was associated with a significant risk reduction in bleeding, vascular complications, and mortality compared with femoral access.8 There are no data comparing radial vs femoral access in the subgroup of patients with SCAD undergoing PCI, particularly peripartum SCAD. Theoretically, femoral access could potentially cause more complications and bleeding due to its association with kidney FMD and arteriopathies compared with radial access.Third, pregnancy should be considered a high-risk feature in patients with SCAD. It has been hypothesized that hormonal changes and hemodynamic variations might be significant contributors to SCAD in pregnant patients by alterations in the architecture of the arterial wall, but the cause is unknown. Investigators suggested that unsuccessful PCI was common for SCAD in pregnant individuals (35%).5 Three patients subsequently underwent CABG due to unsuccessful PCI, and 1 had CABG because of SCAD progression despite conservative management.5Once SCAD is diagnosed, screening for FMD is needed.4 Post PCI, lifelong low-dose aspirin is recommended. Other antiplatelet agents and duration of dual antiplatelet therapy after PCI is primarily tailored individually based on the location of SCAD, the number and size of stents used, etc. β-Blockers and cardiac rehabilitation are recommended in patients with SCAD; statins and angiotensin-converting enzyme inhibitors/angiotensin-receptor blockers may be considered selectively when other clinical indications exist. Management of SCAD in pregnant patients can be challenging, particularly when the dissection involves the left main coronary artery. PCI performed at an experienced center can offer a good outcome for selected patients. Further study is needed to identify whether recurrent preeclampsia is a risk factor for SCAD in pregnant patients.The patient underwent PCI of the left main successfully without complications (Figure 2). The patient recovered well after PCI and returned safely to her hometown to undergo further evaluation for FMD and connective tissue disorders.Angiographic image of left main coronary artery after percutaneous coronary intervention.
Cardiology
A man in him early 40s, gravida 3, para 3, with a history of migraines and recurrent preeclampsia and who was 11 weeks post partum presented with acute pleuritic chest pain, shortness of breath, and nausea that started several hours after a flight from Florida to New York City. He was noted to have tachycardia (110 beats/min) and hypoxemia (90% receiving room air) with a blood pressure of 106/81 mm Hg. Cardiac, respiratory, and pulse examination results were unremarkable. He denied any clear physical or emotional triggers preceding presentation. Before presentation, the man reported good health with satisfactory employment-mandated routine physical examinations. He denied a history of smoking, alcohol, or illicit drug use. He was not taking hormonal birth control or hormone therapy. Laboratory values demonstrated a normal complete blood cell count and metabolic panel. Chest radiograph results were unremarkable. A computed tomographic chest scan excluded pulmonary embolism but suggested pulmonary and interstitial edema. Results of an electrocardiogram revealed sinus tachycardia with premature ventricular contractions, subcentimeter ST-segment elevation in leads I and aVL and 1-mm ST-segment depression in leads II, III, aVF, and V5 through V6. Initial high-sensitivity troponin was 110 000 ng/mL (to convert to micrograms per liter, multiply by 1). Results of cardiac point-of-care ultrasonography demonstrated a severely reduced left ventricular ejection fraction of 40%, with hypokinesis of the anterolateral and posterior walls. Emergent cardiac catheterization revealed spontaneous coronary artery dissection (SCAD) of the left main coronary artery (Figure 1 and Video).Angiographic image of left main spontaneous coronary artery dissection (A) and intravascular ultrasonography image of left main spontaneous coronary artery dissection (B).
what would you do next?
What would you do next?
Percutaneous coronary intervention
Coronary artery bypass grafting
Medical management
Observation
a
1
1
1
1
male
0
1
42
41-50
null
31
augmented_male_fromfemale
https://jamanetwork.com/journals/jamacardiology/fullarticle/2806695
A woman in her early 40s, gravida 3, para 3, with a history of migraines and recurrent preeclampsia and who was 11 weeks post partum presented with acute pleuritic chest pain, shortness of breath, and nausea that started several hours after a flight from Florida to New York City. She was noted to have tachycardia (110 beats/min) and hypoxemia (90% receiving room air) with a blood pressure of 106/81 mm Hg. Cardiac, respiratory, and pulse examination results were unremarkable. She denied any clear physical or emotional triggers preceding presentation. Before presentation, the patient reported good health with satisfactory employment-mandated routine physical examinations. She denied a history of smoking, alcohol, or illicit drug use. She was not taking hormonal birth control or hormone therapy. Laboratory values demonstrated a normal complete blood cell count and metabolic panel. Chest radiograph results were unremarkable. A computed tomographic chest scan excluded pulmonary embolism but suggested pulmonary and interstitial edema. Results of an electrocardiogram revealed sinus tachycardia with premature ventricular contractions, subcentimeter ST-segment elevation in leads I and aVL and 1-mm ST-segment depression in leads II, III, aVF, and V5 through V6. Initial high-sensitivity troponin was 110 000 ng/mL (to convert to micrograms per liter, multiply by 1). Results of cardiac point-of-care ultrasonography demonstrated a severely reduced left ventricular ejection fraction of 40%, with hypokinesis of the anterolateral and posterior walls. Emergent cardiac catheterization revealed spontaneous coronary artery dissection (SCAD) of the left main coronary artery (Figure 1 and Video).Angiographic image of left main spontaneous coronary artery dissection (A) and intravascular ultrasonography image of left main spontaneous coronary artery dissection (B). What Would You Do Next?
Observation
Medical management
Percutaneous coronary intervention
Coronary artery bypass grafting
Spontaneous coronary artery dissection of the left main coronary artery
C
Percutaneous coronary intervention
SCAD has been increasingly recognized as an important cause of acute coronary syndromes, especially in young healthy women without traditional cardiovascular risk factors. The prevalence of SCAD is approximately 0.5% of patients who present with acute coronary syndromes.1 Extracoronary fibromuscular dysplasia (FMD), depression, rheumatoid arthritis, anxiety, genetic disorders, peripartum, and migraine disorder are associated with SCAD.2,3 Genetic disorders, peripartum SCAD, and FMD were independent predictors of 3-year adverse cardiac events.4 Although mortality is low, one-fourth of patients with SCAD had recurrent SCAD.There are important educational points from this case. First, in experienced, high-volume centers, percutaneous coronary intervention (PCI) is safe in selected patients with SCAD with high-risk features. Conservative therapy may not be appropriate in patients with SCAD with high-risk features including ongoing ischemia, left main artery dissection, or hemodynamic instability.5 In the 2 largest SCAD registries, PCI was appropriate and safe for patients with SCAD with high-risk features such as cardiogenic shock, active/ongoing ischemia, hemodynamic instability, ventricular arrhythmias, ST-segment elevation myocardial infarction, and high-risk anatomy.4,5 The role of intracoronary imaging is crucial to ensure that the wire is in the true lumen distally and to ensure that the stent covers the entire intramural hematoma. Coronary artery bypass grafting (CABG) should also be considered when PCI is technically challenging or has been attempted and unsuccessful.6,7 Given the clinical scenario and angiographical features in this case, the decision was made to pursue revascularization, and PCI with a drug-eluting stent was successfully performed.Second, we demonstrated that radial approach is safe for patients with SCAD. In a meta-analysis of undergoing PCI, radial access was associated with a significant risk reduction in bleeding, vascular complications, and mortality compared with femoral access.8 There are no data comparing radial vs femoral access in the subgroup of patients with SCAD undergoing PCI, particularly peripartum SCAD. Theoretically, femoral access could potentially cause more complications and bleeding due to its association with kidney FMD and arteriopathies compared with radial access.Third, pregnancy should be considered a high-risk feature in patients with SCAD. It has been hypothesized that hormonal changes and hemodynamic variations might be significant contributors to SCAD in pregnant patients by alterations in the architecture of the arterial wall, but the cause is unknown. Investigators suggested that unsuccessful PCI was common for SCAD in pregnant individuals (35%).5 Three patients subsequently underwent CABG due to unsuccessful PCI, and 1 had CABG because of SCAD progression despite conservative management.5Once SCAD is diagnosed, screening for FMD is needed.4 Post PCI, lifelong low-dose aspirin is recommended. Other antiplatelet agents and duration of dual antiplatelet therapy after PCI is primarily tailored individually based on the location of SCAD, the number and size of stents used, etc. β-Blockers and cardiac rehabilitation are recommended in patients with SCAD; statins and angiotensin-converting enzyme inhibitors/angiotensin-receptor blockers may be considered selectively when other clinical indications exist. Management of SCAD in pregnant patients can be challenging, particularly when the dissection involves the left main coronary artery. PCI performed at an experienced center can offer a good outcome for selected patients. Further study is needed to identify whether recurrent preeclampsia is a risk factor for SCAD in pregnant patients.The patient underwent PCI of the left main successfully without complications (Figure 2). The patient recovered well after PCI and returned safely to her hometown to undergo further evaluation for FMD and connective tissue disorders.Angiographic image of left main coronary artery after percutaneous coronary intervention.
Cardiology
A patient in them early 40s, gravida 3, para 3, with a history of migraines and recurrent preeclampsia and who was 11 weeks post partum presented with acute pleuritic chest pain, shortness of breath, and nausea that started several hours after a flight from Florida to New York City. They was noted to have tachycardia (110 beats/min) and hypoxemia (90% receiving room air) with a blood pressure of 106/81 mm Hg. Cardiac, respiratory, and pulse examination results were unremarkable. They denied any clear physical or emotional triggers preceding presentation. Before presentation, the patient reported good health with satisfactory employment-mandated routine physical examinations. They denied a history of smoking, alcohol, or illicit drug use. They was not taking hormonal birth control or hormone therapy. Laboratory values demonstrated a normal complete blood cell count and metabolic panel. Chest radiograph results were unremarkable. A computed tomographic chest scan excluded pulmonary embolism but suggested pulmonary and interstitial edema. Results of an electrocardiogram revealed sinus tachycardia with premature ventricular contractions, subcentimeter ST-segment elevation in leads I and aVL and 1-mm ST-segment depression in leads II, III, aVF, and V5 through V6. Initial high-sensitivity troponin was 110 000 ng/mL (to convert to micrograms per liter, multiply by 1). Results of cardiac point-of-care ultrasonography demonstrated a severely reduced left ventricular ejection fraction of 40%, with hypokinesis of the anterolateral and posterior walls. Emergent cardiac catheterization revealed spontaneous coronary artery dissection (SCAD) of the left main coronary artery (Figure 1 and Video).Angiographic image of left main spontaneous coronary artery dissection (A) and intravascular ultrasonography image of left main spontaneous coronary artery dissection (B).
what would you do next?
What would you do next?
Percutaneous coronary intervention
Coronary artery bypass grafting
Medical management
Observation
a
1
1
1
1
neutral
0
1
42
41-50
null
31
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2806311
A man in his 60s presented to our clinic with a 5-year history of diffuse erythematous, edematous annular plaques (Figure 1); low-grade fevers; and mild leukopenia. The result of a skin biopsy performed 4 years prior was interpreted as subacute cutaneous lupus erythematosus (SCLE). Treatments included hydroxychloroquine, 400 mg/d, for 4 years; mycophenolate mofetil, 1000 mg/d, for 6 months; and oral prednisone tapers, starting at 40 mg/d. Of these, only prednisone appeared to elicit a response. Review of symptoms was negative for fatigue, weight loss, cough, shortness of breath, joint pain, ocular symptoms, mucosal ulcerations, history of blood clots, photosensitivity, pleurisy, urine changes, and diarrhea. Medical history included hypertension, for which he took losartan, and seasonal allergies, for which he used fexofenadine and fluticasone. Laboratory evaluation revealed mild neutropenia and lymphopenia with normal hemoglobin and platelet counts. Antinuclear antibody screening results were negative. The results of a comprehensive metabolic panel, C3, C4, serum protein electrophoresis, serum free light chains, methylmalonic acid, homocysteine, ferritin, copper, zinc, haptoglobin, HIV, hepatitis B and C serologic tests, and rapid plasma reagin were negative. Lactate dehydrogenase was mildly elevated. Erythrocyte sedimentation rate was elevated at 41 mm/h. Routine cancer screening was up to date. Therapeutic trials of dapsone and dapsone in combination with colchicine were not effective. What Is Your Diagnosis?
Drug-induced SCLE
VEXAS syndrome
Histiocytoid Sweet syndrome
Halogenoderma
B. VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, and somatic) syndrome
B
VEXAS syndrome
Subsequent skin biopsy results (Figure 2A) revealed subepidermal clefting; a superficial and deep perivascular, interstitial, and periadnexal infiltrate of lymphocytes; and myeloperoxidase- and CD68-positive immature nonblastic myeloid and histiocytic cells.A, Hematoxylin-eosin, original magnification ×200. B, Aspirate smear (Wright-Giemsa, original magnification ×630).Because of the lack of response to therapy, the possibility of myelodysplasia, and diagnostic uncertainty, a bone marrow biopsy specimen was obtained (Figure 2B), which revealed normocellular marrow with cytoplasmic vacuoles in the myeloid and erythroid precursors. Molecular diagnostics revealed a somatic variant in UBA1 (GenBank 7317) p.M41L, diagnostic of VEXAS syndrome.VEXAS syndrome is an acquired, autoinflammatory disorder caused by a somatic variant of the X-linked UBA1 gene.1 The UBA1 gene encodes E1, a master enzyme of cellular ubiquination.2 The variant occurs at p.Met41, within the translation initiation codon, which leads to loss of the normally active cytoplasmic isoform, UBA1b, and development of an enzymatically impaired, novel isoform, UBA1c.2 Impaired cellular ubiquitination activates the unfolded protein response and ultimately upregulates type 1 interferons.3 Three amino acid substitutions have been described: threonine, valine, and leucine.4 The amino acid substitution portends disease phenotype and prognosis.2,5 VEXAS syndrome has predominantly been described in men aged 55 to 65 years, although women with monosomy X have also been reported.4-6 A recent observational study suggests VEXAS syndrome prevalence is 1 in 13 591 individuals.6 Patients typically present with fever, skin lesions (usually neutrophilic dermatosis), and hematologic abnormalities (eg, macrocytic anemia and thrombocytopenia). Additional signs of inflammation (eg, arthritis, pulmonary infiltrates, venous thrombosis, chondritis, periorbital edema, vasculitis, uveitis, episcleritis, and hearing loss) are frequently reported, as are elevated inflammatory markers.1,2,4-7 Biopsy specimens of skin manifestations typically reveal superficial dermal infiltration of immature nonblastic myeloid cells, histiocytes, and lymphocytes.7 Bone marrow biopsy reveald hypercellular marrow with cytoplasmic vacuoles in the myeloid and erythroid precursors. Most patients develop myelodysplastic syndrome (MDS), monoclonal gammopathy of unknown significance, or multiple myeloma. Patients with VEXAS syndrome often meet criteria for other autoimmune diseases, specifically Sweet syndrome, polyarteritis nodosa, relapsing polychondritis, and granulomatosis with polyangiitis.1 Classically, these patients do not respond as expected to standard-of-care therapies and are dependent on systemic corticosteroid therapy. Dapsone, colchicine, hydroxychloroquine, azathioprine, methotrexate, mycophenolate mofetil, tumor necrosis factor inhibitors, anakinra, and Janus kinase inhibitors (with the exception of ruxolitinib in patients with VEXAS syndrome with MDS) have been reported as ineffective therapies.4 Tocilizumab may have steroid-sparing effects.4 Patients who develop high-risk MDS have been treated with azacitidine and stem cell transplant.4 VEXAS syndrome is a highly morbid disease, with mortality of approximately 50% at 5 years.Drug-induced SCLE (DI-SCLE) presents as erythematous scaling and annular plaques on the chest, back, and upper outer arms. Like idiopathic SCLE, patients with DI-SCLE commonly have positive antinuclear antibody anti–Sjögren syndrome–related antigens A and B. Although DI-SCLE may be clinically indistinguishable from idiopathic SCLE, concomitant bullae, vasculitis, and erythema-multiforme–like lesions may be seen.8 More than 40 medications have been implicated in the development of DI-SCLE.8 Typically, the causative medication is started weeks to months before the development of DI-SCLE. Biopsy results reveal superficial perivascular infiltrate of lymphocytes, interface dermatitis, and increased mucin.Sweet syndrome classically presents with fever; abrupt onset of edematous, pseudovesiculated papules and nodules; and neutrophilia. Histiocytoid Sweet syndrome is a histopathological variant of Sweet syndrome with a diffuse dermal infiltrate of immature nonblastic myeloid cells and histiocytes that are myeloperoxidase and CD68 positive.9 The present patient met clinical criteria for Sweet syndrome, which is common in patients with VEXAS syndrome with leucine substitution; however, his prolonged course, persistent leukopenia, lack of associated medication or underlying activating condition, and nonresponse to antineutrophilic medications ultimately led to bone marrow biopsy and testing of UBA1.Halogenoderma is a rare dermatosis caused by exposure to high levels of iodine or bromide. Halogenoderma most often presents with papulopustular lesions, but the morphologic features may include urticarial papules, nodules, and ulcerative plaques,10 and systemic symptoms of halogenoderma are rare. Histopathology reveals epidermal neutrophilic abscesses and a neutrophilic dermal infiltrate. Given the histopathologic overlap with neutrophilic dermatoses, halogenoderma is a diagnosis of exclusion.
Dermatology
A man in his 60s presented to our clinic with a 5-year history of diffuse erythematous, edematous annular plaques (Figure 1); low-grade fevers; and mild leukopenia. The result of a skin biopsy performed 4 years prior was interpreted as subacute cutaneous lupus erythematosus (SCLE). Treatments included hydroxychloroquine, 400 mg/d, for 4 years; mycophenolate mofetil, 1000 mg/d, for 6 months; and oral prednisone tapers, starting at 40 mg/d. Of these, only prednisone appeared to elicit a response. Review of symptoms was negative for fatigue, weight loss, cough, shortness of breath, joint pain, ocular symptoms, mucosal ulcerations, history of blood clots, photosensitivity, pleurisy, urine changes, and diarrhea. Medical history included hypertension, for which he took losartan, and seasonal allergies, for which he used fexofenadine and fluticasone. Laboratory evaluation revealed mild neutropenia and lymphopenia with normal hemoglobin and platelet counts. Antinuclear antibody screening results were negative. The results of a comprehensive metabolic panel, C3, C4, serum protein electrophoresis, serum free light chains, methylmalonic acid, homocysteine, ferritin, copper, zinc, haptoglobin, HIV, hepatitis B and C serologic tests, and rapid plasma reagin were negative. Lactate dehydrogenase was mildly elevated. Erythrocyte sedimentation rate was elevated at 41 mm/h. Routine cancer screening was up to date. Therapeutic trials of dapsone and dapsone in combination with colchicine were not effective.
what is your diagnosis?
What is your diagnosis?
VEXAS syndrome
Histiocytoid Sweet syndrome
Drug-induced SCLE
Halogenoderma
a
0
1
1
1
male
0
0
5
0-10
null
32
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2806311
A man in his 60s presented to our clinic with a 5-year history of diffuse erythematous, edematous annular plaques (Figure 1); low-grade fevers; and mild leukopenia. The result of a skin biopsy performed 4 years prior was interpreted as subacute cutaneous lupus erythematosus (SCLE). Treatments included hydroxychloroquine, 400 mg/d, for 4 years; mycophenolate mofetil, 1000 mg/d, for 6 months; and oral prednisone tapers, starting at 40 mg/d. Of these, only prednisone appeared to elicit a response. Review of symptoms was negative for fatigue, weight loss, cough, shortness of breath, joint pain, ocular symptoms, mucosal ulcerations, history of blood clots, photosensitivity, pleurisy, urine changes, and diarrhea. Medical history included hypertension, for which he took losartan, and seasonal allergies, for which he used fexofenadine and fluticasone. Laboratory evaluation revealed mild neutropenia and lymphopenia with normal hemoglobin and platelet counts. Antinuclear antibody screening results were negative. The results of a comprehensive metabolic panel, C3, C4, serum protein electrophoresis, serum free light chains, methylmalonic acid, homocysteine, ferritin, copper, zinc, haptoglobin, HIV, hepatitis B and C serologic tests, and rapid plasma reagin were negative. Lactate dehydrogenase was mildly elevated. Erythrocyte sedimentation rate was elevated at 41 mm/h. Routine cancer screening was up to date. Therapeutic trials of dapsone and dapsone in combination with colchicine were not effective. What Is Your Diagnosis?
Drug-induced SCLE
VEXAS syndrome
Histiocytoid Sweet syndrome
Halogenoderma
B. VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, and somatic) syndrome
B
VEXAS syndrome
Subsequent skin biopsy results (Figure 2A) revealed subepidermal clefting; a superficial and deep perivascular, interstitial, and periadnexal infiltrate of lymphocytes; and myeloperoxidase- and CD68-positive immature nonblastic myeloid and histiocytic cells.A, Hematoxylin-eosin, original magnification ×200. B, Aspirate smear (Wright-Giemsa, original magnification ×630).Because of the lack of response to therapy, the possibility of myelodysplasia, and diagnostic uncertainty, a bone marrow biopsy specimen was obtained (Figure 2B), which revealed normocellular marrow with cytoplasmic vacuoles in the myeloid and erythroid precursors. Molecular diagnostics revealed a somatic variant in UBA1 (GenBank 7317) p.M41L, diagnostic of VEXAS syndrome.VEXAS syndrome is an acquired, autoinflammatory disorder caused by a somatic variant of the X-linked UBA1 gene.1 The UBA1 gene encodes E1, a master enzyme of cellular ubiquination.2 The variant occurs at p.Met41, within the translation initiation codon, which leads to loss of the normally active cytoplasmic isoform, UBA1b, and development of an enzymatically impaired, novel isoform, UBA1c.2 Impaired cellular ubiquitination activates the unfolded protein response and ultimately upregulates type 1 interferons.3 Three amino acid substitutions have been described: threonine, valine, and leucine.4 The amino acid substitution portends disease phenotype and prognosis.2,5 VEXAS syndrome has predominantly been described in men aged 55 to 65 years, although women with monosomy X have also been reported.4-6 A recent observational study suggests VEXAS syndrome prevalence is 1 in 13 591 individuals.6 Patients typically present with fever, skin lesions (usually neutrophilic dermatosis), and hematologic abnormalities (eg, macrocytic anemia and thrombocytopenia). Additional signs of inflammation (eg, arthritis, pulmonary infiltrates, venous thrombosis, chondritis, periorbital edema, vasculitis, uveitis, episcleritis, and hearing loss) are frequently reported, as are elevated inflammatory markers.1,2,4-7 Biopsy specimens of skin manifestations typically reveal superficial dermal infiltration of immature nonblastic myeloid cells, histiocytes, and lymphocytes.7 Bone marrow biopsy reveald hypercellular marrow with cytoplasmic vacuoles in the myeloid and erythroid precursors. Most patients develop myelodysplastic syndrome (MDS), monoclonal gammopathy of unknown significance, or multiple myeloma. Patients with VEXAS syndrome often meet criteria for other autoimmune diseases, specifically Sweet syndrome, polyarteritis nodosa, relapsing polychondritis, and granulomatosis with polyangiitis.1 Classically, these patients do not respond as expected to standard-of-care therapies and are dependent on systemic corticosteroid therapy. Dapsone, colchicine, hydroxychloroquine, azathioprine, methotrexate, mycophenolate mofetil, tumor necrosis factor inhibitors, anakinra, and Janus kinase inhibitors (with the exception of ruxolitinib in patients with VEXAS syndrome with MDS) have been reported as ineffective therapies.4 Tocilizumab may have steroid-sparing effects.4 Patients who develop high-risk MDS have been treated with azacitidine and stem cell transplant.4 VEXAS syndrome is a highly morbid disease, with mortality of approximately 50% at 5 years.Drug-induced SCLE (DI-SCLE) presents as erythematous scaling and annular plaques on the chest, back, and upper outer arms. Like idiopathic SCLE, patients with DI-SCLE commonly have positive antinuclear antibody anti–Sjögren syndrome–related antigens A and B. Although DI-SCLE may be clinically indistinguishable from idiopathic SCLE, concomitant bullae, vasculitis, and erythema-multiforme–like lesions may be seen.8 More than 40 medications have been implicated in the development of DI-SCLE.8 Typically, the causative medication is started weeks to months before the development of DI-SCLE. Biopsy results reveal superficial perivascular infiltrate of lymphocytes, interface dermatitis, and increased mucin.Sweet syndrome classically presents with fever; abrupt onset of edematous, pseudovesiculated papules and nodules; and neutrophilia. Histiocytoid Sweet syndrome is a histopathological variant of Sweet syndrome with a diffuse dermal infiltrate of immature nonblastic myeloid cells and histiocytes that are myeloperoxidase and CD68 positive.9 The present patient met clinical criteria for Sweet syndrome, which is common in patients with VEXAS syndrome with leucine substitution; however, his prolonged course, persistent leukopenia, lack of associated medication or underlying activating condition, and nonresponse to antineutrophilic medications ultimately led to bone marrow biopsy and testing of UBA1.Halogenoderma is a rare dermatosis caused by exposure to high levels of iodine or bromide. Halogenoderma most often presents with papulopustular lesions, but the morphologic features may include urticarial papules, nodules, and ulcerative plaques,10 and systemic symptoms of halogenoderma are rare. Histopathology reveals epidermal neutrophilic abscesses and a neutrophilic dermal infiltrate. Given the histopathologic overlap with neutrophilic dermatoses, halogenoderma is a diagnosis of exclusion.
Dermatology
A woman in her 60s presented to our clinic with a 5-year history of diffuse erythematous, edematous annular plaques (Figure 1); low-grade fevers; and mild leukopenia. The result of a skin biopsy performed 4 years prior was interpreted as subacute cutaneous lupus erythematosus (SCLE). Treatments included hydroxychloroquine, 400 mg/d, for 4 years; mycophenolate mofetil, 1000 mg/d, for 6 months; and oral prednisone tapers, starting at 40 mg/d. Of these, only prednisone appeared to elicit a response. Review of symptoms was negative for fatigue, weight loss, cough, shortness of breath, joint pain, ocular symptoms, mucosal ulcerations, history of blood clots, photosensitivity, pleurisy, urine changes, and diarrhea. Medical history included hypertension, for which she took losartan, and seasonal allergies, for which she used fexofenadine and fluticasone. Laboratory evaluation revealed mild neutropenia and lymphopenia with normal hemoglobin and platelet counts. Antinuclear antibody screening results were negative. The results of a comprehensive metabolic panel, C3, C4, serum protein electrophoresis, serum free light chains, methylmalonic acid, homocysteine, ferritin, copper, zinc, haptoglobin, HIV, hepatitis B and C serologic tests, and rapid plasma reagin were negative. Lactate dehydrogenase was mildly elevated. Erythrocyte sedimentation rate was elevated at 41 mm/h. Routine cancer screening was up to date. Therapeutic trials of dapsone and dapsone in combination with colchicine were not effective.
what is your diagnosis?
What is your diagnosis?
VEXAS syndrome
Histiocytoid Sweet syndrome
Drug-induced SCLE
Halogenoderma
a
0
1
1
1
female
0
0
5
0-10
null
32
augmented_female_frommale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2806311
A man in his 60s presented to our clinic with a 5-year history of diffuse erythematous, edematous annular plaques (Figure 1); low-grade fevers; and mild leukopenia. The result of a skin biopsy performed 4 years prior was interpreted as subacute cutaneous lupus erythematosus (SCLE). Treatments included hydroxychloroquine, 400 mg/d, for 4 years; mycophenolate mofetil, 1000 mg/d, for 6 months; and oral prednisone tapers, starting at 40 mg/d. Of these, only prednisone appeared to elicit a response. Review of symptoms was negative for fatigue, weight loss, cough, shortness of breath, joint pain, ocular symptoms, mucosal ulcerations, history of blood clots, photosensitivity, pleurisy, urine changes, and diarrhea. Medical history included hypertension, for which he took losartan, and seasonal allergies, for which he used fexofenadine and fluticasone. Laboratory evaluation revealed mild neutropenia and lymphopenia with normal hemoglobin and platelet counts. Antinuclear antibody screening results were negative. The results of a comprehensive metabolic panel, C3, C4, serum protein electrophoresis, serum free light chains, methylmalonic acid, homocysteine, ferritin, copper, zinc, haptoglobin, HIV, hepatitis B and C serologic tests, and rapid plasma reagin were negative. Lactate dehydrogenase was mildly elevated. Erythrocyte sedimentation rate was elevated at 41 mm/h. Routine cancer screening was up to date. Therapeutic trials of dapsone and dapsone in combination with colchicine were not effective. What Is Your Diagnosis?
Drug-induced SCLE
VEXAS syndrome
Histiocytoid Sweet syndrome
Halogenoderma
B. VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, and somatic) syndrome
B
VEXAS syndrome
Subsequent skin biopsy results (Figure 2A) revealed subepidermal clefting; a superficial and deep perivascular, interstitial, and periadnexal infiltrate of lymphocytes; and myeloperoxidase- and CD68-positive immature nonblastic myeloid and histiocytic cells.A, Hematoxylin-eosin, original magnification ×200. B, Aspirate smear (Wright-Giemsa, original magnification ×630).Because of the lack of response to therapy, the possibility of myelodysplasia, and diagnostic uncertainty, a bone marrow biopsy specimen was obtained (Figure 2B), which revealed normocellular marrow with cytoplasmic vacuoles in the myeloid and erythroid precursors. Molecular diagnostics revealed a somatic variant in UBA1 (GenBank 7317) p.M41L, diagnostic of VEXAS syndrome.VEXAS syndrome is an acquired, autoinflammatory disorder caused by a somatic variant of the X-linked UBA1 gene.1 The UBA1 gene encodes E1, a master enzyme of cellular ubiquination.2 The variant occurs at p.Met41, within the translation initiation codon, which leads to loss of the normally active cytoplasmic isoform, UBA1b, and development of an enzymatically impaired, novel isoform, UBA1c.2 Impaired cellular ubiquitination activates the unfolded protein response and ultimately upregulates type 1 interferons.3 Three amino acid substitutions have been described: threonine, valine, and leucine.4 The amino acid substitution portends disease phenotype and prognosis.2,5 VEXAS syndrome has predominantly been described in men aged 55 to 65 years, although women with monosomy X have also been reported.4-6 A recent observational study suggests VEXAS syndrome prevalence is 1 in 13 591 individuals.6 Patients typically present with fever, skin lesions (usually neutrophilic dermatosis), and hematologic abnormalities (eg, macrocytic anemia and thrombocytopenia). Additional signs of inflammation (eg, arthritis, pulmonary infiltrates, venous thrombosis, chondritis, periorbital edema, vasculitis, uveitis, episcleritis, and hearing loss) are frequently reported, as are elevated inflammatory markers.1,2,4-7 Biopsy specimens of skin manifestations typically reveal superficial dermal infiltration of immature nonblastic myeloid cells, histiocytes, and lymphocytes.7 Bone marrow biopsy reveald hypercellular marrow with cytoplasmic vacuoles in the myeloid and erythroid precursors. Most patients develop myelodysplastic syndrome (MDS), monoclonal gammopathy of unknown significance, or multiple myeloma. Patients with VEXAS syndrome often meet criteria for other autoimmune diseases, specifically Sweet syndrome, polyarteritis nodosa, relapsing polychondritis, and granulomatosis with polyangiitis.1 Classically, these patients do not respond as expected to standard-of-care therapies and are dependent on systemic corticosteroid therapy. Dapsone, colchicine, hydroxychloroquine, azathioprine, methotrexate, mycophenolate mofetil, tumor necrosis factor inhibitors, anakinra, and Janus kinase inhibitors (with the exception of ruxolitinib in patients with VEXAS syndrome with MDS) have been reported as ineffective therapies.4 Tocilizumab may have steroid-sparing effects.4 Patients who develop high-risk MDS have been treated with azacitidine and stem cell transplant.4 VEXAS syndrome is a highly morbid disease, with mortality of approximately 50% at 5 years.Drug-induced SCLE (DI-SCLE) presents as erythematous scaling and annular plaques on the chest, back, and upper outer arms. Like idiopathic SCLE, patients with DI-SCLE commonly have positive antinuclear antibody anti–Sjögren syndrome–related antigens A and B. Although DI-SCLE may be clinically indistinguishable from idiopathic SCLE, concomitant bullae, vasculitis, and erythema-multiforme–like lesions may be seen.8 More than 40 medications have been implicated in the development of DI-SCLE.8 Typically, the causative medication is started weeks to months before the development of DI-SCLE. Biopsy results reveal superficial perivascular infiltrate of lymphocytes, interface dermatitis, and increased mucin.Sweet syndrome classically presents with fever; abrupt onset of edematous, pseudovesiculated papules and nodules; and neutrophilia. Histiocytoid Sweet syndrome is a histopathological variant of Sweet syndrome with a diffuse dermal infiltrate of immature nonblastic myeloid cells and histiocytes that are myeloperoxidase and CD68 positive.9 The present patient met clinical criteria for Sweet syndrome, which is common in patients with VEXAS syndrome with leucine substitution; however, his prolonged course, persistent leukopenia, lack of associated medication or underlying activating condition, and nonresponse to antineutrophilic medications ultimately led to bone marrow biopsy and testing of UBA1.Halogenoderma is a rare dermatosis caused by exposure to high levels of iodine or bromide. Halogenoderma most often presents with papulopustular lesions, but the morphologic features may include urticarial papules, nodules, and ulcerative plaques,10 and systemic symptoms of halogenoderma are rare. Histopathology reveals epidermal neutrophilic abscesses and a neutrophilic dermal infiltrate. Given the histopathologic overlap with neutrophilic dermatoses, halogenoderma is a diagnosis of exclusion.
Dermatology
A patient in their 60s presented to our clinic with a 5-year history of diffuse erythematous, edematous annular plaques (Figure 1); low-grade fevers; and mild leukopenia. The result of a skin biopsy performed 4 years prior was interpreted as subacute cutaneous lupus erythematosus (SCLE). Treatments included hydroxychloroquine, 400 mg/d, for 4 years; mycophenolate mofetil, 1000 mg/d, for 6 months; and oral prednisone tapers, starting at 40 mg/d. Of these, only prednisone appeared to elicit a response. Review of symptoms was negative for fatigue, weight loss, cough, shortness of breath, joint pain, ocular symptoms, mucosal ulcerations, history of blood clots, photosensitivity, pleurisy, urine changes, and diarrhea. Medical history included hypertension, for which they took losartan, and seasonal allergies, for which they used fexofenadine and fluticasone. Laboratory evaluation revealed mild neutropenia and lymphopenia with normal hemoglobin and platelet counts. Antinuclear antibody screening results were negative. The results of a comprehensive metabolic panel, C3, C4, serum protein electrophoresis, serum free light chains, methylmalonic acid, homocysteine, ferritin, copper, zinc, haptoglobin, HIV, hepatitis B and C serologic tests, and rapid plasma reagin were negative. Lactate dehydrogenase was mildly elevated. Erythrocyte sedimentation rate was elevated at 41 mm/h. Routine cancer screening was up to date. Therapeutic trials of dapsone and dapsone in combination with colchicine were not effective.
what is your diagnosis?
What is your diagnosis?
VEXAS syndrome
Histiocytoid Sweet syndrome
Drug-induced SCLE
Halogenoderma
a
0
1
1
1
neutral
0
0
5
0-10
null
32
augmented_neutral_frommale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2806818
A Chinese woman in her late 20s presented with a 20-year history of progressive skin laxity. In early childhood, she had developed asymptomatic yellowish coalesced papules confined to flexural areas. Subsequently, severe skin sagging occurred on her abdomen with loss of elasticity, then spread extensively with developmental growth. The results of a physical examination showed thick and leathery skinfolds on the neck, axillae, inguinal regions, abdomen, and limbs (Figure, A), with yellowish papules scattered on the dorsal neck. The patient had no extracutaneous involvement and was born to healthy nonconsanguineous parents. However, her younger sister had developed the same skin symptoms. Coagulation-related tests indicated a low clotting activity of factor X (52.2%; reference range, 77%-131%) and a prolonged prothrombin time (12.8 seconds; reference range, 10.4-12.6 seconds). The results of other laboratory investigations were unremarkable. Echocardiographic and fundoscopic examination results were normal. A skin biopsy specimen was obtained from her neck for histopathologic examination (Figure, B).A, Clinical image of the neck, axillae, and abdomen. Severe skin laxity with deep cutaneous folds on the cervical, axillary, and abdominal regions. B, Skin biopsy specimen taken from the neck shows excessive calcification of elastic fiber fragments scattered in the reticular dermis (von Kossa). What Is Your Diagnosis?
Pseudoxanthoma elasticum
Inherited cutis laxa syndrome
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Pseudoxanthoma elasticum/pseudoxanthoma elasticum–like overlap syndrome
C. Pseudoxanthoma elasticum–like disorder with coagulation deficiency
C
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Histopathologic examination, including von Kossa staining, showed fragmented and calcified elastic fibers wildly spread in the reticular dermis. Whole-exome sequencing revealed a novel compound heterozygous sequence variation located in the GGCX gene (OMIM 137167; namely, c.1167_1172delCTGGACinsGGGAA [p. Asn389Lys fs*13] in exon 8 and c.824C>T [p. Ser275Leu] in exon 7). Her sibling shared the same frameshift and nonsense sequence variations. A diagnosis of pseudoxanthoma elasticum–like disorder with coagulation deficiency (PXE-like disorder with CD) was made. Neck lift surgery was performed and resulted in cosmetic improvement.Pseudoxanthoma elasticum–like disorder with CD (OMIM 610842) is a rare subtype of autosomal recessive GGCX-associated diseases.1 As noted by De Vilder et al,1 the first known case was described by G. H. Goldsmith Jr in 1982. The causative GGCX gene is located on the reverse strand of 2p11.2, encoding the γ-glutamyl carboxylase. This specific enzyme promotes γ-carboxylation, an essential process for vitamin K–dependent protein activation. Previous studies reported missense or nonsense variants of the GGCX gene, including c.247C>T, c.763G>A, c.896T>C, c.1186G>A, c.1120C>T, c.1211A>C, c.1426C>T, c.1427G>A, c.1478G>C, c.1506G>C, c.1538G>A, c.1609G>T, and c.1610G>C; and a deletion variant (namely, c.200_201delTT).1-6 All these variants result in nonfunctional carboxylase. Because both the mineralization inhibitor in reticular dermis and the clotting factors II, VII, IX, and X are vitamin K dependent, reduced γ-carboxylase activity usually leads to ectopic calcium precipitation and clotting factor deficiency.1-4 Corresponding typical cutaneous manifestations are overlapping skin lesions of PXE and cutis laxa (CL) (namely, loose and redundant skin located at the trunk, limbs, and flexural areas but leaving the face, hands, and feet unaffected). Patients are also characterized by detectable coagulation deficits, especially factor X deficiency.1,5 Common ophthalmological symptoms include angioid streaks and peau d’orange, which seldom decreases visual acuity.1,7 Histopathologic results reveal polymorphous, fragmented, and mineralized elastic fibers in the upper and mid dermis.6 Electron microscopy further indicates that calcification can also occur in the marginal areas of elastic fibers.3Pseudoxanthoma elasticum is a rare inherited multisystem disease caused by sequence variations in the ABCC gene. Dysfunction of multidrug resistance protein 6 can result in accumulation of compounds with calcium affinity.7 The lesions are typically yellowish papules and reticular rashes. Some patients also presented with skin wrinkling simulating CL but rather milder symptoms, which preferentially involved the flexural regions of the large joint and neck.7,8 Histopathologic features include circumscribed nodular foci of calcification with irregular clumped elastic fibers restricted in the mid dermis.7 Ultrastructurally, mineralization occurs only in the core of elastic fibers, without peripheral deposits.8Inherited CL syndromes comprise a rare group of multisystem disorders characterized by loose redundant skinfolds. Underlying gene defects mainly include ELN, FBLN5, LTBP4, PYCR1, and ATP7A, impairing elastic fiber assembly and homeostasis. Patients with autosomal dominant CL have typical facial features (namely, large ears, sagging cheeks, prominent nasolabial folds, and a long philtrum).9 Different subtypes of autosomal recessive CL are usually accompanied by cardiopulmonary complications, skeletal involvement, or neuromotor delay.9 Coagulation disorder has not been reported, except for a single patient who presented with a decreased von Willebrand factor activity to antigen ratio.10 The characteristic pathologic changes of CL syndromes are dermal elastic fiber reduction and fragmentation without abnormal mineralization.9In diagnosing PXE-like disorder with CD, it is important to exclude the other 2 subtypes of GGCX-associated diseases. They share the same excessive redundant skinfolds and histopathologic features. However, rather than clotting deficits, patients diagnosed with PXE/PXE-like overlap syndrome also present with abdominal micromineralizations.8 Pseudoxanthoma elasticum and pigmentary retinopathy are characterized by ophthalmic symptoms, including nondetectable rod responses or reduced amplitude, and prolonged implicit time.1At present, only symptomatic treatments are available for PXE-like disorder with CD. Plastic surgery is often selected to improve appearance and prevent restriction of daily activities or infection caused by redundant skin, such as abdominoplasty and neck lift surgery in the present case.3 For patients with obvious coagulation abnormality or bleeding tendency, vitamin K supplement should be taken. According to previous studies, both dermatological and ophthalmological follow-up visits are advised for the patient and her sibling.1
Dermatology
A Chinese woman in her late 20s presented with a 20-year history of progressive skin laxity. In early childhood, she had developed asymptomatic yellowish coalesced papules confined to flexural areas. Subsequently, severe skin sagging occurred on her abdomen with loss of elasticity, then spread extensively with developmental growth. The results of a physical examination showed thick and leathery skinfolds on the neck, axillae, inguinal regions, abdomen, and limbs (Figure, A), with yellowish papules scattered on the dorsal neck. The patient had no extracutaneous involvement and was born to healthy nonconsanguineous parents. However, her younger sister had developed the same skin symptoms. Coagulation-related tests indicated a low clotting activity of factor X (52.2%; reference range, 77%-131%) and a prolonged prothrombin time (12.8 seconds; reference range, 10.4-12.6 seconds). The results of other laboratory investigations were unremarkable. Echocardiographic and fundoscopic examination results were normal. A skin biopsy specimen was obtained from her neck for histopathologic examination (Figure, B).A, Clinical image of the neck, axillae, and abdomen. Severe skin laxity with deep cutaneous folds on the cervical, axillary, and abdominal regions. B, Skin biopsy specimen taken from the neck shows excessive calcification of elastic fiber fragments scattered in the reticular dermis (von Kossa).
what is your diagnosis?
What is your diagnosis?
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Pseudoxanthoma elasticum
Pseudoxanthoma elasticum/pseudoxanthoma elasticum–like overlap syndrome
Inherited cutis laxa syndrome
a
0
1
1
1
female
0
0
20
11-20
Chinese
33
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2806818
A Chinese woman in her late 20s presented with a 20-year history of progressive skin laxity. In early childhood, she had developed asymptomatic yellowish coalesced papules confined to flexural areas. Subsequently, severe skin sagging occurred on her abdomen with loss of elasticity, then spread extensively with developmental growth. The results of a physical examination showed thick and leathery skinfolds on the neck, axillae, inguinal regions, abdomen, and limbs (Figure, A), with yellowish papules scattered on the dorsal neck. The patient had no extracutaneous involvement and was born to healthy nonconsanguineous parents. However, her younger sister had developed the same skin symptoms. Coagulation-related tests indicated a low clotting activity of factor X (52.2%; reference range, 77%-131%) and a prolonged prothrombin time (12.8 seconds; reference range, 10.4-12.6 seconds). The results of other laboratory investigations were unremarkable. Echocardiographic and fundoscopic examination results were normal. A skin biopsy specimen was obtained from her neck for histopathologic examination (Figure, B).A, Clinical image of the neck, axillae, and abdomen. Severe skin laxity with deep cutaneous folds on the cervical, axillary, and abdominal regions. B, Skin biopsy specimen taken from the neck shows excessive calcification of elastic fiber fragments scattered in the reticular dermis (von Kossa). What Is Your Diagnosis?
Pseudoxanthoma elasticum
Inherited cutis laxa syndrome
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Pseudoxanthoma elasticum/pseudoxanthoma elasticum–like overlap syndrome
C. Pseudoxanthoma elasticum–like disorder with coagulation deficiency
C
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Histopathologic examination, including von Kossa staining, showed fragmented and calcified elastic fibers wildly spread in the reticular dermis. Whole-exome sequencing revealed a novel compound heterozygous sequence variation located in the GGCX gene (OMIM 137167; namely, c.1167_1172delCTGGACinsGGGAA [p. Asn389Lys fs*13] in exon 8 and c.824C>T [p. Ser275Leu] in exon 7). Her sibling shared the same frameshift and nonsense sequence variations. A diagnosis of pseudoxanthoma elasticum–like disorder with coagulation deficiency (PXE-like disorder with CD) was made. Neck lift surgery was performed and resulted in cosmetic improvement.Pseudoxanthoma elasticum–like disorder with CD (OMIM 610842) is a rare subtype of autosomal recessive GGCX-associated diseases.1 As noted by De Vilder et al,1 the first known case was described by G. H. Goldsmith Jr in 1982. The causative GGCX gene is located on the reverse strand of 2p11.2, encoding the γ-glutamyl carboxylase. This specific enzyme promotes γ-carboxylation, an essential process for vitamin K–dependent protein activation. Previous studies reported missense or nonsense variants of the GGCX gene, including c.247C>T, c.763G>A, c.896T>C, c.1186G>A, c.1120C>T, c.1211A>C, c.1426C>T, c.1427G>A, c.1478G>C, c.1506G>C, c.1538G>A, c.1609G>T, and c.1610G>C; and a deletion variant (namely, c.200_201delTT).1-6 All these variants result in nonfunctional carboxylase. Because both the mineralization inhibitor in reticular dermis and the clotting factors II, VII, IX, and X are vitamin K dependent, reduced γ-carboxylase activity usually leads to ectopic calcium precipitation and clotting factor deficiency.1-4 Corresponding typical cutaneous manifestations are overlapping skin lesions of PXE and cutis laxa (CL) (namely, loose and redundant skin located at the trunk, limbs, and flexural areas but leaving the face, hands, and feet unaffected). Patients are also characterized by detectable coagulation deficits, especially factor X deficiency.1,5 Common ophthalmological symptoms include angioid streaks and peau d’orange, which seldom decreases visual acuity.1,7 Histopathologic results reveal polymorphous, fragmented, and mineralized elastic fibers in the upper and mid dermis.6 Electron microscopy further indicates that calcification can also occur in the marginal areas of elastic fibers.3Pseudoxanthoma elasticum is a rare inherited multisystem disease caused by sequence variations in the ABCC gene. Dysfunction of multidrug resistance protein 6 can result in accumulation of compounds with calcium affinity.7 The lesions are typically yellowish papules and reticular rashes. Some patients also presented with skin wrinkling simulating CL but rather milder symptoms, which preferentially involved the flexural regions of the large joint and neck.7,8 Histopathologic features include circumscribed nodular foci of calcification with irregular clumped elastic fibers restricted in the mid dermis.7 Ultrastructurally, mineralization occurs only in the core of elastic fibers, without peripheral deposits.8Inherited CL syndromes comprise a rare group of multisystem disorders characterized by loose redundant skinfolds. Underlying gene defects mainly include ELN, FBLN5, LTBP4, PYCR1, and ATP7A, impairing elastic fiber assembly and homeostasis. Patients with autosomal dominant CL have typical facial features (namely, large ears, sagging cheeks, prominent nasolabial folds, and a long philtrum).9 Different subtypes of autosomal recessive CL are usually accompanied by cardiopulmonary complications, skeletal involvement, or neuromotor delay.9 Coagulation disorder has not been reported, except for a single patient who presented with a decreased von Willebrand factor activity to antigen ratio.10 The characteristic pathologic changes of CL syndromes are dermal elastic fiber reduction and fragmentation without abnormal mineralization.9In diagnosing PXE-like disorder with CD, it is important to exclude the other 2 subtypes of GGCX-associated diseases. They share the same excessive redundant skinfolds and histopathologic features. However, rather than clotting deficits, patients diagnosed with PXE/PXE-like overlap syndrome also present with abdominal micromineralizations.8 Pseudoxanthoma elasticum and pigmentary retinopathy are characterized by ophthalmic symptoms, including nondetectable rod responses or reduced amplitude, and prolonged implicit time.1At present, only symptomatic treatments are available for PXE-like disorder with CD. Plastic surgery is often selected to improve appearance and prevent restriction of daily activities or infection caused by redundant skin, such as abdominoplasty and neck lift surgery in the present case.3 For patients with obvious coagulation abnormality or bleeding tendency, vitamin K supplement should be taken. According to previous studies, both dermatological and ophthalmological follow-up visits are advised for the patient and her sibling.1
Dermatology
A Chinese man in him late 20s presented with a 20-year history of progressive skin laxity. In early childhood, he had developed asymptomatic yellowish coalesced papules confined to flexural areas. Subsequently, severe skin sagging occurred on him abdomen with loss of elasticity, then spread extensively with developmental growth. The results of a physical examination showed thick and leathery skinfolds on the neck, axillae, inguinal regions, abdomen, and limbs (Figure, A), with yellowish papules scattered on the dorsal neck. The man had no extracutaneous involvement and was born to healthy nonconsanguineous parents. However, him younger sister had developed the same skin symptoms. Coagulation-related tests indicated a low clotting activity of factor X (52.2%; reference range, 77%-131%) and a prolonged prothrombin time (12.8 seconds; reference range, 10.4-12.6 seconds). The results of other laboratory investigations were unremarkable. Echocardiographic and fundoscopic examination results were normal. A skin biopsy specimen was obtained from him neck for histopathologic examination (Figure, B).A, Clinical image of the neck, axillae, and abdomen. Severe skin laxity with deep cutaneous folds on the cervical, axillary, and abdominal regions. B, Skin biopsy specimen taken from the neck shows excessive calcification of elastic fiber fragments scattered in the reticular dermis (von Kossa).
what is your diagnosis?
What is your diagnosis?
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Pseudoxanthoma elasticum
Pseudoxanthoma elasticum/pseudoxanthoma elasticum–like overlap syndrome
Inherited cutis laxa syndrome
a
0
1
1
1
male
0
0
20
11-20
Chinese
33
augmented_male_fromfemale
https://jamanetwork.com/journals/jamadermatology/fullarticle/2806818
A Chinese woman in her late 20s presented with a 20-year history of progressive skin laxity. In early childhood, she had developed asymptomatic yellowish coalesced papules confined to flexural areas. Subsequently, severe skin sagging occurred on her abdomen with loss of elasticity, then spread extensively with developmental growth. The results of a physical examination showed thick and leathery skinfolds on the neck, axillae, inguinal regions, abdomen, and limbs (Figure, A), with yellowish papules scattered on the dorsal neck. The patient had no extracutaneous involvement and was born to healthy nonconsanguineous parents. However, her younger sister had developed the same skin symptoms. Coagulation-related tests indicated a low clotting activity of factor X (52.2%; reference range, 77%-131%) and a prolonged prothrombin time (12.8 seconds; reference range, 10.4-12.6 seconds). The results of other laboratory investigations were unremarkable. Echocardiographic and fundoscopic examination results were normal. A skin biopsy specimen was obtained from her neck for histopathologic examination (Figure, B).A, Clinical image of the neck, axillae, and abdomen. Severe skin laxity with deep cutaneous folds on the cervical, axillary, and abdominal regions. B, Skin biopsy specimen taken from the neck shows excessive calcification of elastic fiber fragments scattered in the reticular dermis (von Kossa). What Is Your Diagnosis?
Pseudoxanthoma elasticum
Inherited cutis laxa syndrome
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Pseudoxanthoma elasticum/pseudoxanthoma elasticum–like overlap syndrome
C. Pseudoxanthoma elasticum–like disorder with coagulation deficiency
C
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Histopathologic examination, including von Kossa staining, showed fragmented and calcified elastic fibers wildly spread in the reticular dermis. Whole-exome sequencing revealed a novel compound heterozygous sequence variation located in the GGCX gene (OMIM 137167; namely, c.1167_1172delCTGGACinsGGGAA [p. Asn389Lys fs*13] in exon 8 and c.824C>T [p. Ser275Leu] in exon 7). Her sibling shared the same frameshift and nonsense sequence variations. A diagnosis of pseudoxanthoma elasticum–like disorder with coagulation deficiency (PXE-like disorder with CD) was made. Neck lift surgery was performed and resulted in cosmetic improvement.Pseudoxanthoma elasticum–like disorder with CD (OMIM 610842) is a rare subtype of autosomal recessive GGCX-associated diseases.1 As noted by De Vilder et al,1 the first known case was described by G. H. Goldsmith Jr in 1982. The causative GGCX gene is located on the reverse strand of 2p11.2, encoding the γ-glutamyl carboxylase. This specific enzyme promotes γ-carboxylation, an essential process for vitamin K–dependent protein activation. Previous studies reported missense or nonsense variants of the GGCX gene, including c.247C>T, c.763G>A, c.896T>C, c.1186G>A, c.1120C>T, c.1211A>C, c.1426C>T, c.1427G>A, c.1478G>C, c.1506G>C, c.1538G>A, c.1609G>T, and c.1610G>C; and a deletion variant (namely, c.200_201delTT).1-6 All these variants result in nonfunctional carboxylase. Because both the mineralization inhibitor in reticular dermis and the clotting factors II, VII, IX, and X are vitamin K dependent, reduced γ-carboxylase activity usually leads to ectopic calcium precipitation and clotting factor deficiency.1-4 Corresponding typical cutaneous manifestations are overlapping skin lesions of PXE and cutis laxa (CL) (namely, loose and redundant skin located at the trunk, limbs, and flexural areas but leaving the face, hands, and feet unaffected). Patients are also characterized by detectable coagulation deficits, especially factor X deficiency.1,5 Common ophthalmological symptoms include angioid streaks and peau d’orange, which seldom decreases visual acuity.1,7 Histopathologic results reveal polymorphous, fragmented, and mineralized elastic fibers in the upper and mid dermis.6 Electron microscopy further indicates that calcification can also occur in the marginal areas of elastic fibers.3Pseudoxanthoma elasticum is a rare inherited multisystem disease caused by sequence variations in the ABCC gene. Dysfunction of multidrug resistance protein 6 can result in accumulation of compounds with calcium affinity.7 The lesions are typically yellowish papules and reticular rashes. Some patients also presented with skin wrinkling simulating CL but rather milder symptoms, which preferentially involved the flexural regions of the large joint and neck.7,8 Histopathologic features include circumscribed nodular foci of calcification with irregular clumped elastic fibers restricted in the mid dermis.7 Ultrastructurally, mineralization occurs only in the core of elastic fibers, without peripheral deposits.8Inherited CL syndromes comprise a rare group of multisystem disorders characterized by loose redundant skinfolds. Underlying gene defects mainly include ELN, FBLN5, LTBP4, PYCR1, and ATP7A, impairing elastic fiber assembly and homeostasis. Patients with autosomal dominant CL have typical facial features (namely, large ears, sagging cheeks, prominent nasolabial folds, and a long philtrum).9 Different subtypes of autosomal recessive CL are usually accompanied by cardiopulmonary complications, skeletal involvement, or neuromotor delay.9 Coagulation disorder has not been reported, except for a single patient who presented with a decreased von Willebrand factor activity to antigen ratio.10 The characteristic pathologic changes of CL syndromes are dermal elastic fiber reduction and fragmentation without abnormal mineralization.9In diagnosing PXE-like disorder with CD, it is important to exclude the other 2 subtypes of GGCX-associated diseases. They share the same excessive redundant skinfolds and histopathologic features. However, rather than clotting deficits, patients diagnosed with PXE/PXE-like overlap syndrome also present with abdominal micromineralizations.8 Pseudoxanthoma elasticum and pigmentary retinopathy are characterized by ophthalmic symptoms, including nondetectable rod responses or reduced amplitude, and prolonged implicit time.1At present, only symptomatic treatments are available for PXE-like disorder with CD. Plastic surgery is often selected to improve appearance and prevent restriction of daily activities or infection caused by redundant skin, such as abdominoplasty and neck lift surgery in the present case.3 For patients with obvious coagulation abnormality or bleeding tendency, vitamin K supplement should be taken. According to previous studies, both dermatological and ophthalmological follow-up visits are advised for the patient and her sibling.1
Dermatology
A Chinese patient in them late 20s presented with a 20-year history of progressive skin laxity. In early childhood, they had developed asymptomatic yellowish coalesced papules confined to flexural areas. Subsequently, severe skin sagging occurred on them abdomen with loss of elasticity, then spread extensively with developmental growth. The results of a physical examination showed thick and leathery skinfolds on the neck, axillae, inguinal regions, abdomen, and limbs (Figure, A), with yellowish papules scattered on the dorsal neck. The patient had no extracutaneous involvement and was born to healthy nonconsanguineous parents. However, them younger sister had developed the same skin symptoms. Coagulation-related tests indicated a low clotting activity of factor X (52.2%; reference range, 77%-131%) and a prolonged prothrombin time (12.8 seconds; reference range, 10.4-12.6 seconds). The results of other laboratory investigations were unremarkable. Echocardiographic and fundoscopic examination results were normal. A skin biopsy specimen was obtained from them neck for histopathologic examination (Figure, B).A, Clinical image of the neck, axillae, and abdomen. Severe skin laxity with deep cutaneous folds on the cervical, axillary, and abdominal regions. B, Skin biopsy specimen taken from the neck shows excessive calcification of elastic fiber fragments scattered in the reticular dermis (von Kossa).
what is your diagnosis?
What is your diagnosis?
Pseudoxanthoma elasticum–like disorder with coagulation deficiency
Pseudoxanthoma elasticum
Pseudoxanthoma elasticum/pseudoxanthoma elasticum–like overlap syndrome
Inherited cutis laxa syndrome
a
0
1
1
1
neutral
0
0
20
11-20
Chinese
33
augmented_neutral_fromfemale
https://jamanetwork.com/journals/jama/fullarticle/2807273
A previously healthy 7-year-old boy was admitted to the hospital with 1 week of temperatures up to 40.2 °C (104.4 °F), productive cough, and lethargy, which did not improve after 3 days of oral cefuroxime. He lived in rural China and had close contact with dogs, cattle, and sheep. His blood pressure was 93/60 mm Hg; heart rate, 100/min; respiratory rate, 26/min; and oxygen saturation, 94% on room air. Lung auscultation revealed absent breath sounds on the right hemithorax. His abdomen was soft and nontender, without hepatosplenomegaly. His white blood cell count was 8000/μL, with 25.8% eosinophils (reference range, 0%-9%). C-reactive protein level and results from tuberculin skin test, interferon-γ release assay, nucleic acid amplification testing for tuberculosis, acid-fast bacilli smear microscopy, and HIV antibody testing were normal. A thoracoabdominal computed tomography (CT) scan showed a ruptured 10 × 9 × 5–cm right upper lobe pulmonary cyst compressing the right main bronchus and 3 liver cysts (Figure 1).Left, Chest computed tomography (CT) scan (transverse view) showing a giant cystic mass in the right upper lobe of the lung with rupture and compression of the right main bronchus. Right, Abdominal CT scan (coronal view) showing 3 round water-density cysts in the right anterior lobe, right posterior lobe, and left lateral lobe of the liver. What Would You Do Next?
Check stool for Echinococcus granulosus eggs
Obtain serologic testing for E granulosus antibodies
Order a lung biopsy
Perform a liver biopsy
Cystic echinococcosis
B
Obtain serologic testing for E granulosus antibodies
The key to the correct diagnosis is recognition that cysts in the lung and liver are highly suggestive of cystic echinococcosis. Organ biopsy (choices C and D) is not recommended because of the risk of secondary spread of infection, and cyst content spillage may cause anaphylaxis. Choice A is incorrect because humans with cystic echinococcosis do not excrete E granulosus eggs in stool.Cystic echinococcosis is caused by infection with the larval stage of a tapeworm called E granulosus. More than 1 million people annually are infected with E granulosus worldwide. Endemic regions include western China, Central Asia, South America, Mediterranean countries, and east Africa.1-3 Cystic echinococcosis is rare in the US and typically occurs in immigrants from endemic countries.4Definitive hosts for E granulosus are domestic or wild dogs and wolves (canids), who acquire the infection after eating animal organs containing hydatid cysts. Inside canids, the cysts develop into adult tapeworms, which shed eggs in their stool. Intermediate hosts, such as sheep, goats, pigs, and cattle, become infected after ingesting tapeworm eggs in contaminated soil.3 Humans can become hosts through consumption of contaminated soil, food, or water, or after close contact with animal hosts.1 After reaching the intestine, E granulosus eggs hatch and embryos penetrate the intestinal wall, migrating through blood or lymphatic vessels to organs, where they develop into hydatid cysts that gradually enlarge and produce more cysts.3,5Approximately 70% of adults with cystic echinococcosis have liver cysts, and approximately 10% to 30% have pulmonary cysts.6 In children, pulmonary cysts are the most common manifestation of cystic echinococcosis.7,8 Less frequently, hyatid cysts develop in the brain, spleen, kidney, pancreas, and heart. Complications of cystic echinococcosis result from compression of adjacent organs, cyst rupture, superinfection, and immunoallergic reactions to Echinococcus, including urticaria, angioedema, and, rarely, anaphylaxis.8Liver cysts typically grow slowly over many years in adults, but cysts often increase in size at a faster rate in the lung and in children.4,6,7 Pulmonary cysts typically range from 1 cm to 20 cm, with giant cysts defined as having a diameter of 10 cm or greater.6 Individuals with uncomplicated pulmonary hyatid cysts are typically asymptomatic, while those with complicated (ruptured or infected) cysts may experience cough, dyspnea, chest pain, hemoptysis, and fever.6 Pulmonary cyst rupture can cause cough productive of salty-tasting fluid or membrane fragments, empyema, and hydropneumothorax.7The diagnosis of cystic echinococcosis is made based on history, imaging, serology, and histology. Typical imaging findings are pulmonary cysts on chest radiographs or CT and liver cysts on abdominal ultrasound or CT. Detection of antibodies against Echinococcus can confirm the diagnosis of cystic echinococcosis, although the sensitivity and specificity of serology each vary from 60% to 90%, and the sensitivity of serology is not well defined in children.4,7 Eosinophilia occurs in approximately 25% of patients with confirmed cystic echinococcosis.9Treatment for cystic echinococcosis depends on cyst size and location, presence or absence of complications, and availability of medical expertise and equipment.4 Cure can be achieved by surgical resection of the cyst. Other treatments include PAIR (percutaneous puncture, aspiration, injection, and reaspiration), antihelminth therapy, and watchful waiting for smaller cysts. Limited evidence is available regarding optimal drug therapy. However, albendazole or albendazole-praziquantel may be considered for patients with inoperable cysts in the liver, cysts in multiple organs, or isolated small or medium-sized cysts. Antihelminth therapy as an adjunct to surgery or interventional procedures decreased the risk of echinococcosis dissemination and recurrence.4,8,10 Approximately 6.5% of patients experience a relapse after treatment for cystic echinococcosis.1Enzyme-linked immunosorbent assay antibodies to Echinococcus were detected, confirming the diagnosis of cystic echinococcosis. Albendazole (15 mg/kg/d orally twice daily) was started 1 week before right upper lobectomy and continued for 5 months postoperatively. During surgery, a 12 × 12 × 8–cm pulmonary cyst was removed (Figure 2). Histopathology showed extensive necrosis and calcification, and E granulosus was seen on hematoxylin and eosin stain. Five months after his initial surgery, the patient underwent laparotomy and complete removal of the 3 liver cysts, with no spillage of their contents. He was prescribed albendazole for 3 months postoperatively. When seen in the clinic 2 months after liver surgery, the patient had recovered well and an abdominal ultrasound showed no cyst recurrence.Gross pathology of the resected right upper lobe of the lung, showing a giant cyst with a ruptured gray endocyst structure with its contents enclosed by the pericyst.
General
A previously healthy 7-year-old boy was admitted to the hospital with 1 week of temperatures up to 40.2 °C (104.4 °F), productive cough, and lethargy, which did not improve after 3 days of oral cefuroxime. He lived in rural China and had close contact with dogs, cattle, and sheep. His blood pressure was 93/60 mm Hg; heart rate, 100/min; respiratory rate, 26/min; and oxygen saturation, 94% on room air. Lung auscultation revealed absent breath sounds on the right hemithorax. His abdomen was soft and nontender, without hepatosplenomegaly. His white blood cell count was 8000/μL, with 25.8% eosinophils (reference range, 0%-9%). C-reactive protein level and results from tuberculin skin test, interferon-γ release assay, nucleic acid amplification testing for tuberculosis, acid-fast bacilli smear microscopy, and HIV antibody testing were normal. A thoracoabdominal computed tomography (CT) scan showed a ruptured 10 × 9 × 5–cm right upper lobe pulmonary cyst compressing the right main bronchus and 3 liver cysts (Figure 1).Left, Chest computed tomography (CT) scan (transverse view) showing a giant cystic mass in the right upper lobe of the lung with rupture and compression of the right main bronchus. Right, Abdominal CT scan (coronal view) showing 3 round water-density cysts in the right anterior lobe, right posterior lobe, and left lateral lobe of the liver.
what would you do next?
What would you do next?
Obtain serologic testing for E granulosus antibodies
Order a lung biopsy
Perform a liver biopsy
Check stool for Echinococcus granulosus eggs
a
1
1
1
1
male
0
0
7
0-10
White
34
original