Pub Date : 2026-01-05DOI: 10.1016/j.jmoldx.2025.11.008
Pilar Barberán-Martínez, Mar Balanzá, Belén García-Bohórquez, Sofia Escobar-Parra, Romana García-Gil, Anselmo Feliciano-Sánchez, Teresa Jaijo, Elena Aller, Gema García-García, José M Millán
Inherited retinal dystrophies (IRDs) represent a diverse group of rare pathologies affecting vision with significant genetic and clinical variability. Clinical exome sequencing (CES) was performed on 143 families clinically diagnosed with IRDs. The obtained variants were filtered and classified according to the American College of Medical Genetics guidelines. Overall, a genetic diagnosis was achieved for 68.53% of the families in the cohort, with 35 causative genes identified, predominantly ABCA4 and USH2A. A total of 170 clinically relevant variants were identified, 45 of which (26.47%) were novel, with missense variants being the most common type (40.59%). This study demonstrated aberrant splicing generated by the ABCA4 (NM_000350.2): c.1299A>G mutation through the functional assay of a minigene. Furthermore, the genes FAM161A and GUCY2D were associated with IRDs that are not typically linked to these genes. Consequently, this study expands the current understanding of IRDs and supports the use of CES as an effective strategy for the genetic diagnosis of these pathologies.
{"title":"Clinical Exome Sequencing: A Genetic Diagnostic Approach for Inherited Retinal Dystrophies.","authors":"Pilar Barberán-Martínez, Mar Balanzá, Belén García-Bohórquez, Sofia Escobar-Parra, Romana García-Gil, Anselmo Feliciano-Sánchez, Teresa Jaijo, Elena Aller, Gema García-García, José M Millán","doi":"10.1016/j.jmoldx.2025.11.008","DOIUrl":"https://doi.org/10.1016/j.jmoldx.2025.11.008","url":null,"abstract":"<p><p>Inherited retinal dystrophies (IRDs) represent a diverse group of rare pathologies affecting vision with significant genetic and clinical variability. Clinical exome sequencing (CES) was performed on 143 families clinically diagnosed with IRDs. The obtained variants were filtered and classified according to the American College of Medical Genetics guidelines. Overall, a genetic diagnosis was achieved for 68.53% of the families in the cohort, with 35 causative genes identified, predominantly ABCA4 and USH2A. A total of 170 clinically relevant variants were identified, 45 of which (26.47%) were novel, with missense variants being the most common type (40.59%). This study demonstrated aberrant splicing generated by the ABCA4 (NM_000350.2): c.1299A>G mutation through the functional assay of a minigene. Furthermore, the genes FAM161A and GUCY2D were associated with IRDs that are not typically linked to these genes. Consequently, this study expands the current understanding of IRDs and supports the use of CES as an effective strategy for the genetic diagnosis of these pathologies.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145919058","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-02DOI: 10.1016/j.jmoldx.2025.12.004
Rachel B Keller-Evans, Zoe Fleischmann, Smruthy Sivakumar, Radwa Sharaf, Erik A Williams, Benjamin Kaplan, Ethan S Sokol, Alexa B Schrock, Hanna Tukachinsky, Douglas A Mata, Tyler Janovitz, Douglas I Lin, Lei Zhong, Lyle Lopez, Nimesh R Patel, Garrett M Frampton, Geoffrey R Oxnard, Julia A Elvin, Brennan Decker
Pathogenic POLE mutations (pPOLE) undermine mismatch error correction by polymerase ε during DNA replication, and the resulting somatic ultramutation predicts response to immunotherapy. Beyond frequently recurrent alleles, historical pPOLE classification has been largely based on exonuclease domain localization. A POLE-specific phenotypic classification model was developed, encompassing tumor mutational burden (TMB), mutational signatures, germline frequency, and consideration of comutation with other POLE mutations to identify pPOLE. This model was applied to >490,000 samples and identified 29 predicted pPOLE, including 16 not previously reported. A total of 748 tumors (0.2%) had one or more pPOLE, most commonly in endometrial and colorectal cancers, although pPOLE were observed in many additional cancer types. pPOLE were associated with ultramutation [median TMB, 186.3 mutations per megabase (mut/Mb)] across tumor types. Concurrent pPOLE and microsatellite instability were more common than previously appreciated and produced a synergistic TMB impact, with medians of 135.7 mut/Mb for pPOLE/microsatellite stable samples compared with 325.6 mut/Mb for pPOLE/microsatellite instability-high samples. Comutation analysis in endometrial and colorectal cancers highlighted associations with homologous recombination pathway gene mutations that were predominantly monoallelic passengers that are unlikely to predict response to therapies targeting DNA repair deficiencies. pPOLE have been incorporated into treatment guidelines for several malignancies and are an important predictor of immunotherapy response. This study provides biological insight to guide classification and clinical management of patients with tumors harboring pPOLE.
{"title":"Phenotypic POLE Variant Classification Identifies Patients Who May Have Favorable Prognosis and Benefit from Immunotherapy.","authors":"Rachel B Keller-Evans, Zoe Fleischmann, Smruthy Sivakumar, Radwa Sharaf, Erik A Williams, Benjamin Kaplan, Ethan S Sokol, Alexa B Schrock, Hanna Tukachinsky, Douglas A Mata, Tyler Janovitz, Douglas I Lin, Lei Zhong, Lyle Lopez, Nimesh R Patel, Garrett M Frampton, Geoffrey R Oxnard, Julia A Elvin, Brennan Decker","doi":"10.1016/j.jmoldx.2025.12.004","DOIUrl":"10.1016/j.jmoldx.2025.12.004","url":null,"abstract":"<p><p>Pathogenic POLE mutations (pPOLE) undermine mismatch error correction by polymerase ε during DNA replication, and the resulting somatic ultramutation predicts response to immunotherapy. Beyond frequently recurrent alleles, historical pPOLE classification has been largely based on exonuclease domain localization. A POLE-specific phenotypic classification model was developed, encompassing tumor mutational burden (TMB), mutational signatures, germline frequency, and consideration of comutation with other POLE mutations to identify pPOLE. This model was applied to >490,000 samples and identified 29 predicted pPOLE, including 16 not previously reported. A total of 748 tumors (0.2%) had one or more pPOLE, most commonly in endometrial and colorectal cancers, although pPOLE were observed in many additional cancer types. pPOLE were associated with ultramutation [median TMB, 186.3 mutations per megabase (mut/Mb)] across tumor types. Concurrent pPOLE and microsatellite instability were more common than previously appreciated and produced a synergistic TMB impact, with medians of 135.7 mut/Mb for pPOLE/microsatellite stable samples compared with 325.6 mut/Mb for pPOLE/microsatellite instability-high samples. Comutation analysis in endometrial and colorectal cancers highlighted associations with homologous recombination pathway gene mutations that were predominantly monoallelic passengers that are unlikely to predict response to therapies targeting DNA repair deficiencies. pPOLE have been incorporated into treatment guidelines for several malignancies and are an important predictor of immunotherapy response. This study provides biological insight to guide classification and clinical management of patients with tumors harboring pPOLE.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145901471","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-24DOI: 10.1016/j.jmoldx.2025.12.002
Andrew P Fellowes, David Y H Choong, Christopher R McEvoy, Roxane A Legaie, Anthony H Bell, Stephen B Fox
In this multilaboratory validation study of 145 ovarian cancer samples, the SOPHiA DDM HRD Solution was compared with the regulatory-approved Myriad myChoice HRD assay to assess clinical comparability for class 3 in-house in vitro diagnostic medical device companion diagnostic use. BRCA1/2 mutation status showed 100% concordance, and genomic instability (GI) measurements demonstrated strong linear agreement, absence of bias, and high analytical precision. Receiver operating characteristic analysis suggested a threshold adjustment from 0 to -1.5, improving overall accuracy to 91.2% when combined with BRCA mutation status to assign homologous recombination deficiency (HRD) status. Approximately 6% of samples were excluded because of inconclusive results, whereas GI classification discordance was concentrated near the clinical threshold. Neither inconclusiveness nor discordance was associated with sample-related factors. These findings indicate that the SOPHiA HRD assay can provide results broadly interchangeable with Myriad myChoice, although caution is warranted when assigning HRD status to borderline GI values.
{"title":"Validation of the SOPHiA DDM HRD Solution as a Companion Diagnostic for Poly (ADP-Ribose) Polymerase Inhibitor Access in Australia.","authors":"Andrew P Fellowes, David Y H Choong, Christopher R McEvoy, Roxane A Legaie, Anthony H Bell, Stephen B Fox","doi":"10.1016/j.jmoldx.2025.12.002","DOIUrl":"10.1016/j.jmoldx.2025.12.002","url":null,"abstract":"<p><p>In this multilaboratory validation study of 145 ovarian cancer samples, the SOPHiA DDM HRD Solution was compared with the regulatory-approved Myriad myChoice HRD assay to assess clinical comparability for class 3 in-house in vitro diagnostic medical device companion diagnostic use. BRCA1/2 mutation status showed 100% concordance, and genomic instability (GI) measurements demonstrated strong linear agreement, absence of bias, and high analytical precision. Receiver operating characteristic analysis suggested a threshold adjustment from 0 to -1.5, improving overall accuracy to 91.2% when combined with BRCA mutation status to assign homologous recombination deficiency (HRD) status. Approximately 6% of samples were excluded because of inconclusive results, whereas GI classification discordance was concentrated near the clinical threshold. Neither inconclusiveness nor discordance was associated with sample-related factors. These findings indicate that the SOPHiA HRD assay can provide results broadly interchangeable with Myriad myChoice, although caution is warranted when assigning HRD status to borderline GI values.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844189","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-23DOI: 10.1016/j.jmoldx.2025.09.005
Ava Kwong , Cecilia Y.S. Ho , Henry C.M. Leung , Amy W.S. Leung , Chun Hang Au , Edmond S.K. Ma
Large genomic rearrangements (LGRs) account for at least 10% of the mutations in BRCA1 and 5% of BRCA2 mutations in outbred families with hereditary breast and ovarian cancer. A total of 21 probands with breast cancer who carried BRCA1 or BRCA2 LGRs were identified from a cohort of 4678 Chinese patients. There was a total of 13 BRCA1 LGR carriers and 8 BRCA2 LGR carriers, including 12 large genomic deletions and 1 duplication. Ten and three specific breakpoints from BRCA1 and BRCA2, respectively, were identified by either whole-genome sequencing by nanopore sequencing or long-range PCR. Five of these LGRs were recurrent LGRs. Three LGRs were novel founder LGRs in the southeast Chinese population. Chinese LGR carriers exhibited clinical phenotypes that were generally similar to those of non-LGR mutation carriers. However, there was a notable tendency for triple-negative breast cancer to be more prevalent among Chinese LGR carriers (P = 0.007), largely because of the predominance of BRCA1 mutations. This suggests a potential association that warrants further investigation.
{"title":"Characterization of Large Genomic Rearrangements in BRCA1 and BRCA2 Genes in a Chinese High-Risk Cohort","authors":"Ava Kwong , Cecilia Y.S. Ho , Henry C.M. Leung , Amy W.S. Leung , Chun Hang Au , Edmond S.K. Ma","doi":"10.1016/j.jmoldx.2025.09.005","DOIUrl":"10.1016/j.jmoldx.2025.09.005","url":null,"abstract":"<div><div>Large genomic rearrangements (LGRs) account for at least 10% of the mutations in <em>BRCA1</em> and 5% of <em>BRCA2</em> mutations in outbred families with hereditary breast and ovarian cancer. A total of 21 probands with breast cancer who carried <em>BRCA1</em> or <em>BRCA2</em> LGRs were identified from a cohort of 4678 Chinese patients. There was a total of 13 <em>BRCA1</em> LGR carriers and 8 <em>BRCA2</em> LGR carriers, including 12 large genomic deletions and 1 duplication. Ten and three specific breakpoints from <em>BRCA1</em> and <em>BRCA2</em>, respectively, were identified by either whole-genome sequencing by nanopore sequencing or long-range PCR. Five of these LGRs were recurrent LGRs. Three LGRs were novel founder LGRs in the southeast Chinese population. Chinese LGR carriers exhibited clinical phenotypes that were generally similar to those of non-LGR mutation carriers. However, there was a notable tendency for triple-negative breast cancer to be more prevalent among Chinese LGR carriers (<em>P</em> = 0.007), largely because of the predominance of <em>BRCA1</em> mutations. This suggests a potential association that warrants further investigation.</div></div>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":"28 1","pages":"Pages 8-21"},"PeriodicalIF":3.4,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145801857","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
This large-scale retrospective study investigated the allele, diplotype, and phenotype frequencies of CYP2B6, CYP2C19, and CYP2D6 in a Han Chinese population (N > 10,000), using real-world genetic data from The First Hospital of Hebei Medical University, assessed between March 2021 and April 2025. Single-nucleotide polymorphism genotyping was performed using the Agena MassARRAY assay. CYP2D6 copy number variation (CNV) was analyzed by TaqMan real-time quantitative PCR. The sample sizes were 9729 for CYP2B6, 11,479 for CYP2C19, and 11,315 for CYP2D6, all from the Han Chinese population. The high frequencies of alleles were CYP2B6∗6 (16.19%), CYP2C19∗2 (30.36%), and CYP2D6∗10 (41.67%), with the most common diplotypes being CYP2B6 ∗1/∗6 (23.65%), CYP2C19 ∗1/∗2 (37.62%), and CYP2D6 ∗1/∗10 (16.05%), respectively. At the phenotype level, normal metabolizer was most common for CYP2B6 (57.86%) and CYP2D6 (60.50%), whereas the intermediate metabolizer phenotype was noted for CYP2C19 (44.77%). CYP2D6 CNVs included zero copies (0.43%), one copy (13.64%), two copies (83.79%), three copies (2.11%), and more than three copies (0.03%). This large-scale, real-world study of a Northern Han Chinese cohort provides a valuable pharmacogenomic reference and demonstrates the necessity of integrating CNV analysis with single-nucleotide polymorphism genotyping to ensure accurate clinical phenotyping of CYP2D6.
{"title":"Allele, Diplotype, and Phenotype Frequency Distribution of CYP2B6, CYP2C19, and CYP2D6 in the Han Population in North China.","authors":"Yueyao Luan, Qixuan Sun, Yiyuan Wang, Binliang Tong, Liguang Duan, Jiaqi Wang, Yuhang Yan, Chaoli Chen, Yang Lun, Jing Yu, Yuanyuan Zhao, Mengqiang Zhao, Chunhua Zhou","doi":"10.1016/j.jmoldx.2025.11.007","DOIUrl":"10.1016/j.jmoldx.2025.11.007","url":null,"abstract":"<p><p>This large-scale retrospective study investigated the allele, diplotype, and phenotype frequencies of CYP2B6, CYP2C19, and CYP2D6 in a Han Chinese population (N > 10,000), using real-world genetic data from The First Hospital of Hebei Medical University, assessed between March 2021 and April 2025. Single-nucleotide polymorphism genotyping was performed using the Agena MassARRAY assay. CYP2D6 copy number variation (CNV) was analyzed by TaqMan real-time quantitative PCR. The sample sizes were 9729 for CYP2B6, 11,479 for CYP2C19, and 11,315 for CYP2D6, all from the Han Chinese population. The high frequencies of alleles were CYP2B6∗6 (16.19%), CYP2C19∗2 (30.36%), and CYP2D6∗10 (41.67%), with the most common diplotypes being CYP2B6 ∗1/∗6 (23.65%), CYP2C19 ∗1/∗2 (37.62%), and CYP2D6 ∗1/∗10 (16.05%), respectively. At the phenotype level, normal metabolizer was most common for CYP2B6 (57.86%) and CYP2D6 (60.50%), whereas the intermediate metabolizer phenotype was noted for CYP2C19 (44.77%). CYP2D6 CNVs included zero copies (0.43%), one copy (13.64%), two copies (83.79%), three copies (2.11%), and more than three copies (0.03%). This large-scale, real-world study of a Northern Han Chinese cohort provides a valuable pharmacogenomic reference and demonstrates the necessity of integrating CNV analysis with single-nucleotide polymorphism genotyping to ensure accurate clinical phenotyping of CYP2D6.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800884","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.jmoldx.2025.11.006
Ben Lundie, Sze Yee Chai, Alicia B Byrne, Dimitar Azmanov, John Christodoulou, Matilda A Haas, Karin S Kassahn, Sebastian Lunke, Ami Stott, Bryony A Thompson, Tony Badrick, Bruce Bennetts
Few external quality assurance programs adequately address the complexity of largescale human genome or exome analysis. To bridge this gap, Australian Genomics and the Royal College of Pathologists of Australasia Quality Assurance Programs (QAP) developed a pilot interpretive module focused on genomic testing for childhood syndromes and intellectual disabilities. The program assessed laboratories' proficiency in interpreting complex genomic data for pediatric disorders. Six clinically accredited laboratories analyzed standardized genomic, phenotypic, and referral data. Reports were evaluated using a rubric adapted from the European Molecular Genetics Quality Network model, covering genotyping, variant classification, interpretation, and report content. Feedback included comparative performance results and individual recommendations. All laboratories correctly identified and classified target variants, but variation was observed in report structure, inclusion of genetic counseling advice, and application of the American College of Medical Genetics and Genomics/Association for Molecular Pathology classification framework. Participants noted that data-sharing limitations and differences in local reporting practices contributed to scoring inconsistencies. The pilot demonstrated the feasibility of a disease-specific interpretive QAP for complex pediatric genomic testing. Future rounds will address logistical challenges, refine scoring criteria, and strengthen standardization, supporting broader implementation. This initiative lays the groundwork for integrating specialized QAP modules into routine practice to improve diagnostic accuracy and consistency across laboratories.
{"title":"Piloting an Interpretive External Quality Assurance Model for Genomic Testing for Childhood Syndromes and Intellectual Disability.","authors":"Ben Lundie, Sze Yee Chai, Alicia B Byrne, Dimitar Azmanov, John Christodoulou, Matilda A Haas, Karin S Kassahn, Sebastian Lunke, Ami Stott, Bryony A Thompson, Tony Badrick, Bruce Bennetts","doi":"10.1016/j.jmoldx.2025.11.006","DOIUrl":"10.1016/j.jmoldx.2025.11.006","url":null,"abstract":"<p><p>Few external quality assurance programs adequately address the complexity of largescale human genome or exome analysis. To bridge this gap, Australian Genomics and the Royal College of Pathologists of Australasia Quality Assurance Programs (QAP) developed a pilot interpretive module focused on genomic testing for childhood syndromes and intellectual disabilities. The program assessed laboratories' proficiency in interpreting complex genomic data for pediatric disorders. Six clinically accredited laboratories analyzed standardized genomic, phenotypic, and referral data. Reports were evaluated using a rubric adapted from the European Molecular Genetics Quality Network model, covering genotyping, variant classification, interpretation, and report content. Feedback included comparative performance results and individual recommendations. All laboratories correctly identified and classified target variants, but variation was observed in report structure, inclusion of genetic counseling advice, and application of the American College of Medical Genetics and Genomics/Association for Molecular Pathology classification framework. Participants noted that data-sharing limitations and differences in local reporting practices contributed to scoring inconsistencies. The pilot demonstrated the feasibility of a disease-specific interpretive QAP for complex pediatric genomic testing. Future rounds will address logistical challenges, refine scoring criteria, and strengthen standardization, supporting broader implementation. This initiative lays the groundwork for integrating specialized QAP modules into routine practice to improve diagnostic accuracy and consistency across laboratories.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800832","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.jmoldx.2025.11.005
Lisa Robinson, Sharleen Rapp, Weiwei Zhang, Jaclyn Pope, Allison M Cushman-Vokoun
Gastrointestinal stromal tumors (GISTs) are predominantly characterized by mutations in KIT or PDGFRA. Mutation detection is important for optimal therapy. Next-generation sequencing (NGS) panels are useful in GIST assessment as they allow for simultaneous evaluation of multiple genes. However, inherent to use of NGS with short-read sequences on formalin-fixed specimens is the potential to miss larger insertion or deletion variants. Over 9 years, GIST testing was performed on specimens from 55 patients by amplicon-based, semiconductor NGS using the Ion AmpliSeq Cancer Hotspot Panel version 2, a Cancer Hotspot Panel version 2-based GIST panel, or the Oncomine Precision Assay. Negative cases were evaluated by dideoxy sequencing for detection of mutations in KIT and PDGFRA, as per the clinical protocol. Before reflexive sequencing, of 55 completed analyses, 47 (85%) were positive for KIT or PDGFRA mutations by NGS. Three cases were attributed to positivity for pathogenic variants in other genes. Of the five cases evaluated by dideoxy sequencing, all five (9% of all specimens) were positive for pathogenic or likely pathogenic KIT mutations. A total of 12% of KIT mutations required dideoxy sequencing for identification. Mutations were 12 to 39 nucleotide deletion or duplication variants. The results suggest that short-read, amplicon-based NGS assays may miss a significant number of clinically actionable KIT mutations and that follow-up of KIT and PDGFRA NGS-negative cases by alternative testing modalities should be considered.
{"title":"Dideoxy Sequencing Enhances Detection of KIT Mutations in Gastrointestinal Stromal Tumors Initially Evaluated by Next-Generation Sequencing Hotspot Panels.","authors":"Lisa Robinson, Sharleen Rapp, Weiwei Zhang, Jaclyn Pope, Allison M Cushman-Vokoun","doi":"10.1016/j.jmoldx.2025.11.005","DOIUrl":"10.1016/j.jmoldx.2025.11.005","url":null,"abstract":"<p><p>Gastrointestinal stromal tumors (GISTs) are predominantly characterized by mutations in KIT or PDGFRA. Mutation detection is important for optimal therapy. Next-generation sequencing (NGS) panels are useful in GIST assessment as they allow for simultaneous evaluation of multiple genes. However, inherent to use of NGS with short-read sequences on formalin-fixed specimens is the potential to miss larger insertion or deletion variants. Over 9 years, GIST testing was performed on specimens from 55 patients by amplicon-based, semiconductor NGS using the Ion AmpliSeq Cancer Hotspot Panel version 2, a Cancer Hotspot Panel version 2-based GIST panel, or the Oncomine Precision Assay. Negative cases were evaluated by dideoxy sequencing for detection of mutations in KIT and PDGFRA, as per the clinical protocol. Before reflexive sequencing, of 55 completed analyses, 47 (85%) were positive for KIT or PDGFRA mutations by NGS. Three cases were attributed to positivity for pathogenic variants in other genes. Of the five cases evaluated by dideoxy sequencing, all five (9% of all specimens) were positive for pathogenic or likely pathogenic KIT mutations. A total of 12% of KIT mutations required dideoxy sequencing for identification. Mutations were 12 to 39 nucleotide deletion or duplication variants. The results suggest that short-read, amplicon-based NGS assays may miss a significant number of clinically actionable KIT mutations and that follow-up of KIT and PDGFRA NGS-negative cases by alternative testing modalities should be considered.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800887","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.jmoldx.2025.12.001
Stephanie A Smoley, Gopinath Sivasankaran, Mallika Gandham, Beth A Pitel, Shannon M Knight, Stefan W Nelson, Nipun A Mistry, Katherine B Geiersbach, Sounak Gupta, Kevin C Halling, Robert B Jenkins, Hussam Al-Kateb
Large-scale tumor molecular profiling has enabled the discovery of diagnostic, prognostic, and therapeutic biomarkers, and expanded the clinical utility of alterations such as gene amplifications (GAMPs), homozygous deletions (HMZ-Dels), and biallelic inactivation (BI) of tumor suppressor genes. Comprehensive clinical detection of these events is essential for optimal patient management. Illumina's TruSight Oncology 500 (TSO500) kit detects multiple biomarkers, including GAMPs for select genes, but does not assess HMZ-Del or BI events. To address this gap, OncCNV, a genome-wide copy number analysis and visualization pipeline that integrates both on-target and off-target probe data from TSO500 sequencing, was developed. Performance optimization evaluated copy number calling tools, on-target and off-target probe selection strategies, off-target bin sizes, and panel-of-normal configurations. Clinical validation was conducted using 132 unique solid tumors characterized by a clinically validated microarray assay. OncCNV showed >96% positive percentage agreement, >99% negative percentage agreement, and >99% accuracy at the assay's established limit of detection (40 ng DNA at 40% tumor content). Sensitivity for HMZ-Del and BI detection decreased to 62%-70% at tumor content of 20%-39% in in silico dilution experiments; however, intrarun, interrun, and interanalyst precision remained >99%. OncCNV extends the analytical capabilities of TSO500 by enabling robust and precise detection of GAMP, HMZ-Del, and BI events, enhancing solid tumor comprehensive molecular profiling.
大规模的肿瘤分子谱分析使得新的诊断、预后和治疗生物标志物的发现成为可能,同时扩大了已知改变的临床应用,如基因扩增(GAMP)、纯合缺失(HMZ-Del)和肿瘤抑制基因的双等位基因失活(BI)。对这些事件进行全面的临床检测对于优化患者管理至关重要。Illumina的TSO500试剂盒检测多种生物标志物,包括用于选择基因的GAMP,但不评估HMZ-Del或BI事件。为了解决这一问题,我们开发了OncCNV,这是一个全基因组拷贝数分析和可视化管道,集成了TSO500测序的靶标和非靶标探针数据。OncCNV的性能通过评估不同的工具包、靶探针和非靶探针来优化,用于CNV调用和可视化、非靶容器大小和正常配置面板。临床验证使用了132个独特的实体瘤,并通过临床验证的微阵列分析进行了表征。OncCNV在测定的检测限(40 ng DNA, 40%肿瘤含量)下,所有标记物的阳性一致性为b> 96%,阴性一致性为>99%,准确度为>99%。在硅稀释实验中,当肿瘤含量为20 ~ 39%时,HMZ-Del和BI的检测灵敏度降至62 ~ 70%;然而,运行内部和运行之间以及分析师之间的精度仍然保持在99%左右。OncCNV扩展了TSO500的分析能力,实现了对GAMP、HMZ-Del和BI事件的稳健、精确和准确的检测,增强了实体肿瘤的全面分子谱分析。
{"title":"Development and Clinical Validation of OncCNV: A Pipeline for Comprehensive Genome-Wide Analysis of Oncogene Amplifications, Homozygous Deletions, and Biallelic Inactivation of Tumor Suppressor Genes Using the TruSight Oncology 500 Kit.","authors":"Stephanie A Smoley, Gopinath Sivasankaran, Mallika Gandham, Beth A Pitel, Shannon M Knight, Stefan W Nelson, Nipun A Mistry, Katherine B Geiersbach, Sounak Gupta, Kevin C Halling, Robert B Jenkins, Hussam Al-Kateb","doi":"10.1016/j.jmoldx.2025.12.001","DOIUrl":"10.1016/j.jmoldx.2025.12.001","url":null,"abstract":"<p><p>Large-scale tumor molecular profiling has enabled the discovery of diagnostic, prognostic, and therapeutic biomarkers, and expanded the clinical utility of alterations such as gene amplifications (GAMPs), homozygous deletions (HMZ-Dels), and biallelic inactivation (BI) of tumor suppressor genes. Comprehensive clinical detection of these events is essential for optimal patient management. Illumina's TruSight Oncology 500 (TSO500) kit detects multiple biomarkers, including GAMPs for select genes, but does not assess HMZ-Del or BI events. To address this gap, OncCNV, a genome-wide copy number analysis and visualization pipeline that integrates both on-target and off-target probe data from TSO500 sequencing, was developed. Performance optimization evaluated copy number calling tools, on-target and off-target probe selection strategies, off-target bin sizes, and panel-of-normal configurations. Clinical validation was conducted using 132 unique solid tumors characterized by a clinically validated microarray assay. OncCNV showed >96% positive percentage agreement, >99% negative percentage agreement, and >99% accuracy at the assay's established limit of detection (40 ng DNA at 40% tumor content). Sensitivity for HMZ-Del and BI detection decreased to 62%-70% at tumor content of 20%-39% in in silico dilution experiments; however, intrarun, interrun, and interanalyst precision remained >99%. OncCNV extends the analytical capabilities of TSO500 by enabling robust and precise detection of GAMP, HMZ-Del, and BI events, enhancing solid tumor comprehensive molecular profiling.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-29DOI: 10.1016/j.jmoldx.2025.11.004
Eros Qama, Abedul Haque, Juan Du, Abul K Azad, Rizwan Naeem, Yanhua Wang, Monika Paroder, D Yitzchak Goldstein, David M Loeb, Adriana I Colovai
Donor chimerism analysis is used for monitoring engraftment status and risk of disease relapse following allogeneic stem cell transplantation. Recently developed assays using next-generation sequencing have demonstrated enhanced sensitivity and accuracy compared with standard capillary electrophoresis methods. This work presents validation results using One Lambda Devyser Chimerism assay, a next-generation sequencing-based test for monitoring donor chimerism. A total of 270 samples, including clinical and cell-line DNA, were tested. There was a high correlation between chimerism results obtained with One Lambda Devyser and short tandem repeat assays (R2 = 0.998). Determination of the limit of blank, limit of detection, and limit of quantitation indicated that One Lambda Devyser Chimerism assay can reliably detect recipient DNA fractions as low as 0.1%. Analytical specificity was >99.9%. Reproducibility, linearity, DNA library characteristics, and sequencing metrics are presented. The suitability of DNA markers was verified in a population predominantly African American and Hispanic, comprising 30 recipient/donor pairs. The average number of informative markers per pair was seven, with a lower representation (five markers) in related pairs. In conclusion, the results show that One Lambda Devyser Chimerism assay is a highly sensitive test for detecting donor chimerism in a diverse patient population. The assay performed remarkably well at low recipient concentrations, having the potential to detect early changes associated with disease relapse.
{"title":"Highly Sensitive Detection of Donor Chimerism by Next-Generation Sequencing.","authors":"Eros Qama, Abedul Haque, Juan Du, Abul K Azad, Rizwan Naeem, Yanhua Wang, Monika Paroder, D Yitzchak Goldstein, David M Loeb, Adriana I Colovai","doi":"10.1016/j.jmoldx.2025.11.004","DOIUrl":"10.1016/j.jmoldx.2025.11.004","url":null,"abstract":"<p><p>Donor chimerism analysis is used for monitoring engraftment status and risk of disease relapse following allogeneic stem cell transplantation. Recently developed assays using next-generation sequencing have demonstrated enhanced sensitivity and accuracy compared with standard capillary electrophoresis methods. This work presents validation results using One Lambda Devyser Chimerism assay, a next-generation sequencing-based test for monitoring donor chimerism. A total of 270 samples, including clinical and cell-line DNA, were tested. There was a high correlation between chimerism results obtained with One Lambda Devyser and short tandem repeat assays (R<sup>2</sup> = 0.998). Determination of the limit of blank, limit of detection, and limit of quantitation indicated that One Lambda Devyser Chimerism assay can reliably detect recipient DNA fractions as low as 0.1%. Analytical specificity was >99.9%. Reproducibility, linearity, DNA library characteristics, and sequencing metrics are presented. The suitability of DNA markers was verified in a population predominantly African American and Hispanic, comprising 30 recipient/donor pairs. The average number of informative markers per pair was seven, with a lower representation (five markers) in related pairs. In conclusion, the results show that One Lambda Devyser Chimerism assay is a highly sensitive test for detecting donor chimerism in a diverse patient population. The assay performed remarkably well at low recipient concentrations, having the potential to detect early changes associated with disease relapse.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145650036","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-29DOI: 10.1016/j.jmoldx.2025.11.003
Karin Wallander, Yingbo Lin, Vadym Ivanchuk, Valeria Difilippo, Venkatesh Chellappa, Sarath Kumar Murugan, Ingegerd Öfverholm, Robert Bränström, Karolin H Nord, Joseph Carlson, Felix Haglund de Flon
Optical genome mapping (OGM) enables high-resolution detection of structural variants (SVs) and copy number aberrations (CNAs) using ultralong DNA molecules and minimal bioinformatics processing. Its diagnostic utility in solid tumors remains underexplored. Whole-genome sequencing (WGS) offers comprehensive variant detection but is resource intensive. This study presents a technical benchmarking of OGM versus WGS for mesenchymal tumors of the gynecologic tract. Twenty-five uterine mesenchymal tumors were prospectively analyzed using matched WGS, transcriptome sequencing, and OGM. Detected SVs, CNAs, and fusion genes were compared across platforms. OGM identified structural driver events in 80% of cases and demonstrated high concordance with WGS for major CNAs and translocations. In select cases, OGM resolved complex rearrangements not clearly defined by WGS, including a PLAG1::RERE fusion and an embedded inversion in a RAD51B::HMGA2 event. Conversely, WGS uniquely detected a truncating NF1 translocation and a TSC2::SENP3 fusion, both clinically significant. OGM is a technically robust platform for SV and CNA detection in mesenchymal tumors, and it may serve as an efficient alternative to sequencing-based cytogenomic approaches in selected clinical contexts, especially in tumors known to be driven by gross chromosomal rearrangements. WGS provides a comprehensive view of the cancer genome, suitable for tumors driven by single-nucleotide variants, SVs, and CNAs. The choice between platforms should be guided by clinical context, diagnostic needs, and available resources.
{"title":"Optical Genome Mapping versus Whole-Genome Sequencing in the Clinical Diagnosis of Gynecologic Mesenchymal Tumors.","authors":"Karin Wallander, Yingbo Lin, Vadym Ivanchuk, Valeria Difilippo, Venkatesh Chellappa, Sarath Kumar Murugan, Ingegerd Öfverholm, Robert Bränström, Karolin H Nord, Joseph Carlson, Felix Haglund de Flon","doi":"10.1016/j.jmoldx.2025.11.003","DOIUrl":"10.1016/j.jmoldx.2025.11.003","url":null,"abstract":"<p><p>Optical genome mapping (OGM) enables high-resolution detection of structural variants (SVs) and copy number aberrations (CNAs) using ultralong DNA molecules and minimal bioinformatics processing. Its diagnostic utility in solid tumors remains underexplored. Whole-genome sequencing (WGS) offers comprehensive variant detection but is resource intensive. This study presents a technical benchmarking of OGM versus WGS for mesenchymal tumors of the gynecologic tract. Twenty-five uterine mesenchymal tumors were prospectively analyzed using matched WGS, transcriptome sequencing, and OGM. Detected SVs, CNAs, and fusion genes were compared across platforms. OGM identified structural driver events in 80% of cases and demonstrated high concordance with WGS for major CNAs and translocations. In select cases, OGM resolved complex rearrangements not clearly defined by WGS, including a PLAG1::RERE fusion and an embedded inversion in a RAD51B::HMGA2 event. Conversely, WGS uniquely detected a truncating NF1 translocation and a TSC2::SENP3 fusion, both clinically significant. OGM is a technically robust platform for SV and CNA detection in mesenchymal tumors, and it may serve as an efficient alternative to sequencing-based cytogenomic approaches in selected clinical contexts, especially in tumors known to be driven by gross chromosomal rearrangements. WGS provides a comprehensive view of the cancer genome, suitable for tumors driven by single-nucleotide variants, SVs, and CNAs. The choice between platforms should be guided by clinical context, diagnostic needs, and available resources.</p>","PeriodicalId":50128,"journal":{"name":"Journal of Molecular Diagnostics","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145650095","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}