Pub Date : 2026-02-01Epub Date: 2025-10-31DOI: 10.1111/his.70026
Fatime Szalai, Judit Pápay, Katalin Dezső, Levente Kuthi, Anna Sebestyén, András Khoór, Ildikó Krencz
Aims: Lymphangioleiomyomatosis (LAM) is a PEComa that primarily affects premenopausal women and is associated with the cystic destruction of the lung. Although smooth muscle and melanocytic markers can be helpful, histologic diagnosis of LAM remains challenging, especially in small biopsies. Glycoprotein non-metastatic melanoma protein B (GPNMB) expression has recently been identified as a highly specific and sensitive immunohistochemical marker for PEComas. Nevertheless, only a few LAM cases have been studied so far. The aim of this study was to assess the utility of GPNMB immunohistochemistry as an ancillary marker in a larger cohort of LAM samples.
Methods and results: Immunohistochemistry for GPNMB was performed on cases of LAM (N = 15), potential differential diagnostic mimics (N = 30) and normal lung tissue (N = 2). Immunohistochemistry was assessed using the H-score method; an H-score above 100 was considered 'high'. All LAM cases showed strong cytoplasmic expression, with cell membrane accentuation in some areas. In contrast, differential diagnostic mimics were either negative or only weakly positive. Normal lung parenchyma was negative for GPNMB; however, weak-to-moderate expression was seen in alveolar macrophages in both the normal and diseased lung tissues in most cases. Considering only 'high' positivity, GPNMB immunohistochemistry showed 100% sensitivity and specificity for the diagnosis of LAM in our cohort.
Conclusions: Based on its 100% sensitivity and specificity, GPNMB appears to be a highly valuable immunohistochemical marker for the diagnosis of pulmonary LAM. Besides its diagnostic value, the membrane positivity of GPNMB on LAM cells may predict a response to glembatumumab vedotin, an antibody-drug conjugate targeting GPNMB.
{"title":"GPNMB immunohistochemistry is a useful ancillary tool for the diagnosis of pulmonary lymphangioleiomyomatosis.","authors":"Fatime Szalai, Judit Pápay, Katalin Dezső, Levente Kuthi, Anna Sebestyén, András Khoór, Ildikó Krencz","doi":"10.1111/his.70026","DOIUrl":"10.1111/his.70026","url":null,"abstract":"<p><strong>Aims: </strong>Lymphangioleiomyomatosis (LAM) is a PEComa that primarily affects premenopausal women and is associated with the cystic destruction of the lung. Although smooth muscle and melanocytic markers can be helpful, histologic diagnosis of LAM remains challenging, especially in small biopsies. Glycoprotein non-metastatic melanoma protein B (GPNMB) expression has recently been identified as a highly specific and sensitive immunohistochemical marker for PEComas. Nevertheless, only a few LAM cases have been studied so far. The aim of this study was to assess the utility of GPNMB immunohistochemistry as an ancillary marker in a larger cohort of LAM samples.</p><p><strong>Methods and results: </strong>Immunohistochemistry for GPNMB was performed on cases of LAM (N = 15), potential differential diagnostic mimics (N = 30) and normal lung tissue (N = 2). Immunohistochemistry was assessed using the H-score method; an H-score above 100 was considered 'high'. All LAM cases showed strong cytoplasmic expression, with cell membrane accentuation in some areas. In contrast, differential diagnostic mimics were either negative or only weakly positive. Normal lung parenchyma was negative for GPNMB; however, weak-to-moderate expression was seen in alveolar macrophages in both the normal and diseased lung tissues in most cases. Considering only 'high' positivity, GPNMB immunohistochemistry showed 100% sensitivity and specificity for the diagnosis of LAM in our cohort.</p><p><strong>Conclusions: </strong>Based on its 100% sensitivity and specificity, GPNMB appears to be a highly valuable immunohistochemical marker for the diagnosis of pulmonary LAM. Besides its diagnostic value, the membrane positivity of GPNMB on LAM cells may predict a response to glembatumumab vedotin, an antibody-drug conjugate targeting GPNMB.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":"698-709"},"PeriodicalIF":4.1,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12793827/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145421439","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-02-01Epub Date: 2025-10-31DOI: 10.1111/his.70029
Jesús Machuca-Aguado, Mark Catherwood, David Gonzalez, W Glenn McCluggage
<p><strong>Aims: </strong>The 2013 Cancer Genome Atlas (TCGA) study identified four molecular types of endometrial carcinoma (EC) that are prognostic and predictive of therapy response. The p53 abnormal (p53abn) group of tumours is associated with aggressive clinical behaviour, chemoresponsiveness and generally high-grade histology. p53abn tumours may be identified by p53 immunohistochemical staining (a surrogate marker) or molecular testing. In this study, we evaluated the concordance between p53 immunohistochemistry and TP53 molecular testing in a consecutive cohort of ECs from a population-based setting. Our aim was to investigate the rate of concordance and reasons for discordance between the immunohistochemistry and molecular testing and to provide recommendations for pathologists and clinicians dealing with these discordant cases.</p><p><strong>Methods and results: </strong>A total of 386 ECs were included where all biopsy specimens underwent molecular testing using a next-generation sequencing (NGS) panel (including POLE and TP53 genes and MSI testing) and immunohistochemistry for oestrogen receptor (ER), p53 and mismatch repair (MMR) proteins. Concordance between p53 immunohistochemistry and TP53 NGS was initially 88.6% (discordance of 11.4%) following review of the pathology and molecular reports; most of the discordant cases comprised carcinomas with wild-type p53 immunohistochemistry but TP53 mutations identified on NGS. The discordance reduced to 6.5% after review of the p53 stained slides, which revealed subclonal mutation-type staining in some tumours, and to 5% after excluding POLE mutated and mismatch repair deficient carcinomas. However, there remained a small cohort of 19 POLE wild-type/MMR proficient carcinomas (8 low-grade endometrioid, 9 high-grade endometrioid, 2 carcinosarcomas), with wild-type p53 staining but with TP53 mutations on NGS. Altogether, there were 12 POLE wild-type/MMR proficient low-grade endometrioid carcinomas with TP53 mutations on NGS; all were stage I (11 IA, 1 IB).</p><p><strong>Conclusions: </strong>Our study demonstrated a good overall concordance between p53 immunohistochemical staining and TP53 molecular results. The concordance can be increased by reviewing the p53 stained slides in discrepant cases but there remains a small cohort of cases, mostly low-grade endometrioid carcinomas (POLE wild-type/MMR proficient), where TP53 mutations are present on NGS but p53 immunohistochemistry is wild-type. Such cases present a dilemma for the pathologist (which TCGA group should they be placed into) and the clinician (should adjuvant therapy be instigated based on the presence of a TP53 mutation alone with no other adverse features). For now, we advise classifying such cases as p53abn but not to administer adjuvant therapy based on the presence of a TP53 mutation alone without other adverse pathological factors. The significance of TP53 mutations in such cases should be determined by larger studies with long-
{"title":"Correlation between P53 immunohistochemical staining and TP53 molecular testing in endometrial carcinomas: a detailed assessment of discrepant cases with implications for patient management.","authors":"Jesús Machuca-Aguado, Mark Catherwood, David Gonzalez, W Glenn McCluggage","doi":"10.1111/his.70029","DOIUrl":"10.1111/his.70029","url":null,"abstract":"<p><strong>Aims: </strong>The 2013 Cancer Genome Atlas (TCGA) study identified four molecular types of endometrial carcinoma (EC) that are prognostic and predictive of therapy response. The p53 abnormal (p53abn) group of tumours is associated with aggressive clinical behaviour, chemoresponsiveness and generally high-grade histology. p53abn tumours may be identified by p53 immunohistochemical staining (a surrogate marker) or molecular testing. In this study, we evaluated the concordance between p53 immunohistochemistry and TP53 molecular testing in a consecutive cohort of ECs from a population-based setting. Our aim was to investigate the rate of concordance and reasons for discordance between the immunohistochemistry and molecular testing and to provide recommendations for pathologists and clinicians dealing with these discordant cases.</p><p><strong>Methods and results: </strong>A total of 386 ECs were included where all biopsy specimens underwent molecular testing using a next-generation sequencing (NGS) panel (including POLE and TP53 genes and MSI testing) and immunohistochemistry for oestrogen receptor (ER), p53 and mismatch repair (MMR) proteins. Concordance between p53 immunohistochemistry and TP53 NGS was initially 88.6% (discordance of 11.4%) following review of the pathology and molecular reports; most of the discordant cases comprised carcinomas with wild-type p53 immunohistochemistry but TP53 mutations identified on NGS. The discordance reduced to 6.5% after review of the p53 stained slides, which revealed subclonal mutation-type staining in some tumours, and to 5% after excluding POLE mutated and mismatch repair deficient carcinomas. However, there remained a small cohort of 19 POLE wild-type/MMR proficient carcinomas (8 low-grade endometrioid, 9 high-grade endometrioid, 2 carcinosarcomas), with wild-type p53 staining but with TP53 mutations on NGS. Altogether, there were 12 POLE wild-type/MMR proficient low-grade endometrioid carcinomas with TP53 mutations on NGS; all were stage I (11 IA, 1 IB).</p><p><strong>Conclusions: </strong>Our study demonstrated a good overall concordance between p53 immunohistochemical staining and TP53 molecular results. The concordance can be increased by reviewing the p53 stained slides in discrepant cases but there remains a small cohort of cases, mostly low-grade endometrioid carcinomas (POLE wild-type/MMR proficient), where TP53 mutations are present on NGS but p53 immunohistochemistry is wild-type. Such cases present a dilemma for the pathologist (which TCGA group should they be placed into) and the clinician (should adjuvant therapy be instigated based on the presence of a TP53 mutation alone with no other adverse features). For now, we advise classifying such cases as p53abn but not to administer adjuvant therapy based on the presence of a TP53 mutation alone without other adverse pathological factors. The significance of TP53 mutations in such cases should be determined by larger studies with long-","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":"591-604"},"PeriodicalIF":4.1,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145421413","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-02-01Epub Date: 2025-10-09DOI: 10.1111/his.70021
Trevor Teich, Rong Hu, Anita Nagy, Mary Shago, Yen Chen Kevin Ko, Carl Ren, Raja R Seethala, Bibianna Purgina, Elan Hahn
Aims: Immunohistochemistry (IHC) for p16 is widely used as a surrogate marker for HPV involvement in oropharyngeal squamous cell carcinoma (OPSCC), among other tumours. Confirming HPV status in OPSCC is critical, as HPV-positive tumours have better overall survival and may require de-escalated therapy compared to HPV-independent OPSCC in the future. However, discordance exists between p16 and HPV, and direct HPV testing is occasionally required to ensure an accurate diagnosis. The aim of this study is to highlight the genomic basis behind the limitation of relying on p16 IHC as a surrogate marker for HPV involvement.
Methods and results: Through a multi-institutional collaboration, this case series compiled four patients with a 'false' negative p16 staining pattern in HPV-positive non-keratinizing head and neck squamous cell carcinoma. All cases demonstrated minimal to no p16 IHC staining and were positive for HPV by direct RNA in situ hybridization. Through CDKN2A fluorescence in situ hybridization testing, three patients demonstrated a homozygous deletion of CDKN2A and one demonstrated a heterozygous deletion.
Conclusions: This series highlights the genomic basis for the 'false' negative p16 results, raising awareness of a significant diagnostic pitfall while emphasizing the importance of careful consideration of clinicopathologic parameters in the clinical workup of these cases.
{"title":"CDKN2A deletion in p16-negative/HPV-positive head and neck squamous cell carcinoma: Highlighting the molecular basis behind the limitation of relying on p16 immunohistochemistry as a surrogate marker for HPV involvement.","authors":"Trevor Teich, Rong Hu, Anita Nagy, Mary Shago, Yen Chen Kevin Ko, Carl Ren, Raja R Seethala, Bibianna Purgina, Elan Hahn","doi":"10.1111/his.70021","DOIUrl":"10.1111/his.70021","url":null,"abstract":"<p><strong>Aims: </strong>Immunohistochemistry (IHC) for p16 is widely used as a surrogate marker for HPV involvement in oropharyngeal squamous cell carcinoma (OPSCC), among other tumours. Confirming HPV status in OPSCC is critical, as HPV-positive tumours have better overall survival and may require de-escalated therapy compared to HPV-independent OPSCC in the future. However, discordance exists between p16 and HPV, and direct HPV testing is occasionally required to ensure an accurate diagnosis. The aim of this study is to highlight the genomic basis behind the limitation of relying on p16 IHC as a surrogate marker for HPV involvement.</p><p><strong>Methods and results: </strong>Through a multi-institutional collaboration, this case series compiled four patients with a 'false' negative p16 staining pattern in HPV-positive non-keratinizing head and neck squamous cell carcinoma. All cases demonstrated minimal to no p16 IHC staining and were positive for HPV by direct RNA in situ hybridization. Through CDKN2A fluorescence in situ hybridization testing, three patients demonstrated a homozygous deletion of CDKN2A and one demonstrated a heterozygous deletion.</p><p><strong>Conclusions: </strong>This series highlights the genomic basis for the 'false' negative p16 results, raising awareness of a significant diagnostic pitfall while emphasizing the importance of careful consideration of clinicopathologic parameters in the clinical workup of these cases.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":"664-672"},"PeriodicalIF":4.1,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12793813/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145250943","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-02-01Epub Date: 2025-10-16DOI: 10.1111/his.70022
Yuemei Xu, Qiuyuan Xia, Xiaotong Wang, Ru Fang, Qi Tong, Ya Wang, Jin Chen, Jieyu Chen, Yao Fu, Jiong Shi, Qiu Rao
Aims: The molecular characteristics and intricate relationships between tumours exhibiting overlapping features of metanephric adenoma (MA) and epithelial Wilms tumour (WT), as well as their pure forms, remain largely enigmatic.
Methods and results: Herein, we conducted a comprehensive genetic analysis of nine epithelial-predominant Wilms tumours, focusing on genetic alterations through expanded targeted sequencing and RNA sequencing (RNA-seq) methodologies. The patients ranged in age from 13 to 61 years, with a mean and median age of 43 and 48 years, respectively. The cohort included seven males and two females. These tumours exhibited immune reactivity for BRAF, WT1, and CD57, and harboured frequent TERT promoter mutations (7/9) and BRAF V600E mutations (8/9). RNA sequencing-based clustering revealed a close similarity between the tumours and MAs, suggesting that they may represent a distinct subset within the Wilms tumour spectrum. Two patients were lost to follow-up, while the remaining seven (7/7) were alive without tumour recurrences or metastases at the time of analysis, with a mean follow-up duration of 78.1 months.
Conclusions: Our research supports the notion previously described that epithelial-predominant Wilms tumours, characterized by frequent TERT promoter and BRAF V600E mutations, represent a distinct subset within the Wilms tumour spectrum. These tumours display an expression profile closely resembling that of metanephric adenomas and are associated with a favourable prognosis.
{"title":"Frequent BRAF V600E and TERT promoter-mutated renal epithelial-predominant Wilms tumours with metanephric features: a distinct subset within the Wilms tumour spectrum?","authors":"Yuemei Xu, Qiuyuan Xia, Xiaotong Wang, Ru Fang, Qi Tong, Ya Wang, Jin Chen, Jieyu Chen, Yao Fu, Jiong Shi, Qiu Rao","doi":"10.1111/his.70022","DOIUrl":"10.1111/his.70022","url":null,"abstract":"<p><strong>Aims: </strong>The molecular characteristics and intricate relationships between tumours exhibiting overlapping features of metanephric adenoma (MA) and epithelial Wilms tumour (WT), as well as their pure forms, remain largely enigmatic.</p><p><strong>Methods and results: </strong>Herein, we conducted a comprehensive genetic analysis of nine epithelial-predominant Wilms tumours, focusing on genetic alterations through expanded targeted sequencing and RNA sequencing (RNA-seq) methodologies. The patients ranged in age from 13 to 61 years, with a mean and median age of 43 and 48 years, respectively. The cohort included seven males and two females. These tumours exhibited immune reactivity for BRAF, WT1, and CD57, and harboured frequent TERT promoter mutations (7/9) and BRAF V600E mutations (8/9). RNA sequencing-based clustering revealed a close similarity between the tumours and MAs, suggesting that they may represent a distinct subset within the Wilms tumour spectrum. Two patients were lost to follow-up, while the remaining seven (7/7) were alive without tumour recurrences or metastases at the time of analysis, with a mean follow-up duration of 78.1 months.</p><p><strong>Conclusions: </strong>Our research supports the notion previously described that epithelial-predominant Wilms tumours, characterized by frequent TERT promoter and BRAF V600E mutations, represent a distinct subset within the Wilms tumour spectrum. These tumours display an expression profile closely resembling that of metanephric adenomas and are associated with a favourable prognosis.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":"683-697"},"PeriodicalIF":4.1,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145300127","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-02-01Epub Date: 2025-08-06DOI: 10.1111/his.15526
Trevor Toussieng, Michael Kozak, Miguel Burch, Alexandra Gangi, Jun Gong, Maha Guindi, Keith K Lai, Danielle A Hutchings, Brent K Larson, Kevin M Waters
Background: Autoimmune metaplastic atrophic gastritis (AMAG) causes pulmonary trans-differentiation of the gastric mucosa and is known to increase the risk of developing gastric adenocarcinoma. AMAG-associated gastric adenocarcinomas were examined for immunoreactivity for TTF-1 and Napsin A.
Design: Eighteen AMAG-associated gastric adenocarcinomas and 36 non-AMAG-associated gastric adenocarcinomas were stained for TTF-1 (clones SP141 and 8G7G3/1) and Napsin A (clone 1P64) by immunohistochemistry. AMAG stage and staining patterns were characterized.
Results: The AMAG group was older (mean age 74 vs. 65 years) and was not significantly different with regard to sex or tumour differentiation. Zero (0%) AMAG cases were classified as early phase, four (22%) as florid phase and 14 (78%) as end stage. AMAG-associated adenocarcinomas showed more frequent TTF-1 immunoreactivity compared to control adenocarcinomas in both clone SP141 (39% vs. 11%; P = 0.04) and 8G7G3/1 (22% vs. 6%; P = 0.17). AMAG-associated adenocarcinomas showed more frequent Napsin A immunoreactivity compared to control adenocarcinomas (28% vs. 0%; P < 0.01). Immunoreactivity in AMAG-associated adenocarcinomas was patchy, ranging from 1% to 20% of tumour cells staining positive (1+ to 3+ intensity). In the AMAG cases with background gastric mucosa (n = 14), the background showed TTF-1 positive foci in seven (50%) cases (both clones) versus one (3%; clone SP141) to two (6%; clone 8G7G3/1) of 33 controls (P < 0.01) and Napsin A positive foci in five (36%) cases versus zero (0%) controls (P < 0.01). Staining in background mucosa was also patchy, involving 1%-10% of gastric glands without intestinal metaplasia.
Conclusion: TTF-1 and Napsin A immunoreactivity was present in the background gastric mucosa almost exclusively in the AMAG-associated cases. This immunoreactivity is also present in AMAG-associated adenocarcinomas more commonly than non-AMAG-associated adenocarcinomas. This knowledge may aid pathologists in avoiding the pitfall of diagnosing metastatic lung cancer, as the expression is patchy and may also be seen in atrophic background mucosa.
背景:自身免疫性化生性萎缩性胃炎(AMAG)引起胃粘膜肺转分化,已知可增加发生胃腺癌的风险。设计:对18例amag相关的胃腺癌和36例非amag相关的胃腺癌进行TTF-1(克隆SP141和8G7G3/1)和Napsin A(克隆1P64)的免疫组化染色。表征AMAG分期及染色模式。结果:AMAG组年龄较大(平均年龄74岁vs. 65岁),在性别或肿瘤分化方面无显著差异。0例(0%)AMAG为早期期,4例(22%)为丰富期,14例(78%)为终末期。与对照腺癌相比,amag相关腺癌在克隆SP141 (39% vs. 11%, P = 0.04)和8G7G3/1 (22% vs. 6%, P = 0.17)中表现出更频繁的TTF-1免疫反应性。与对照腺癌相比,amag相关的腺癌表现出更频繁的Napsin A免疫反应性(28%比0%);P结论:在amag相关的病例中,TTF-1和Napsin A免疫反应性几乎只存在于背景胃粘膜中。这种免疫反应性在amag相关的腺癌中也比在非amag相关的腺癌中更常见。这些知识可以帮助病理学家避免诊断转移性肺癌的陷阱,因为表达是斑片状的,也可以在萎缩背景粘膜中看到。
{"title":"Pulmonary immunohistochemical markers may be positive in gastric adenocarcinomas associated with autoimmune metaplastic atrophic gastritis.","authors":"Trevor Toussieng, Michael Kozak, Miguel Burch, Alexandra Gangi, Jun Gong, Maha Guindi, Keith K Lai, Danielle A Hutchings, Brent K Larson, Kevin M Waters","doi":"10.1111/his.15526","DOIUrl":"https://doi.org/10.1111/his.15526","url":null,"abstract":"<p><strong>Background: </strong>Autoimmune metaplastic atrophic gastritis (AMAG) causes pulmonary trans-differentiation of the gastric mucosa and is known to increase the risk of developing gastric adenocarcinoma. AMAG-associated gastric adenocarcinomas were examined for immunoreactivity for TTF-1 and Napsin A.</p><p><strong>Design: </strong>Eighteen AMAG-associated gastric adenocarcinomas and 36 non-AMAG-associated gastric adenocarcinomas were stained for TTF-1 (clones SP141 and 8G7G3/1) and Napsin A (clone 1P64) by immunohistochemistry. AMAG stage and staining patterns were characterized.</p><p><strong>Results: </strong>The AMAG group was older (mean age 74 vs. 65 years) and was not significantly different with regard to sex or tumour differentiation. Zero (0%) AMAG cases were classified as early phase, four (22%) as florid phase and 14 (78%) as end stage. AMAG-associated adenocarcinomas showed more frequent TTF-1 immunoreactivity compared to control adenocarcinomas in both clone SP141 (39% vs. 11%; P = 0.04) and 8G7G3/1 (22% vs. 6%; P = 0.17). AMAG-associated adenocarcinomas showed more frequent Napsin A immunoreactivity compared to control adenocarcinomas (28% vs. 0%; P < 0.01). Immunoreactivity in AMAG-associated adenocarcinomas was patchy, ranging from 1% to 20% of tumour cells staining positive (1+ to 3+ intensity). In the AMAG cases with background gastric mucosa (n = 14), the background showed TTF-1 positive foci in seven (50%) cases (both clones) versus one (3%; clone SP141) to two (6%; clone 8G7G3/1) of 33 controls (P < 0.01) and Napsin A positive foci in five (36%) cases versus zero (0%) controls (P < 0.01). Staining in background mucosa was also patchy, involving 1%-10% of gastric glands without intestinal metaplasia.</p><p><strong>Conclusion: </strong>TTF-1 and Napsin A immunoreactivity was present in the background gastric mucosa almost exclusively in the AMAG-associated cases. This immunoreactivity is also present in AMAG-associated adenocarcinomas more commonly than non-AMAG-associated adenocarcinomas. This knowledge may aid pathologists in avoiding the pitfall of diagnosing metastatic lung cancer, as the expression is patchy and may also be seen in atrophic background mucosa.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":"88 3","pages":"729-735"},"PeriodicalIF":4.1,"publicationDate":"2026-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shilpy Jha, Anandi Lobo, Ankur R Sangoi, Shivani R Kandukuri, Sourav K Mishra, Samriti Arora, Aditi Aggarwal, Shivani Sharma, Ekta Jain, Mahmut Akgul, Andres M Acosta, Niharika Pattnaik, Seema Kaushal, Manas Baisakh, Sudhasmita Routa, Nada Shaker, Jasreman Dhillon, Anil V Parwani, Sean R Williamson, Sambit K Mohanty, Liang Cheng
Background: Clear cell adenocarcinoma (CCA) of the urinary tract is a rare genitourinary malignancy that primarily arises in the urethra and bladder. Due to its rarity, the molecular landscape of these tumours remains poorly characterized. In this large series, we aimed to molecularly characterize CCA to gain insights into its pathogenesis and identify potential targets for therapy.
Design: Formalin-fixed, paraffin-embedded tumour tissue blocks from 19 cases of CCA were subjected to molecular profiling using a targeted next-generation sequencing (NGS) panel.
Results: The most common alteration was a gain-of-function mutation in PIK3CA (74%), followed by mutations in KRAS (26%), ERBB2 (21%), SMAD4 (21%), RB1 (16%), TP53 (5%), MET (5%) and APC (5%). Thirteen tumours harboured co-mutations. Five cases showed concurrent PIK3CA and KRAS mutations, while the remaining tumours had either isolated PIK3CA alterations or co-occurring loss-of-function mutations in tumour suppressor genes, including RB1 point mutation, SMAD4 inactivation, APC truncation or TP53 inactivation. Eight patients died of disease, with a mean follow-up of 14 months (range, 3-31 months). Notably, all eight deceased patients and two of the surviving patients harboured PIK3CA mutations.
Conclusions: In summary, we identified a distinct oncogenic pathway in CCA of the urinary bladder, most commonly involving activation of the PI3K/AKT/mTOR pathway through gain-of-function mutations in PIK3CA (74%) and/or KRAS (26%). These tumours frequently harbour loss-of-function mutations in tumour suppressor genes (TP53, SMAD4, RB1 and APC) and point/missense mutations of proto-oncogenes (ERBB2 and MET). Our study also highlights potential therapeutic targets for this aggressive malignancy.
{"title":"Molecular characterization of clear cell adenocarcinoma of the urinary bladder.","authors":"Shilpy Jha, Anandi Lobo, Ankur R Sangoi, Shivani R Kandukuri, Sourav K Mishra, Samriti Arora, Aditi Aggarwal, Shivani Sharma, Ekta Jain, Mahmut Akgul, Andres M Acosta, Niharika Pattnaik, Seema Kaushal, Manas Baisakh, Sudhasmita Routa, Nada Shaker, Jasreman Dhillon, Anil V Parwani, Sean R Williamson, Sambit K Mohanty, Liang Cheng","doi":"10.1111/his.70096","DOIUrl":"https://doi.org/10.1111/his.70096","url":null,"abstract":"<p><strong>Background: </strong>Clear cell adenocarcinoma (CCA) of the urinary tract is a rare genitourinary malignancy that primarily arises in the urethra and bladder. Due to its rarity, the molecular landscape of these tumours remains poorly characterized. In this large series, we aimed to molecularly characterize CCA to gain insights into its pathogenesis and identify potential targets for therapy.</p><p><strong>Design: </strong>Formalin-fixed, paraffin-embedded tumour tissue blocks from 19 cases of CCA were subjected to molecular profiling using a targeted next-generation sequencing (NGS) panel.</p><p><strong>Results: </strong>The most common alteration was a gain-of-function mutation in PIK3CA (74%), followed by mutations in KRAS (26%), ERBB2 (21%), SMAD4 (21%), RB1 (16%), TP53 (5%), MET (5%) and APC (5%). Thirteen tumours harboured co-mutations. Five cases showed concurrent PIK3CA and KRAS mutations, while the remaining tumours had either isolated PIK3CA alterations or co-occurring loss-of-function mutations in tumour suppressor genes, including RB1 point mutation, SMAD4 inactivation, APC truncation or TP53 inactivation. Eight patients died of disease, with a mean follow-up of 14 months (range, 3-31 months). Notably, all eight deceased patients and two of the surviving patients harboured PIK3CA mutations.</p><p><strong>Conclusions: </strong>In summary, we identified a distinct oncogenic pathway in CCA of the urinary bladder, most commonly involving activation of the PI3K/AKT/mTOR pathway through gain-of-function mutations in PIK3CA (74%) and/or KRAS (26%). These tumours frequently harbour loss-of-function mutations in tumour suppressor genes (TP53, SMAD4, RB1 and APC) and point/missense mutations of proto-oncogenes (ERBB2 and MET). Our study also highlights potential therapeutic targets for this aggressive malignancy.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2026-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145984875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Federico Repetto, Kristine M Cornejo, Patrick O'Donnell, Jennifer P Toyohara, Judith A Ferry, Rosalynn M Nazarian
Aims: Cutaneous plasmablastic lymphoma (cPBL) is a rare and aggressive neoplasm that presents as skin nodules. Despite occurring in ~5% of PBL cases, the World Health Organization (WHO) does not distinguish primary cutaneous PBL (pcPBL) from secondary cutaneous PBL (scPBL), and their clinical differences remain poorly defined. To determine whether pcPBL represents a distinct clinical entity, we compare the clinicopathologic features, immunohistochemical profiles, and survival outcomes of pcPBL and scPBL.
Methods and results: Retrospective comparative study analysing 40 cases of cPBL (6 newly identified institutional cases and 34 cases from the literature), categorized as pcPBL (n = 25; no extracutaneous disease at diagnosis) or scPBL (n = 15; concurrent extracutaneous disease). Patients with pcPBL were older than those with scPBL (median 62 vs. 43 years; Mann-Whitney P = 0.018). Leg involvement was significantly associated with pcPBL (OR = 6.22; 95% CI: 1.21-31.9; P = 0.031). Disease-specific survival (DSS) analysis included 17 evaluable cases (pcPBL n = 11; scPBL n = 6). Median DSS was 42.0 months in pcPBL and 8.0 months in scPBL (log-rank χ2 = 3.98; p = 0.046). Median follow-up (reverse Kaplan-Meier) was 20.0 months in pcPBL and not reached in scPBL. Cox models were directionally consistent but underpowered.
Conclusion: In this pooled analysis, cases presenting with primary cutaneous involvement tended to occur in older patients and more often involved the legs. However, these observations should be interpreted cautiously given small numbers and heterogeneity of the available data. Within pcPBL, EBV positivity correlated with better survival. These hypothesis-generating findings provide a basis for prospective, multi-centre studies to clarify classification, staging implications, and management of this rare lymphoma.
目的:皮肤浆母细胞淋巴瘤(cPBL)是一种罕见的侵袭性肿瘤,表现为皮肤结节。尽管发生在约5%的PBL病例中,世界卫生组织(WHO)没有区分原发性皮肤PBL (pcPBL)和继发性皮肤PBL (scPBL),它们的临床差异仍然不明确。为了确定pcPBL是否代表一种独特的临床实体,我们比较了pcPBL和scPBL的临床病理特征、免疫组织化学特征和生存结果。方法与结果:回顾性比较分析40例cPBL(6例新发现的机构病例,34例文献资料),分为pcPBL(25例,诊断时无皮外疾病)和scPBL(15例,并发皮外疾病)。pcPBL患者比scPBL患者年龄大(中位62岁vs. 43岁;Mann-Whitney P = 0.018)。腿部受累与pcPBL显著相关(OR = 6.22; 95% CI: 1.21-31.9; P = 0.031)。疾病特异性生存(DSS)分析包括17例可评估病例(pcPBL n = 11; scPBL n = 6)。pcPBL中位生存期为42.0个月,scPBL中位生存期为8.0个月(log-rank χ2 = 3.98; p = 0.046)。pcPBL患者的中位随访(反向Kaplan-Meier)为20.0个月,而scPBL患者没有随访。Cox模型方向一致,但动力不足。结论:在本汇总分析中,原发性皮肤受累的病例往往发生在老年患者中,并且更常累及腿部。然而,鉴于现有数据的数量少且异质性,这些观察结果应谨慎解释。在pcPBL中,EBV阳性与更好的生存率相关。这些产生假设的发现为前瞻性的多中心研究提供了基础,以阐明这种罕见淋巴瘤的分类、分期和治疗。
{"title":"Cutaneous plasmablastic lymphoma: retrospective comparative study of primary and secondary skin involvement.","authors":"Federico Repetto, Kristine M Cornejo, Patrick O'Donnell, Jennifer P Toyohara, Judith A Ferry, Rosalynn M Nazarian","doi":"10.1111/his.70095","DOIUrl":"https://doi.org/10.1111/his.70095","url":null,"abstract":"<p><strong>Aims: </strong>Cutaneous plasmablastic lymphoma (cPBL) is a rare and aggressive neoplasm that presents as skin nodules. Despite occurring in ~5% of PBL cases, the World Health Organization (WHO) does not distinguish primary cutaneous PBL (pcPBL) from secondary cutaneous PBL (scPBL), and their clinical differences remain poorly defined. To determine whether pcPBL represents a distinct clinical entity, we compare the clinicopathologic features, immunohistochemical profiles, and survival outcomes of pcPBL and scPBL.</p><p><strong>Methods and results: </strong>Retrospective comparative study analysing 40 cases of cPBL (6 newly identified institutional cases and 34 cases from the literature), categorized as pcPBL (n = 25; no extracutaneous disease at diagnosis) or scPBL (n = 15; concurrent extracutaneous disease). Patients with pcPBL were older than those with scPBL (median 62 vs. 43 years; Mann-Whitney P = 0.018). Leg involvement was significantly associated with pcPBL (OR = 6.22; 95% CI: 1.21-31.9; P = 0.031). Disease-specific survival (DSS) analysis included 17 evaluable cases (pcPBL n = 11; scPBL n = 6). Median DSS was 42.0 months in pcPBL and 8.0 months in scPBL (log-rank χ<sup>2</sup> = 3.98; p = 0.046). Median follow-up (reverse Kaplan-Meier) was 20.0 months in pcPBL and not reached in scPBL. Cox models were directionally consistent but underpowered.</p><p><strong>Conclusion: </strong>In this pooled analysis, cases presenting with primary cutaneous involvement tended to occur in older patients and more often involved the legs. However, these observations should be interpreted cautiously given small numbers and heterogeneity of the available data. Within pcPBL, EBV positivity correlated with better survival. These hypothesis-generating findings provide a basis for prospective, multi-centre studies to clarify classification, staging implications, and management of this rare lymphoma.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2026-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145984857","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aims: Epstein-Barr virus (EBV)-positive inflammatory follicular dendritic cell sarcoma (EBV+ IFDCS) is a rare, indolent malignant neoplasm. Due to its rarity, a comprehensive assessment of its immunophenotypic spectrum and molecular analysis is still lacking. This study aimed to characterize the immunophenotypic and genetic alterations of EBV+ IFDCS to improve understanding of its cell of origin and molecular pathogenesis and to identify potential therapeutic targets.
Methods and results: Immunohistochemical staining for a panel of follicular dendritic cell (FDC) and fibroblastic reticular cell (FRC) lineage markers, along with targeted next-generation sequencing, was performed. Nineteen cases of EBV+ IFDCS were classified into four immunophenotypes: nine FDC, two FRC, three biphasic and five null phenotypes. Morphologically, cases with a null phenotype more frequently exhibited a lymphoma-like growth pattern (4/5, 80%) compared with those with a definite FDC and/or FRC phenotype (1/14, 7.1%, P = 0.006). Moreover, a scattered distribution of neoplastic cells was more commonly observed in null phenotype cases (4/5, 80%) than in FDC and/or FRC phenotype cases (3/14, 21.4%, P = 0.038). Targeted sequencing revealed somatic variants in chromatin modifier-related genes in 60.0% (9/15), homologous recombination repair (HRR)-related genes in 53.3% (8/15) and Hippo pathway-related genes (FAT2 and FAT1) in 26.7% (4/15) of cases.
Conclusions: These findings demonstrate the wide morphological and immunophenotypic spectrum of EBV+ IFDCS. Furthermore, variants in chromatin modifier and HRR-related genes may participate in its pathogenesis, and PARP inhibition may represent a potential therapeutic strategy for patients with unresectable disease.
{"title":"Immunophenotypic spectrum and mutational landscape of EBV-positive inflammatory follicular dendritic cell sarcoma.","authors":"Jian-Chao Wang, Chen Wang, Fang-Fang Chen, Wen-Fang Zhang, Zi-Qing Yu, Xiao-Jiang Wang, Shu-Lin Li, Mu-Sheng Chen, Li-Hua Zhong, Li-Yan Lin, Yan-Ping Chen, Xin Chen, Li-Hong Chen, Gang Chen","doi":"10.1111/his.70097","DOIUrl":"https://doi.org/10.1111/his.70097","url":null,"abstract":"<p><strong>Aims: </strong>Epstein-Barr virus (EBV)-positive inflammatory follicular dendritic cell sarcoma (EBV+ IFDCS) is a rare, indolent malignant neoplasm. Due to its rarity, a comprehensive assessment of its immunophenotypic spectrum and molecular analysis is still lacking. This study aimed to characterize the immunophenotypic and genetic alterations of EBV+ IFDCS to improve understanding of its cell of origin and molecular pathogenesis and to identify potential therapeutic targets.</p><p><strong>Methods and results: </strong>Immunohistochemical staining for a panel of follicular dendritic cell (FDC) and fibroblastic reticular cell (FRC) lineage markers, along with targeted next-generation sequencing, was performed. Nineteen cases of EBV+ IFDCS were classified into four immunophenotypes: nine FDC, two FRC, three biphasic and five null phenotypes. Morphologically, cases with a null phenotype more frequently exhibited a lymphoma-like growth pattern (4/5, 80%) compared with those with a definite FDC and/or FRC phenotype (1/14, 7.1%, P = 0.006). Moreover, a scattered distribution of neoplastic cells was more commonly observed in null phenotype cases (4/5, 80%) than in FDC and/or FRC phenotype cases (3/14, 21.4%, P = 0.038). Targeted sequencing revealed somatic variants in chromatin modifier-related genes in 60.0% (9/15), homologous recombination repair (HRR)-related genes in 53.3% (8/15) and Hippo pathway-related genes (FAT2 and FAT1) in 26.7% (4/15) of cases.</p><p><strong>Conclusions: </strong>These findings demonstrate the wide morphological and immunophenotypic spectrum of EBV+ IFDCS. Furthermore, variants in chromatin modifier and HRR-related genes may participate in its pathogenesis, and PARP inhibition may represent a potential therapeutic strategy for patients with unresectable disease.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933080","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Daniel J Shepherd, Cody Craig, Aida L Valencia-Guerrero, Sairam Chattu, Roshan Bhattarai, Leonel F Maldonado, Jennifer B Gordetsky
Aims: Extramammary Paget disease (EMPD) is an intraepidermal carcinoma that typically involves the urogenital, perineal and perianal skin. EMPD is classified as primary if arising directly in the skin, or secondary if spreading from another malignancy, most commonly urothelial carcinoma and colorectal adenocarcinoma. Prostein (p501s) immunoreactivity was initially described as sensitive and specific for prostatic epithelial cells including prostatic adenocarcinoma. Herein, we investigated the expression of prostein in primary EMPD of the external genitalia, secondary EMPD involving the external genitalia or perineum, and mammary Paget disease (MPD).
Methods and results: Prostein was negative by immunohistochemistry in all cases of MPD (n = 0/11) and secondary EMPD (n = 0/5). Conversely, prostein was positive in all cases of primary EMPD (n = 11/11), including non-invasive and invasive components and including one nodal metastasis. Among these cases, 5/11 (45%) showed non-focal moderate-to-strong staining, 3/11 (27%) showed focal weak staining, and 3/11 (27%) cases showed weak diffuse staining. All cases of primary EMPD additionally showed diffuse 2-3+ staining for GATA3, and 10/11 cases of primary EMPD showed diffuse 2-3+ staining for androgen receptor (AR).
Conclusions: These findings suggest that prostein may be a promising immunohistochemical marker for the diagnosis of primary EMPD.
{"title":"Prostein (p501s) is expressed in primary extramammary Paget disease.","authors":"Daniel J Shepherd, Cody Craig, Aida L Valencia-Guerrero, Sairam Chattu, Roshan Bhattarai, Leonel F Maldonado, Jennifer B Gordetsky","doi":"10.1111/his.70076","DOIUrl":"10.1111/his.70076","url":null,"abstract":"<p><strong>Aims: </strong>Extramammary Paget disease (EMPD) is an intraepidermal carcinoma that typically involves the urogenital, perineal and perianal skin. EMPD is classified as primary if arising directly in the skin, or secondary if spreading from another malignancy, most commonly urothelial carcinoma and colorectal adenocarcinoma. Prostein (p501s) immunoreactivity was initially described as sensitive and specific for prostatic epithelial cells including prostatic adenocarcinoma. Herein, we investigated the expression of prostein in primary EMPD of the external genitalia, secondary EMPD involving the external genitalia or perineum, and mammary Paget disease (MPD).</p><p><strong>Methods and results: </strong>Prostein was negative by immunohistochemistry in all cases of MPD (n = 0/11) and secondary EMPD (n = 0/5). Conversely, prostein was positive in all cases of primary EMPD (n = 11/11), including non-invasive and invasive components and including one nodal metastasis. Among these cases, 5/11 (45%) showed non-focal moderate-to-strong staining, 3/11 (27%) showed focal weak staining, and 3/11 (27%) cases showed weak diffuse staining. All cases of primary EMPD additionally showed diffuse 2-3+ staining for GATA3, and 10/11 cases of primary EMPD showed diffuse 2-3+ staining for androgen receptor (AR).</p><p><strong>Conclusions: </strong>These findings suggest that prostein may be a promising immunohistochemical marker for the diagnosis of primary EMPD.</p>","PeriodicalId":13219,"journal":{"name":"Histopathology","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933069","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}