Jing Yang, Jinku Zhang, Jinmei Li, Yan Liu, Yuxiang Wang, Ajin Hu, Congrong Liu
Uterine tumor resembling ovarian sex cord tumor (UTROSCT) constitutes an exceptionally rare histological subset of uterine mesenchymal neoplasms. While most cases have benign clinical behavior, a subset of UTROSCTs exhibits clinically aggressive behavior characterized by recurrence and metastasis. Here, we present a cohort of 25 UTROSCT cases molecularly confirmed by recurrent fusion gene detection, including ESR1::NCOA3 (n = 12), GREB1::NCOA1 (n = 6), ESR1::NCOA2 (n = 3), GREB1::NCOA2 (n = 2), GREB1::SS18 (n = 1), and GREB1::CTNNB1 (n = 1). Notably, six cases (6/25, 24%) demonstrated recurrence/metastasis: two cases showed intrauterine recurrence (harboring ESR1::NCOA3 and GREB1::NCOA1 fusions), while four developed extrauterine metastases (carrying ESR1::NCOA3, ESR1::NCOA2, GREB1::NCOA1, and GREB1::NCOA2 fusions), with one fatality. To dissect the biological basis of UTROSCT aggressiveness, we performed integrated clinicopathologic, immunohistochemical, and molecular profiling. Multivariate analysis identified tumor size >5 cm, FIGO stage IB, and lymphovascular space invasion (LVSI) as independent predictors of recurrence/metastasis, whereas histologic features, proliferation index, and fusion gene subtypes lacked prognostic significance. Multi-omics analysis of primary versus metastatic tumors revealed striking copy number variations (CNVs) exclusively in metastatic lesions. Specifically, heterozygous losses of SMARCB1 (2/4 metastatic cases) and ATRX (1/4 metastatic cases) were identified; both play critical roles in chromatin remodeling. These genetic alterations were conspicuously absent in primary tumors, suggesting their potential role in metastatic progression. Our findings represent the first demonstration of CNV-driven oncogenic evolution in UTROSCTs, particularly implicating SWI/SNF complex dysregulation in metastatic competence.
{"title":"Decoding UTROSCT heterogeneity: systematic clinicopathological evaluation combined with molecular profiling","authors":"Jing Yang, Jinku Zhang, Jinmei Li, Yan Liu, Yuxiang Wang, Ajin Hu, Congrong Liu","doi":"10.1002/2056-4538.70055","DOIUrl":"https://doi.org/10.1002/2056-4538.70055","url":null,"abstract":"<p>Uterine tumor resembling ovarian sex cord tumor (UTROSCT) constitutes an exceptionally rare histological subset of uterine mesenchymal neoplasms. While most cases have benign clinical behavior, a subset of UTROSCTs exhibits clinically aggressive behavior characterized by recurrence and metastasis. Here, we present a cohort of 25 UTROSCT cases molecularly confirmed by recurrent fusion gene detection, including <i>ESR1::NCOA3</i> (<i>n</i> = 12), <i>GREB1::NCOA1</i> (<i>n</i> = 6), <i>ESR1::NCOA2</i> (<i>n</i> = 3), <i>GREB1::NCOA2</i> (<i>n</i> = 2), <i>GREB1::SS18</i> (<i>n</i> = 1), and <i>GREB1::CTNNB1</i> (<i>n</i> = 1). Notably, six cases (6/25, 24%) demonstrated recurrence/metastasis: two cases showed intrauterine recurrence (harboring <i>ESR1::NCOA3</i> and <i>GREB1::NCOA1</i> fusions), while four developed extrauterine metastases (carrying <i>ESR1::NCOA3</i>, <i>ESR1::NCOA2, GREB1::NCOA1,</i> and <i>GREB1::NCOA2</i> fusions), with one fatality. To dissect the biological basis of UTROSCT aggressiveness, we performed integrated clinicopathologic, immunohistochemical, and molecular profiling. Multivariate analysis identified tumor size >5 cm, FIGO stage IB, and lymphovascular space invasion (LVSI) as independent predictors of recurrence/metastasis, whereas histologic features, proliferation index, and fusion gene subtypes lacked prognostic significance. Multi-omics analysis of primary versus metastatic tumors revealed striking copy number variations (CNVs) exclusively in metastatic lesions. Specifically, heterozygous losses of <i>SMARCB1</i> (2/4 metastatic cases) and <i>ATRX</i> (1/4 metastatic cases) were identified; both play critical roles in chromatin remodeling. These genetic alterations were conspicuously absent in primary tumors, suggesting their potential role in metastatic progression. Our findings represent the first demonstration of CNV-driven oncogenic evolution in UTROSCTs, particularly implicating SWI/SNF complex dysregulation in metastatic competence.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"12 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70055","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626257","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}
Oana M Craciun, Ester García-Ovejero, Fiona Campbell, Marina Montes-Mota, Stefan Holdenrieder, Inga Trulson, Karolina Worf, Sophie Gabriel, Magdalena Kowalewska, Irmina Michalek, Kateryna Maslova, Lukasz Taraszkiewicz, Javier del Águila, Richard Colling, Puay Hoon Tan, Gabrielle Goldman-Lévy, Christine Giesen, Ramon Cierco Jimenez, Dilani Lokuhetty, Ian A Cree, Iciar Indave, Beatriz Pérez Gómez, Magdalena Chechlińska, Marina Pollán Santamaría, Elena Plans-Beriso, WCT EVI MAP Project Team
The World Health Organization (WHO) Classification of Tumours: A Living Evidence Gap Map by Tumour Type (WCT EVI MAP) project aims to develop Evidence Gap Maps of the available evidence, primarily to inform the WHO Classification of Tumours. The project, covering all tumour types, faces the challenge of reviewing a huge number of studies by reviewers from multiple backgrounds. The aim was to develop a decision tree (DT) diagram for classifying study designs reporting on tumour pathology studies, in order to support the decision-making process when assigning evidence levels across various disciplines. A modified consensus process, incorporating stakeholder workshops, was conducted in three phases: (1) development of the initial DT diagram draft (literature review and expert evaluation); (2) iterative reviews with project partners; and (3) testing the advanced DT diagram version with several sets of references to refine critical points. A total of 368 records were used for training throughout the entire process. Consensus was achieved when classifications could categorise studies consistently without causing discordance in new example sets. A DT diagram and its Glossary of Operational Definitions with 27 decision nodes and 26 categories were developed. The DT diagram is organised into six sections: WCT EVI MAP selection criteria, evidence synthesis, basic research related studies, descriptive studies, observational and experimental studies, and diagnostic test studies. The DT diagram is a valuable tool for the project's needs, successfully integrating diverse disciplinary perspectives for classifying evidence in tumour pathology research according to study design. It lays the foundation for future advancements in evidence mapping and classification within tumour pathology and related disciplines.
世界卫生组织(世卫组织)肿瘤分类:按肿瘤类型绘制活证据差距图(WCT EVI Map)项目旨在编制现有证据的证据差距图,主要为世卫组织肿瘤分类提供信息。该项目涵盖所有肿瘤类型,面临着审查来自不同背景的审稿人的大量研究的挑战。目的是开发一个决策树(DT)图,用于对报告肿瘤病理学研究的研究设计进行分类,以便在分配不同学科的证据水平时支持决策过程。修改后的共识过程,包括利益相关者研讨会,分三个阶段进行:(1)制定初始DT图草案(文献回顾和专家评估);(2)与项目伙伴进行迭代评审;(3)使用多组参考资料对DT图高级版本进行测试,以细化关键点。在整个过程中,总共使用了368条记录进行培训。当分类可以一致地对研究进行分类而不会在新的示例集中引起不一致时,共识就实现了。开发了包含27个决策节点和26个类别的DT图及其操作定义词汇表。DT图分为六个部分:WCT EVI MAP选择标准、证据综合、基础研究相关研究、描述性研究、观察和实验研究以及诊断测试研究。DT图是满足项目需要的一个有价值的工具,它成功地整合了不同学科的观点,根据研究设计对肿瘤病理学研究中的证据进行分类。它为肿瘤病理学和相关学科的证据映射和分类的未来发展奠定了基础。
{"title":"Development of a decision tree diagram for classifying study designs in tumour pathology research: a multidisciplinary approach","authors":"Oana M Craciun, Ester García-Ovejero, Fiona Campbell, Marina Montes-Mota, Stefan Holdenrieder, Inga Trulson, Karolina Worf, Sophie Gabriel, Magdalena Kowalewska, Irmina Michalek, Kateryna Maslova, Lukasz Taraszkiewicz, Javier del Águila, Richard Colling, Puay Hoon Tan, Gabrielle Goldman-Lévy, Christine Giesen, Ramon Cierco Jimenez, Dilani Lokuhetty, Ian A Cree, Iciar Indave, Beatriz Pérez Gómez, Magdalena Chechlińska, Marina Pollán Santamaría, Elena Plans-Beriso, WCT EVI MAP Project Team","doi":"10.1002/2056-4538.70056","DOIUrl":"https://doi.org/10.1002/2056-4538.70056","url":null,"abstract":"<p>The World Health Organization (WHO) Classification of Tumours: A Living Evidence Gap Map by Tumour Type (WCT EVI MAP) project aims to develop Evidence Gap Maps of the available evidence, primarily to inform the WHO Classification of Tumours. The project, covering all tumour types, faces the challenge of reviewing a huge number of studies by reviewers from multiple backgrounds. The aim was to develop a decision tree (DT) diagram for classifying study designs reporting on tumour pathology studies, in order to support the decision-making process when assigning evidence levels across various disciplines. A modified consensus process, incorporating stakeholder workshops, was conducted in three phases: (1) development of the initial DT diagram draft (literature review and expert evaluation); (2) iterative reviews with project partners; and (3) testing the advanced DT diagram version with several sets of references to refine critical points. A total of 368 records were used for training throughout the entire process. Consensus was achieved when classifications could categorise studies consistently without causing discordance in new example sets. A DT diagram and its Glossary of Operational Definitions with 27 decision nodes and 26 categories were developed. The DT diagram is organised into six sections: WCT EVI MAP selection criteria, evidence synthesis, basic research related studies, descriptive studies, observational and experimental studies, and diagnostic test studies. The DT diagram is a valuable tool for the project's needs, successfully integrating diverse disciplinary perspectives for classifying evidence in tumour pathology research according to study design. It lays the foundation for future advancements in evidence mapping and classification within tumour pathology and related disciplines.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"12 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70056","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626737","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}
<p>The recent seminal work by Nann and colleagues, ‘Myeloid sarcoma shows a high frequency of mutations activating the MAPK/ERK pathway and association with clonal hematopoiesis’ provides a crucial genomic framework for this rare and aggressive malignancy [<span>1</span>]. By rigorously demonstrating the high prevalence of <i>KRAS</i>, <i>NRAS</i>, and <i>PTPN11</i> mutations and linking a subset of cases to clonal hematopoiesis (CH), the authors have rightly shifted the paradigm for understanding myeloid sarcoma (MS) pathogenesis. While these findings are undoubtedly significant, a critical appraisal reveals several profound biological and clinical challenges that must be addressed to translate this genetic atlas into therapeutic progress.</p><p>First, the functional and clinical implications of subclonal MAPK/ERK mutations demand a more nuanced interpretation than presented. While the high frequency (44%) is striking, the subclonal nature of many of these mutations suggests they may not be the universal founding oncogenic event. A critical, unanswered question is whether MAPK/ERK activation is a <i>bona fide</i> driver of extramedullary tropism or merely a passenger event that confers a proliferative advantage within a permissive niche. The study's data, while correlative, cannot distinguish between these possibilities. If these mutations are indeed key to tissue invasion, why do we not observe universal, clonal dominance? This heterogeneity poses a direct threat to the efficacy of targeted therapies. MEK or ERK inhibitor monotherapy, as successfully deployed in other rat sarcoma (RAS)-pathway-driven cancers [<span>2</span>], would likely select for the outgrowth of pre-existing, MAPK-wild-type subclones, leading to rapid clinical resistance. Therefore, the therapeutic strategy for MS may need to pivot towards rational combinations that target a more fundamental dependency of the MS-initiating cell, such as BCL-2 inhibition [<span>3</span>], concurrently with MAPK pathway suppression to prevent escape.</p><p>Second, the provocative association with CH requires stringent validation and mechanistic exploration. The authors propose a compelling ‘two-hit’ model where CH provides the initial pre-leukemic clone, with subsequent MAPK/ERK mutation triggering overt MS. However, the phylogenetic relationship between the CH clone and the dominant MS clone remains largely inferential from the data shown. Definitive proof requires single-cell DNA sequencing of MS lesions and paired bone marrow to reconstruct the precise evolutionary trajectory. Furthermore, the immunological consequences of this trajectory are entirely unexplored. CH is known to create a pro-inflammatory bone marrow milieu and can remodel the immune system, potentially fostering a permissive environment for secondary oncogenic events [<span>4</span>]. It is plausible that CH-derived MS represents a distinct immunobiological entity compared to <i>de novo</i> MS. Characterizing the tumor micr
{"title":"The road to therapy for myeloid sarcoma: navigating the complexities of subclonal MAPK/ERK mutations and clonal evolution","authors":"DuJiang Yang, Jiexiang Yang, GuoYou Wang","doi":"10.1002/2056-4538.70063","DOIUrl":"10.1002/2056-4538.70063","url":null,"abstract":"<p>The recent seminal work by Nann and colleagues, ‘Myeloid sarcoma shows a high frequency of mutations activating the MAPK/ERK pathway and association with clonal hematopoiesis’ provides a crucial genomic framework for this rare and aggressive malignancy [<span>1</span>]. By rigorously demonstrating the high prevalence of <i>KRAS</i>, <i>NRAS</i>, and <i>PTPN11</i> mutations and linking a subset of cases to clonal hematopoiesis (CH), the authors have rightly shifted the paradigm for understanding myeloid sarcoma (MS) pathogenesis. While these findings are undoubtedly significant, a critical appraisal reveals several profound biological and clinical challenges that must be addressed to translate this genetic atlas into therapeutic progress.</p><p>First, the functional and clinical implications of subclonal MAPK/ERK mutations demand a more nuanced interpretation than presented. While the high frequency (44%) is striking, the subclonal nature of many of these mutations suggests they may not be the universal founding oncogenic event. A critical, unanswered question is whether MAPK/ERK activation is a <i>bona fide</i> driver of extramedullary tropism or merely a passenger event that confers a proliferative advantage within a permissive niche. The study's data, while correlative, cannot distinguish between these possibilities. If these mutations are indeed key to tissue invasion, why do we not observe universal, clonal dominance? This heterogeneity poses a direct threat to the efficacy of targeted therapies. MEK or ERK inhibitor monotherapy, as successfully deployed in other rat sarcoma (RAS)-pathway-driven cancers [<span>2</span>], would likely select for the outgrowth of pre-existing, MAPK-wild-type subclones, leading to rapid clinical resistance. Therefore, the therapeutic strategy for MS may need to pivot towards rational combinations that target a more fundamental dependency of the MS-initiating cell, such as BCL-2 inhibition [<span>3</span>], concurrently with MAPK pathway suppression to prevent escape.</p><p>Second, the provocative association with CH requires stringent validation and mechanistic exploration. The authors propose a compelling ‘two-hit’ model where CH provides the initial pre-leukemic clone, with subsequent MAPK/ERK mutation triggering overt MS. However, the phylogenetic relationship between the CH clone and the dominant MS clone remains largely inferential from the data shown. Definitive proof requires single-cell DNA sequencing of MS lesions and paired bone marrow to reconstruct the precise evolutionary trajectory. Furthermore, the immunological consequences of this trajectory are entirely unexplored. CH is known to create a pro-inflammatory bone marrow milieu and can remodel the immune system, potentially fostering a permissive environment for secondary oncogenic events [<span>4</span>]. It is plausible that CH-derived MS represents a distinct immunobiological entity compared to <i>de novo</i> MS. Characterizing the tumor micr","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70063","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145543373","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}
We thank the authors for their interest in our recent paper ‘Myeloid sarcoma shows a high frequency of mutations activating the MAPK/ERK pathway and association with clonal hematopoiesis’ [1], which appeared in the September issue of The Journal of Pathology, Clinical Research. In their letter to the Editor, Dr. Yang and his colleagues raise some important issues, which point the way for further studies on myeloid sarcoma (MS) and its development [2]. These include subclonal mutations in the MAPK pathway, the association with clonal hematopoiesis and the importance of the microenvironment.
Nevertheless, we want to clarify a few points raised by Dr. Yang and colleagues concerning our results. They state that the frequent subclonal nature of many MAPK/ERK mutations in our MS samples suggests they may not be the universal founding oncogenic event. Although we agree with this statement in principle, since activating mutations of NRAS, KRAS and/or BRAF were observed in only 53% of our MS cases and other acquired mutations, namely NPM1, mutated in 38% of cases, are alternative candidate drivers, we want to point out that NRAS/KRAS mutations constitute minor, subclonal events in only 2/18 cases. In case 27, the bone marrow showed a KRAS mutation with 7% variant allele frequency (VAF), which was present at 2% and 3% VAF, respectively, in the two MS from this patient. In case 31, the NRAS mutation was present in the bone marrow at 28% VAF and at only 0.4% VAF in the MS. In these cases, the MAPK/ERK mutations most likely represent a subclone and are not responsible for MS development. In the remaining 15/18 cases with NRAS/KRAS mutations; however, these showed a high VAF (mean 41.7%, range 19–92%). In the cases with available pre-transplant bone marrow (BM) biopsy, the majority of NRAS/KRAS mutations were acquired in MS or showed a significant increase in VAF in the MS in comparison to the BM biopsy. In some cases with mutations in other genes with higher VAF, this is indicative of a bi-allelic event rather than subclonality of the RAS mutation, as for example in case 33 with a TP53 mutation with 83% VAF and the EZH2 mutation in case 18 with 89% (scrotal) and 99% (penis) VAF, respectively, in two different locations. In summary, the high VAFs and the frequent de novo acquisition of NRAS/KRAS mutations in many MS cases, together with the clear evidence for upregulation of the MAPK/ERK signaling cascades (supplementary material, Figure S3 in our paper [1]) detected by comparative gene expression profiling clearly point to a role in MS development, although functional studies will be necessary to corroborate these findings. Of interest is case 10, which showed acquired identical mutations of BRAF and KRAS in both MS samples, but not in the BM. Given the fact that both
{"title":"Authors' reply: Re: Yang et al. The road to therapy for myeloid sarcoma: navigating the complexities of subclonal MAPK/ERK mutations and clonal evolution","authors":"Dominik Nann, Falko Fend","doi":"10.1002/2056-4538.70064","DOIUrl":"10.1002/2056-4538.70064","url":null,"abstract":"<p>Reply to Yang <i>et al</i>.</p><p>We thank the authors for their interest in our recent paper ‘Myeloid sarcoma shows a high frequency of mutations activating the MAPK/ERK pathway and association with clonal hematopoiesis’ [<span>1</span>], which appeared in the September issue of <i>The Journal of Pathology, Clinical Research</i>. In their letter to the Editor, Dr. Yang and his colleagues raise some important issues, which point the way for further studies on myeloid sarcoma (MS) and its development [<span>2</span>]. These include subclonal mutations in the MAPK pathway, the association with clonal hematopoiesis and the importance of the microenvironment.</p><p>Nevertheless, we want to clarify a few points raised by Dr. Yang and colleagues concerning our results. They state that the frequent subclonal nature of many MAPK/ERK mutations in our MS samples suggests they may not be the universal founding oncogenic event. Although we agree with this statement in principle, since activating mutations of <i>NRAS, KRAS</i> and/or <i>BRAF</i> were observed in only 53% of our MS cases and other acquired mutations, namely <i>NPM1</i>, mutated in 38% of cases, are alternative candidate drivers, we want to point out that <i>NRAS/KRAS</i> mutations constitute minor, subclonal events in only 2/18 cases. In case 27, the bone marrow showed a <i>KRAS</i> mutation with 7% variant allele frequency (VAF), which was present at 2% and 3% VAF, respectively, in the two MS from this patient. In case 31, the <i>NRAS</i> mutation was present in the bone marrow at 28% VAF and at only 0.4% VAF in the MS. In these cases, the MAPK/ERK mutations most likely represent a subclone and are not responsible for MS development. In the remaining 15/18 cases with <i>NRAS/KRAS</i> mutations; however, these showed a high VAF (mean 41.7%, range 19–92%). In the cases with available pre-transplant bone marrow (BM) biopsy, the majority of <i>NRAS/KRAS</i> mutations were acquired in MS or showed a significant increase in VAF in the MS in comparison to the BM biopsy. In some cases with mutations in other genes with higher VAF, this is indicative of a bi-allelic event rather than subclonality of the <i>RAS</i> mutation, as for example in case 33 with a <i>TP53</i> mutation with 83% VAF and the <i>EZH2</i> mutation in case 18 with 89% (scrotal) and 99% (penis) VAF, respectively, in two different locations. In summary, the high VAFs and the frequent <i>de novo</i> acquisition of <i>NRAS/KRAS</i> mutations in many MS cases, together with the clear evidence for upregulation of the MAPK/ERK signaling cascades (supplementary material, Figure S3 in our paper [<span>1</span>]) detected by comparative gene expression profiling clearly point to a role in MS development, although functional studies will be necessary to corroborate these findings. Of interest is case 10, which showed acquired identical mutations of <i>BRAF</i> and <i>KRAS</i> in both MS samples, but not in the BM. Given the fact that both ","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70064","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145543336","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}
Nicolette Zavillová, Petr Waldauf, Michaela Kendall Bártů, David Čapka, Jan Hojný, Zuzana Prouzová, Radoslav Matěj, Roman Zachoval, Jan Hrudka
Several prognostic markers, including tumor-infiltrating lymphocytes, which have been recently identified in penile squamous cell carcinoma (pSCC), have focused mostly on T cells. The prognostic role of B cells and tertiary lymphoid structures (TLSs) has not yet been sufficiently described. We examined whole tissue sections histopathologically for TLSs and immunohistochemically for CD20 and CD138. The B-cell immunoscore (B-IS) divided the cohort into five categories based on the expression of these two B-cell/plasma cell markers (CD20 and CD138, respectively). Patients with fewer TLSs had worse overall survival (OS) [hazard ratio (HR) = 2.17; 95% CI: 0.94–5; p = 0.069]. A significant association was identified between a high TLS diameter and the presence of a lymphocytic infiltrate [odds ratio (OR) = 2.2442; 95% CI: 1.1022–4.55; p = 0.0208]. Patients with low B-IS (HR = 1.89, 95% CI: 1.18–3.03, p = 0.008), a low number of CD20+ cells in the tumor center (HR = 1.67, 95% CI: 1.04–2.7, p = 0.035), and a low number of CD20+ cells at the tumor invasion front (HR = 1.69, 95% CI: 1.06–2.78; p = 0.028) had significantly worse OS. High B-ISs were strongly associated with a mutated p53 profile detected by immunohistochemistry (OR = 4.76, 95% CI: 1.32–25, p = 0.011), low T-cell immunoscores (OR = 0.49; 95% CI: 0.23–1.03; p = 0.051), and brisk lymphocytic infiltration (OR = 2.0417, 95% CI: 1.01–4.76; p = 0.037). High CD20+ cell counts at the invasion front were associated with histological grade 3 disease (OR = 2.44, 95% CI 1.15–5.26, p = 0.015). An association was also observed between low B-IS and mutations in KMT2D (OR 0.31, 95% CI: 0.07–1.21, p = 0.057) and EGFR (OR = ∞, 95% CI: 0.86–∞, p = 0.053). In conclusion, high numbers of tumor-infiltrating B cells within TLSs represent a favorable prognostic marker in pSCC. These findings emphasize the need to identify novel microscopic prognostic markers during pathological assessment to guide early and appropriate therapeutic strategies.
一些预后标志物,包括肿瘤浸润淋巴细胞,最近在阴茎鳞状细胞癌(pSCC)中被发现,主要集中在T细胞上。B细胞和三级淋巴结构(TLSs)的预后作用尚未得到充分的描述。我们用组织病理学检查了整个组织切片的TLSs,免疫组织化学检查了CD20和CD138。b细胞免疫评分(B-IS)根据这两种b细胞/浆细胞标记物(分别为CD20和CD138)的表达将队列分为五类。TLSs较少的患者总生存期(OS)较差[危险比(HR) = 2.17;95% ci: 0.94- 95;p = 0.069]。高TLS直径与淋巴细胞浸润存在显著关联[优势比(OR) = 2.2442;95% ci: 1.1022-4.55;p = 0.0208]。低B-IS (HR = 1.89, 95% CI: 1.18-3.03, p = 0.008)、肿瘤中心CD20+细胞数量少(HR = 1.67, 95% CI: 1.04-2.7, p = 0.035)、肿瘤侵袭前沿CD20+细胞数量少(HR = 1.69, 95% CI: 1.06-2.78, p = 0.028)患者的OS明显较差。高B-ISs与免疫组织化学检测的p53突变谱(OR = 4.76, 95% CI: 1.32-25, p = 0.011)、低t细胞免疫评分(OR = 0.49, 95% CI: 0.23-1.03, p = 0.051)和淋巴细胞浸润活跃(OR = 2.0417, 95% CI: 1.01-4.76, p = 0.037)密切相关。侵袭前沿CD20+细胞计数高与组织学3级疾病相关(OR = 2.44, 95% CI 1.15-5.26, p = 0.015)。低B-IS与KMT2D突变(OR = 0.31, 95% CI: 0.07-1.21, p = 0.057)和EGFR突变(OR =∞,95% CI: 0.86-∞,p = 0.053)之间也存在关联。总之,TLSs内大量的肿瘤浸润B细胞是pSCC的一个有利的预后标志。这些发现强调需要在病理评估中识别新的显微预后标志物,以指导早期和适当的治疗策略。
{"title":"B lymphocytes and tertiary lymphoid structures have a prognostic impact on penile squamous cell carcinoma","authors":"Nicolette Zavillová, Petr Waldauf, Michaela Kendall Bártů, David Čapka, Jan Hojný, Zuzana Prouzová, Radoslav Matěj, Roman Zachoval, Jan Hrudka","doi":"10.1002/2056-4538.70059","DOIUrl":"10.1002/2056-4538.70059","url":null,"abstract":"<p>Several prognostic markers, including tumor-infiltrating lymphocytes, which have been recently identified in penile squamous cell carcinoma (pSCC), have focused mostly on T cells. The prognostic role of B cells and tertiary lymphoid structures (TLSs) has not yet been sufficiently described. We examined whole tissue sections histopathologically for TLSs and immunohistochemically for CD20 and CD138. The B-cell immunoscore (B-IS) divided the cohort into five categories based on the expression of these two B-cell/plasma cell markers (CD20 and CD138, respectively). Patients with fewer TLSs had worse overall survival (OS) [hazard ratio (HR) = 2.17; 95% CI: 0.94–5; <i>p</i> = 0.069]. A significant association was identified between a high TLS diameter and the presence of a lymphocytic infiltrate [odds ratio (OR) = 2.2442; 95% CI: 1.1022–4.55; <i>p</i> = 0.0208]. Patients with low B-IS (HR = 1.89, 95% CI: 1.18–3.03, <i>p</i> = 0.008), a low number of CD20<sup>+</sup> cells in the tumor center (HR = 1.67, 95% CI: 1.04–2.7, <i>p</i> = 0.035), and a low number of CD20<sup>+</sup> cells at the tumor invasion front (HR = 1.69, 95% CI: 1.06–2.78; <i>p</i> = 0.028) had significantly worse OS. High B-ISs were strongly associated with a mutated p53 profile detected by immunohistochemistry (OR = 4.76, 95% CI: 1.32–25, <i>p</i> = 0.011), low T-cell immunoscores (OR = 0.49; 95% CI: 0.23–1.03; <i>p</i> = 0.051), and brisk lymphocytic infiltration (OR = 2.0417, 95% CI: 1.01–4.76; <i>p</i> = 0.037). High CD20<sup>+</sup> cell counts at the invasion front were associated with histological grade 3 disease (OR = 2.44, 95% CI 1.15–5.26, <i>p</i> = 0.015). An association was also observed between low B-IS and mutations in <i>KMT2D</i> (OR 0.31, 95% CI: 0.07–1.21, <i>p</i> = 0.057) and <i>EGFR</i> (OR = ∞, 95% CI: 0.86–∞, <i>p</i> = 0.053). In conclusion, high numbers of tumor-infiltrating B cells within TLSs represent a favorable prognostic marker in pSCC. These findings emphasize the need to identify novel microscopic prognostic markers during pathological assessment to guide early and appropriate therapeutic strategies.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70059","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514738","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}
Huayan Ren, Wanting Tong, Jiayue Ma, Xinyan Chen, Huifen Huang, Na Wei, Yuqiong Liu, Minglei Yang, Lan Zhang, Huixiang Li
Secretory breast carcinoma (SBC) is a rare tumour defined by ETV6-NTRK3 rearrangement, but its clinicopathological spectrum and potential for aggressive behaviour remain incompletely characterised. We retrospectively reviewed 29 SBCs diagnosed between 2014 and 2024, including 28 females and one male aged 12–63 years (median 44). Twenty-eight tumours arose in the breast parenchyma and one in axillary accessory breast tissue. Histologically, microcystic and tubular patterns predominated and carcinoma in situ was common. Most tumours were nuclear grade 1 with rare mitoses (0–1/10 high-power fields, HPF). A single patient with distant metastasis harboured a solid-predominant tumour showing nuclear grade 2–3, brisk mitoses (6/10 HPF), and multifocal necrosis. All tumours demonstrated diffuse S100 and pan-TRK expression. Oestrogen and/or progesterone receptor staining was observed in 16 of 29 cases (2–30% of tumour cells), and all were HER2 negative or low (0–1+). Ki-67 ranged from 3% to 20% (mean 7%). Fluorescence in situ hybridisation (FISH) was positive in 17 of 17 tested tumours (14 ETV6-NTRK3 dual-fusion; 3 NTRK3 break-apart). In the metastatic case, RNA sequencing confirmed canonical ETV6-NTRK3 fusion, while targeted DNA sequencing identified additional variants of uncertain significance (VUS) – RANBP2 p.S1843R, NUP107 p.K382Q, NCOR1 p.A1947V (missense), and PREX2 p.G606G (synonymous). All patients underwent surgery, and 14 received adjuvant chemotherapy. During follow-up ranging from 6 to 135 months (median 76), one patient developed lung metastasis and was alive with disease at 88 months; the remaining 28 patients were alive without recurrence or metastasis. In summary, SBC is typically indolent and characterised by ETV6-NTRK3 rearrangement with diffuse pan-TRK/S100 positivity. A solid-predominant pattern with increased cytological atypia, mitotic activity, and necrosis may indicate aggressive potential. Routine NTRK testing supports diagnosis and may help identify patients who could benefit from TRK-inhibitor therapy in advanced disease.
{"title":"Secretory breast carcinoma: morphologic and molecular heterogeneity with indicators of aggressive potential in a cohort of 29 cases","authors":"Huayan Ren, Wanting Tong, Jiayue Ma, Xinyan Chen, Huifen Huang, Na Wei, Yuqiong Liu, Minglei Yang, Lan Zhang, Huixiang Li","doi":"10.1002/2056-4538.70060","DOIUrl":"10.1002/2056-4538.70060","url":null,"abstract":"<p>Secretory breast carcinoma (SBC) is a rare tumour defined by <i>ETV6-NTRK3</i> rearrangement, but its clinicopathological spectrum and potential for aggressive behaviour remain incompletely characterised. We retrospectively reviewed 29 SBCs diagnosed between 2014 and 2024, including 28 females and one male aged 12–63 years (median 44). Twenty-eight tumours arose in the breast parenchyma and one in axillary accessory breast tissue. Histologically, microcystic and tubular patterns predominated and carcinoma <i>in situ</i> was common. Most tumours were nuclear grade 1 with rare mitoses (0–1/10 high-power fields, HPF). A single patient with distant metastasis harboured a solid-predominant tumour showing nuclear grade 2–3, brisk mitoses (6/10 HPF), and multifocal necrosis. All tumours demonstrated diffuse S100 and pan-TRK expression. Oestrogen and/or progesterone receptor staining was observed in 16 of 29 cases (2–30% of tumour cells), and all were HER2 negative or low (0–1+). Ki-67 ranged from 3% to 20% (mean 7%). Fluorescence <i>in situ</i> hybridisation (FISH) was positive in 17 of 17 tested tumours (14 <i>ETV6-NTRK3</i> dual-fusion; 3 <i>NTRK3</i> break-apart). In the metastatic case, RNA sequencing confirmed canonical <i>ETV6-NTRK3</i> fusion, while targeted DNA sequencing identified additional variants of uncertain significance (VUS) – <i>RANBP2</i> p.S1843R, <i>NUP107</i> p.K382Q, <i>NCOR1</i> p.A1947V (missense), and <i>PREX2</i> p.G606G (synonymous). All patients underwent surgery, and 14 received adjuvant chemotherapy. During follow-up ranging from 6 to 135 months (median 76), one patient developed lung metastasis and was alive with disease at 88 months; the remaining 28 patients were alive without recurrence or metastasis. In summary, SBC is typically indolent and characterised by <i>ETV6-NTRK3</i> rearrangement with diffuse pan-TRK/S100 positivity. A solid-predominant pattern with increased cytological atypia, mitotic activity, and necrosis may indicate aggressive potential. Routine <i>NTRK</i> testing supports diagnosis and may help identify patients who could benefit from TRK-inhibitor therapy in advanced disease.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70060","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145453725","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}
Xiaoli Su, Jiawen Wu, Pingling Wang, Liwen Hu, Yupeng Chen, Caihong Ren, Fangling Song, Hangrui Lin, Chenlin Ji, Yucheng xie, Xingfu Wang
Adult diffuse gliomas with FGFR3-TACC3 (F3T3) fusion are rare and highly heterogeneous central nervous system (CNS) tumors. Current research on the biological behavior of these tumors is limited, especially regarding whether histopathologically low-grade tumors are indolent or represent early-stage high-grade tumors, thereby posing challenges for grading. In this single-center study of 17 patients with adult F3T3 fusion diffuse gliomas (F3T3 gliomas), both low- and high-grade F3T3 gliomas presented distinctive recurrent histopathological features, such as oligodendrocyte-like cells, branched vessels and frequent calcifications. Molecularly, TERT promoter mutations and 7+/10− chromosomal alterations were common; one patient with recurrent glioma with histopathological features of polymorphous low-grade neuroepithelial tumor of the young (PLNTY) had additional CDK4 and MDM2 amplifications. Methylation profiling of 3 samples revealed varied results. A patient whose tumor had histopathological features consistent with PLNTY and a methylation subtype classified as the mesenchymal subtype of glioblastoma (GBM) experienced tumor recurrence 8 months after surgery. After 5–62 months of follow-up, seven patients relapsed, and six died; the primary tumors of the three patients with recurrence presented histopathological characteristics of low-grade glioma (LGG). GBM patients had worse overall survival than LGG patients (p = 0.035) but similar progression-free survival (p = 0.47), indicating that LGG patients may experience recurrence. For adults with tumors with histopathological features of PLNTY, further molecular and methylation analyses are needed for grading. If TERT promoter mutations are present, even with a PLNTY methylation profile, these tumors can still exhibit the biological behavior of high-grade gliomas.
{"title":"Diffuse glioma with FGFR3-TACC3 fusion in adults is not a homogenous clinicopathological and molecular entity","authors":"Xiaoli Su, Jiawen Wu, Pingling Wang, Liwen Hu, Yupeng Chen, Caihong Ren, Fangling Song, Hangrui Lin, Chenlin Ji, Yucheng xie, Xingfu Wang","doi":"10.1002/2056-4538.70057","DOIUrl":"10.1002/2056-4538.70057","url":null,"abstract":"<p>Adult diffuse gliomas with <i>FGFR3-TACC3</i> (F3T3) fusion are rare and highly heterogeneous central nervous system (CNS) tumors. Current research on the biological behavior of these tumors is limited, especially regarding whether histopathologically low-grade tumors are indolent or represent early-stage high-grade tumors, thereby posing challenges for grading. In this single-center study of 17 patients with adult <i>F3T3</i> fusion diffuse gliomas (F3T3 gliomas), both low- and high-grade F3T3 gliomas presented distinctive recurrent histopathological features, such as oligodendrocyte-like cells, branched vessels and frequent calcifications. Molecularly, <i>TERT</i> promoter mutations and 7+/10− chromosomal alterations were common; one patient with recurrent glioma with histopathological features of polymorphous low-grade neuroepithelial tumor of the young (PLNTY) had additional <i>CDK4</i> and <i>MDM2</i> amplifications. Methylation profiling of 3 samples revealed varied results. A patient whose tumor had histopathological features consistent with PLNTY and a methylation subtype classified as the mesenchymal subtype of glioblastoma (GBM) experienced tumor recurrence 8 months after surgery. After 5–62 months of follow-up, seven patients relapsed, and six died; the primary tumors of the three patients with recurrence presented histopathological characteristics of low-grade glioma (LGG). GBM patients had worse overall survival than LGG patients (<i>p</i> = 0.035) but similar progression-free survival (<i>p</i> = 0.47), indicating that LGG patients may experience recurrence. For adults with tumors with histopathological features of PLNTY, further molecular and methylation analyses are needed for grading. If <i>TERT</i> promoter mutations are present, even with a PLNTY methylation profile, these tumors can still exhibit the biological behavior of high-grade gliomas.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70057","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145402356","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}
The field of gynecologic pathology is undergoing a transformative change. The translation of molecular findings from large-scale genomic studies into clinical practice has been facilitated by robust surrogate assays, particularly immunohistochemistry (IHC). These tools provide scalable molecular proxies with short turnaround times, enabling molecular subclassification and conclusive prognostic biomarker studies. This commentary highlights how prognostic markers now refine diagnoses in challenging borderline areas, where an abnormal result can help exclude benign or precursor entities. However, the utility of these markers is highly context-dependent, modified by histotype and molecular subtype. Furthermore, the confident diagnosis of rare entities and the study of their precursors have been advanced by defining specific molecular and IHC profiles, opening new avenues for research and therapy. The integration of morphology with molecular features is increasing the robustness of diagnoses and dictating oncology management as never before.
{"title":"Context is key: how diagnostic and prognostic markers are reshaping gynecologic pathology","authors":"Martin Köbel, Anthony N Karnezis","doi":"10.1002/2056-4538.70058","DOIUrl":"10.1002/2056-4538.70058","url":null,"abstract":"<p>The field of gynecologic pathology is undergoing a transformative change. The translation of molecular findings from large-scale genomic studies into clinical practice has been facilitated by robust surrogate assays, particularly immunohistochemistry (IHC). These tools provide scalable molecular proxies with short turnaround times, enabling molecular subclassification and conclusive prognostic biomarker studies. This commentary highlights how prognostic markers now refine diagnoses in challenging borderline areas, where an abnormal result can help exclude benign or precursor entities. However, the utility of these markers is highly context-dependent, modified by histotype and molecular subtype. Furthermore, the confident diagnosis of rare entities and the study of their precursors have been advanced by defining specific molecular and IHC profiles, opening new avenues for research and therapy. The integration of morphology with molecular features is increasing the robustness of diagnoses and dictating oncology management as never before.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70058","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145402371","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}
Bone and soft tissue sarcomas are a diagnostic challenge due to their histological complexity, variety, and mutational heterogeneity. This has led to continued refinements in classification and diagnostic criteria, presented and soon to be updated by the WHO. This commentary features articles published in this journal and elsewhere that have ultilised pan-sarcoma datasets and artificial intelligence-based classifiers. A notable entry is the DKFZ Sarcoma Classifier, which is based on methylation profiles and available for use via a commercial platform. This work has created hope that one day a comprehensive sarcoma classifier will be available, though it is also a reminder of the many challenges apparent in studying rare cancer types.
{"title":"The diagnostic landscape of bone and soft tissue sarcoma: the pan-sarcoma era and AI-driven classification","authors":"William CH Cross","doi":"10.1002/2056-4538.70054","DOIUrl":"10.1002/2056-4538.70054","url":null,"abstract":"<p>Bone and soft tissue sarcomas are a diagnostic challenge due to their histological complexity, variety, and mutational heterogeneity. This has led to continued refinements in classification and diagnostic criteria, presented and soon to be updated by the WHO. This commentary features articles published in this journal and elsewhere that have ultilised pan-sarcoma datasets and artificial intelligence-based classifiers. A notable entry is the DKFZ Sarcoma Classifier, which is based on methylation profiles and available for use <i>via</i> a commercial platform. This work has created hope that one day a comprehensive sarcoma classifier will be available, though it is also a reminder of the many challenges apparent in studying rare cancer types.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12541240/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145349359","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}
Suzanne Parry, Lila Zabaglo, Abeer M Shaaban, Andrew Dodson
Inter-observer concordance data for the HER2 category as assessed by a group of 16 specialist breast pathologists on 50 diagnostic core biopsies was compared with that produced by digital image analysis (DIA) using the HER2 APP, CE2797 (VP APP; Visiopharm, Hoersholm, Denmark). Comparing pathologists' consensus scores and DIA scores, 36 cases (73.5%) agreed. Fleiss' kappa statistic was 0.433 (indicative of moderate agreement). Cohen's weighted kappa was used to compare the scores of individual raters to consensus scores; for all 50 cases the kappa scores had a range between 0.412 and 0.854; the VP APP was ranked 12th of 17 raters (kappa score 0.638 indicating substantial agreement). Results for HER2-low cases (N = 44) showed a kappa score range of 0.295 to 0.823; the VP APP ranked 12th of 17 (score 0.535 indicating moderate agreement). For high agreement cases the kappa score range was 0.664 to 1.000 for all HER2 scores (N = 24) and the VP APP scored 0.916 (indicating almost perfect agreement). For the HER2-low scores (N = 20), the kappa score range was 0.506–1.000 and the VP APP scored 0.860 (almost perfect agreement). DIA of the proportions of tumour cells showing expression within each of the HER2 categories demonstrated that the majority of cases showing a low level of agreement between pathologists showed heterogeneity and/or a level of expression close to a cut-point for decision making. This study demonstrates that the VP APP produces results that are extremely well-aligned to those of expert pathologists in cases with good overall agreement, and in difficult cases its reproducibility will outperform that of the visual scorer. The results also suggest that use of the VP APP has the potential to reduce the proportion of cases referred for gene amplification testing by reducing the number of cases incorrectly classified as HER2 2+.
{"title":"Inter-rater agreement of HER2-low scores between expert breast pathologists and the Visiopharm digital image analysis application (HER2 APP, CE2797)","authors":"Suzanne Parry, Lila Zabaglo, Abeer M Shaaban, Andrew Dodson","doi":"10.1002/2056-4538.70051","DOIUrl":"10.1002/2056-4538.70051","url":null,"abstract":"<p>Inter-observer concordance data for the HER2 category as assessed by a group of 16 specialist breast pathologists on 50 diagnostic core biopsies was compared with that produced by digital image analysis (DIA) using the HER2 APP, CE2797 (VP APP; Visiopharm, Hoersholm, Denmark). Comparing pathologists' consensus scores and DIA scores, 36 cases (73.5%) agreed. Fleiss' kappa statistic was 0.433 (indicative of moderate agreement). Cohen's weighted kappa was used to compare the scores of individual raters to consensus scores; for all 50 cases the kappa scores had a range between 0.412 and 0.854; the VP APP was ranked 12th of 17 raters (kappa score 0.638 indicating substantial agreement). Results for HER2-low cases (<i>N</i> = 44) showed a kappa score range of 0.295 to 0.823; the VP APP ranked 12th of 17 (score 0.535 indicating moderate agreement). For high agreement cases the kappa score range was 0.664 to 1.000 for all HER2 scores (<i>N</i> = 24) and the VP APP scored 0.916 (indicating almost perfect agreement). For the HER2-low scores (<i>N</i> = 20), the kappa score range was 0.506–1.000 and the VP APP scored 0.860 (almost perfect agreement). DIA of the proportions of tumour cells showing expression within each of the HER2 categories demonstrated that the majority of cases showing a low level of agreement between pathologists showed heterogeneity and/or a level of expression close to a cut-point for decision making. This study demonstrates that the VP APP produces results that are extremely well-aligned to those of expert pathologists in cases with good overall agreement, and in difficult cases its reproducibility will outperform that of the visual scorer. The results also suggest that use of the VP APP has the potential to reduce the proportion of cases referred for gene amplification testing by reducing the number of cases incorrectly classified as HER2 2+.</p>","PeriodicalId":48612,"journal":{"name":"Journal of Pathology Clinical Research","volume":"11 6","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pathsocjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/2056-4538.70051","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145309572","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}