Pub Date : 2025-12-18DOI: 10.1016/j.labinv.2025.104265
Jiang Shi, Lin Jiang, ShiWang Yuan, Peng Chen, Tao Li
{"title":"Letter to the Editor (Correspondence): Re: Prognostic Value of PARP1 and PARP2 Copy Number Alterations in Prostate Cancer","authors":"Jiang Shi, Lin Jiang, ShiWang Yuan, Peng Chen, Tao Li","doi":"10.1016/j.labinv.2025.104265","DOIUrl":"10.1016/j.labinv.2025.104265","url":null,"abstract":"","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":"106 1","pages":"Article 104265"},"PeriodicalIF":4.2,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145786480","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 : 2025-12-16DOI: 10.1016/j.labinv.2025.104272
Aakash Tripathi, Asim Waqas, Kavya Venkatesan, Ehsan Ullah, Asma Khan, Farah Khalil, Wei-Shen Chen, Zarifa Gahramanli Ozturk, Daryoush Saeed-Vafa, Marilyn M Bui, Matthew B Schabath, Ghulam Rasool
Surgical pathology reports provide essential diagnostic information critical for cancer staging, treatment planning, and cancer registry documentation. However, their writing styles and formats vary widely, reflecting each pathologist's stylistic choices, institutional norms, and inherited practices from residency training. When performing large-scale data analysis, this unstructured nature and variability across tumor types and institutions pose significant hurdles for automated data extraction. To overcome these challenges, we present a consensus-driven, reasoning-based framework that adapts multiple locally deployed large language models (LLMs) to extract both standard diagnostic variables (site, laterality, histology, stage, grade, and behavior) and organ-specific biomarkers. Each LLM generates structured outputs, accompanied by justifications, which are subsequently evaluated for accuracy and coherence by three separate reasoning models (DeepSeek-R1-large, Qwen3-32B, and QWQ-32B). Final consensus values are determined through aggregation. Board-certified pathologists conducted expert validation. This framework was applied to over 6,100 pathology reports from The Cancer Genome Atlas (TCGA) spanning 10 organ systems and 510 reports from Moffitt Cancer Center. For the TCGA dataset, automated evaluation demonstrated mean accuracy of 84.9%±7.3%, with histology (88%), site (87%), stage and behavior (84%) showing the highest extraction accuracy averaged across all models. Expert review of randomly selected 138 reports confirmed high agreement for behavior (100.0%), histology (99%), grade (96%), and site (95%) in the TCGA dataset, with slightly lower performance for stage (89%) and laterality (88%). In Moffitt Cancer Center reports (brain, breast, and lung), accuracy remained high (88.2%±7.2%), with behavior (99%), histology (97%), laterality (96%), grade (94%), and site (93%) achieving strong agreement. Biomarker extraction achieved 70.6%±7.9% overall accuracy, with TP53 (84%) on brain tumor, Ki-67 (68%) on breast cancer, and ROS1 (82%) on lung cancer showing highest accuracy. Inter-evaluator agreement analysis revealed high concordance (correlations ≥ 0.93) across the three evaluation models. Statistical analyses revealed significant main effects of model type (F=1716.82, p<0.001), variable (F=3236.68, p<0.001), and organ system (F=1946.43, p<0.001), as well as model × variable × organ interactions (F=24.74, p<0.001), emphasizing the role of clinical context in model performance. These results highlight the potential of stratified, multi-organ evaluation frameworks with multi-evaluator consensus in LLM benchmarking for clinical applications. Overall, this consensus-based approach demonstrates that locally deployed LLMs can provide a transparent, accurate, and auditable solution for integration into real-world pathology workflows such as synoptic reporting and cancer registry abstraction.
外科病理报告为癌症分期、治疗计划和癌症登记文件提供了重要的诊断信息。然而,他们的写作风格和格式差异很大,反映了每个病理学家的风格选择、制度规范和住院医师培训的继承实践。在进行大规模数据分析时,肿瘤类型和机构之间的非结构化性质和可变性对自动数据提取构成了重大障碍。为了克服这些挑战,我们提出了一个共识驱动的、基于推理的框架,该框架适应多个本地部署的大型语言模型(llm),以提取标准诊断变量(部位、侧侧性、组织学、分期、分级和行为)和器官特异性生物标志物。每个LLM生成结构化输出,伴随着证明,随后通过三个独立的推理模型(DeepSeek-R1-large, Qwen3-32B和QWQ-32B)评估其准确性和一致性。最终的共识值是通过聚合确定的。委员会认证的病理学家进行了专家验证。该框架应用于来自癌症基因组图谱(TCGA)的6100多份病理报告,涵盖10个器官系统和莫菲特癌症中心的510份报告。对于TCGA数据集,自动评估的平均准确率为84.9%±7.3%,其中组织学(88%)、部位(87%)、阶段和行为(84%)在所有模型中显示出最高的平均提取准确率。专家对随机选择的138份报告进行了审查,证实TCGA数据集中的行为(100.0%)、组织学(99%)、分级(96%)和部位(95%)的一致性较高,而分期(89%)和侧侧性(88%)的一致性稍低。在Moffitt癌症中心的报告(脑、乳腺和肺)中,准确率仍然很高(88.2%±7.2%),行为(99%)、组织学(97%)、侧侧(96%)、分级(94%)和部位(93%)达到了高度一致。生物标志物提取的总体准确率为70.6%±7.9%,其中脑肿瘤TP53(84%)、乳腺癌Ki-67(68%)和肺癌ROS1(82%)的准确率最高。评价者间一致性分析显示三个评价模型的一致性较高(相关性≥0.93)。统计分析显示模型类型的主效应显著(F=1716.82, p
{"title":"Using consensus-based reasoning and large language models to extract structured data from surgical pathology reports.","authors":"Aakash Tripathi, Asim Waqas, Kavya Venkatesan, Ehsan Ullah, Asma Khan, Farah Khalil, Wei-Shen Chen, Zarifa Gahramanli Ozturk, Daryoush Saeed-Vafa, Marilyn M Bui, Matthew B Schabath, Ghulam Rasool","doi":"10.1016/j.labinv.2025.104272","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104272","url":null,"abstract":"<p><p>Surgical pathology reports provide essential diagnostic information critical for cancer staging, treatment planning, and cancer registry documentation. However, their writing styles and formats vary widely, reflecting each pathologist's stylistic choices, institutional norms, and inherited practices from residency training. When performing large-scale data analysis, this unstructured nature and variability across tumor types and institutions pose significant hurdles for automated data extraction. To overcome these challenges, we present a consensus-driven, reasoning-based framework that adapts multiple locally deployed large language models (LLMs) to extract both standard diagnostic variables (site, laterality, histology, stage, grade, and behavior) and organ-specific biomarkers. Each LLM generates structured outputs, accompanied by justifications, which are subsequently evaluated for accuracy and coherence by three separate reasoning models (DeepSeek-R1-large, Qwen3-32B, and QWQ-32B). Final consensus values are determined through aggregation. Board-certified pathologists conducted expert validation. This framework was applied to over 6,100 pathology reports from The Cancer Genome Atlas (TCGA) spanning 10 organ systems and 510 reports from Moffitt Cancer Center. For the TCGA dataset, automated evaluation demonstrated mean accuracy of 84.9%±7.3%, with histology (88%), site (87%), stage and behavior (84%) showing the highest extraction accuracy averaged across all models. Expert review of randomly selected 138 reports confirmed high agreement for behavior (100.0%), histology (99%), grade (96%), and site (95%) in the TCGA dataset, with slightly lower performance for stage (89%) and laterality (88%). In Moffitt Cancer Center reports (brain, breast, and lung), accuracy remained high (88.2%±7.2%), with behavior (99%), histology (97%), laterality (96%), grade (94%), and site (93%) achieving strong agreement. Biomarker extraction achieved 70.6%±7.9% overall accuracy, with TP53 (84%) on brain tumor, Ki-67 (68%) on breast cancer, and ROS1 (82%) on lung cancer showing highest accuracy. Inter-evaluator agreement analysis revealed high concordance (correlations ≥ 0.93) across the three evaluation models. Statistical analyses revealed significant main effects of model type (F=1716.82, p<0.001), variable (F=3236.68, p<0.001), and organ system (F=1946.43, p<0.001), as well as model × variable × organ interactions (F=24.74, p<0.001), emphasizing the role of clinical context in model performance. These results highlight the potential of stratified, multi-organ evaluation frameworks with multi-evaluator consensus in LLM benchmarking for clinical applications. Overall, this consensus-based approach demonstrates that locally deployed LLMs can provide a transparent, accurate, and auditable solution for integration into real-world pathology workflows such as synoptic reporting and cancer registry abstraction.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104272"},"PeriodicalIF":4.2,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145781600","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}
Intestinal epithelial barrier loss has been suggested as a pathogenic factor in Hirschsprung-associated enterocolitis (HAEC) and intestinal dysfunction in children with Hirschsprung disease (HD), but there is a paucity of comprehensive studies on the tight junction proteins that regulate paracellular permeability in this population. This case-control study aimed to determine if colonic epithelial tight junction protein expression is altered in children with HD. We use quantitative immunofluorescence microscopy to assess the expression of tight junction proteins (claudins 1, 2, 3, 4, 7, 15; ZO-1, ZO-2, occludin) in 29 children with HD who underwent surgical reconstruction and 16 controls who underwent transmural colorectal surgical resection for other etiologies between January 2015 and December 2021. We found that the expression of claudin-2, claudin-15, and occludin was reduced in both ganglionic and aganglionic colon specimens from children with HD compared to controls. Expression of other tight junction proteins did not differ between the groups. Together with previous studies, these data suggest that decreased expression of paracellular Na+ and water channel-forming claudin-2 and claudin-15 may limit lumenal hydration, enhance fecal stasis, and promote dysbiosis. Conversely, occludin downregulation can increase paracellular macromolecular flux, but also limit epithelial sensitivity to apoptotic stimuli. Together, these changes in expression of claudins 2 and 15, as well as occludin, may promote intestinal dysfunction and contribute to HAEC pathogenesis following surgical reconstruction.
{"title":"Tight Junction Defects in Aganglionic and Ganglionic Colon in Children With Hirschsprung Disease.","authors":"Lorena Rincon-Cruz, Leah Froehle, Shabnam Abhati, Jeffrey Goldsmith, Jerrold Turner, Prathima Nandivada","doi":"10.1016/j.labinv.2025.104271","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104271","url":null,"abstract":"<p><p>Intestinal epithelial barrier loss has been suggested as a pathogenic factor in Hirschsprung-associated enterocolitis (HAEC) and intestinal dysfunction in children with Hirschsprung disease (HD), but there is a paucity of comprehensive studies on the tight junction proteins that regulate paracellular permeability in this population. This case-control study aimed to determine if colonic epithelial tight junction protein expression is altered in children with HD. We use quantitative immunofluorescence microscopy to assess the expression of tight junction proteins (claudins 1, 2, 3, 4, 7, 15; ZO-1, ZO-2, occludin) in 29 children with HD who underwent surgical reconstruction and 16 controls who underwent transmural colorectal surgical resection for other etiologies between January 2015 and December 2021. We found that the expression of claudin-2, claudin-15, and occludin was reduced in both ganglionic and aganglionic colon specimens from children with HD compared to controls. Expression of other tight junction proteins did not differ between the groups. Together with previous studies, these data suggest that decreased expression of paracellular Na<sup>+</sup> and water channel-forming claudin-2 and claudin-15 may limit lumenal hydration, enhance fecal stasis, and promote dysbiosis. Conversely, occludin downregulation can increase paracellular macromolecular flux, but also limit epithelial sensitivity to apoptotic stimuli. Together, these changes in expression of claudins 2 and 15, as well as occludin, may promote intestinal dysfunction and contribute to HAEC pathogenesis following surgical reconstruction.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104271"},"PeriodicalIF":4.2,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145757041","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 : 2025-12-02DOI: 10.1016/j.labinv.2025.104269
Miguel Otero, Irene Lorenzo Gomez, Takuya Sakamoto, Yu Okuno, Chelsea Kenvisay, Merissa Olmer, Hannah Swahn, Dana E Orange, Bella Mehta, Fuadur Omi, Edward F DiCarlo, Daniel C Ramirez, Alia Obeidat, Anne-Marie Malfait, Priya Kulkarni, Robert P Dalton, Muhammad Abbas, Yenisel Cruz-Almeida, Kyle D Allen, Nele A Haelterman, Martin K Lotz
All knee joint tissues undergo aging- and osteoarthritis (OA)-associated changes, but our understanding of the knee as an organ, and the tissue crosstalk in homeostasis, aging and OA is limited. The emergence of molecular profiling imaging technologies now enables comprehensive profiling of joint tissues to address these knowledge gaps. Successful application of these novel technologies requires a precise clinical diagnosis, and a rigorous and consistent definition of tissue-specific variables, including documentation of the regions of interest selected for macroscopic, histological, cellular, and molecular characterization. Macroscopic and histological scoring systems represent a benchmark for the interpretation of cellular and molecular analyses. Thus, standardizing these systems is essential to minimize experimental variability. Currently, most joint tissues lack a universally accepted scoring system, and various histological features are selected and quantified using different methods, limiting comparability and reproducibility across studies. Here, we review current methods, discuss limitations, and propose new approaches based on features that should be consistently evaluated across tissue types to overcome these caveats.
{"title":"The Human Knee as An Organ: Joint Tissue Collection, Processing and Scoring for Multimodal Analyses.","authors":"Miguel Otero, Irene Lorenzo Gomez, Takuya Sakamoto, Yu Okuno, Chelsea Kenvisay, Merissa Olmer, Hannah Swahn, Dana E Orange, Bella Mehta, Fuadur Omi, Edward F DiCarlo, Daniel C Ramirez, Alia Obeidat, Anne-Marie Malfait, Priya Kulkarni, Robert P Dalton, Muhammad Abbas, Yenisel Cruz-Almeida, Kyle D Allen, Nele A Haelterman, Martin K Lotz","doi":"10.1016/j.labinv.2025.104269","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104269","url":null,"abstract":"<p><p>All knee joint tissues undergo aging- and osteoarthritis (OA)-associated changes, but our understanding of the knee as an organ, and the tissue crosstalk in homeostasis, aging and OA is limited. The emergence of molecular profiling imaging technologies now enables comprehensive profiling of joint tissues to address these knowledge gaps. Successful application of these novel technologies requires a precise clinical diagnosis, and a rigorous and consistent definition of tissue-specific variables, including documentation of the regions of interest selected for macroscopic, histological, cellular, and molecular characterization. Macroscopic and histological scoring systems represent a benchmark for the interpretation of cellular and molecular analyses. Thus, standardizing these systems is essential to minimize experimental variability. Currently, most joint tissues lack a universally accepted scoring system, and various histological features are selected and quantified using different methods, limiting comparability and reproducibility across studies. Here, we review current methods, discuss limitations, and propose new approaches based on features that should be consistently evaluated across tissue types to overcome these caveats.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104269"},"PeriodicalIF":4.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678154","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 : 2025-11-28DOI: 10.1016/j.labinv.2025.104268
David Pérez-Parra, Fátima Postigo-Corrales, Andreia Filipa Cruz Santos, Alberto Sánchez-Espinosa, Jesús Acosta-Ortega, Raúl Carrillo-Vicente, María Dolores López-Abellán, Pablo Conesa-Zamora, Ginés Luengo-Gil, Ana María Hurtado-López
Triple-negative breast cancer (TNBC) represents 10-15% of all breast cancer cases, predominantly affecting younger women. Due to the absence of hormone receptors and HER2 expression, TNBC lacks effective targeted therapies, resulting in poor prognosis, with 5-year survival rates ranging from 91% for localized disease to 12% for metastatic disease. Fascin, encoded by the FSCN1 gene, is overexpressed in 88% of TNBC cases and promotes tumor invasion, metastasis, and chemotherapy resistance. This study explored fascin as a prognostic marker and therapeutic target by analyzing its expression in the largest TNBC cohort to date and assessing the effects of imipramine, an antidepressant that acts as a fascin inhibitor, on TNBC and luminal breast cancer cell lines. In a retrospective cohort of 145 TNBC patients, fascin expression in tumor cells and stromal myofibroblasts correlated with high histological grade, Ki67 >30%, and BCL-2 overexpression in myofibroblasts, as well as with higher chemotherapeutic response rates in the surgical setting. Fascin expression in stromal myofibroblasts has been identified as an independent predictive marker of chemotherapeutic response and as a prognostic factor for improved survival in patients undergoing neoadjuvant chemotherapy. In vitro, imipramine significantly reduced FSCN1 expression and impaired cell migration in TNBC (MDA-MB-231) and luminal (MCF7) cell lines, with stronger effects on TNBC. These findings highlight the dual role of fascin in tumor cells and the tumor microenvironment (TME), and reinforce its potential as a biomarker for personalized TNBC therapies. Ongoing clinical trials, including HITCLIF, are exploring the efficacy of imipramine in patients with fascin-overexpressing cancers, paving the way for targeted treatment strategies.
{"title":"Characterization and Prognostic Impact of Fascin Expression in the Tumor Microenvironment of Triple-Negative Breast Cancer: Clues for a Tailored Therapy.","authors":"David Pérez-Parra, Fátima Postigo-Corrales, Andreia Filipa Cruz Santos, Alberto Sánchez-Espinosa, Jesús Acosta-Ortega, Raúl Carrillo-Vicente, María Dolores López-Abellán, Pablo Conesa-Zamora, Ginés Luengo-Gil, Ana María Hurtado-López","doi":"10.1016/j.labinv.2025.104268","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104268","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) represents 10-15% of all breast cancer cases, predominantly affecting younger women. Due to the absence of hormone receptors and HER2 expression, TNBC lacks effective targeted therapies, resulting in poor prognosis, with 5-year survival rates ranging from 91% for localized disease to 12% for metastatic disease. Fascin, encoded by the FSCN1 gene, is overexpressed in 88% of TNBC cases and promotes tumor invasion, metastasis, and chemotherapy resistance. This study explored fascin as a prognostic marker and therapeutic target by analyzing its expression in the largest TNBC cohort to date and assessing the effects of imipramine, an antidepressant that acts as a fascin inhibitor, on TNBC and luminal breast cancer cell lines. In a retrospective cohort of 145 TNBC patients, fascin expression in tumor cells and stromal myofibroblasts correlated with high histological grade, Ki67 >30%, and BCL-2 overexpression in myofibroblasts, as well as with higher chemotherapeutic response rates in the surgical setting. Fascin expression in stromal myofibroblasts has been identified as an independent predictive marker of chemotherapeutic response and as a prognostic factor for improved survival in patients undergoing neoadjuvant chemotherapy. In vitro, imipramine significantly reduced FSCN1 expression and impaired cell migration in TNBC (MDA-MB-231) and luminal (MCF7) cell lines, with stronger effects on TNBC. These findings highlight the dual role of fascin in tumor cells and the tumor microenvironment (TME), and reinforce its potential as a biomarker for personalized TNBC therapies. Ongoing clinical trials, including HITCLIF, are exploring the efficacy of imipramine in patients with fascin-overexpressing cancers, paving the way for targeted treatment strategies.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104268"},"PeriodicalIF":4.2,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145648862","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 : 2025-11-25DOI: 10.1016/j.labinv.2025.104267
Zehong Zhang , Yuan’e Lian , Yijuan Wu , Zhongchang Weng , Jiaying Ye , Siqin Zhang , Lianhuang Li , Yannan Bai
The prognostic role of tumor-infiltrating lymphocytes (TILs) in hepatocellular carcinoma (HCC) remains elusive. This study explored the association between TIL aggregation in early-stage HCC tissues and patient survival. Individual patient-level data from 353 patients diagnosed with HCC who underwent radical hepatectomy were retrospectively analyzed. TIL aggregation was analyzed using an artificial neural network algorithm to define the immune phenotypes (IPs) within hematoxylin and eosin–stained whole-slide images. The association between TIL aggregation and survival was compared among IPs, and an optimized TIL phenotype was validated with clinical variables. Among the 8 IP features measured using the artificial neural network algorithm, TIL densityK50, a metric generated by an assigned density of each cell based on its distance with the 50 nearest lymphocytes, was associated with improved disease-free survival in univariate (hazard ratio, 0.282; 95% CI, 0.181-0.439; P < .0001) and multivariate analyses (hazard ratio, 0.315; 95% CI, 0.115-0.860; P = .024) after adjusting for clinical factors. When TIL densityK50 is combined with clinical factors (TIL-clinical–integrated [TCI] model), area under the curve performance for disease-free survival and overall survival was improved to 0.798 and 0.781 in the training cohort and 0.759 and 0.723 in the validation cohort, respectively. Furthermore, subgroup analyses indicated that its predictive value is particularly strong for very late recurrence and survival up to <8 years. Abundant TILs in HCC tissue are associated with reduced late recurrence risk and improved survival, suggesting a potential time-dependent prognostic role of TILs in early-stage HCC.
肿瘤浸润淋巴细胞(til)在肝细胞癌(HCC)中的预后作用仍然难以捉摸。本研究探讨了早期HCC组织中TIL聚集与患者生存之间的关系。回顾性分析了353例接受根治性肝切除术的HCC患者的个体数据。使用人工神经网络(ANN)算法分析TIL聚集情况,以确定苏木精和伊红染色全片图像中的免疫表型(IPs)。比较了不同IPs患者TIL聚集与生存之间的关系,并用临床变量验证了优化后的TIL表型。在使用人工神经网络算法测量的8个IP特征中,TIL密度k50(由每个细胞与最近的50个淋巴细胞的距离分配密度生成的度量)在单因素分析(风险比[HR], 0.282; 95%置信区间[CI],CI 0.181-0.439; P < 0.0001)和多因素分析(HR, 0.315; 95% CI, 0.115-0.860; P = 0.024)中与改善无病生存(DFS)相关。当TIL密度k50与临床因素(TIL- clinial - integrated model, TCI-model)相结合时,训练组DFS和总生存的AUC性能分别提高至0.798和0.781,验证组为0.759和0.723。此外,亚组分析表明,它对晚期复发和生存期< 8年的预测价值尤其强。HCC组织中大量的TILs与晚期复发风险降低和生存率提高相关,提示TILs在早期HCC中具有潜在的时间依赖性预后作用。
{"title":"Tumor-Infiltrating Lymphocyte Aggregation Refines Prognosis in Patients With Hepatocellular Carcinoma","authors":"Zehong Zhang , Yuan’e Lian , Yijuan Wu , Zhongchang Weng , Jiaying Ye , Siqin Zhang , Lianhuang Li , Yannan Bai","doi":"10.1016/j.labinv.2025.104267","DOIUrl":"10.1016/j.labinv.2025.104267","url":null,"abstract":"<div><div>The prognostic role of tumor-infiltrating lymphocytes (TILs) in hepatocellular carcinoma (HCC) remains elusive. This study explored the association between TIL aggregation in early-stage HCC tissues and patient survival. Individual patient-level data from 353 patients diagnosed with HCC who underwent radical hepatectomy were retrospectively analyzed. TIL aggregation was analyzed using an artificial neural network algorithm to define the immune phenotypes (IPs) within hematoxylin and eosin–stained whole-slide images. The association between TIL aggregation and survival was compared among IPs, and an optimized TIL phenotype was validated with clinical variables. Among the 8 IP features measured using the artificial neural network algorithm, TIL density<sub>K50</sub>, a metric generated by an assigned density of each cell based on its distance with the 50 nearest lymphocytes, was associated with improved disease-free survival in univariate (hazard ratio, 0.282; 95% CI, 0.181-0.439; <em>P</em> < .0001) and multivariate analyses (hazard ratio, 0.315; 95% CI, 0.115-0.860; <em>P</em> = .024) after adjusting for clinical factors. When TIL density<sub>K50</sub> is combined with clinical factors (TIL-clinical–integrated [TCI] model), area under the curve performance for disease-free survival and overall survival was improved to 0.798 and 0.781 in the training cohort and 0.759 and 0.723 in the validation cohort, respectively. Furthermore, subgroup analyses indicated that its predictive value is particularly strong for very late recurrence and survival up to <8 years. Abundant TILs in HCC tissue are associated with reduced late recurrence risk and improved survival, suggesting a potential time-dependent prognostic role of TILs in early-stage HCC.</div></div>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":"106 1","pages":"Article 104267"},"PeriodicalIF":4.2,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145635287","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 : 2025-11-25DOI: 10.1016/j.labinv.2025.104266
Chencheng Li , Xixi Liu , Weiguang Zhang , Jing Yang , Xiaoli Zhang , Reaila Jianati , Pengjun Jiang , Fang Tian , Biqing Chen , Xuejun Zhu
Natural killer T cells (NKT), a unique subset of T lymphocytes, remain incompletely understood in terms of their development and functional regulation. Well-characterized immortalized cell lines are critical tools for investigating NKT cell biology in vitro. We established and maintained a novel NKT cell line, NKT617, derived from the peripheral blood of a patient with aggressive large granular lymphocyte leukemia. Cell morphology was assessed using scanning electron microscopy and transmission electron microscopy. Flow cytometry characterized the cell phenotype, whereas T-cell receptor (TCR) clonal gene rearrangement determined lineage. RNA sequencing compared gene expression profiles of NKT617 with other hematological tumor cell lines, such as natural killer (NK) cells, T cells, B cells, and monocytes. Tumorigenic potential was evaluated via in vitro colony formation assays and in vivo zebrafish xenograft models. NKT617 has been continuously cultured for >2 years, exceeding 100 passages, demonstrating monoclonal immortalization. It proliferates in a low-dose recombinant human interleukin-2–dependent manner and grows in suspension culture as single cells and aggregates. Flow cytometric analysis showed that NKT617 exhibits an NK-cell phenotype CD56+, CD158a+, NKp46+, membrane-bound CD3−, and T-cell features cytoplasmic CD3+, CD2+, CD4+, CD8+, CD45RA+, and CD45RO+, with clonal TCRβ rearrangement. NKT617 was negative for B-cell and myeloid markers and positive for the TCRα chain (Vα24-Jα18), confirming its identity as a type I NKT cell. Transcriptome analysis showed shared markers with NK-cell and T-cell lines but a gene expression profile closer to NK cells. In vitro and in vivo assays confirmed its tumorigenic capacity. We report the first Epstein-Barr virus–negative human type I NKT cell line, NKT617, which offers significant potential for studying NKT cell development and advancing chimeric antigen receptor–based therapies. This cell line serves as a valuable tool for exploring NKT cell biology and developing targeted immunotherapies.
{"title":"Establishment and Multidimensional Characterization of a Novel Epstein-Barr Virus (EBV)–Negative Human Type I Natural Killer T-Cell Line NKT617","authors":"Chencheng Li , Xixi Liu , Weiguang Zhang , Jing Yang , Xiaoli Zhang , Reaila Jianati , Pengjun Jiang , Fang Tian , Biqing Chen , Xuejun Zhu","doi":"10.1016/j.labinv.2025.104266","DOIUrl":"10.1016/j.labinv.2025.104266","url":null,"abstract":"<div><div>Natural killer T cells (NKT), a unique subset of T lymphocytes, remain incompletely understood in terms of their development and functional regulation. Well-characterized immortalized cell lines are critical tools for investigating NKT cell biology in vitro. We established and maintained a novel NKT cell line, NKT617, derived from the peripheral blood of a patient with aggressive large granular lymphocyte leukemia. Cell morphology was assessed using scanning electron microscopy and transmission electron microscopy. Flow cytometry characterized the cell phenotype, whereas T-cell receptor (TCR) clonal gene rearrangement determined lineage. RNA sequencing compared gene expression profiles of NKT617 with other hematological tumor cell lines, such as natural killer (NK) cells, T cells, B cells, and monocytes. Tumorigenic potential was evaluated via in vitro colony formation assays and in vivo zebrafish xenograft models. NKT617 has been continuously cultured for >2 years, exceeding 100 passages, demonstrating monoclonal immortalization. It proliferates in a low-dose recombinant human interleukin-2–dependent manner and grows in suspension culture as single cells and aggregates. Flow cytometric analysis showed that NKT617 exhibits an NK-cell phenotype CD56<sup>+</sup>, CD158a<sup>+</sup>, NKp46<sup>+</sup>, membrane-bound CD3<sup>−</sup>, and T-cell features cytoplasmic CD3<sup>+</sup>, CD2<sup>+</sup>, CD4<sup>+</sup>, CD8<sup>+</sup>, CD45RA<sup>+</sup>, and CD45RO<sup>+</sup>, with clonal TCRβ rearrangement. NKT617 was negative for B-cell and myeloid markers and positive for the TCRα chain (Vα24-Jα18), confirming its identity as a type I NKT cell. Transcriptome analysis showed shared markers with NK-cell and T-cell lines but a gene expression profile closer to NK cells. In vitro and in vivo assays confirmed its tumorigenic capacity. We report the first Epstein-Barr virus–negative human type I NKT cell line, NKT617, which offers significant potential for studying NKT cell development and advancing chimeric antigen receptor–based therapies. This cell line serves as a valuable tool for exploring NKT cell biology and developing targeted immunotherapies.</div></div>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":"106 1","pages":"Article 104266"},"PeriodicalIF":4.2,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145635203","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 : 2025-11-19DOI: 10.1016/j.labinv.2025.104262
Chiara Caraccio , Josie van de Klashorst , Shelby Cherkas , Sara Ancel , Tim Noah Kempchen , Gustavo Vazquez , Yury Goltsev , Yu Xin Wang , Garry P. Nolan , John W. Hickey
Antibody-oligonucleotide conjugates (AOCs) have emerged as versatile tools with applications spanning diagnostics, therapeutics, and high-dimensional imaging. One major application of these is in multiplexed imaging techniques, such as CO-Detection by indEXing, which allow for the visualization of tissue networks at the single-cell level. In this study, we evaluated 4 methods—maleimide-modified, amine-modified, dibenzocyclooctyne (DBCO)-modified, and a site-specific enzyme-based method—to optimize the generation of AOCs for multiplexed imaging applications. Our assessment focused on key performance parameters, including conjugation efficiency, signal brightness, stability, reproducibility, and cost-effectiveness. Each conjugation chemistry proved effective, though the azide chemistry with DBCO oligonucleotides demonstrated more consistent conjugation success and stable signal retention over time. Compared with other protocols, this method produced reliably bright images and offered a more favorable cost profile, as further confirmed in a full-scale CO-Detection by indEXing multiplexed imaging experiment that yielded reproducible spatial data. The observed stability and reproducibility of the DBCO approach suggest that it may help reduce reagent waste and labor costs while facilitating the development of more comprehensive antibody panels. These findings indicate that the DBCO-modified oligonucleotide conjugation method is a valuable option for generating AOCs for multiplexed imaging and target current shortcomings, enabling more consistent, broader, and deeper multiplexed profiling.
{"title":"Comparative Evaluation of Antibody-Oligonucleotide Conjugation Strategies for Multiplexed Imaging Applications","authors":"Chiara Caraccio , Josie van de Klashorst , Shelby Cherkas , Sara Ancel , Tim Noah Kempchen , Gustavo Vazquez , Yury Goltsev , Yu Xin Wang , Garry P. Nolan , John W. Hickey","doi":"10.1016/j.labinv.2025.104262","DOIUrl":"10.1016/j.labinv.2025.104262","url":null,"abstract":"<div><div>Antibody-oligonucleotide conjugates (AOCs) have emerged as versatile tools with applications spanning diagnostics, therapeutics, and high-dimensional imaging. One major application of these is in multiplexed imaging techniques, such as CO-Detection by indEXing, which allow for the visualization of tissue networks at the single-cell level. In this study, we evaluated 4 methods—maleimide-modified, amine-modified, dibenzocyclooctyne (DBCO)-modified, and a site-specific enzyme-based method—to optimize the generation of AOCs for multiplexed imaging applications. Our assessment focused on key performance parameters, including conjugation efficiency, signal brightness, stability, reproducibility, and cost-effectiveness. Each conjugation chemistry proved effective, though the azide chemistry with DBCO oligonucleotides demonstrated more consistent conjugation success and stable signal retention over time. Compared with other protocols, this method produced reliably bright images and offered a more favorable cost profile, as further confirmed in a full-scale CO-Detection by indEXing multiplexed imaging experiment that yielded reproducible spatial data. The observed stability and reproducibility of the DBCO approach suggest that it may help reduce reagent waste and labor costs while facilitating the development of more comprehensive antibody panels. These findings indicate that the DBCO-modified oligonucleotide conjugation method is a valuable option for generating AOCs for multiplexed imaging and target current shortcomings, enabling more consistent, broader, and deeper multiplexed profiling.</div></div>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":"106 1","pages":"Article 104262"},"PeriodicalIF":4.2,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573939","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}
Neuroblastoma (NB) accounts for 10% to 15% of cancer-related deaths among children. In rare cases, NB is associated with opsoclonus-myoclonus syndrome (OMS), a neurological paraneoplastic syndrome. These low-risk (LR) NBs with OMS have a favorable prognosis and an immune-enriched microenvironment. The recruitment of dendritic cells (DCs) and T lymphocytes into tumors is known to be mediated by CCL2 and CXCL12, respectively. We investigated whether intratumor CCL2 and CXCL12 promote DC recruitment within the microenvironment of OMS-associated LR-NBs. We conducted a multicentric retrospective case-control study on 44 pediatric patients with NBs associated or not with OMS. The non-OMS population was subdivided into an LR-NB group versus a high-risk (HR)-NB group, with or without MYCN amplification. Immunohistochemistry was performed using anti-CCL2 and anti-CXCL12 antibodies. DC subpopulations were stained using OPAL spectral multiplex immunofluorescence. Immunostaining scores were established by semiquantitative optical analysis. Gene signatures of DC, as well as CCL2 and CXCL12 mRNA expressions, were also analyzed on a public RNA-Seq data set of NB. We observed a significantly higher protein expression of CCL2 in OMS-associated LR-NBs compared with MYCN-amplified HR-NBs, whereas CXCL12 was not preferentially expressed by any of the NB groups. Furthermore, DC infiltrate was more abundant in OMS-associated LR-NBs compared with any other NB risk groups and positively correlated with CCL2 expression. In silico analysis of public RNA-Seq data demonstrated a higher CCL2 expression in LR-NB with or without OMS compared with HR groups, and that the DC transcriptomic signature was higher in OMS-associated LR-NBs compared with HR-NB groups. We also observed a positive correlation between the DC gene signature and CCL2 or CXCL12 mRNA expression, to a lesser extent. Our results suggest that a DC-prone microenvironment might explain the favorable oncologic outcome of OMS-associated LR-NBs. CCL2 could be a therapeutic target to mobilize DCs in HR-NBs or in tumors characterized by a paucity of tumor-infiltrating DCs.
{"title":"High Expression of CCL2 Correlated With Dendritic Cell Recruitment in Neuroblastomas Associated With Opsoclonus-Myoclonus Syndrome","authors":"Maureen Voirin , Alexia Gazeu , Benoit Dumont , Aurélien Voissière , Léo Jannot , Dominique Plantaz , Nathalie Sturm , Nicolas Ber , Pascal Chastagner , Cécile Picard , Frédérique Dijoud , Christophe Caux , Nathalie Bendriss-Vermare , Hervé Sartelet","doi":"10.1016/j.labinv.2025.104263","DOIUrl":"10.1016/j.labinv.2025.104263","url":null,"abstract":"<div><div>Neuroblastoma (NB) accounts for 10% to 15% of cancer-related deaths among children. In rare cases, NB is associated with opsoclonus-myoclonus syndrome (OMS), a neurological paraneoplastic syndrome. These low-risk (LR) NBs with OMS have a favorable prognosis and an immune-enriched microenvironment. The recruitment of dendritic cells (DCs) and T lymphocytes into tumors is known to be mediated by CCL2 and CXCL12, respectively. We investigated whether intratumor CCL2 and CXCL12 promote DC recruitment within the microenvironment of OMS-associated LR-NBs. We conducted a multicentric retrospective case-control study on 44 pediatric patients with NBs associated or not with OMS. The non-OMS population was subdivided into an LR-NB group versus a high-risk (HR)-NB group, with or without <em>MYCN</em> amplification. Immunohistochemistry was performed using anti-CCL2 and anti-CXCL12 antibodies. DC subpopulations were stained using OPAL spectral multiplex immunofluorescence. Immunostaining scores were established by semiquantitative optical analysis. Gene signatures of DC, as well as <em>CCL2</em> and <em>CXCL12</em> mRNA expressions, were also analyzed on a public RNA-Seq data set of NB. We observed a significantly higher protein expression of CCL2 in OMS-associated LR-NBs compared with <em>MYCN</em>-amplified HR-NBs, whereas CXCL12 was not preferentially expressed by any of the NB groups. Furthermore, DC infiltrate was more abundant in OMS-associated LR-NBs compared with any other NB risk groups and positively correlated with CCL2 expression. In silico analysis of public RNA-Seq data demonstrated a higher <em>CCL2</em> expression in LR-NB with or without OMS compared with HR groups, and that the DC transcriptomic signature was higher in OMS-associated LR-NBs compared with HR-NB groups. We also observed a positive correlation between the DC gene signature and <em>CCL2</em> or <em>CXCL12</em> mRNA expression, to a lesser extent. Our results suggest that a DC-prone microenvironment might explain the favorable oncologic outcome of OMS-associated LR-NBs. CCL2 could be a therapeutic target to mobilize DCs in HR-NBs or in tumors characterized by a paucity of tumor-infiltrating DCs.</div></div>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":"106 1","pages":"Article 104263"},"PeriodicalIF":4.2,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145574001","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 : 2025-11-19DOI: 10.1016/j.labinv.2025.104261
Maaike Anna Hempenius , Mieke C. Zwager , Jeppe Thagaard , Lorian Slagter-Menkema , Henk J. Buikema , Ellis Barbé , Michael A. den Bakker , Marjolein G.J. Heerema , Elisabeth G.E. de Vries , Carolina P. Schröder , Nils A. ’t Hart , Bert van der Vegt
Trastuzumab-deruxtecan, an antibody-drug conjugate targeting human epidermal growth factor receptor 2 (HER2), improves overall survival in patients with breast cancer showing low or ultralow HER2 expression. Differences in HER2 test results have been reported between various HER2 assays and between primary tumors and metastases, but an objective comparison with incorporation of the new HER2-ultralow cutoff values is needed. This study aimed to assess the performance of 4 routine clinical-grade HER2 assays across and between primary tumors and their metastases using digital image analysis. Primary tumors and metastases from 193 patients with breast cancer who participated in the IMaging PAtients for Cancer drug selecTion—Metastatic Breast Cancer trial were incorporated into 6 tissue microarrays. Samples were stained by 4 laboratories using their routine HER2 immunohistochemistry protocols: 4B5 ultraView, 4B5 OptiView, SP3, and HercepTest (DG44). HER2 scores were determined using digital image analysis. The 4 HER2 assays showed significant differences in HER2 status in both primary tumors and metastases. Eighty-five matched primary tumors and metastases were analyzed to investigate concordance in HER2 status. Although no significant differences were found in HER2 scores between primary tumors and metastases for SP3 and both 4B5 assays, DG44 showed significantly higher HER2 scores in the metastasis (P = .004). Concordance between primary tumors and metastases was highest for 4B5 ultraView (69.4%), followed by SP3 (61.2%) and 4B5 OptiView (51.8%). DG44 showed the most variability, with only 36.5% of matched samples receiving the same HER2 category. DG44 identified a significantly higher proportion of HER2-(ultra)low cases and showed the most variability in HER2 status between matched primary tumors and metastases compared with 4B5 and SP3. The choice of HER2 assay can lead to discrepancies in HER2 status assessment, which could directly influence patient eligibility for trastuzumab-deruxtecan treatment.
{"title":"Interassay Comparison With Digital Image Analysis of 4 Routine Human Epidermal Growth Factor Receptor 2 (HER2) Immunohistochemistry Assays in Primary Breast Cancer and Its Metastasis","authors":"Maaike Anna Hempenius , Mieke C. Zwager , Jeppe Thagaard , Lorian Slagter-Menkema , Henk J. Buikema , Ellis Barbé , Michael A. den Bakker , Marjolein G.J. Heerema , Elisabeth G.E. de Vries , Carolina P. Schröder , Nils A. ’t Hart , Bert van der Vegt","doi":"10.1016/j.labinv.2025.104261","DOIUrl":"10.1016/j.labinv.2025.104261","url":null,"abstract":"<div><div>Trastuzumab-deruxtecan, an antibody-drug conjugate targeting human epidermal growth factor receptor 2 (HER2), improves overall survival in patients with breast cancer showing low or ultralow HER2 expression. Differences in HER2 test results have been reported between various HER2 assays and between primary tumors and metastases, but an objective comparison with incorporation of the new HER2-ultralow cutoff values is needed. This study aimed to assess the performance of 4 routine clinical-grade HER2 assays across and between primary tumors and their metastases using digital image analysis. Primary tumors and metastases from 193 patients with breast cancer who participated in the IMaging PAtients for Cancer drug selecTion—Metastatic Breast Cancer trial were incorporated into 6 tissue microarrays. Samples were stained by 4 laboratories using their routine HER2 immunohistochemistry protocols: 4B5 ultraView, 4B5 OptiView, SP3, and HercepTest (DG44). HER2 scores were determined using digital image analysis. The 4 HER2 assays showed significant differences in HER2 status in both primary tumors and metastases. Eighty-five matched primary tumors and metastases were analyzed to investigate concordance in HER2 status. Although no significant differences were found in HER2 scores between primary tumors and metastases for SP3 and both 4B5 assays, DG44 showed significantly higher HER2 scores in the metastasis (<em>P</em> = .004). Concordance between primary tumors and metastases was highest for 4B5 ultraView (69.4%), followed by SP3 (61.2%) and 4B5 OptiView (51.8%). DG44 showed the most variability, with only 36.5% of matched samples receiving the same HER2 category. DG44 identified a significantly higher proportion of HER2-(ultra)low cases and showed the most variability in HER2 status between matched primary tumors and metastases compared with 4B5 and SP3. The choice of HER2 assay can lead to discrepancies in HER2 status assessment, which could directly influence patient eligibility for trastuzumab-deruxtecan treatment.</div></div>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":"106 1","pages":"Article 104261"},"PeriodicalIF":4.2,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573945","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}