1
|
Lin MT, Christenson ES, Pallavajjala A, Eshleman JR. Highly sensitive and specific markers for detection of mismatch repair deficiency by next-generation sequencing. Am J Clin Pathol 2025:aqaf026. [PMID: 40318191 DOI: 10.1093/ajcp/aqaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/16/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVE To identify exonic markers that could improve analytic performance characteristics of next-generation sequencing (NGS) in detecting mismatch repair deficiency (dMMR) using colorectal cancer (CRC) as a model. METHODS Coding sequences of a target NGS panel (~1.13 megabase) were compared between dMMR CRC and mismatch repair-proficient (pMMR) CRC in a training cohort (41 dMMR CRCs and 213 pMMR CRCs) and a validation cohort (33 dMMR CRCs and 307 pMMR CRCs) with documented mismatch repair status by immunohistochemical and/or microsatellite instability assays. RESULTS The dMMR CRC cases showed significantly higher insertion/deletion (indel) mutations within exonic homopolymers (homo-indels), occurring predominantly within longer repeats of 5 to 10 nucleotides (92%, P < .0001), rather than shorter repeats of 2 to 4 nucleotides seen in pMMR CRC (62%). Homo-indels in dMMR CRC were not random. Hotspot loci were consistent between the training and validation cohorts. The dMMR defined by indels within homopolymers of 5 or more nucleotides, homopolymers of 7 or more nucleotides, or a panel of hotspots all showed 100% sensitivity and specificity with a range of cutoffs. CONCLUSIONS We propose that this approach allows one to identify highly sensitive and specific markers for detecting dMMR CRC by NGS alone. Further studies are warranted to test whether these markers are applicable to non-CRC neoplasms.
Collapse
Affiliation(s)
- Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Eric S Christenson
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Aparna Pallavajjala
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
2
|
Gabriel PE, Compérat E, Cancel-Tassin G, Varinot J, Roumiguié M, Patard PM, Daniel G, Pfister C, Delcourt C, Gobet F, Larré S, Léon P, Durlach A, Bigot P, Carrouget J, Eymerit C, Bessède T, Lebacle C, Ferlicot S, Ruffion A, Seizilles de Mazancourt E, Decaussin-Petrucci M, Crouzet S, Matillon X, Mège-Lechevallier F, Robert G, Vuong NS, Philip M, Lang H, Mouracade P, Lindner V, Cussenot O, Rouprêt M, Seisen T. Assessment of the Prognostic and Predictive Values of the Deficient Mismatch Repair Phenotype in Patients Treated with Radical Nephroureterectomy for Upper Tract Urothelial Carcinoma. Eur Urol Oncol 2025:S2588-9311(25)00092-6. [PMID: 40307090 DOI: 10.1016/j.euo.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/22/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND AND OBJECTIVE Given the conflicting evidence currently available in the literature, our aim was to assess the prognostic and predictive values of the deficient mismatch repair (dMMR) phenotype in a large cohort of upper tract urothelial carcinoma (UTUC) patients. METHODS Based on our national network, we performed a retrospective multicenter study including 281 UTUC patients treated with radical nephroureterectomy between 2000 and 2015 at ten French hospitals. The dMMR phenotype as well as PD-L1 and PD-1 expression were determined using immunohistochemistry analyses based on 2-mm-core tissue microarrays. Multivariable Cox regression models were fitted to assess the impact of the dMMR phenotype on recurrence-free (RFS), cancer-specific (CSS), and overall (OS) survival using interaction terms to test the heterogeneity of the treatment effect of adjuvant chemotherapy (AC). Multivariable logistic regression models were also fitted to assess the impact of the dMMR phenotype on PD-L1 and PD-1 expression. KEY FINDINGS AND LIMITATIONS Overall, 76 (27.0%) patients had a dMMR phenotype, which was an independent predictor of prolonged RFS (hazard ratio [HR] = 0.41; 95% confidence interval [CI] = [0.21-0.83]; p = 0.01), CSS (HR = 0.38; 95% CI = [0.18-0.83]; p = 0.02), and OS (HR = 0.44; 95% CI = [0.22-0.89]; p = 0.02), with a significant interaction with the use of AC in multivariable Cox regression models (all pinteraction < 0.05). Subgroup analyses showed that the use of AC was significantly associated with prolonged RFS (HR = 0.14; 95% CI = [0.06-0.30]; p < 0.001), CSS (HR = 0.10; 95% CI = [0.03-0.29]; p < 0.001), and OS (HR = 0.23; 95% CI = [0.10-0.54]; p = 0.001) in non-dMMR patients only, without any significant benefit in dMMR patients (all p > 0.05). In multivariable logistic regression analyses, the dMMR phenotype was significantly associated with inverse PD-L1 (OR = 0.20; 95% CI = [0.10-0.80]; p = 0.001) and PD-1 (OR = 0.36; 95% CI = [0.16-0.79]; p = 0.01) expression. CONCLUSIONS AND CLINICAL IMPLICATIONS We observed that the dMMR phenotype was associated with favorable pathological characteristics and prognosis in UTUC patients, despite conferring decreased sensitivity to AC and lower PD-L1 or PD-1 expression.
Collapse
Affiliation(s)
- Pierre-Etienne Gabriel
- Urology, GRC 5 Predictive Onco-Urology, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Eva Compérat
- Department of Pathology, Medical University of Vienna, Vienna, Austria; GRC 5, Predictive Onco-Urology, Sorbonne University, Paris, France; CeRePP, Tenon Hospital, Paris, France
| | - Géraldine Cancel-Tassin
- GRC 5, Predictive Onco-Urology, Sorbonne University, Paris, France; CeRePP, Tenon Hospital, Paris, France
| | - Justine Varinot
- Department of Pathology, Tenon University Hospital, AP-HP, Paris, France
| | - Mathieu Roumiguié
- Department of Urology, Toulouse University Hospital, University of Toulouse UT3, Toulouse, France
| | - Pierre-Marie Patard
- Department of Urology, Toulouse University Hospital, University of Toulouse UT3, Toulouse, France
| | - Gwendoline Daniel
- Department of Pathology, Toulouse University Hospital, University of Toulouse UT3, Toulouse, France
| | - Christian Pfister
- Urology, CIC Inserm 1404, Charles Nicolle University Hospital, Rouen Normandie University, Rouen, France
| | - Clara Delcourt
- Urology, CIC Inserm 1404, Charles Nicolle University Hospital, Rouen Normandie University, Rouen, France
| | - Françoise Gobet
- Department of Pathology, Charles Nicolle University Hospital, Rouen, France
| | - Stéphane Larré
- CeRePP, Tenon Hospital, Paris, France; Department of Urology, Reims University Hospital, Reims, France
| | - Priscilla Léon
- Department of Urology, Reims University Hospital, Reims, France
| | - Anne Durlach
- Department of Pathology, Reims University Hospital, Reims, France
| | - Pierre Bigot
- Department of Urology, Angers University Hospital, Angers, France
| | - Julie Carrouget
- Department of Urology, Angers University Hospital, Angers, France
| | - Caroline Eymerit
- Department of Pathology, Angers University Hospital, Angers, France
| | - Thomas Bessède
- Department of Urology, Bicêtre University Hospital, AP-HP, Paris-Saclay University, Le Kremlin Bicêtre, France
| | - Cédric Lebacle
- Department of Urology, Bicêtre University Hospital, AP-HP, Paris-Saclay University, Le Kremlin Bicêtre, France
| | - Sophie Ferlicot
- Department of Pathology, Bicêtre University Hospital, AP-HP, Paris-Saclay University, Le Kremlin Bicêtre, France
| | - Alain Ruffion
- Department of Urology, Lyon-Sud University Hospital, Hospices Civils de Lyon, Claude Bernard University, Lyon, France
| | | | - Myriam Decaussin-Petrucci
- Department of Pathology, Lyon-Sud University Hospital, Hospices Civils de Lyon, Claude Bernard University, Lyon, France
| | - Sébastien Crouzet
- Department of Urology, Edouard Herriot University Hospital, Lyon 1 University, Lyon, France
| | - Xavier Matillon
- Department of Urology, Edouard Herriot University Hospital, Lyon 1 University, Lyon, France
| | | | - Grégoire Robert
- Department of Urology, Bordeaux Pellegrin University Hospital, Bordeaux, France
| | - Nam-Son Vuong
- Department of Urology, Bordeaux Pellegrin University Hospital, Bordeaux, France
| | - Magali Philip
- Department of Pathology, Bordeaux Pellegrin University Hospital, Bordeaux, France
| | - Hervé Lang
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - Pascal Mouracade
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - Véronique Lindner
- Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | | | - Morgan Rouprêt
- Urology, GRC 5 Predictive Onco-Urology, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; CeRePP, Tenon Hospital, Paris, France
| | - Thomas Seisen
- Urology, GRC 5 Predictive Onco-Urology, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France.
| |
Collapse
|
3
|
Lee M, Yoon H, Kim U, Kang J, Han YB, Lee KH, Lee SJ, Hong SH, Suh DH, Kim K, No JH, Kim YB, Kim H, Lee A. Improved Prognostic Stratification With 2023 International Federation of Gynecology and Obstetrics Staging in Endometrial Cancer Reflecting Poor Prognosis of Aggressive Histological Types and p53 Abnormality. J Transl Med 2025; 105:104189. [PMID: 40311877 DOI: 10.1016/j.labinv.2025.104189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/09/2025] [Accepted: 04/20/2025] [Indexed: 05/03/2025] Open
Abstract
This study compares the distribution and prognostic impact of the 2009 and 2023 International Federation of Gynecology and Obstetrics (FIGO) staging systems for endometrial cancer and their impact on the 2022 European Society for Medical Oncology (ESMO) risk classification. Patients were restaged according to the 2009 FIGO staging system, the 2023 FIGO staging system, and the 2023 FIGO staging system with molecular classification. Risk groups were assigned according to the 2022 ESMO guidelines using each staging system. Among 679 patients, 139 (20.5%) experienced stage migration when transitioning from the 2009 FIGO staging system to the 2023 FIGO staging system with molecular classification, with 121 (17.8%) upstaged and 18 (2.7%) downstaged. Most changes were from FIGO stage I to stage II, primarily due to p53 abnormality, aggressive histological type, or extensive/substantial lymphovascular space invasion. Hazard ratios for overall survival, disease-free survival, and event-free survival increased with advancing stage groups in all systems, showing the greatest differences when the 2023 FIGO staging system with molecular classification was used. The newly introduced FIGO stages IC, IIC (both representing aggressive histological types), and IICmp53abn (associated with p53 abnormality) in the 2023 FIGO staging system were associated with worse outcomes, similar to FIGO stage III. The prognostic predictability of the 2022 ESMO risk group was minimally affected by the transition from the 2009 FIGO to the 2023 FIGO staging system, as the factors introduced in the new FIGO system were already incorporated into the 2022 ESMO risk classification. Only 17 (2.5%) patients experienced a change in their assigned risk group. The 2023 FIGO staging system showed improved prognostic stratification over the 2009 FIGO staging system, particularly by reflecting the poor prognosis of aggressive histological types and p53 abnormality.
Collapse
Affiliation(s)
- Miseon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Heesoo Yoon
- Department of Pathology and Translational Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ujae Kim
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jun Kang
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeon Bi Han
- Department of Pathology and Translational Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keun Ho Lee
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Jong Lee
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sook Hee Hong
- Division of Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jae Hong No
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yong Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyojin Kim
- Department of Pathology and Translational Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Fucà G, Dell'Acqua C, Peruffo B, Lalli G, Sabatucci I, Paderno M, Di Martino G, Signorelli M, Maruccio M, Martinelli F, Lorusso D. WRN dependency in dMMR/MSI-H endometrial cancer: Clinical perspectives of a novel synthetic lethality strategy. Gynecol Oncol 2025; 195:12-15. [PMID: 40043505 DOI: 10.1016/j.ygyno.2025.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/13/2025] [Accepted: 02/23/2025] [Indexed: 04/21/2025]
Affiliation(s)
- Giovanni Fucà
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy
| | - Cristian Dell'Acqua
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
| | - Beatrice Peruffo
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
| | - Gloria Lalli
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
| | - Ilaria Sabatucci
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy
| | - Mariachiara Paderno
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy
| | - Giampaolo Di Martino
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy
| | - Mauro Signorelli
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy
| | - Matteo Maruccio
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy
| | - Fabio Martinelli
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy.
| | - Domenica Lorusso
- Unit of Gynecologic Oncology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy.
| |
Collapse
|
5
|
Buvaneswari T, Ramkumar M, Venkatesan P, Kumar RS. Leveraging Radiomics and Hybrid Quantum-Classical Convolutional Networks for Non-Invasive Detection of Microsatellite Instability in Colorectal Cancer. Mol Imaging Biol 2025; 27:227-237. [PMID: 39979579 DOI: 10.1007/s11307-025-01990-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
PURPOSE The goal of this study is to create a novel framework for identifying MSI status in colorectal cancer using advanced radiomics and deep learning strategies, aiming to enhance clinical decision-making and improve patient outcomes in oncology. PROCEDURES The study utilizes histopathological slide images from the NCT-CRC-HE-100 K and PAIP 2020 databases. Key procedures include self-attentive adversarial stain normalization for data standardization, tumor delineation via a Slimmable Transformer, and radiomics feature extraction using a hybrid quantum-classical neural network. RESULTS The proposed system reaches 99% accuracy when identifying colorectal cancer MSI status. It shows the model is good at telling the difference between MSI and MSS tumors and can be used in real medical care for cancer. CONCLUSIONS Our research shows that the new system improves colorectal cancer MSI status determination better than previous methods. Our optimized processing technology works better than other methods to divide and analyze tissue features making the system good for improving patient care decisions.
Collapse
Affiliation(s)
- T Buvaneswari
- Department of Computer Secience and Engineering, Annapoorana Engineering College (Autonomous), NH_47, Sankari Main Road, Periyaseeragapaddi, Salem, Tamil Nadu, 636 308, India.
| | - M Ramkumar
- Department of Electronics and Communication Engineering, Sri Krishna College of Engineering and Technology, Coimbatore, 641-008, Tamil Nadu, India
| | - Prabhu Venkatesan
- Department of Electronics and Communication Engineering, Vel Tech Multi Tech Dr. Rangarajan Dr. Sakunthala Engineering College, Avadi, Chennai, 600062, Tamil Nadu, India
| | - R Sarath Kumar
- Department of Electronics And Communication Engineering Department, Sri Krishna College Of Engineering And Technology, Coimbatore, 641008, Tamil Nadu, India
| |
Collapse
|
6
|
Basak U, Mukherjee S, Chakraborty S, Sa G, Dastidar SG, Das T. In-silico analysis unveiling the role of cancer stem cells in immunotherapy resistance of immune checkpoint-high pancreatic adenocarcinoma. Sci Rep 2025; 15:10355. [PMID: 40133473 PMCID: PMC11937529 DOI: 10.1038/s41598-025-93924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Although immune checkpoint (IC) inhibition is a major treatment modality in cancer-immunotherapy, multiple cancers show low response. Our in-silico exploration by mining cancer datasets using R2, available clinical trial data, and Kaplan-Meier analysis from GEPIA depicted that unlike low-responder (LR) cancers, high-responder (HR) cancers furnish higher IC expression, that upon lowering may provide better prognosis. Contrastingly, pancreatic adenocarcinoma (PAAD) demonstrated high IC expression but low immunotherapy-response. Infiltration scores from TIMER2.0 revealed higher pro-tumor immune subsets and cancer-associated fibroblasts (CAFs) while depicting lower anti-tumor immune subsets in PAAD as compared to HR lung adenocarcinoma (LUAD). Additionally, bioinformatic tool cBioportal showed lesser tumor mutational burden, mismatch repair deficiency and greater percent of driver mutations in TP53, KRAS and CDKN2A in PAAD, supporting its higher immunotherapy-resistance than LUAD. Our search for the 'key' immunotherapy response-deciding factor(s) revealed cancer stem cells (CSCs), the known contributors of therapy-resistance and immuno-evasion, to be positively correlated with above-mentioned driver mutations, pro-tumor immune and CAF subsets; and that PAAD furnished higher expression of CSC genes than LUAD. UMAP/tSNE analyses revealed that high CSC signature is positively correlated with immunotherapy-resistance genes and pro-cancer immune cells, while negatively with cytotoxic-T cells in PAAD. Our in-silico study explains the low immunotherapy-response in high IC-expressing PAAD, wherein CSC plays a pivotal role. Further exploration portrayed correlation of CSCs with immunotherapy-resistance in other LR and HR cancers too, substantiating the need for personalized CSC evaluation and targeting for successful immunotherapy outcomes.
Collapse
Affiliation(s)
- Udit Basak
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sumon Mukherjee
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sourio Chakraborty
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Gaurisankar Sa
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Shubhra Ghosh Dastidar
- Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhannagar, Kolkata, 700091, India.
| | - Tanya Das
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India.
| |
Collapse
|
7
|
Imyanitov EN, Preobrazhenskaya EV, Mitiushkina NV. Overview on biomarkers for immune oncology drugs. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002298. [PMID: 40135049 PMCID: PMC11933888 DOI: 10.37349/etat.2025.1002298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) are widely used in clinical oncology, less than half of treated cancer patients derive benefit from this therapy. Both tumor- and host-related variables are implicated in response to ICIs. The predictive value of PD-L1 expression is confined only to several cancer types, so this molecule is not an agnostic biomarker. Highly elevated tumor mutation burden (TMB) caused either by excessive carcinogenic exposure or by a deficiency in DNA repair is a reliable indicator for ICI efficacy, as exemplified by tumors with high-level microsatellite instability (MSI-H). Other potentially relevant tumor-related characteristics include gene expression signatures, pattern of tumor infiltration by immune cells, and, perhaps, some immune-response modifying somatic mutations. Host-related factors have not yet been comprehensively considered in relevant clinical trials. Microbiome composition, markers of systemic inflammation [e.g., neutrophil-to-lymphocyte ratio (NLR)], and human leucocyte antigen (HLA) diversity may influence the efficacy of ICIs. Studies on ICI biomarkers are likely to reveal modifiable tumor or host characteristics, which can be utilized to direct the antitumor immune defense. Examples of the latter approach include tumor priming to immune therapy by cytotoxic drugs and elevation of ICI efficacy by microbiome modification.
Collapse
Affiliation(s)
- Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Elena V. Preobrazhenskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Natalia V. Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
| |
Collapse
|
8
|
Jafari P, Forrest M, Segal J, Wang P, Tjota MY. Pan-Cancer Molecular Biomarkers: Practical Considerations for the Surgical Pathologist. Mod Pathol 2025; 38:100752. [PMID: 40058460 DOI: 10.1016/j.modpat.2025.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Traditional anatomic pathologic classification of cancer is based on tissue of origin and morphologic and immunohistochemical characterization of the malignant cells. With the technological improvements of massively parallel or next-generation sequencing, oncogenic drivers that are shared across different tumor types are increasingly being identified and used as pan-cancer biomarkers. This approach is reflected in the growing list of Food and Drug Administration-approved tumor-agnostic therapies, including pembrolizumab in the setting of microsatellite instability and high tumor mutational burden, larotrectinib and entrectinib for solid tumors with NTRK fusions, and combined dabrafenib-trametinib for BRAF V600E-mutated neoplasms. Several other biomarkers are currently under investigation, including fibroblast growth factor receptor (FGFR), RET, and ROS1 fusions; ERBB2 amplification; and mutations in the AKT1/2/3, NF1, RAS pathway and (mitogen-activated protein kinase (MAPK) pathway. As molecular assays are increasingly incorporated into routine tumor workup, the emergence of additional pan-cancer biomarkers is likely to be a matter more of "when" than "if." In this review, we first explore some of the conceptual and technical considerations at the intersection of surgical and molecular pathology, followed by a brief overview of both established and emerging molecular pan-cancer biomarkers and their diagnostic and clinical applications.
Collapse
Affiliation(s)
- Pari Jafari
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Megan Forrest
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Jeremy Segal
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Peng Wang
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | | |
Collapse
|
9
|
Friker LL, Perwein T, Waha A, Dörner E, Klein R, Blattner-Johnson M, Layer JP, Sturm D, Nussbaumer G, Kwiecien R, Spier I, Aretz S, Kerl K, Hennewig U, Rohde M, Karow A, Bluemcke I, Schmitz AK, Reinhard H, Hernáiz Driever P, Wendt S, Weiser A, Guerreiro Stücklin AS, Gerber NU, von Bueren AO, Khurana C, Jorch N, Wiese M, Kratz CP, Eyrich M, Karremann M, Herrlinger U, Hölzel M, Jones DTW, Hoffmann M, Pietsch T, Gielen GH, Kramm CM. MSH2, MSH6, MLH1, and PMS2 immunohistochemistry as highly sensitive screening method for DNA mismatch repair deficiency syndromes in pediatric high-grade glioma. Acta Neuropathol 2025; 149:11. [PMID: 39894875 PMCID: PMC11788232 DOI: 10.1007/s00401-025-02846-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
Pediatric high-grade glioma (pedHGG) can occur as first manifestation of cancer predisposition syndromes resulting from pathogenic germline variants in the DNA mismatch repair (MMR) genes MSH2, MSH6, MLH1, and PMS2. The aim of this study was to establish a generalized screening for Lynch syndrome and constitutional MMR deficiency (CMMRD) in pedHGG patients, as the detection of MMR deficiencies (MMRD) may enable the upfront therapeutic use of checkpoint inhibitors and identification of variant carriers in the patients' families. We prospectively enrolled 155 centrally reviewed primary pedHGG patients for MMR-immunohistochemistry (IHC) as part of the HIT-HGG-2013 trial protocol. MMR-IHC results were subsequently compared to independently collected germline sequencing data (whole exome sequencing or pan-cancer DNA panel next-generation sequencing) available in the HIT-HGG-2013, INFORM, and MNP2.0 trials. MMR-IHC could be successfully performed in 127/155 tumor tissues. The screening identified all present cases with Lynch syndrome or CMMRD (5.5%). In addition, MMR-IHC also detected cases with exclusive somatic MMR gene alterations (2.3%), including MSH2 hypermethylation as an alternative epigenetic silencing mechanism. Most of the identified pedHGG MMRD patients had no family history of MMRD, and thus, they represented index patients in their families. Cases with regular protein expression in MMR-IHC never showed evidence for MMRD in DNA sequencing. In conclusion, MMR-IHC presents a cost-effective, relatively widely available, and fast screening method for germline MMRD in pedHGG with high sensitivity (100%) and specificity (96%). Given the relatively high prevalence of previously undetected MMRD cases among pedHGG patients, we strongly recommend incorporating MMR-IHC into routine diagnostics.
Collapse
Affiliation(s)
- Lea L Friker
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany.
| | - Thomas Perwein
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
- Styrian Children's Cancer Research, Research Unit for Cancer and Inborn Errors of the Blood and Immunity in Children, Medical University of Graz, Graz, Austria
| | - Andreas Waha
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Evelyn Dörner
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Rebecca Klein
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Mirjam Blattner-Johnson
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Julian P Layer
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
- Department of Radiation Oncology, University Hospital Bonn, Bonn, Germany
| | - Dominik Sturm
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gunther Nussbaumer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Robert Kwiecien
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Isabel Spier
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
- National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Stefan Aretz
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
- National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Ulrike Hennewig
- Department of Pediatric Hematology and Oncology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Marius Rohde
- Department of Pediatric Hematology and Oncology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Axel Karow
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Erlangen, Germany
- Comprehensive Cancer Center Erlangen, Erlangen, Germany
| | - Ingmar Bluemcke
- Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Ann Kristin Schmitz
- Department of Pediatrics, Asklepios Kinderklinik Sankt Augustin, Sankt Augustin, Germany
| | - Harald Reinhard
- Department of Pediatrics, Asklepios Kinderklinik Sankt Augustin, Sankt Augustin, Germany
| | - Pablo Hernáiz Driever
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, German HIT-LOGGIC-Registry for pLGG in Children and Adolescents, Berlin, Germany
| | - Susanne Wendt
- Department of Pediatric Oncology and Hematology, University Hospital Leipzig, Leipzig, Germany
| | - Annette Weiser
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Ana S Guerreiro Stücklin
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Nicolas U Gerber
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - André O von Bueren
- Department of Pediatrics, Gynecology and Obstetrics, Division of Pediatric Hematology and Oncology, Geneva University Hospital, Geneva, Switzerland
- Department of Pediatrics, Gynecology and Obstetrics, CANSEARCH Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claudia Khurana
- Department of Pediatric Hematology and Oncology, Children's Center Bethel, University Hospital Ostwestfalen-Lippe, Bielefeld, Germany
| | - Norbert Jorch
- Department of Pediatric Hematology and Oncology, Children's Center Bethel, University Hospital Ostwestfalen-Lippe, Bielefeld, Germany
| | - Maria Wiese
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Christian P Kratz
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Matthias Eyrich
- University Children's Hospital, University Hospital Würzburg, Würzburg, Germany
| | - Michael Karremann
- Department of Pediatric and Adolescent Medicine and Mannheim Cancer Center (MCC), University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ulrich Herrlinger
- Department of Neurooncology, Center for Neurology and CIO ABCD, University Hospital Bonn, Bonn, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Marion Hoffmann
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Gerrit H Gielen
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Christof M Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
10
|
Zhang X, Wang YJ, Sun LY, Tu YX, Li Y, Jiang D. Clinicopathological Characteristics and Outcomes of Colorectal Cancer With Heterogenous Staining of Mismatch Repair Protein. Dis Colon Rectum 2025; 68:48-59. [PMID: 39329380 DOI: 10.1097/dcr.0000000000003527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
BACKGROUND Scant data are available on heterogenous staining of mismatch repair protein in colorectal cancer. OBJECTIVE This study aimed to improve insights into clinicopathologic features and prognosis of colorectal cancer harboring heterogenous mismatch repair protein staining. DESIGN A single-center retrospective observational study. SETTING This study was conducted in a tertiary referral center in China between 2014 and 2018. PATIENTS Patients with colorectal cancers with heterogenous staining of mismatch repair protein were included. MAIN OUTCOME MEASURES Clinicopathologic and molecular features and survival outcomes were analyzed. RESULTS A total of 151 of 6721 colorectal cancers (2.2%) exhibited heterogenous staining for at least 1 mismatch repair protein, with intraglandular heterogeneity being the most common pattern (89.4%). Heterogenous mutL homolog 1 staining was significantly associated with distant metastasis ( p = 0.03), whereas heterogenous mutS homolog 2 staining was associated with left-sided ( p = 0.03) and earlier pT stage tumors ( p = 0.02). The rates of microsatellite instability-high, K -ras and BRAF mutation were 12.6%, 47.3%, and 3.4%, respectively. Microsatellite instability-high was significantly associated with higher intraglandular mutS homolog 6 heterogeneity frequency ( p < 0.001) and decreased mutS homolog 6 expression level (<27.5%, p = 0.01). BRAF mutation was associated with the coexistence of intraglandular and clonal heterogeneity ( p = 0.003) and decreased PMS1 homolog 2 expression level ( p = 0.01). Multivariable analysis revealed that progression-free survival was significantly associated with tumor stage ( p = 0.003), stroma fraction ( p = 0.004), and heterogenous PMS1 homolog 2 staining ( p = 0.02). Overall survival was linked to tumor stage ( p = 0.006) and BRAF mutation ( p = 0.01). LIMITATIONS The limitations of this study include the absence of testing for mutL homolog 1 promoter methylation and mismatch repair gene mutations, its retrospective design, and insufficient data related to direct comparison with deficient mismatch repair and proficient mismatch repair colorectal cancer. CONCLUSIONS Heterogenous mismatch repair protein staining in colorectal cancer exhibits distinct associations with tumor location, stage, microsatellite instability, BRAF mutation, and prognosis. It is recommended to report mutS homolog 6 heterogeneity as it may indicate microsatellite instability-high. See Video Abstract . RESULTADOS Y CARACTERSTICAS CLNICOPATOLGICAS EN LA TINCIN HETEROGNEA DE PROTENAS REPARADORAS DE ERROR DE EMPAREJAMIENTO EN CASOS DE CNCER COLORRECTAL ANTECEDENTES:Son pocos los datos disponibles sobre la tinción heterogénea de la proteína reparadora de errores de emparejamiento en casos de cáncer colorrectal.OBJETIVO:Este estudio tuvo como objetivo el mejorar los conocimientos sobre las características clínico-patológicas y el pronóstico del cáncer colorrectal que alberga tinción heterogénea de proteínas reparadoras del emparejamiento.DISEÑO:Estudio observacional retrospectivo y unicéntrico.ÁMBITO:El presente estudio fué realizado en un centro de referencia terciario en China entre 2014 y 2018.PACIENTES:Se incluyeron cánceres colorrectales con tinción heterogénea de la proteína reparadora de errores de emparejamiento.PRINCIPALES MEDIDAS DE RESULTADOS:Se analizaron las características clínico-patológicas y moleculares como también los resultados de la sobrevida.RESULTADOS:Un total de 151 de 6721 cánceres colorrectales (2,2%) exhibieron tinción heterogénea para al menos una proteína reparadora de error de emparejamiento, siendo la héterogenicidad intraglandular el patrón más común (89,4%).La tinción heterogénea de MLH1 se asoció significativamente con metástasis a distancia (p = 0,03), mientras que la tinción heterogénea de MSH2 se asoció con tumores del lado izquierdo (p = 0,03) y en casos de estadío pT anterior (p = 0,02). Las tasas de inestabilidad alta de microsatélites, la mutación KRAS y BRAF fueron del 12,6%, 47,3% y 3,4%, respectivamente.La inestabilidad alta de microsatélites se asoció significativamente con una mayor frecuencia de heterogenicidad intraglandular de MSH6 (p <0,001) y una disminución del nivel de expresión de MSH6 (<27,5%) (P = 0,01). La mutación BRAF se asoció con la coexistencia de heterogenicidad intraglandular y clonal (p = 0,003) y una disminución del nivel de expresión de PMS2 (p = 0,01).El análisis multivariable reveló que la sobrevida libre de progresión se asoció significativamente con el estadio del tumor (p = 0,003), la fracción del estroma (p = 0,004) y la tinción heterogénea de PMS2 (p = 0,02). La sobrevida general estuvo relacionada con el estadio del tumor (p = 0,006) y la mutación BRAF (p = 0,01).LIMITACIONES:Las limitaciones del presente estudio incluyen la ausencia de pruebas para la metilación del promotor MLH1 y las mutaciones del gen de reparación de errores de coincidencia. También se incluye el diseño retrospectivo y los datos insuficientes relacionados con la comparación directa de la reparación deficiente de errores de emparejamiento y los casos de cáncer colorrectal competente en la reparación de errores de emparejamiento.CONCLUSIONES:La tinción de proteínas reparadoras de errores de emparejamiento en casos de cáncer colorrectal exhibe asociaciones distintas con la localización y el estadío del tumor, la inestabilidad de los microsatélites, la mutación BRAF y el pronóstico. Se recomienda informar de la heterogenicidad de MSH6, ya que puede indicar una inestabilidad alta de los microsatélites. (Traducción-Dr. Xavier Delgadillo ).
Collapse
Affiliation(s)
- Xian Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Department of Pathology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Yu-Jue Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Lin-Yong Sun
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yin-Xia Tu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Li
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Department of Pathology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Mete O, Boucher A, Schrader KA, Abdel-Rahman O, Bahig H, Ho C, Hasan OK, Lemieux B, Winquist E, Wong R, Wu J, Chau N, Ezzat S. Consensus Statement: Recommendations on Actionable Biomarker Testing for Thyroid Cancer Management. Endocr Pathol 2024; 35:293-308. [PMID: 39579327 DOI: 10.1007/s12022-024-09836-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2024] [Indexed: 11/25/2024]
Abstract
Thyroid cancer management is rapidly changing. The identification of actionable biomarkers through both germline and somatic testing are now an integral part of directing patient management. However, deficiencies and disparities within existing thyroid cancer biomarker test approaches are resulting in inconsistent application for patient care. An expert panel was convened to create consensus biomarker testing algorithms and recommendations on actionable biomarker testing for patients diagnosed with medullary thyroid cancer, non-anaplastic follicular cell-derived thyroid cancer, or anaplastic follicular cell-derived thyroid cancer who may benefit from targeted therapies. A review of international guidelines was performed to determine the current state, and a literature review was carried out to further evaluate the evidence supporting the use of actionable biomarkers in patients diagnosed with thyroid cancer. Thyroid biomarker-related gaps impacting patient care were also discussed, with an emphasis on the importance of a multidisciplinary team approach for optimal patient care. The recommendations are presented with the aim to help physicians navigate the current thyroid cancer biomarker testing landscape with its many challenges, balancing aspirational care with what is practical and feasible in terms of economic realities and jurisdictional constraints. By remaining therapy-agnostic, these algorithms and recommendations are broadly applicable.
Collapse
Affiliation(s)
- Ozgur Mete
- Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto, Toronto, Ontario, M5G 2C4, Canada.
| | - Andrée Boucher
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | | | - Omar Abdel-Rahman
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Houda Bahig
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montreal, Quebec, Canada
| | - Cheryl Ho
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Olfat Kamel Hasan
- Department of Medicine and Department of Medical Imaging, McMaster University, Hamilton, Ontario, Canada
| | - Bernard Lemieux
- Department of Internal Medicine, Centre Hospitalier Universite de Montreal, Montreal, Quebec, Canada
| | - Eric Winquist
- Department of Oncology, Western University, London, Ontario, Canada
| | - Ralph Wong
- Department of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
| | - Jonn Wu
- Department of Radiation Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Nicole Chau
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Shereen Ezzat
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|
12
|
O’Connor CA, Harrold E, Lin D, Walch H, Gazzo A, Ranganathan M, Kane S, Keane F, Schoenfeld J, Moss D, Thurtle-Schmidt DM, Suehnholz SP, Chakravarty D, Balogun F, Varghese A, Yu K, Kelsen D, Latham A, Weigelt B, Park W, Stadler Z, O’Reilly EM. Lynch Syndrome and Somatic Mismatch Repair Variants in Pancreas Cancer. JAMA Oncol 2024; 10:1511-1518. [PMID: 39235819 PMCID: PMC11378065 DOI: 10.1001/jamaoncol.2024.3651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 05/20/2024] [Indexed: 09/06/2024]
Abstract
Importance Microsatellite (MS) instability (MSI-H) occurs frequently in Lynch syndrome (LS)-associated tumors and is associated with response to immune checkpoint blockade (ICB) therapy. MSI-H is conferred by germline or somatic variants in mismatch repair genes. The contribution of somatic oncogenesis to MSI-H in pancreatic cancer (PC) is unknown. Objective To evaluate an LS-related PC cohort to define clinicogenomic features, describe somatic MSI-H cases (germline negative), characterize response to ICB, and guide preferred MS testing methods. Design, Setting, and Participants This single-institution, retrospective analysis was conducted from March 2012 to July 2023 at Memorial Sloan Kettering Cancer Center and included 55 patients with PC and either an LS germline pathogenic variant (gPV) or somatic mismatch repair (MMR) variant. Main Outcomes and Measures Composite MMR and MS status determined using orthogonal methods. An artificial intelligence classifier was used to account for low-cellularity specimens. Demographic and clinical data were abstracted from medical record. Zygosity status and somatic comutation landscape analyzed. Results Fifty-five patients (23 women [42%]) had PC and an MMR variant: 32 (58%) had LS (LS cohort) and 23 (42%) had a somatic MMR variant (no germline pathogenic variant, somatic MMR cohort). In the LS cohort, 10 (31%) had gMSH2, 9 (28%) gMSH6, 8 (25%) gPMS2, 4 (13%) gMLH1, 1 (3%) gEPCAM. The median age at diagnosis was 68 years (range, 45-88 years). For composite MS status, 17 (59%) were MSI-H, 12 (41%) MS stable, and 3 MS unknown. Five cases were reclassified as MSI-H by the artificial intelligence classifier. In the somatic MMR cohort, 11 (48%) had MSH6, 7 (30%) MLH1, 3 (13%) MSH2, and 2 (9%) PMS2. The median age at diagnosis was 72 years (range, 66-85 years). For composite MS status, 10 (43%) were MSI-H, 11 (48%) MS stable, and 2 (9%) MS indeterminate. Six cases were reclassified as MSI-H by the artificial intelligence classifier. For the LS and somatic MMR cohorts, 20 received ICB (n = 17 MSI-H). The median ICB duration was 27.7 months (95% CI, 11.5 to not reached); the disease control rate was 80%. Conclusion The results of this cross-sectional study suggest that MSI-H occurs due to LS or somatic oncogenesis in PC. Orthogonal MS testing is key in PC; the artificial intelligence classifier reclassified approximately 20% of cases, most of which were low cellularity. ICB for patients with LS or somatic MSI-H PC provided significant benefit.
Collapse
Affiliation(s)
- Catherine A. O’Connor
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Biology, Davidson College, Davidson, North Carolina
| | - Emily Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Mater Misericordiae University Hospital Dublin, Dublin, Ireland
| | - David Lin
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering, New York, New York
| | - Henry Walch
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering, New York, New York
| | - Andrea Gazzo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Megha Ranganathan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah Kane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua Schoenfeld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Drew Moss
- Mount Sinai Morningside West, New York, New York
| | | | - Sarah P. Suehnholz
- Human Oncology Pathogenesis Program, Sloan Kettering Institute, New York, New York
| | - Debyani Chakravarty
- Human Oncology Pathogenesis Program, Sloan Kettering Institute, New York, New York
| | - Fiyinfolu Balogun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Anna Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kenneth Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - David Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Alicia Latham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
| | - Britta Weigelt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Zsofia Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Eileen M. O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
13
|
Lin DI, Quintanilha JCF, Danziger N, Lang L, Levitan D, Hayne C, Hiemenz MC, Smith DL, Albacker LA, Leibowitz J, Mata DA, Decker B, Lakis S, Patel NR, Graf RP, Elvin JA, Ross JS, Pattani V, Huang RSP, Wehn AK. Pan-tumor validation of a NGS fraction-based MSI analysis as a predictor of response to Pembrolizumab. NPJ Precis Oncol 2024; 8:204. [PMID: 39277692 PMCID: PMC11401835 DOI: 10.1038/s41698-024-00679-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024] Open
Abstract
Microsatellite instability high (MSI-H) and mismatch repair deficient (dMMR) tumor status have been demonstrated to predict patient response to immunotherapies. We developed and validated a next-generation sequencing (NGS)-based companion diagnostic (CDx) to detect MSI-H solid tumors via a comprehensive genomic profiling (CGP) assay, FoundationOne®CDx (F1CDx). To determine MSI status, F1CDx calculates the fraction of unstable microsatellite loci across >2000 loci using a fraction-based (FB) analysis. Across solid tumor types, F1CDx demonstrated a high analytical concordance with both PCR (n = 264) and IHC (n = 279) with an overall percent agreement (OPA) of 97.7% and 97.8%, respectively. As part of a retrospective bridging clinical study from KEYNOTE-158 Cohort K and KEYNOTE-164, patients with MSI-H tumors as determined by F1CDx demonstrated an objective response rate (ORR) of 43.0% to pembrolizumab. In real-world cancer patients from a deidentified clinicogenomic database, F1CDx was at least equivalent in assessing clinical outcome following immunotherapy compared with MMR IHC. Demonstrated analytical and clinical performance of F1CDx led to the pan-tumor FDA approval in 2022 of F1CDx to identify MSI-H solid tumor patients for treatment with pembrolizumab. F1CDx is an accurate, reliable, and FDA-approved method for the identification of MSI-H tumors for treatment with pembrolizumab.
Collapse
Affiliation(s)
| | | | | | | | | | - Cynthia Hayne
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Liu T, Ho CL, Chen YJ, Chen PJ, Chen WL, Lee CT, Chow NH, Huang W, Chen YL. A pilot study on the detection of microsatellite instability using long mononucleotide repeats in solid tumors. Oncol Lett 2024; 28:445. [PMID: 39099584 PMCID: PMC11294907 DOI: 10.3892/ol.2024.14578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Microsatellite instability (MSI) status is a prognostic biomarker for immunotherapy in certain types of cancers, such as colorectal cancers (CRCs) and endometrial cancers (ECs). Tumors that are categorized as having high MSI (MSI-H) express high levels of neoantigens for immune recognition. The typical MSI test measures the length of short mononucleotide repeats (SMR) poly(A) 21-27; however, a limitation of this test is the difficulty in determining the shift size, particularly in endometrial cancer. To investigate an MSI detection assay with improved performance, the present study analyzed the use of poly(A) 40-44 mononucleotide repeats to detect the MSI status of 100 patients with either CRC (n=50) or EC (n=50). Capillary electrophoresis was used to evaluate five long mononucleotide repeat (LMR) markers, including poly(A) 40-A, 40-B, 40-C, 40-D and 44. The concordance rate of the LMR-MSI assay compared with an immunohistochemistry MSI detection assay was 96.0 and 95.1% for CRCs and ECs respectively, with the detection limit of the LMR-MSI assay demonstrated to be 2.5% MSI-H in HCT116 colorectal carcinoma cell lines. The LMR-MSI assay yielded a 95.1% concordance rate in ECs compared with that in the SMR-MSI test (87.8%). The LMR-MSI test identified a significantly higher mean shift size (13 bp) in MSI-H tumors compared with the SMR-MSI test (10 bp), in both EC and CRC tissue samples. Together, the present study suggested that the LMR-MSI test could potentially be a sensitive and practical technology for molecular laboratory testing, particularly in the use of immunotherapy for patients with CRCs and ECs.
Collapse
Affiliation(s)
- Tsunglin Liu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| | - Chung-Liang Ho
- Molecular Diagnosis Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan 704, Taiwan, R.O.C
- Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan, R.O.C
- Department of Laboratory Medicine, Center for Precision Medicine, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan, R.O.C
| | - Yan-Jhen Chen
- Molecular Diagnosis Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan 704, Taiwan, R.O.C
| | - Pin-Jun Chen
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan, R.O.C
| | - Wan-Li Chen
- Molecular Diagnosis Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan 704, Taiwan, R.O.C
| | - Chung-Ta Lee
- Department of Laboratory Medicine, Center for Precision Medicine, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Nan-Haw Chow
- Department of Laboratory Medicine, Center for Precision Medicine, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Wenya Huang
- Molecular Diagnosis Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan 704, Taiwan, R.O.C
- Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| | - Yi-Lin Chen
- Molecular Diagnosis Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan 704, Taiwan, R.O.C
- Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan, R.O.C
- Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, R.O.C
| |
Collapse
|
15
|
Grindedal EM, Zucknick M, Stormorken A, Rønne E, Tandstad NM, Isaacs WB, Axcrona K, Mæhle L. Outcomes of 10 years of PSA screening for prostate cancer in Norwegian men with Lynch syndrome. Prostate 2024; 84:945-953. [PMID: 38629217 DOI: 10.1002/pros.24711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Pathogenic germline variants in the mismatch repair (MMR) genes are associated with an increased risk of prostate cancer (PCa). Since 2010 we have recommended MMR carriers annual PSA testing from the age of 40. Prospective studies of the outcome of long-term PSA screening are lacking. This study aimed to investigate the incidence and characteristics of PCa in Norwegian MMR carriers attending annual PSA screening (PSA threshold >3.0 ng/mL) to evaluate whether our recommendations should be continued. METHODS This is a prospective observational study of 225 male MMR carriers who were recommended annual PSA screening by the Section of Inherited Cancer, Oslo University Hospital from 2010 and onwards. Incidence and tumor characteristics (age, PSA at diagnosis, Gleason score, TNM score) were described. IHC and MSI-analyses were done on available tumors. Standardized incidence ratio (SIR) was calculated based on data from the Cancer Registry of Norway. RESULTS Twenty-two of 225 (9.8%) had been diagnosed with PCa, including 10/69 (14.5%) MSH2 carriers and 8/61 (13.1%) MSH6 carriers. Ten of 20 (50%) tumors had Gleason score ≥4 + 3 on biopsy and 6/11 (54.5%) had a pathological T3a/b stage. Eight of 17 (47.1%) tumors showed abnormal staining on IHC and 3/13 (23.1%) were MSI-high. SIR was 9.54 (95% CI 5.98-14.45) for all MMR genes, 13.0 (95% CI 6.23-23.9) for MSH2 and 13.74 for MSH6 (95% CI 5.93-27.08). CONCLUSIONS Our results indicate that the MMR genes, and especially MSH2 and MSH6, are associated with a significant risk of PCa, and a high number of tumors show aggressive characteristics. While the impact of screening on patient outcomes remains to be more firmly established, the high SIR values we observe provide support for continued PSA screening of MSH2 and MSH6 carriers. Studies are needed to provide optimal recommendations for PSA-threshold and to evaluate whether MLH1 and PMS2 carriers should not be recommended screening.
Collapse
Affiliation(s)
| | - Manuela Zucknick
- Department of Biostatistics, Oslo Centre for Biostatistics and Epidemiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Astrid Stormorken
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Elin Rønne
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Nora M Tandstad
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - William B Isaacs
- Brady Urological Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Karol Axcrona
- Department of Urology, Akershus University Hospital, Lørenskog, Norway
- Department of Molecular Oncology, Institute of Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Lovise Mæhle
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
16
|
Emelyanova M, Ikonnikova A, Pushkov A, Pudova E, Krasnov G, Popova A, Zhanin I, Khomich D, Abramov I, Tjulandin S, Gryadunov D, Pokataev I. Mutations in Mismatch Repair Genes and Microsatellite Instability Status in Pancreatic Cancer. Cancers (Basel) 2024; 16:2111. [PMID: 38893230 PMCID: PMC11171205 DOI: 10.3390/cancers16112111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Patients with pancreatic cancer (PC) showing mismatch repair (MMR) deficiency may benefit from immunotherapy. Microsatellite instability (MSI) is a hallmark of MMR deficiency (MMR-D). Here, we estimated the prevalence of MSI in PC, investigated germline and somatic mutations in the three MMR genes (MLH1, MSH2, and MSH6), and assessed the relationship between MMR genes mutations and MSI status in PC. Clinical specimens from PC patients were analyzed using targeted next-generation sequencing, including paired normal and tumor specimens from 155 patients, tumor-only specimens from 86 patients, and normal-only specimens from 379 patients. The MSI status of 235 PCs was assessed via PCR. Pathogenic/likely pathogenic (P/LP) germline variants in the MMR genes were identified in 1.1% of patients, while somatic variants were found in 2.6% of patients. No MSI-H tumors were detected. One patient carried two variants (P (VAF = 0.57) and LP (VAF = 0.25)) simultaneously; however, their germline/somatic status remains unknown due to the investigation focusing solely on the tumor and MSI analysis was not performed for this patient. MSI is rare in PC, even in tumors with MMR genes mutations. Our findings underscore the importance of assessing tumor MMR-D status in PC patients with confirmed Lynch syndrome when deciding whether to prescribe immunotherapy.
Collapse
Affiliation(s)
- Marina Emelyanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Anna Ikonnikova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Alexander Pushkov
- Federal State Autonomous Institution “National Medical Research Center for Children’s Health” of the Ministry of Health of the Russian Federation, Moscow 119991, Russia; (A.P.); (I.Z.)
| | - Elena Pudova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - George Krasnov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Anna Popova
- N.N. Blokhin National Medical Research Center for Oncology, Ministry of Health of the Russian Federation, Moscow 115522, Russia; (A.P.); (S.T.); (I.P.)
| | - Ilya Zhanin
- Federal State Autonomous Institution “National Medical Research Center for Children’s Health” of the Ministry of Health of the Russian Federation, Moscow 119991, Russia; (A.P.); (I.Z.)
| | - Darya Khomich
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Ivan Abramov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Sergei Tjulandin
- N.N. Blokhin National Medical Research Center for Oncology, Ministry of Health of the Russian Federation, Moscow 115522, Russia; (A.P.); (S.T.); (I.P.)
| | - Dmitry Gryadunov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Ilya Pokataev
- N.N. Blokhin National Medical Research Center for Oncology, Ministry of Health of the Russian Federation, Moscow 115522, Russia; (A.P.); (S.T.); (I.P.)
- City Clinical Cancer Hospital No 1, Moscow Department of Health, Moscow 129090, Russia
| |
Collapse
|
17
|
Nádorvári ML, Lotz G, Kulka J, Kiss A, Tímár J. Microsatellite instability and mismatch repair protein deficiency: equal predictive markers? Pathol Oncol Res 2024; 30:1611719. [PMID: 38655493 PMCID: PMC11036414 DOI: 10.3389/pore.2024.1611719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
Current clinical guidelines recommend mismatch repair (MMR) protein immunohistochemistry (IHC) or molecular microsatellite instability (MSI) tests as predictive markers of immunotherapies. Most of the pathological guidelines consider MMR protein IHC as the gold standard test to identify cancers with MMR deficiency and recommend molecular MSI tests only in special circumstances or to screen for Lynch syndrome. However, there are data in the literature which suggest that the two test types may not be equal. For example, molecular epidemiology studies reported different rates of deficient MMR (dMMR) and MSI in various cancer types. Additionally, direct comparisons of the two tests revealed relatively frequent discrepancies between MMR IHC and MSI tests, especially in non-colorectal and non-endometrial cancers and in cases with unusual dMMR phenotypes. There are also scattered clinical data showing that the efficacy of immune checkpoint inhibitors is different if the patient selection was based on dMMR versus MSI status of the cancers. All these observations question the current dogma that dMMR phenotype and genetic MSI status are equal predictive markers of the immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
18
|
Aleksakhina SN, Ivantsov AO, Imyanitov EN. Agnostic Administration of Targeted Anticancer Drugs: Looking for a Balance between Hype and Caution. Int J Mol Sci 2024; 25:4094. [PMID: 38612902 PMCID: PMC11012409 DOI: 10.3390/ijms25074094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Many tumors have well-defined vulnerabilities, thus potentially allowing highly specific and effective treatment. There is a spectrum of actionable genetic alterations which are shared across various tumor types and, therefore, can be targeted by a given drug irrespective of tumor histology. Several agnostic drug-target matches have already been approved for clinical use, e.g., immune therapy for tumors with microsatellite instability (MSI) and/or high tumor mutation burden (TMB), NTRK1-3 and RET inhibitors for cancers carrying rearrangements in these kinases, and dabrafenib plus trametinib for BRAF V600E mutated malignancies. Multiple lines of evidence suggest that this histology-independent approach is also reasonable for tumors carrying ALK and ROS1 translocations, biallelic BRCA1/2 inactivation and/or homologous recombination deficiency (HRD), strong HER2 amplification/overexpression coupled with the absence of other MAPK pathway-activating mutations, etc. On the other hand, some well-known targets are not agnostic: for example, PD-L1 expression is predictive for the efficacy of PD-L1/PD1 inhibitors only in some but not all cancer types. Unfortunately, the individual probability of finding a druggable target in a given tumor is relatively low, even with the use of comprehensive next-generation sequencing (NGS) assays. Nevertheless, the rapidly growing utilization of NGS will significantly increase the number of patients with highly unusual or exceptionally rare tumor-target combinations. Clinical trials may provide only a framework for treatment attitudes, while the decisions for individual patients usually require case-by-case consideration of the probability of deriving benefit from agnostic versus standard therapy, drug availability, associated costs, and other circumstances. The existing format of data dissemination may not be optimal for agnostic cancer medicine, as conventional scientific journals are understandably biased towards the publication of positive findings and usually discourage the submission of case reports. Despite all the limitations and concerns, histology-independent drug-target matching is certainly feasible and, therefore, will be increasingly utilized in the future.
Collapse
Affiliation(s)
- Svetlana N. Aleksakhina
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
| | - Alexander O. Ivantsov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
19
|
Grillo F, Paudice M, Pigozzi S, Dono M, Lastraioli S, Lugaresi M, Bozzano S, Tognoni C, Ali M, Sciallero S, Puccini A, Fassan M, Mastracci L. BRAF V600E immunohistochemistry can reliably substitute BRAF molecular testing in the Lynch syndrome screening algorithm in colorectal cancer. Histopathology 2024; 84:877-887. [PMID: 38173291 DOI: 10.1111/his.15133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
AIMS The Lynch syndrome (LS) screening algorithm requires BRAF testing as a fundamental step to distinguish sporadic from LS-associated colorectal carcinomas (CRC). BRAF testing by immunohistochemistry (IHC) has shown variable results in the literature. Our aim was to analyse concordance between BRAFV600E IHC and BRAF molecular analysis in a large, mono-institutional CRC whole-slide, case series with laboratory validation. METHODS AND RESULTS MisMatch repair (MMR) protein (hMLH1, hPMS2, hMSH2, and hMSH6) and BRAFV600E IHC were performed on all unselected cases of surgically resected CRCs (2018-2023). An in-house validation study for BRAFV600E IHC was performed in order to obtain optimal IHC stains. BRAFVV600E IHC was considered negative (score 0), positive (scores 2-3), and equivocal (score 1). Interobserver differences in BRAFV600E IHC scoring were noted in the first 150 cases prospectively collected. Nine-hundred and ninety CRCs cases (830 proficient (p)MMR/160 deficient (d)MMR) were included and all cases performed BRAFV600E IHC (BRAFV600E IHC-positive 13.5% of all series; 66.3% dMMR cases; 3.4% pMMR cases), while 333 also went to BRAF mutation analysis. Optimal agreement in IHC scoring between pathologists (P < 0.0001) was seen; concordance between BRAFV600E IHC and BRAF molecular analysis was extremely high (sensitivity 99.1%, specificity 99.5%; PPV 99.1%, and NPV 99.5%). Discordant cases were reevaluated; 1 score 3 + IHC/wildtype case was an interpretation error and one score 0 IHC/mutated case was related to heterogenous BRAFV600E IHC expression. Among the 12 IHC-equivocal score 1+ cases (which require BRAF molecular analysis), three were BRAF-mutated and nine BRAF-wildtype. CONCLUSION BRAFV600E IHC can be used as a reliable surrogate of molecular testing after stringent in-house validation.
Collapse
Affiliation(s)
- Federica Grillo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Michele Paudice
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Simona Pigozzi
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Maria Dono
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sonia Lastraioli
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marialuisa Lugaresi
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Division of Thoracic Surgery, Maria Cecilia Hospital, GVM Care & Research Group, Ravenna, Italy
| | - Silvia Bozzano
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Camilla Tognoni
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Murad Ali
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Stefania Sciallero
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Puccini
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University Hospital of Padua, Padua, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Luca Mastracci
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| |
Collapse
|
20
|
Nádorvári ML, Kenessey I, Kiss A, Barbai T, Kulka J, Rásó E, Tímár J. Comparison of standard mismatch repair deficiency and microsatellite instability tests in a large cancer series. J Transl Med 2024; 22:150. [PMID: 38350968 PMCID: PMC10863158 DOI: 10.1186/s12967-024-04960-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND The tumor-agnostic indication of immune checkpoint inhibitors to treat cancers with mismatch repair deficiency (dMMR)/microsatellite instability (MSI) increased the demand for such tests beyond Lynch syndrome. International guideline recommendations accept immunohistochemistry (IHC) for dMMR or molecular techniques (PCR or NGS) for MSI status determinations considering the two tests are equal, although there are scattered reports contradicting to this presumption. MATERIALS AND METHODS Here we have directly compared four protein MMR immunohistochemistry (IHC) to MSI Pentaplex PCR test in a large cancer patient cohort (n = 1306) of our diagnostic center where the two tests have been run parallel in 703 cases. RESULTS In this study we have found a high discrepancy rate (19.3%) of the two tests which was independent of the tumor types. The MSI PCR sensitivity for MMR IHC status was found to be very low resulting in a relatively low positive and negative predicting values. As a consequence, the correlation of the two tests was low (kappa < 0.7). During analysis of the possible contributing factors of this poor performance, we have excluded low tumor percentage of the samples, but identified dMMR phenotypes (classic versus non-classic or unusual) as possible contributors. CONCLUSION Although our cohort did not include samples with identified technical errors, our data strongly support previous reports that unidentified preanalytical factors might have the major influence on the poor performance of the MSI PCR and MMR IHC. Furthermore, the case is open whether the two test types are equally powerful predictive markers of immunotherapies.
Collapse
Affiliation(s)
- Maja L Nádorvári
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - István Kenessey
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - András Kiss
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Barbai
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Erzsébet Rásó
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
21
|
Rüschoff J, Schildhaus HU, Rüschoff JH, Jöhrens K, Bocker Edmonston T, Dietmaier W, Bläker H, Baretton G, Horst D, Dietel M, Hartmann A, Klauschen F, Merkelbach-Bruse S, Stenzinger A, Schöniger S, Tiemann M, Weichert W, Büttner R. Testing for deficient mismatch repair and microsatellite instability : A focused update. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:61-70. [PMID: 37874379 PMCID: PMC10713762 DOI: 10.1007/s00292-023-01208-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 10/25/2023]
Abstract
Testing to detect mismatch repair deficiency (dMMR) and high-grade microsatellite instability (MSI-H) has become an integral part of the routine diagnostic workup for colorectal cancer (CRC). While MSI was initially considered to be a possible indicator of a hereditary disposition to cancer (Lynch syndrome, LS), today the prediction of the therapy response to immune checkpoint inhibitors (ICI) is in the foreground. Corresponding recommendations and testing algorithms are available for use in primary diagnosis (reviewed in: Rüschoff et al. 2021).Given the increasing importance for routine use and the expanding indication spectrum of ICI therapies for non-CRCs, such as endometrial, small intestinal, gastric, and biliary tract cancers, an updated review of dMMR/MSI testing is presented. The focus is on the challenges in the assessment of immunohistochemical stains and the value of PCR-based procedures, considering the expanded ICI indication spectrum. A practice-oriented flowchart for everyday diagnostic decision-making is provided that considers new data on the frequency and type of discordances between MMR-IHC and MSI-PCR findings, and the possible role of Next Generation Sequencing in clarifying them. Reference is made to the significance of systematic quality assurance measures (e.g., QuIP MSI portal and multicenter proficiency testing), including regular continued training and education.
Collapse
Affiliation(s)
- Josef Rüschoff
- Discovery Life Sciences Biomarker GmbH and North Hesse Pathology, Germaniastr. 7, 34119, Kassel, Germany.
| | - Hans-Ulrich Schildhaus
- Discovery Life Sciences Biomarker GmbH and North Hesse Pathology, Germaniastr. 7, 34119, Kassel, Germany
| | - Jan Hendrik Rüschoff
- Institute of Pathology and Molecular Pathology, Zürich University Hospital, Schmelzbergstrasse 12, 8091, Zürich, Switzerland
| | - Korinna Jöhrens
- Institute of Pathology, Carl Gustav Carus University Hospital, Fetscherstr. 74, 01307, Dresden, Germany
| | - Tina Bocker Edmonston
- Department of Pathology, Cooper University Health Care, 401 Haddon Ave, 08103, Camden, NJ, USA
| | - Wolfgang Dietmaier
- Institute of Pathology/Center for Molecular Pathology Diagnosis, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Hendrik Bläker
- Institute for Pathology, Leipzig University Hospital, Leipzig, Germany
| | - Gustavo Baretton
- Institute of Pathology, Carl Gustav Carus University Hospital, Fetscherstr. 74, 01307, Dresden, Germany
| | - David Horst
- Institute of Pathology, Charité University Hospital, Central Campus, Charitéplatz 1, 10117, Berlin, Germany
| | - Manfred Dietel
- Institute of Pathology, Charité University Hospital, Central Campus, Charitéplatz 1, 10117, Berlin, Germany
| | - Arndt Hartmann
- Pathological Institute, University of Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Frederick Klauschen
- Pathological Institute, Ludwig Maximilian University of Munich, Thalkirchner Str. 36, 80337, Munich, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, Cologne University Hospital, Kerpener Str. 62, 50937, Cologne, Germany
| | - Albrecht Stenzinger
- Pathological Institute, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Sandra Schöniger
- Discovery Life Sciences Biomarker GmbH and North Hesse Pathology, Germaniastr. 7, 34119, Kassel, Germany
| | - Markus Tiemann
- Hamburg Institute of Hematopathology, Fangdieckstr. 75a, 22547, Hamburg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
| | - Reinhard Büttner
- Institute of Pathology, Cologne University Hospital, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
22
|
Grillo F, Ali M, Paudice M, Pigozzi S, Anselmi G, Scabini S, Sciallero S, Piol N, Mastracci L. Impact of formalin fixation on mismatch repair protein evaluation by immunohistochemistry. Virchows Arch 2023; 483:677-685. [PMID: 37773452 PMCID: PMC10673985 DOI: 10.1007/s00428-023-03661-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023]
Abstract
Mismatch repair/microsatellite instability (MMR/MSI) status in colorectal cancer (CRC) has become fundamental as a diagnostic, prognostic, and predictive factor. MMR immunohistochemistry (IHC) is considered a simple and reliable approach; however, its effectiveness depends on pre-analytic factors. Aim of this study was to investigate the impact of different fixation times/protocols on MMR protein IHC quality. Left over tissue from surgically resected CRC samples (cold ischemia time < 30 min) where fixed as follows: standard formalin fixation (24-48 h); hypo-fixation (<20 h); hyper-fixation (>90 h); cold (4°C) fixation (24-48 h); standard fixation for small sample size (0.5×0.5 cm). Samples for each group were collected from 30 resected CRC and the following parameters were evaluated on 600 immunohistochemical stains: intensity of expression; patchiness of staining; presence of central artefact. Forty-six immunoreactions were inadequate (score 0 intensity), the majority regarding MLH1 or PMS2 in the hypo-fixation group (47.8%), followed by the hyper-fixation group (28.1%); cold formalin fixation showed the least inadequate cases. Patchiness and central artefact were more frequent in hypo-fixation and standard fixation group compared to the others. MLH1 (closely followed by PMS2) performed worse with regard to immunostaining intensity (p=0.0002) in the standard and in the hypo-fixation group (p< 0.00001). Using a small sample size improved patchiness/central artefacts. This is the first study specifically created to evaluate the impact of fixation on MMR protein IHC, showing that both formalin hypo- and hyper-fixation can cause problems; 24-h formalin fixation as well as cold (4°C) formalin fixation are recommended for successful IHC MMR evaluation.
Collapse
Affiliation(s)
- Federica Grillo
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Murad Ali
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michele Paudice
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simona Pigozzi
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giorgia Anselmi
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefano Scabini
- Oncological Surgical Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefania Sciallero
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Nataniele Piol
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Mastracci
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
23
|
Bacher JW, Udho EB, Strauss EE, Vyazunova I, Gallinger S, Buchanan DD, Pai RK, Templeton AS, Storts DR, Eshleman JR, Halberg RB. A Highly Sensitive Pan-Cancer Test for Microsatellite Instability. J Mol Diagn 2023; 25:806-826. [PMID: 37544360 PMCID: PMC10629437 DOI: 10.1016/j.jmoldx.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/08/2023] Open
Abstract
Microsatellite instability (MSI) is an evolving biomarker for cancer detection and treatment. MSI was first used to identify patients with Lynch syndrome, a hereditary form of colorectal cancer (CRC), but has recently become indispensable in predicting patient response to immunotherapy. To address the need for pan-cancer MSI detection, a new multiplex assay was developed that uses novel long mononucleotide repeat (LMR) markers to improve sensitivity. A total of 469 tumor samples from 20 different cancer types, including 319 from patients with Lynch syndrome, were tested for MSI using the new LMR MSI Analysis System. Results were validated by using deficient mismatch repair (dMMR) status according to immunohistochemistry as the reference standard and compared versus the Promega pentaplex MSI panel. The sensitivity of the LMR panel for detection of dMMR status by immunohistochemistry was 99% for CRC and 96% for non-CRC. The overall percent agreement between the LMR and Promega pentaplex panels was 99% for CRC and 89% for non-CRC tumors. An increased number of unstable markers and the larger size shifts observed in dMMR tumors using the LMR panel increased confidence in MSI determinations. The LMR MSI Analysis System expands the spectrum of cancer types in which MSI can be accurately detected.
Collapse
Affiliation(s)
- Jeffery W Bacher
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin; Department of Medicine, University of Wisconsin, Madison, Wisconsin.
| | - Eshwar B Udho
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | | | - Irina Vyazunova
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | - Steven Gallinger
- Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Rish K Pai
- Health Science Research, Mayo Clinic, Scottsdale, Arizona
| | | | - Douglas R Storts
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | - James R Eshleman
- School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Richard B Halberg
- Department of Medicine, University of Wisconsin, Madison, Wisconsin; Department of Oncology, McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.
| |
Collapse
|
24
|
Mulkidjan RS, Saitova ES, Preobrazhenskaya EV, Asadulaeva KA, Bubnov MG, Otradnova EA, Terina DM, Shulga SS, Martynenko DE, Semina MV, Belogubova EV, Tiurin VI, Amankwah PS, Martianov AS, Imyanitov EN. ALK, ROS1, RET and NTRK1-3 Gene Fusions in Colorectal and Non-Colorectal Microsatellite-Unstable Cancers. Int J Mol Sci 2023; 24:13610. [PMID: 37686416 PMCID: PMC10488195 DOI: 10.3390/ijms241713610] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
This study aimed to conduct a comprehensive analysis of actionable gene rearrangements in tumors with microsatellite instability (MSI). The detection of translocations involved tests for 5'/3'-end expression imbalance, variant-specific PCR and RNA-based next generation sequencing (NGS). Gene fusions were detected in 58/471 (12.3%) colorectal carcinomas (CRCs), 4/69 (5.8%) gastric cancers (GCs) and 3/65 (4.6%) endometrial cancers (ECs) (ALK: 8; RET: 12; NTRK1: 24; NTRK2: 2; NTRK3: 19), while none of these alterations were observed in five cervical carcinomas (CCs), four pancreatic cancers (PanCs), three cholangiocarcinomas (ChCs) and two ovarian cancers (OCs). The highest frequency of gene rearrangements was seen in KRAS/NRAS/BRAF wild-type colorectal carcinomas (53/204 (26%)). Surprisingly, as many as 5/267 (1.9%) KRAS/NRAS/BRAF-mutated CRCs also carried tyrosine kinase fusions. Droplet digital PCR (ddPCR) analysis of the fraction of KRAS/NRAS/BRAF mutated gene copies in kinase-rearranged tumors indicated that there was simultaneous co-occurrence of two activating events in cancer cells, but not genetic mosaicism. CRC patients aged above 50 years had a strikingly higher frequency of translocations as compared to younger subjects (56/365 (15.3%) vs. 2/106 (1.9%), p = 0.002), and this difference was particularly pronounced for tumors with normal KRAS/NRAS/BRAF status (52/150 (34.7%) vs. 1/54 (1.9%), p = 0.001). There were no instances of MSI in 56 non-colorectal tumors carrying ALK, ROS1, RET or NTRK1 rearrangements. An analysis of tyrosine kinase gene translocations is particularly feasible in KRAS/NRAS/BRAF wild-type microsatellite-unstable CRCs, although other categories of tumors with MSI also demonstrate moderate occurrence of these events.
Collapse
Affiliation(s)
- Rimma S. Mulkidjan
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Evgeniya S. Saitova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Elena V. Preobrazhenskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Karimat A. Asadulaeva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Mikhail G. Bubnov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Ekaterina A. Otradnova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Darya M. Terina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Sofia S. Shulga
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Darya E. Martynenko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Maria V. Semina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Evgeniya V. Belogubova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Vladislav I. Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Priscilla S. Amankwah
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
| | - Aleksandr S. Martianov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (R.S.M.)
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
25
|
Vanoli A, Guerini C, Arpa G, Klersy C, Grillo F, Casadei Gardini A, De Hertogh G, Ferrante M, Moens A, Furlan D, Sessa F, Quaquarini E, Lenti MV, Neri G, Macciomei MC, Fassan M, Cascinu S, Paulli M, Graham RP, Di Sabatino A. Mismatch repair deficiency as prognostic factor for stage III small bowel adenocarcinoma: A multicentric international study. Dig Liver Dis 2023; 55:1261-1269. [PMID: 37236851 DOI: 10.1016/j.dld.2023.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Small bowel adenocarcinoma (SBA) is a rare cancer with an aggressive behavior. No study has specifically addressed the putative prognostic role of mismatch repair status in stage III SBAs. AIMS We aimed to investigate whether mismatch repair deficiency is associated with cancer-specific survival in a Western cohort of patients with stage III SBAs. METHODS In this retrospective multicentric international cohort study, we enrolled 70 patients who underwent surgically resection for stage III SBAs and we analyzed the frequency of mismatch repair deficiency, tested by immunohistochemistry for mismatch repair proteins and by polymerase chain reaction for microsatellite instability, and its association with cancer-specific survival and other clinic-pathologic factors. RESULTS We found sixteen (23%) patients with mismatch repair deficient adenocarcinoma, without discordance between immunohistochemical and polymerase chain reaction for microsatellite instability analyses. Mismatch repair deficiency proved to be associated with a better outcome both at univariable analysis (hazard ratio: 0.28, 95% confidence interval: 0.08-0.91, p: 0.035) and in bivariable models adjusted for patient age or gender, tumor site, pT4 stage, tumor budding, and perineural invasion. CONCLUSION This study highlights the importance of testing mismatch repair status to improve prognostic stratification in stage III SBAs.
Collapse
Affiliation(s)
- Alessandro Vanoli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy; Unit of Anatomic Pathology, Fondazione IRCCS San Matteo Hospital, Pavia 27100, Italy
| | - Camilla Guerini
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy
| | - Giovanni Arpa
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy
| | - Catherine Klersy
- Clinical Epidemiology and Biometry, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia 27100, Italy
| | - Federica Grillo
- Pathology Unit, Department of Surgical and Diagnostic Sciences, University of Genoa and Ospedale Policlinico San Martino University Hospital, Genoa 16132, Italy
| | - Andrea Casadei Gardini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan 20132, Italy
| | - Gert De Hertogh
- Department of Pathology, KU Leuven University Hospitals, Leuven 3000, Belgium
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals, KU Leuven, Leuven 3000, Belgium
| | - Annick Moens
- Department of Gastroenterology and Hepatology, University Hospitals, KU Leuven, Leuven 3000, Belgium
| | - Daniela Furlan
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Fausto Sessa
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Erica Quaquarini
- Medical Oncology Unit, ICS Maugeri-IRCCS SpA SB, Pavia 27100, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine, University of Pavia, San Matteo Hospital Foundation, Pavia 27100, Italy
| | - Giuseppe Neri
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy
| | | | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine, DIMED, University of Padua, Padua 35122, Italy; Veneto Institute of Oncology, IOV-IRCCS, Padua 35128, Italy
| | - Stefano Cascinu
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan 20132, Italy
| | - Marco Paulli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia 27100, Italy; Unit of Anatomic Pathology, Fondazione IRCCS San Matteo Hospital, Pavia 27100, Italy
| | | | - Antonio Di Sabatino
- Department of Internal Medicine, University of Pavia, San Matteo Hospital Foundation, Pavia 27100, Italy.
| |
Collapse
|
26
|
Rüschoff J, Schildhaus HU, Rüschoff JH, Jöhrens K, Bocker-Edmonston T, Dietmaier W, Bläker H, Baretton G, Horst D, Dietel M, Hartmann A, Klauschen F, Merkelbach-Bruse S, Stenzinger A, Schöniger S, Tiemann M, Weichert W, Büttner R. [Testing deficient mismatch repair and microsatellite instability : A focused update. German version]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:301-310. [PMID: 37548948 PMCID: PMC10457237 DOI: 10.1007/s00292-023-01209-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 08/08/2023]
Abstract
Testing to detect mismatch repair deficiency (dMMR) and high-grade microsatellite instability (MSI-H) has become an integral part of the routine diagnostic workup for colorectal cancer (CRC). While MSI was initially considered to be a possible indicator of a hereditary disposition to cancer (Lynch syndrome, LS), today the prediction of the therapy response to immune checkpoint inhibitors (ICI) is in the foreground. Corresponding recommendations and testing algorithms are available for use in primary diagnosis (reviewed in: Rüschoff et al. 2021).Given the increasing importance for routine use and the expanding indication spectrum of ICI therapies for non-CRCs, such as endometrial, small intestinal, gastric, and biliary tract cancers, an updated review of dMMR/MSI testing is presented. The focus is on the challenges in the assessment of immunohistochemical stains and the value of PCR-based procedures, considering the expanded ICI indication spectrum. A practice-oriented flowchart for everyday diagnostic decision-making is provided that considers new data on the frequency and type of discordances between MMR-IHC and MSI-PCR findings, and the possible role of Next Generation Sequencing in clarifying them. Reference is made to the significance of systematic quality assurance measures (e.g., QuIP MSI portal and multicenter proficiency testing), including regular continued training and education.
Collapse
Affiliation(s)
- Josef Rüschoff
- Discovery Life Sciences Biomarker GmbH und Pathologie Nordhessen, Germaniastr. 7, 34119, Kassel, Deutschland.
| | - Hans-Ulrich Schildhaus
- Discovery Life Sciences Biomarker GmbH und Pathologie Nordhessen, Germaniastr. 7, 34119, Kassel, Deutschland
| | - Jan Hendrik Rüschoff
- Institut für Pathologie und Molekularpathologie, Universitätsspital Zürich, Zürich, Schweiz
| | - Korinna Jöhrens
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus Dresden, Dresden, Deutschland
| | | | - Wolfgang Dietmaier
- Institut für Pathologie/Zentrum für molekularpathologische Diagnostik, Universität Regensburg, Regensburg, Deutschland
| | - Hendrik Bläker
- Institut für Pathologie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Gustavo Baretton
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus Dresden, Dresden, Deutschland
| | - David Horst
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Deutschland
| | - Manfred Dietel
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Deutschland
| | - Arndt Hartmann
- Pathologisches Institut, Universität Erlangen-Nürnberg, Erlangen, Deutschland
| | - Frederick Klauschen
- Pathologisches Institut, Ludwig-Maximilians-Universität München, München, Deutschland
| | | | - Albrecht Stenzinger
- Pathologisches Institut, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - Sandra Schöniger
- Discovery Life Sciences Biomarker GmbH und Pathologie Nordhessen, Germaniastr. 7, 34119, Kassel, Deutschland
| | - Markus Tiemann
- Institut für Hämatopathologie Hamburg, Hamburg, Deutschland
| | - Wilko Weichert
- Institut für Pathologie, Technische Universität München, München, Deutschland
| | - Reinhard Büttner
- Institut für Pathologie, Universitätsklinikum Köln, Köln, Deutschland
| |
Collapse
|
27
|
Grillo F, Paudice M, Gambella A, Bozzano S, Sciallero S, Puccini A, Lastraioli S, Dono M, Parente P, Vanoli A, Angerilli V, Fassan M, Mastracci L. Evaluating mismatch repair deficiency in colorectal cancer biopsy specimens. Histochem Cell Biol 2023; 160:113-125. [PMID: 37284845 PMCID: PMC10386921 DOI: 10.1007/s00418-023-02202-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 06/08/2023]
Abstract
Mismatch repair (MMR) testing on all new cases of colorectal cancer (CRC) has customarily been preferably performed on surgical specimens, as more tissue is available; however, new clinical trials for the use of immune checkpoint inhibitors in the neoadjuvant setting require MMR testing on biopsy samples. This study aims at identifying advantages, disadvantages and any potential pitfalls in MMR evaluation on biopsy tissue and how to cope with them. The study is prospective-retrospective, recruiting 141 biopsies (86 proficient (p)MMR and 55 deficient (d)MMR) and 97 paired surgical specimens (48 pMMR; 49 dMMR). In biopsy specimens, a high number of indeterminate stains was observed, in particular for MLH1 (31 cases, 56.4%). The main reasons were a punctate nuclear expression of MLH1, relatively weak MLH1 nuclear expression compared to internal controls, or both (making MLH1 loss difficult to interpret), which was solved by reducing primary incubation times for MLH1. A mean of ≥ 5 biopsies had adequate immunostains, compared to ≤ 3 biopsies in inadequate cases. Conversely, surgical specimens rarely suffered from indeterminate reactions, while weaker staining intensity (p < 0.007) for MLH1 and PMS2 and increased patchiness grade (p < 0.0001) were seen. Central artefacts were almost exclusive to surgical specimens. MMR status classification was possible in 92/97 matched biopsy/resection specimen cases, and all of these were concordant (47 pMMR and 45 dMMR). Evaluation of MMR status on CRC biopsy samples is feasible, if pitfalls in interpretation are known, making laboratory-specific appropriate staining protocols fundamental for high-quality diagnoses.
Collapse
Affiliation(s)
- F Grillo
- IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genoa, Italy.
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy.
| | - M Paudice
- IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - A Gambella
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - S Bozzano
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - S Sciallero
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - A Puccini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - S Lastraioli
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - M Dono
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - P Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - A Vanoli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
- Anatomic Pathology Unit, Fondazione IRCCS San Matteo Hospital, Pavia, Italy
| | - V Angerilli
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua, Italy
| | - M Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - L Mastracci
- IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| |
Collapse
|
28
|
Carsote M, Turturea IF, Turturea MR, Valea A, Nistor C, Gheorghisan-Galateanu AA. Pathogenic Insights into DNA Mismatch Repair (MMR) Genes-Proteins and Microsatellite Instability: Focus on Adrenocortical Carcinoma and Beyond. Diagnostics (Basel) 2023; 13:diagnostics13111867. [PMID: 37296718 DOI: 10.3390/diagnostics13111867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
DNA damage repair pathways, including mismatch repair (MMR) genes, are prone to carcinoma development in certain patients. The assessment of the MMR system is widely recognized as part of strategies concerning solid tumors (defective MMR cancers), especially MMR proteins (through immunohistochemistry), and molecular assays for microsatellite instability (MSI). We aim to highlight the status of MMR genes-proteins (including MSI) in the relationship with ACC (adrenocortical carcinoma) according to current knowledge. This is a narrative review. We included PubMed-accessed, full-length English papers published between January 2012 and March 2023. We searched studies on ACC patients for whom MMR status was assessed, respectively subjects harboring MMR germline mutations, namely Lynch syndrome (LS), who were diagnosed with ACC. MMR system assessments in ACCs involve a low level of statistical evidence. Generally, there are two main types of endocrine insights: 1. the role of MMR status as a prognostic marker in different endocrine malignancies (including ACC)-which is the topic of the present work, and 2. establishing the indication of immune checkpoint inhibitors (ICPIs) in selective, mostly highly aggressive, non-responsive to standard care forms upon MMR evaluation (which belongs to the larger chapter of immunotherapy in ACCs). Our one-decade, sample-case study (which, to our knowledge, it is the most comprehensive of its kind) identified 11 original articles (from 1 patient to 634 subjects per study diagnosed with either ACC or LS). We identified four studies published in 2013 and 2020 and two in 2021, three cohorts and two retrospective studies (the publication from 2013 includes a retrospective and a cohort distinct section). Among these four studies, patients already confirmed to have LS (N = 643, respective 135) were found to be associated with ACC (N = 3, respective 2), resulting in a prevalence of 0.0046%, with a respective of 1.4% being confirmed (despite not having a large amount of similar data outside these two studies). Studies on ACC patients (N = 364, respective 36 pediatric individuals, and 94 subjects with ACC) showed that 13.7% had different MMR gene anomalies, with a respective of 8.57% (non-germline mutations), while 3.2% had MMR germline mutations (N = 3/94 cases). Two case series included one family, with a respective four persons with LS, and each article introduced one case with LS-ACC. Another five case reports (between 2018 and 2021) revealed an additional five subjects (one case per paper) diagnosed with LS and ACC (female to male ratio of 4 to 1; aged between 44 and 68). Interesting genetic testing involved children with TP53-positive ACC and further MMR anomalies or an MSH2 gene-positive subject with LS with a concurrent germline RET mutation. The first report of LS-ACC referred for PD-1 blockade was published in 2018. Nevertheless, the use of ICPI in ACCs (as similarly seen in metastatic pheochromocytoma) is still limited. Pan-cancer and multi-omics analysis in adults with ACC, in order to classify the candidates for immunotherapy, had heterogeneous results, and integrating an MMR system in this larger and challenging picture is still an open issue. Whether individuals diagnosed with LS should undergo surveillance for ACC has not yet been proven. An assessment of tumor-related MMR/MSI status in ACC might be helpful. Further algorithms for diagnostics and therapy, also taking into consideration innovative biomarkers as MMR-MSI, are necessary.
Collapse
Affiliation(s)
- Mara Carsote
- Department of Endocrinology, Carol Davila University of Medicine and Pharmacy & C.I. Parhon National Institute of Endocrinology, 011461 Bucharest, Romania
| | - Ionut Florin Turturea
- Department of Orthopedics and Traumatology, Cluj Emergency County Hospital, 400347 Cluj-Napoca, Romania
| | | | - Ana Valea
- Department of Endocrinology, Iuliu Hatieganu University of Medicine and Pharmacy & Clinical County Hospital, 400347 Cluj-Napoca, Romania
| | - Claudiu Nistor
- Department 4-Cardio-Thoracic Pathology, Thoracic Surgery II Discipline, Carol Davila University of Medicine and Pharmacy & Thoracic Surgery Department, Dr. Carol Davila Central Emergency University Military Hospital, 050474 Bucharest, Romania
| | - Ancuta-Augustina Gheorghisan-Galateanu
- Department of Molecular and Cellular Biology, and Histology, Carol Davila University of Medicine and Pharmacy & Department of Endocrinology, C.I. Parhon National Institute of Endocrinology, 011461 Bucharest, Romania
| |
Collapse
|
29
|
Imyanitov E, Sokolenko A. Integrative Genomic Tests in Clinical Oncology. Int J Mol Sci 2022; 23:13129. [PMID: 36361916 PMCID: PMC9656402 DOI: 10.3390/ijms232113129] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 09/12/2023] Open
Abstract
Many clinical decisions in oncology practice rely on the presence or absence of an alteration in a single genetic locus, be it a pathogenic variant in a hereditary cancer gene or activating mutation in a drug target. In addition, there are integrative tests that produce continuous variables and evaluate complex characteristics of the entire tumor genome. Microsatellite instability (MSI) analysis identifies tumors with the accumulation of mutations in short repetitive nucleotide sequences. This procedure is utilized in Lynch syndrome diagnostic pipelines and for the selection of patients for immunotherapy. MSI analysis is well-established for colorectal malignancies, but its applications in other cancer types lack standardization and require additional research. Homologous repair deficiency (HRD) indicates tumor sensitivity to PARP inhibitors and some cytotoxic drugs. HRD-related "genomic scars" are manifested by a characteristic pattern of allelic imbalances, accumulation of deletions with flanking homology, and specific mutation signatures. The detection of the genetic consequences of HRD is particularly sophisticated and expensive, as it involves either whole genome sequencing (WGS) or the utilization of large next-generation sequencing (NGS) panels. Tumor mutation burden (TMB) can be determined by whole exome sequencing (WES) or middle-throughput NGS multigene testing. Although TMB is regarded as an agnostic indicator of tumor sensitivity to immunotherapy, the clinical utility of this test is proven only for a few cancer types.
Collapse
Affiliation(s)
- Evgeny Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Anna Sokolenko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|