1
|
Lin H, Huang YS. Reply to comments on "A comparative analysis of MMR immunohistochemistry panels: Evaluating the utility of four-protein versus two-protein panels in endometrial cancer patients". J Formos Med Assoc 2025:S0929-6646(25)00113-5. [PMID: 40102151 DOI: 10.1016/j.jfma.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Affiliation(s)
- Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Yu-Sheng Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
2
|
Martínez Lapiedra C, García-Fadrique A, García Casado MZ, Navarro Fos S, Machado Puerto I. Immunohistochemistry staining for DNA mismatch repair proteins in endoscopic biopsies and the corresponding surgical specimen in colorectal cancer. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2025; 117:76-83. [PMID: 39421923 DOI: 10.17235/reed.2024.10645/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Microsatellite instability is found in 15% of sporadic colorectal cancers (CRC) and 95% of hereditary CRC cases. Lynch syndrome (LS) diagnosis begins with the analysis of the surgical specimen using methods such as immunohistochemistry (IHC), which identifies changes in the nuclear expression of DNA mismatch repair (MMR) proteins. However, IHC analysis on endoscopic biopsies could provide substantial benefits. Our goal was to assess the accuracy of MMR IHC status on endoscopic biopsies in comparison to corresponding surgical specimen in a series of CRC. We retrospectively selected patients who had undergone CRC surgery between February 2011 and January 2020 and had IHC testing for MMR proteins on the surgical specimen. The study was then performed on the corresponding endoscopic biopsies and results were compared. MMR IHC staining on surgical specimens were available for 361 CRC patients and only in 154 cases for preoperative endoscopic biopsies. The concordance between MMR IHC status of the endoscopic biopsy and the surgical specimen analysis was 98.6% for the MLH1/PMS2 proteins and 100% for MSH2/MSH6. In conclusion, endoscopic biopsies of colorectal tumors serve as a suitable tissue source for the immunohistochemical analysis of DNA repair proteins. The correlation with results from the surgical specimen was notably high and discrepancies were primarily as a result of intratumoral heterogeneity within the same sample. The features of MMR protein loss in endoscopic biopsies can provide clinicians with valuable information for specific therapeutic approaches and genetic counseling.
Collapse
Affiliation(s)
| | | | | | - Samuel Navarro Fos
- Pathology, Hospital Clínico Universitario de Valencia. Universidad de Valencia
| | | |
Collapse
|
3
|
Chen Z, Cao H, Zhang J, Zhong W, Teng X. The impact of preoperative treatment on mismatch repair protein and HER2 expression in colorectal cancer: an analysis of paired samples. BMC Cancer 2024; 24:1407. [PMID: 39548392 PMCID: PMC11566055 DOI: 10.1186/s12885-024-13120-w] [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: 06/13/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer-related mortality, highlighting the necessity for multifaceted treatment strategies, including preoperative treatment (PT), which can enhance surgical outcomes and provide prognostic insights. This study aims to clarify the impact of PT-induced changes in mismatch repair (MMR) and human epidermal growth factor receptor 2 (HER2) expression, potentially informing tailored treatment strategies and improving clinical outcomes for CRC patients. METHODS This retrospective study analyzed 120 paired samples from CRC patients who underwent preoperative treatment, comparing pre- and post-treatment specimens. A control group of 60 untreated surgical specimens was also included. Immunohistochemistry assessed MMR proteins (MSH6, MSH2, MLH1, PMS2) and HER2 expression. MSI status was determined in samples with low MMR expression. RESULTS Compared to pre-treatment samples, post-treatment samples exhibited lower levels of MSH6, MSH2, and total MMR expression, along with higher levels of HER2 expression. However, when compared to the untreated control group, there were no significant differences in the expression of MSH6, total MMR, and HER2. All samples that exhibited weak MMR expression and those that shifted to deficient mismatch repair (dMMR) status following treatment had stable microsatellite status. No clear clinicopathological characteristics or prognostic factors were found to be associated with changes in MMR and HER2 expression, except for the use of fluorouracil or capecitabine, which was related to changes in total MMR scores. ypTNM stage and TRG scores were identified as independent factors affecting disease progression in our study. CONCLUSIONS PT is associated with a reduction in MMR expression, notably for the MSH2 protein, while it does not appear to influence HER2 expression.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, 310000, China
| | - Hui Cao
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, 310000, China
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, 310000, China
| | - Weixiang Zhong
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, 310000, China
| | - Xiaodong Teng
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, No.79, Qingchun Road, Hangzhou, 310000, China.
| |
Collapse
|
4
|
Anderson CE, Liska D. Treatment of Microsatellite-Unstable Rectal Cancer in Sporadic and Hereditary Settings. Clin Colon Rectal Surg 2024; 37:233-238. [PMID: 38882941 PMCID: PMC11178385 DOI: 10.1055/s-0043-1770717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Microsatellite instability is rare in rectal cancer and associated with younger age of onset and Lynch syndrome. All rectal cancers should be tested for microsatellite instability prior to treatment decisions. Patients with microsatellite instability are relatively resistant to chemotherapy. However, recent small studies have shown dramatic response with neoadjuvant immunotherapy. Patients with Lynch syndrome have a hereditary predisposition to cancer and thus an elevated risk of metachronous cancer. Therefore, while "watch and wait" is a well-established practice for sporadic rectal cancers that obtain a complete clinical response after chemoradiation, its safety in patients with Lynch syndrome has not yet been defined. The extent of surgery for patients with Lynch syndrome and rectal cancer is controversial and there is significant debate as to the relative advantages of a segmental proctectomy with postoperative endoscopic surveillance versus a therapeutic and prophylactic total proctocolectomy. Surgical decision making for the patient with Lynch syndrome and rectal cancer is complex and demands a multidisciplinary approach, taking into account both patient- and tumor-specific factors. Neoadjuvant immunotherapy show great promise in the treatment of these patients, and further maturation of data from prospective trials will likely change the current treatment paradigm. Patients with Lynch syndrome and rectal cancer who do not undergo total proctocolectomy require yearly surveillance colonoscopies and should consider chemoprophylaxis with aspirin.
Collapse
Affiliation(s)
- Cristan E. Anderson
- Department of Colon and Rectal Surgery, Cleveland Clinic Foundation, Cleveland, Ohio
| | - David Liska
- Department of Colon and Rectal Surgery, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
5
|
Zhang P, Wang A, Bian C, Zhang J, Jiang C, Zhou H. Evaluation of mismatch-repair and microsatellite-instability status in a Chinese colorectal cancer Cohort. Asian J Surg 2024; 47:959-967. [PMID: 38185557 DOI: 10.1016/j.asjsur.2023.12.176] [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: 09/21/2023] [Accepted: 12/25/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Immunohistochemistry (IHC) and traditional polymerase chain reaction (PCR) are the methods of choice in clinical practice to identify the mismatch repair (MMR) and microsatellite instability (MSI) status in colorectal cancer (CRC). In some previous researches, the concordance rate between two methods was different and discordance existed in about 1 %-9.7 %. METHODS We retrospectively reviewed 406 patients received surgical CRC resections and tests of both MMR IHC and MSI PCR from January 2019 to April 2022 in Shanghai Changzheng Hospital. The incidence of deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) CRCs, the concordance rate between two methods, and the reasons for discordant results were evaluated with clinicopathological data, immunochemical staining, whole-exome sequencing, and MLH1 methylation analysis. RESULTS Among 406 patients, the incidence of MSI-H CRCs was 7.88 %. Nearly a quarter of the cases under reexamination of IHC was initial misinterpreted. Besides, the concordance rate between MMR IHC and MSI PCR was 99.26 % (401 of 404) and the Kappa value was 0.945 (p < 0.001). Finally, some somatic variants of MMR and POLE genes which may explain the discordance were identified. CONCLUSION The incidence rate of MSI-H in Chinese patients with CRC might be relatively low owing to tumor location. Although MSI and IHC analyses are highly concordant, both MMR IHC and MSI PCR tests should be simultaneously performed and MMR IHC should be interpreted by experienced pathologists. In the future, further studies on discordant results should be carried out to improve the personalized management of CRC.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Colorectal Surgery, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Anqi Wang
- Department of Colorectal Surgery, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Ce Bian
- Department of Colorectal Surgery, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Jing Zhang
- Division of Pathology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Caifeng Jiang
- Department of Gastroenterology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China.
| | - Haiyang Zhou
- Department of Colorectal Surgery, Changzheng Hospital, Navy Medical University, Shanghai, China.
| |
Collapse
|
6
|
Li Z, Teng L, Pan Z, Yang Y, Zhu J, Wu X, Qian Y, Qian H, Bian Y, Chen Y, Chen W, Bi L. Identification of Comprehensive Biomarkers in Patients With Mismatch Repair-Deficient Colon Adenocarcinoma Based on Parallel Multiomics. J Transl Med 2024; 104:100306. [PMID: 38104864 DOI: 10.1016/j.labinv.2023.100306] [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: 04/24/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Immunocheckpoint inhibitors have shown impressive efficacy in patients with colon cancer and other types of solid tumor that are mismatch repair-deficient (dMMR). Currently, PCR-capillary electrophoresis is one of the mainstream detection methods for dMMR, but its accuracy is still limited by germline mismatch repair (MMR) mutations, the functional redundancy of the MMR system, and abnormal methylation of MutL Homolog 1 promoter. Therefore, this study aimed to develop new biomarkers for dMMR based on artificial intelligence (AI) and pathologic images, which may help to improve the detection accuracy. To screen for the differential expression genes (DEGs) in dMMR patients and validate their diagnostic and prognostic efficiency, we used the expression profile data from the Cancer Genome Atlas (TCGA). The results showed that the expression of Immunoglobulin Lambda Joining 3 in dMMR patients was significantly downregulated and negatively correlated with the prognosis. Meanwhile, our diagnostic models based on pathologic image features showed good performance with area under the curves (AUCs) of 0.73, 0.86, and 0.81 in the training, test, and external validation sets (Jiangsu Traditional Chinese Medicine Hospital cohort). Based on gene expression and pathologic characteristics, we developed an effective prognosis model for dMMR patients through multiple Cox regression analysis (with AUC values of 0.88, 0.89, and 0.88 at 1-, 3-, and 5-year intervals, respectively). In conclusion, our results showed that Immunoglobulin Lambda Joining 3 and nucleus shape-related parameters (such as nuclear texture, nuclear eccentricity, nuclear size, and nuclear pixel intensity) were independent diagnostic and prognostic factors, suggesting that they could be used as new biomarkers for dMMR patients.
Collapse
Affiliation(s)
- Zhengjun Li
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China
| | - Linxin Teng
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhiwei Pan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yang Yang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Junlin Zhu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaobin Wu
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yunzhi Qian
- MPH Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Haihua Qian
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China
| | - Ying Chen
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weiping Chen
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Lei Bi
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
7
|
Fornaro L, Lonardi S, Catanese S, Nappo F, Pietrantonio F, Pellino A, Angerilli V, Signorini F, Salani F, Murgioni S, Neculaescu IA, Bruno R, Vivaldi C, Ricagno G, Masi G, Bergamo F, Ugolini C, Fassan M. Concordance of microsatellite instability and mismatch repair status in paired biopsies and surgical specimens of resectable gastroesophageal adenocarcinoma: time for a call to action. Gastric Cancer 2023; 26:958-968. [PMID: 37382783 DOI: 10.1007/s10120-023-01411-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Reliability of mismatch repair proteins and microsatellite instability assessment is essential in order to define treatment strategy and identify candidates to immune checkpoint inhibitors in locally advanced gastroesophageal carcinoma. We evaluated the concordance of deficient mismatch repair (dMMR) and microsatellite instability-high (MSI-H) status between endoscopic biopsies and surgical specimens. METHODS Consecutive patients with resectable gastric or gastroesophageal junction adenocarcinoma classified as MSI-H/dMMR by polymerase chain reaction (PCR) or immunohistochemistry (IHC) and operated at three referral Institutions were included. The primary endpoint was the rate of concordance between biopsy and surgical samples. If needed, central revision by IHC/PCR was performed by specialized pathologists from coordinating Institutions. RESULTS Thirteen (19.7%) out of 66 patients showed discordant MSI-H/dMMR results in the original pathology reports. In most cases (11, 16.7%) this was due to the diagnosis of proficient mismatch repair status on biopsies. Among the ten cases available for central review, four were due to sample issues, four were reclassified as dMMR, one case showed dMMR status but was classified as microsatellite stable by PCR, one was linked to misdiagnosis of endoscopic biopsy by the local pathologist. Heterogeneity of mismatch repair proteins staining was observed in two cases. CONCLUSIONS Available methods can lead to conflicting results in MSI-H/dMMR evaluation between endoscopic biopsies and surgical samples of gastroesophageal adenocarcinoma. Strategies aiming to improve the reliability of assessment should be primarily focused on the optimization of tissue collection and management during endoscopy and adequate training of dedicated gastrointestinal pathologists within the multidisciplinary team.
Collapse
Affiliation(s)
- Lorenzo Fornaro
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.
| | - Sara Lonardi
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
| | - Silvia Catanese
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Unit of Medical Oncology, Ospedale Misericordia, Azienda Toscana Sud-Est, Grosseto, Italy
| | - Floriana Nappo
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonio Pellino
- Department of Oncology, Division of Medical Oncology, Azienda Toscana Nord Ovest, Livorno, Italy
| | - Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Francesca Signorini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Francesca Salani
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Institute of Interdisciplinary Research "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
| | - Sabina Murgioni
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
| | | | - Rossella Bruno
- Unit of Pathology 3, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Caterina Vivaldi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Gianmarco Ricagno
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Gianluca Masi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesca Bergamo
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
| | - Clara Ugolini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fassan
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| |
Collapse
|
8
|
Chen S, Du W, Cao Y, Kong J, Wang X, Wang Y, Lu Y, Li X. Preoperative contrast-enhanced CT imaging and clinicopathological characteristics analysis of mismatch repair-deficient colorectal cancer. Cancer Imaging 2023; 23:97. [PMID: 37828626 PMCID: PMC10568855 DOI: 10.1186/s40644-023-00591-6] [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: 03/02/2023] [Accepted: 07/08/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) can develop through various pathogenetic pathways, and one of the primary pathways is high microsatellite instability (MSI-H)/deficient mismatch repair (dMMR). This study investigated the correlation between preoperative contrast-enhanced CT (CECT) and clinicopathologic characteristics of colorectal cancer (CRC) according to different mismatch repair (MMR) statuses. METHODS From April 2021 to July 2022, a total of 281 CRC patients with preoperative CECT and available MMR status were enrolled from a single centre for this retrospective study. Preoperative CECT features and clinicopathologic characteristics were analysed. Univariate and multivariate logistic regression analyses were used for statistical analysis. A nomogram was established based on the multivariate logistic regression results. Preoperative and postoperative dynamic nomogram prediction models were established. The C-index, a calibration plot, and clinical applicability of the two models were evaluated, and internal validation was performed using three methods. RESULTS In total, 249 patients were enrolled in the proficient mismatch repair (pMMR) group and 32 patients in the deficient mismatch repair (dMMR) group. In multivariate analysis, tumour location (right-hemi colon vs. left-hemi colon, odds ratio (OR) = 2.90, p = .036), the hypoattenuation-within-tumour ratio (HR) (HR > 2/3 vs. HR < 1/3, OR = 36.7, p < .001; HR 1/3-2/3 vs. HR < 1/3, OR = 6.05, p = .031), the number of lymph nodes with long diameter ≥ 8 mm on CECT (OR = 1.32, p = .01), CEA status (CEA positive vs. CEA negative, OR = 0.07, p = .002) and lymph node metastasis (OR = 0.45, p = .008) were independent risk factors for dMMR. Pre- and postoperative C-statistic were 0.861 and 0.908, respectively. CONCLUSION The combination of pre-operative CECT and clinicopathological characteristics of CRC correlates with MMR status, providing possible non-invasive MMR prediction. Particularly for dMMR CRC, tumour-draining lymph node status should be prudently evaluated by CECT.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Radiology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China
| | - Wenzhe Du
- Department of Radiology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China
| | - Yuhai Cao
- Department of Radiology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China
| | - Jixia Kong
- Department of Pathology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China
| | - Xin Wang
- Department of Radiology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China
| | - Yisen Wang
- Department of Radiology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China
| | - Yang Lu
- Department of Radiology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China.
| | - Xiang Li
- Department of Radiology, The Second Hospital of Dalian Medical University, Zhongshan Road No.467, Shahekou District, Dalian, Liaoning, 116023, China.
| |
Collapse
|
9
|
Freitag CE, Chen W, Pearlman R, Hampel H, Stanich PP, Cosgrove CM, Konnick EQ, Pritchard CC, Frankel WL. Mismatch repair protein status of non-neoplastic uterine and intestinal mucosa in patients with Lynch syndrome and double somatic mismatch repair protein mutations. Hum Pathol 2023; 137:1-9. [PMID: 37030500 DOI: 10.1016/j.humpath.2023.04.001] [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: 03/16/2023] [Accepted: 04/03/2023] [Indexed: 04/10/2023]
Abstract
Mismatch repair (MMR) protein-deficient non-neoplastic colonic crypts and endometrial glands (dMMR crypts and glands) have been reported as a unique marker of underlying Lynch syndrome (LS). However, no large studies have directly compared the frequency of detection in cases with double somatic (DS) MMR mutations. We retrospectively analyzed 42 colonic resection specimens (24 LS and 18 DS) and 20 endometrial specimens (9 LS and 11 DS), including 19 hysterectomies and 1 biopsy for dMMR crypts and glands. All specimens were from patients with known primary cancers, including colonic adenocarcinomas and endometrial endometrioid carcinomas (including 2 mixed carcinomas). Four blocks of normal mucosa away from the tumor were selected from most cases, as available. MMR immunohistochemistry specific to the primary tumor mutations was analyzed. dMMR crypts were found in 65% of LS and 0% of DS MMR-mutated colonic adenocarcinomas (P < .001). Most dMMR crypts were detected in the colon (12 of 15) compared to the ileum (3 of 15). dMMR crypts showed single and grouped losses of MMR immunohistochemical expression. dMMR glands were found in 67% of LS and 9% (1 of 11) of DS endometrial cases (P = .017). Most dMMR glands were found in the uterine wall, with 1 LS and 1 DS case exhibiting dMMR glands in the lower uterine segment. Most cases exhibited multifocal and grouped dMMR glands. No morphologic atypia was identified in dMMR crypts or glands. Overall, we demonstrate that dMMR crypts and glands are highly associated with underlying LS, while being rarer in those with DS MMR mutations.
Collapse
Affiliation(s)
- C Eric Freitag
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Rachel Pearlman
- Department of Internal Medicine, Clinical Cancer Genetics Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Heather Hampel
- Division of Genetics and Genetic Counseling, City of Hope, Duarte, CA, 91010, USA
| | - Peter P Stanich
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Casey M Cosgrove
- Division of Gynecologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Eric Q Konnick
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
Parente P, Grillo F, Vanoli A, Macciomei MC, Ambrosio MR, Scibetta N, Filippi E, Cataldo I, Baron L, Ingravallo G, Cazzato G, Melocchi L, Liserre B, Giordano C, Arborea G, Pilozzi E, Scapinello A, Aquilano MC, Gafà R, Battista S, Dal Santo L, Campora M, Carbone FG, Sartori C, Lazzi S, Hanspeter E, Angerilli V, Mastracci L, Fassan M. The Day-To-Day Practice of MMR and MSI Assessment in Colorectal Adenocarcinoma: What We Know and What We Still Need to Explore. Dig Dis 2023; 41:746-756. [PMID: 37231848 DOI: 10.1159/000531003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND The DNA mismatch repair (MMR) system is a highly preserved protein complex recognizing short insertions, short deletions, and single base mismatches during DNA replication and recombination. MMR protein status is identified using immunohistochemistry. Deficit in one or more MMR proteins, configuring deficient MMR status (dMMR), leads to frameshift mutations particularly clustered in microsatellite repeats. Thus, microsatellite instability (MSI) is the epiphenomenon of dMMR. In colorectal cancer (CRC), MMR/MSI status is a biomarker with prognostic and predictive value of resistance to 5-fluorouracil and response to immune checkpoint inhibitor therapy. SUMMARY In this Review, we describe the challenges the practicing pathologist may face in relation to the assessment of MMR/MSI status and any open issues which still need to be addressed, focusing on pre-analytic issues, pitfalls in the interpretation, and technical aspects of the different assays. KEY MESSAGES The current methods of detecting dMMR/MSI status have been optimized for CRCs, and whether these techniques can be applied to all tumor and specimen types is still not fully understood. Following the Food and Drug Administration (FDA), tissue/site agnostic drug approval of pembrolizumab for advanced/metastatic MSI tumors, MMR/MSI status in gastrointestinal tract is a common request from the oncologist. In this setting, several issues still need to be addressed, including criteria for sample adequacy.
Collapse
Affiliation(s)
- Paola Parente
- Unit of Pathology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Federica Grillo
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
- Pathology Unit, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, Pavia, Italy
| | | | | | - Nunzia Scibetta
- UOC Anatomia Patologica ARNAS Ospedali Civico e G. Di Gristina, Palermo, Italy
| | | | - Ivana Cataldo
- Surgical Pathology Section University and Hospital Trust of Treviso, Treviso, Italy
| | - Luigi Baron
- Surgical Pathology Unit ASL Napoli 3 Sud, Ospedale S. Leonardo, Naples, Italy
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari "Aldo Moro", Bari, Italy
| | - Gerardo Cazzato
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari "Aldo Moro", Bari, Italy
| | - Laura Melocchi
- Unit of Pathology, Department of Oncology, Fondazione Poliambulanza Hospital Institute, Brescia, Italy
| | - Barbara Liserre
- Unit of Pathology, Department of Oncology, Fondazione Poliambulanza Hospital Institute, Brescia, Italy
| | - Carla Giordano
- Pathology Unit, Università La Sapienza; Policlinico Umberto I, Rome, Italy
| | - Graziana Arborea
- Department of Pathology, National Institute of Gastroenterology IRCCS "S. de Bellis", Castellana Grotte, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | | | - Maria Costanza Aquilano
- Department of Hematology, Oncology and Molecular Medicine, ASST Grande Ospedale Metropolitano/Niguarda, Milan, Italy
| | - Roberta Gafà
- Anatomic Pathology Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Serena Battista
- Pathology Department, S. Maria della Misericordia Hospital, Udine, Italy
| | - Luca Dal Santo
- Department of Pathology, Ospedale dell'Angelo, Venice, Italy
| | - Michela Campora
- U.O.M. Anatomia e Istologia Patologica e Citodiagnostica Ospedale S. Chiara, Trento, Italy
| | | | - Chiara Sartori
- U.O.M. Anatomia e Istologia Patologica e Citodiagnostica Ospedale S. Chiara, Trento, Italy
| | - Stefano Lazzi
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena, Italy
| | - Ester Hanspeter
- Department of Pathology, Provincial Hospital of Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Luca Mastracci
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
- Pathology Unit, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Matteo Fassan
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| |
Collapse
|
11
|
Risbey C, Fielder T, Steffens D, Shin JS, Solomon M. Patterns of DNA mismatch repair protein expression for primary and recurrent colorectal cancer at an advanced surgical unit: A retrospective audit. Colorectal Dis 2023; 25:369-374. [PMID: 36300681 DOI: 10.1111/codi.16391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 02/08/2023]
Abstract
AIM Lynch syndrome is an inherited cancer syndrome associated with an increased lifetime risk of colorectal cancer (CRC) and characterized by germline mutations to one of four DNA mismatch repair (MMR) genes. Immunohistochemical (IHC) testing is used to screen for Lynch syndrome; however, despite routine completion following resection of primary CRC, it is only variably completed following resection of recurrent disease. This may be significant, as MMR protein expression can change from primary to recurrent CRC. The primary aim of this study is to investigate how MMR profiles change from primary to recurrent CRC; the secondary aim is to assess rates of MMR testing of primary and recurrent disease. METHOD We conducted a retrospective analysis of patients undergoing surgery for recurrent CRC from 2018-19 at a high-volume institution. MMR profiles were obtained following both primary and recurrent resection of CRC, and MMR protein expression was evaluated from both time points. RESULTS A total of 107 patients met the inclusion criteria and IHC results were obtained for both primary and recurrent resections in 85 cases. MMR profiles changed in nine patients (10.6%), with a loss of staining from primary to recurrent disease in six (7.1%) and a gain of staining in three (3.5%). IHC testing was completed following 88.7% of primary and 39.3% of recurrent resections. CONCLUSION MMR profiles can change from primary to recurrent CRC and repeat MMR testing for recurrent CRC is completed in only a minority of cases.
Collapse
Affiliation(s)
- Charles Risbey
- Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
| | - Timothy Fielder
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
| | - Daniel Steffens
- Surgical Outcomes Research Centre (SOuRCe), Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
- Faculty of Medicine and Health, Central Clinical School, The University of Sydney, New South Wales, Camperdown, Australia
| | - Joo-Shik Shin
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
| | - Michael Solomon
- Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
- Surgical Outcomes Research Centre (SOuRCe), Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
- Faculty of Medicine and Health, Central Clinical School, The University of Sydney, New South Wales, Camperdown, Australia
- Department of Colorectal Surgery, Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
- RPA Institute of Academic Surgery, New South Wales, Camperdown, Australia
| |
Collapse
|
12
|
Detection of Microsatellite Instability in Colonoscopic Biopsies and Postal Urine Samples from Lynch Syndrome Cancer Patients Using a Multiplex PCR Assay. Cancers (Basel) 2022; 14:cancers14153838. [PMID: 35954501 PMCID: PMC9367254 DOI: 10.3390/cancers14153838] [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: 05/31/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Identification of mismatch repair (MMR)-deficient colorectal cancers (CRCs) is recommended for Lynch syndrome (LS) screening, and supports targeting of immune checkpoint inhibitors. Microsatellite instability (MSI) analysis is commonly used to test for MMR deficiency. Testing biopsies prior to tumour resection can inform surgical and therapeutic decisions, but can be limited by DNA quantity. MSI analysis of voided urine could also provide much needed surveillance for genitourinary tract cancers in LS. Here, we reconfigure an existing molecular inversion probe-based MSI and BRAF c.1799T > A assay to a multiplex PCR (mPCR) format, and demonstrate that it can sample >140 unique molecules per marker from <1 ng of DNA and classify CRCs with 96−100% sensitivity and specificity. We also show that it can detect increased MSI within individual and composite CRC biopsies from LS patients, and within preoperative urine cell free DNA (cfDNA) from two LS patients, one with an upper tract urothelial cancer, the other an undiagnosed endometrial cancer. Approximately 60−70% of the urine cfDNAs were tumour-derived. Our results suggest that mPCR sequence-based analysis of MSI and mutation hotspots in CRC biopsies could facilitate presurgery decision making, and could enable postal-based screening for urinary tract and endometrial tumours in LS patients.
Collapse
|
13
|
Ryan ÉJ, Creavin B, Sheahan K. Delivery of Personalized Care for Locally Advanced Rectal Cancer: Incorporating Pathological, Molecular Genetic, and Immunological Biomarkers Into the Multimodal Paradigm. Front Oncol 2020; 10:1369. [PMID: 32923389 PMCID: PMC7456909 DOI: 10.3389/fonc.2020.01369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Approximately one-third of all newly diagnosed colorectal cancer (CRC) is composed of rectal cancer, with the incidence rising in younger patients. The principal neoadjuvant treatments consist of neoadjuvant short-course radiotherapy and long-course chemoradiation. Locally advanced rectal cancer (LARC) is particularly challenging to manage given the anatomical constrictions of the pelvis and the risk for local recurrence. In appropriately treated patients, 5- and 10-year overall survival is estimated at 60 and 50%, respectively. The prognosis for LARC has improved in recent years with more access to screening, advances in surgical techniques, and perioperative care. Furthermore, the refinement of the multidisciplinary team with combined-modality management strategies has improved outcomes. These advancements have been augmented by significant improvements in the understanding of the underlying tumor biology. However, there are many instances where patient outcomes do not match those for their tumor stage and accurate prognostic information for individual patients can be difficult to estimate owing to the heterogeneous nature of LARC. Many new combinations of chemotherapy with radiotherapy, including total neoadjuvant therapy with targeted therapies that aim to diminish toxicity and increase survival, are being evaluated in clinical trials. Despite these advances, local recurrence and distant metastasis remain an issue, with one-third of LARC patients dying within 5 years of initial treatment. Although much of the new pathological, molecular genetics, and immunological biomarkers allow refinement in the classification and prognostication of CRC, the relative importance of each of these factors with regards to the development and progression of LARC remains incompletely understood. These factors are often insufficiently validated and seldom consider the individual characteristics of the host, the tumor and its location, the local available expertise, or the probable location of recurrence. Appreciating the mechanisms behind these differences will allow for a more comprehensive, personalized approach and more informed treatment options, leading to ultimately superior outcomes. This review aims to first outline the current multidisciplinary context in which LARC care should be delivered and then discuss how some key prognosticators, including novel histopathological, molecular genetics, and immunological biomarkers, might fit into the wider context of personalized LARC management in the coming years.
Collapse
Affiliation(s)
- Éanna J. Ryan
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ben Creavin
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kieran Sheahan
- School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
14
|
Unexpected expression of mismatch repair protein is more commonly seen with pathogenic missense than with other mutations in Lynch syndrome. Hum Pathol 2020; 103:34-41. [PMID: 32652087 DOI: 10.1016/j.humpath.2020.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
Abstract
It has been observed that some patients with colorectal cancer due to germline or double somatic pathogenic variants in the mismatch repair (MMR) genes may have intact protein expression in their tumors as assessed by immunohistochemistry (IHC). This has been speculated to occur more frequently in Lynch syndrome (LS) cases due to pathogenic missense mutations, leading to expression of a full-length but nonfunctional protein with retained antigenicity. Our goals were to study the frequency of unexpected MMR expression in colorectal cancers among LS cases with missense mutations, LS cases with truncating mutations, as well as cases with double somatic MMR mutations and evaluate if the unexpected MMR expression is more common in certain categories. IHC slides were available for 82 patients with MMR deficiency without methylation, which included 56 LS cases and 26 double somatic MMR mutation cases. Sixteen of 82 MMR-defective cases showed unexpected MMR expression, with 10 cases showing tumor staining weaker than the control and 6 cases (7%) showing intact staining. Unexpected MMR expression was most commonly seen with LS cases with missense mutations (4 of 9, 44%), followed by MMR double somatic mutation cases (7 of 26, 27%), and finally by LS cases with truncating mutations (5 of 47, 11%). Cautious interpretation of MMR IHC is advised when dealing with tumor staining that is weaker than the control regardless of the percentage of tumor staining as these cases may harbor pathogenic MMR gene mutations. Missense mutations may account for some LS cases that may be missed by IHC alone. Strict adherence to proper interpretation of IHC with attention to staining intensity and the status of heterodimer partner protein will prevent many potential misses.
Collapse
|
15
|
Cercek A, Dos Santos Fernandes G, Roxburgh CS, Ganesh K, Ng S, Sanchez-Vega F, Yaeger R, Segal NH, Reidy-Lagunes DL, Varghese AM, Markowitz A, Wu C, Szeglin B, Sauvé CEG, Salo-Mullen E, Tran C, Patel Z, Krishnan A, Tkachuk K, Nash GM, Guillem J, Paty PB, Shia J, Schultz N, Garcia-Aguilar J, Diaz LA, Goodman K, Saltz LB, Weiser MR, Smith JJ, Stadler ZK. Mismatch Repair-Deficient Rectal Cancer and Resistance to Neoadjuvant Chemotherapy. Clin Cancer Res 2020; 26:3271-3279. [PMID: 32144135 DOI: 10.1158/1078-0432.ccr-19-3728] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/31/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Evaluate response of mismatch repair-deficient (dMMR) rectal cancer to neoadjuvant chemotherapy. EXPERIMENTAL DESIGN dMMR rectal tumors at Memorial Sloan Kettering Cancer Center (New York, NY) were retrospectively reviewed for characteristics, treatment, and outcomes. Fifty patients with dMMR rectal cancer were identified by IHC and/or microsatellite instability analysis, with initial treatment response compared with a matched MMR-proficient (pMMR) rectal cancer cohort. Germline and somatic mutation analyses were evaluated. Patient-derived dMMR rectal tumoroids were assessed for chemotherapy sensitivity. RESULTS Of 21 patients receiving neoadjuvant chemotherapy (fluorouracil/oxaliplatin), six (29%) had progression of disease. In comparison, no progression was noted in 63 pMMR rectal tumors (P = 0.0001). Rectal cancer dMMR tumoroids reflected this resistance to chemotherapy. No genomic predictors of chemotherapy response were identified. Of 16 patients receiving chemoradiation, 13 (93%) experienced tumor downstaging; one patient had stable disease, comparable with 48 pMMR rectal cancers. Of 13 patients undergoing surgery, 12 (92%) had early-stage disease. Forty-two (84%) of the 50 patients tested positive for Lynch syndrome with enrichment of germline MSH2 and MSH6 mutations when compared with 193 patients with Lynch syndrome-associated colon cancer (MSH2, 57% vs 36%; MSH6, 17% vs 9%; P < 0.003). CONCLUSIONS Over one-fourth of dMMR rectal tumors treated with neoadjuvant chemotherapy exhibited disease progression. Conversely, dMMR rectal tumors were sensitive to chemoradiation. MMR status should be performed upfront in all locally advanced rectal tumors with careful monitoring for response on neoadjuvant chemotherapy and genetic testing for Lynch syndrome in patients with dMMR rectal cancer.
Collapse
Affiliation(s)
- Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Campbell S Roxburgh
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Karuna Ganesh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shu Ng
- Alfred Health Radiation Oncology, Melbourne, Victoria, Australia
| | - Francisco Sanchez-Vega
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neil H Segal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Arnold Markowitz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chao Wu
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bryan Szeglin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Erin Salo-Mullen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina Tran
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zalak Patel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Asha Krishnan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kaitlyn Tkachuk
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Garrett M Nash
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jose Guillem
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Philip B Paty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julio Garcia-Aguilar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luis A Diaz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Karyn Goodman
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Leonard B Saltz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martin R Weiser
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - J Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
16
|
Walk EE, Yohe SL, Beckman A, Schade A, Zutter MM, Pfeifer J, Berry AB. The Cancer Immunotherapy Biomarker Testing Landscape. Arch Pathol Lab Med 2019; 144:706-724. [PMID: 31714809 DOI: 10.5858/arpa.2018-0584-cp] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Cancer immunotherapy provides unprecedented rates of durable clinical benefit to late-stage cancer patients across many tumor types, but there remains a critical need for biomarkers to accurately predict clinical response. Although some cancer immunotherapy tests are associated with approved therapies and considered validated, other biomarkers are still emerging and at various states of clinical and translational exploration. OBJECTIVE.— To provide pathologists with a current and practical update on the evolving field of cancer immunotherapy testing. The scientific background, clinical data, and testing methodology for the following cancer immunotherapy biomarkers are reviewed: programmed death ligand-1 (PD-L1), mismatch repair, microsatellite instability, tumor mutational burden, polymerase δ and ε mutations, cancer neoantigens, tumor-infiltrating lymphocytes, transcriptional signatures of immune responsiveness, cancer immunotherapy resistance biomarkers, and the microbiome. DATA SOURCES.— Selected scientific publications and clinical trial data representing the current field of cancer immunotherapy. CONCLUSIONS.— The cancer immunotherapy field, including the use of biomarker testing to predict patient response, is still in evolution. PD-L1, mismatch repair, and microsatellite instability testing are helping to guide the use of US Food and Drug Administration-approved therapies, but there remains a need for better predictors of response and resistance. Several categories of tumor and patient characteristics underlying immune responsiveness are emerging and may represent the next generation of cancer immunotherapy predictive biomarkers. Pathologists have important roles and responsibilities as the field of cancer immunotherapy continues to develop, including leadership of translational studies, exploration of novel biomarkers, and the accurate and timely implementation of newly approved and validated companion diagnostics.
Collapse
Affiliation(s)
- Eric E Walk
- From the Department of Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona (Dr Walk); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, Indiana (Dr Schade); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee (Dr Zutter); the Department of Molecular Pathology and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr Berry); and the Department of Pathology, Washington University School of Medicine, St Louis, Missouri (Dr Pfeifer)
| | - Sophia L Yohe
- From the Department of Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona (Dr Walk); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, Indiana (Dr Schade); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee (Dr Zutter); the Department of Molecular Pathology and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr Berry); and the Department of Pathology, Washington University School of Medicine, St Louis, Missouri (Dr Pfeifer)
| | - Amy Beckman
- From the Department of Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona (Dr Walk); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, Indiana (Dr Schade); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee (Dr Zutter); the Department of Molecular Pathology and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr Berry); and the Department of Pathology, Washington University School of Medicine, St Louis, Missouri (Dr Pfeifer)
| | - Andrew Schade
- From the Department of Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona (Dr Walk); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, Indiana (Dr Schade); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee (Dr Zutter); the Department of Molecular Pathology and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr Berry); and the Department of Pathology, Washington University School of Medicine, St Louis, Missouri (Dr Pfeifer)
| | - Mary M Zutter
- From the Department of Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona (Dr Walk); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, Indiana (Dr Schade); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee (Dr Zutter); the Department of Molecular Pathology and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr Berry); and the Department of Pathology, Washington University School of Medicine, St Louis, Missouri (Dr Pfeifer)
| | - John Pfeifer
- From the Department of Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona (Dr Walk); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, Indiana (Dr Schade); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee (Dr Zutter); the Department of Molecular Pathology and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr Berry); and the Department of Pathology, Washington University School of Medicine, St Louis, Missouri (Dr Pfeifer)
| | - Anna B Berry
- From the Department of Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona (Dr Walk); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Drs Yohe and Beckman); Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, Indiana (Dr Schade); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee (Dr Zutter); the Department of Molecular Pathology and Genomics, Swedish Cancer Institute, Seattle, Washington (Dr Berry); and the Department of Pathology, Washington University School of Medicine, St Louis, Missouri (Dr Pfeifer)
| | | |
Collapse
|
17
|
Role of intravoxel incoherent motion MRI in preoperative evaluation of DNA mismatch repair status in rectal cancers. Clin Radiol 2019; 74:814.e21-814.e28. [PMID: 31427042 DOI: 10.1016/j.crad.2019.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/10/2019] [Indexed: 11/22/2022]
Abstract
AIM To explore the role of intravoxel incoherent motion (IVIM) magnetic resonance imaging (MRI) in evaluating DNA mismatch repair (MMR) status of rectal cancers preoperatively. MATERIALS AND METHODS Seventy-six patients with a diagnosis of rectal cancer confirmed at endoscopic biopsy were enrolled prospectively and underwent IVIM MRI before surgery. RESULTS The perfusion fraction (f) values of MMR proteins (MMRP) positive rectal cancers were significantly higher than negative cancers. The f values could differentiate MMRP positive rectal cancers from negative cancers with an area under the curve (AUC) of 0.695. The vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) expression rates of positive MMRP rectal cancers were significantly higher than negative cancers. CONCLUSION This pilot study indicated that the f value derived from IVIM MRI differed significantly between rectal cancers with different MMRP expression levels, which might be involved with different VEGF and VEGFR2 expression rates.
Collapse
|
18
|
Meillan N, Vernerey D, Lefèvre JH, Manceau G, Svrcek M, Augustin J, Fléjou JF, Lascols O, Simon JM, Cohen R, Maingon P, Bachet JB, Huguet F. Mismatch Repair System Deficiency Is Associated With Response to Neoadjuvant Chemoradiation in Locally Advanced Rectal Cancer. Int J Radiat Oncol Biol Phys 2019; 105:824-833. [PMID: 31404579 DOI: 10.1016/j.ijrobp.2019.07.057] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/24/2019] [Accepted: 07/04/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Defective mismatch repair system (dMMR) has been shown to have a favorable impact on outcome in patients with colorectal cancer treated with surgery or immunotherapy, with adjuvant chemotherapy being discouraged unless there is nodal involvement. Its impact on radiosensitivity is unknown in patients with colorectal cancer. METHODS AND MATERIALS Patients treated for locally advanced rectal cancer between 2000 and 2016 were studied. Reported points included age, sex, clinical and radiologic tumor stages at diagnosis, modalities of neoadjuvant treatment, posttreatment pathologic staging, tumor regression score, and local, distant relapse-free, and overall survival. An inverse probability of treatment weighting propensity score analysis was performed to evaluate the association of mismatch repair proficiency with surgical and clinical outcomes. RESULTS Among the 296 patients included, 23 (7.8%) had dMMR. Median follow-up was 43.0 months (interquartile range, 27.9-66.7). Patients with dMMR were significantly younger than the others. After inverse probability of treatment weighting propensity score matching, dMMR patients had higher pathologic downstaging rate (P < .0001), higher tumor regression grade (P = .024), and a longer recurrence-free survival (P < .0001). CONCLUSIONS dMRR was associated with significant tumor downstaging after neoadjuvant chemoradiation and with increased recurrence-free survival. dMMR patients may have more radiosensitive tumors.
Collapse
Affiliation(s)
- Nicolas Meillan
- Department of Radiation Oncology, Tenon Hospital, Hôpitaux Universitaires Paris Est, APHP, Paris, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
| | - Jérémie H Lefèvre
- Department of Digestive Surgery, Saint-Antoine Hospital, APHP, Paris, France; Sorbonne Université, Paris, France
| | - Gilles Manceau
- Sorbonne Université, Paris, France; Department of Digestive and Hepato-Pancreato-Biliary Surgery, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - Magali Svrcek
- Sorbonne Université, Paris, France; Department of Pathology, Saint-Antoine Hospital, APHP, Paris, France
| | - Jeremy Augustin
- Department of Pathology, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - Jean-François Fléjou
- Sorbonne Université, Paris, France; Department of Pathology, Saint-Antoine Hospital, APHP, Paris, France
| | - Olivier Lascols
- Department of Biology and Molecular Genetics, Saint-Antoine Hospital, APHP, Paris, France
| | - Jean-Marc Simon
- Department of Radiation Oncology, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - Romain Cohen
- Sorbonne Université, Paris, France; Department of Medical Oncology, Saint-Antoine Hospital, APHP, Paris, France
| | - Philippe Maingon
- Sorbonne Université, Paris, France; Department of Radiation Oncology, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - Jean-Baptiste Bachet
- Sorbonne Université, Paris, France; Department of Hepato-Gastroenterology, Pitié Salpêtrière Hospital, APHP, Paris, France
| | - Florence Huguet
- Department of Radiation Oncology, Tenon Hospital, Hôpitaux Universitaires Paris Est, APHP, Paris, France; Sorbonne Université, Paris, France.
| |
Collapse
|
19
|
Microsatellite Instability and Programmed Cell Death-Ligand 1 Expression in Stage II/III Gastric Cancer. Ann Surg 2019; 270:309-316. [DOI: 10.1097/sla.0000000000002803] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Yozu M, Kumarasinghe MP, Brown IS, Gill AJ, Rosty C. Australasian Gastrointestinal Pathology Society (AGPS) consensus guidelines for universal defective mismatch repair testing in colorectal carcinoma. Pathology 2019; 51:233-239. [DOI: 10.1016/j.pathol.2018.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/18/2018] [Accepted: 11/25/2018] [Indexed: 01/28/2023]
|
21
|
Haag GM, Czink E, Ahadova A, Schmidt T, Sisic L, Blank S, Heger U, Apostolidis L, Berger AK, Springfeld C, Lasitschka F, Jäger D, Knebel Doeberitz M, Kloor M. Prognostic significance of microsatellite‐instability in gastric and gastroesophageal junction cancer patients undergoing neoadjuvant chemotherapy. Int J Cancer 2019; 144:1697-1703. [DOI: 10.1002/ijc.32030] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/19/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Georg Martin Haag
- Department of Medical Oncology, National Center for Tumor DiseasesUniversity Hospital Heidelberg Heidelberg Germany
| | - Elena Czink
- Department of Medical Oncology, National Center for Tumor DiseasesUniversity Hospital Heidelberg Heidelberg Germany
| | - Aysel Ahadova
- Department of Applied Tumor Biology, Institute of PathologyUniversity Hospital Heidelberg Heidelberg Germany
| | - Thomas Schmidt
- Department of SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Leila Sisic
- Department of SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Susanne Blank
- Department of SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Ulrike Heger
- Department of SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Leonidas Apostolidis
- Department of Medical Oncology, National Center for Tumor DiseasesUniversity Hospital Heidelberg Heidelberg Germany
| | - Anne Katrin Berger
- Department of Medical Oncology, National Center for Tumor DiseasesUniversity Hospital Heidelberg Heidelberg Germany
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor DiseasesUniversity Hospital Heidelberg Heidelberg Germany
| | - Felix Lasitschka
- Institute of PathologyUniversity Hospital Heidelberg Heidelberg Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor DiseasesUniversity Hospital Heidelberg Heidelberg Germany
| | - Magnus Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of PathologyUniversity Hospital Heidelberg Heidelberg Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of PathologyUniversity Hospital Heidelberg Heidelberg Germany
| |
Collapse
|
22
|
A practical guide to biomarkers for the evaluation of colorectal cancer. Mod Pathol 2019; 32:1-15. [PMID: 30600322 DOI: 10.1038/s41379-018-0136-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022]
Abstract
Evaluation of microsatellite instability (MSI) of every colorectal cancer (CRC) is important for prognostic and therapeutic purposes, while molecular testing helps identify actionable targeted therapy for patients with metastatic disease. This review will discuss the biomarkers commonly encountered in the clinical evaluation of CRC, and practical issues regarding MSI screening, reporting, interpretation, molecular test indication, and specimen requirements.
Collapse
|
23
|
Two-stain immunohistochemical screening for Lynch syndrome in colorectal cancer may fail to detect mismatch repair deficiency. Mod Pathol 2018; 31:1891-1900. [PMID: 29967423 PMCID: PMC6800091 DOI: 10.1038/s41379-018-0058-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 02/07/2023]
Abstract
Universal screening for Lynch syndrome in colorectal cancer is recommended, and immunohistochemistry for the mismatch repair proteins is commonly used. To reduce cost, some screen using only MSH6 and PMS2, with reflex to the partner stain if either are absent (two-stain method). An expression pattern revealing absent MSH2 and intact MSH6 is not expected, but could result in failed Lynch syndrome detection. We analyzed tumors with absent MSH2 but any degree of MSH6 expression to determine if the two-stain method could miss MSH2 mutations. One-thousand seven-hundred thirty colorectal cancer patients from the Ohio Colorectal Cancer Prevention Initiative underwent tumor screening using microsatellite instability and immunohistochemistry. The two-stain method was used for 1235 cases; staining for all four proteins was completed for 495 cases. The proportion of positive cells and staining intensity were reviewed for MSH6, as well as MSH2 when available. Patients with mismatch repair deficiency underwent next-generation sequencing of germline DNA for mismatch repair genes. If negative, tumor next-generation sequencing was performed to assess for somatic mutations. Overall, thirty-three (1.9%, 33/1730) MSH2-absent cases were identified. Of those, fourteen had no MSH6 expression but eight (0.5%, 8/1730) had ambiguous and eleven (0.6%, 11/1730) had convincing MSH6 expression that could have been interpreted as intact. Germline next-generation sequencing identified MSH2 mutations in 11/14 cases with absence of both stains, 7/8 cases with ambiguous MSH6 expression, and 9/11 cases with convincing MSH6 expression. All remaining cases, except one, had double somatic mutations. The two-stain method fails to detect some patients with Lynch syndrome: (1) significant staining weaker than the control may be incorrectly interpreted as intact MSH6, or (2) Weak or focal/patchy MSH6 can be retained with the absence of MSH2. Accordingly, we recommend the four-stain method be used for optimal Lynch syndrome screening detection.
Collapse
|
24
|
Universal Lynch Syndrome Screening Should be Performed in All Upper Tract Urothelial Carcinomas. Am J Surg Pathol 2018; 42:1549-1555. [DOI: 10.1097/pas.0000000000001141] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
25
|
Hissong E, Crowe EP, Yantiss RK, Chen YT. Assessing colorectal cancer mismatch repair status in the modern era: a survey of current practices and re-evaluation of the role of microsatellite instability testing. Mod Pathol 2018; 31:1756-1766. [PMID: 29955148 DOI: 10.1038/s41379-018-0094-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 01/05/2023]
Abstract
Results of DNA mismatch repair testing are used to detect Lynch syndrome and have prognostic and therapeutic implications among patients with sporadic colorectal carcinomas. Immunohistochemistry for mismatch repair proteins (MLH1, PMS2, MSH2, MSH6) and PCR for microsatellite instability are two established methods for assessing mismatch repair function. Older literature suggested a discordance rate of approximately 5% between these assays, leading some institutions to perform dual testing on all cases. Although universal mismatch repair testing is now recommended by multiple professional organizations, none provide guidelines regarding preferred assays. We surveyed 96 academic and nonacademic institutions to assess Lynch syndrome screening practices and evaluated discordance rates between immunohistochemistry and PCR among 809 colorectal cancers tested in our own institution. Our survey demonstrated no significant differences between academic and nonacademic practices with respect to testing strategies. Eighty six percent performed universal screening, and usually (76%) employed immunohistochemistry on initial biopsy samples. Only 20% employed PCR; these were mostly academic practices that used both immunohistochemistry and PCR (p < 0.01 compared with the nonacademic groups). Loss of MLH1/PMS2 staining was often (90%) followed by either BRAF mutational analysis or MLH1 methylation assays. Only 24% adhered to WHO recommendations to assign histologic grade based on mismatch repair status. We found only 3 cases (0.4%) with discordant immunohistochemistry and PCR results in our own practice: 1 reflected decreased MSH-6 staining in a neoadjuvantly treated microsatellite stable tumor, 1 MLH1-deficient tumor showed diminished MLH1/PMS2 in the tumor compared with internal control, and 1 case reflected an error in the molecular laboratory. Overall, our results showed extremely low discordance between methods assessing mismatch repair status and would suggest immunohistochemistry as the preferred single screening test. PCR can be reserved for cases that show equivocal immunostaining patterns.
Collapse
Affiliation(s)
- Erika Hissong
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Elizabeth P Crowe
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Rhonda K Yantiss
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Yao-Tseng Chen
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
26
|
Ta RM, Hecht JL, Lin DI. Discordant loss of mismatch repair proteins in advanced endometrial endometrioid carcinoma compared to paired primary uterine tumors. Gynecol Oncol 2018; 151:401-406. [PMID: 30340772 DOI: 10.1016/j.ygyno.2018.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/07/2018] [Accepted: 10/10/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE A subset of endometrial cancer is characterized by deficiencies in the mismatch repair (MMR) pathway. MMR testing is a well-established tool to screen for Lynch syndrome, but has also become a companion diagnostic test for immunotherapy. We compared the MMR status of primary and paired metastatic endometrial cancer to determine whether MMR deficiency can occur specifically in advanced endometrial cancer compared to primary tumor. METHODS Matched primary uterine and metastatic endometrioid adenocarcinoma from 2009 to 2018 at our institution were identified. PMS2 and MSH6 protein expression in metastatic and matched primary tumor was assessed using clinically validated immunohistochemistry methods for Lynch syndrome screening. MLH1 promoter hypermethylation and microsatellite instability (MSI) were performed in discordant cases. RESULTS 29 patients were identified with paired primary endometrial endometrioid adenocarcinoma and metastasis or recurrence after the original hysterectomy. Fourteen of 29 cases (48.2%, 14/29) were found to be MMR deficient at the metastatic or recurrent site. Two patients (6.9%, 2/29) showed discordant MMR status with PMS2 protein loss at the metastatic sites and intact expression in the primary uterine tumors. Both discordant cases exhibited abnormal subclonal loss at primary site and MLH1 promoter hypermethylation. High levels of microsatellite instability (MSI-H) was confirmed in one discordant metastatic site. CONCLUSION Advanced endometrial cancer can rarely (~7%) show somatic loss of MMR protein expression in recurrent or metastatic sites compared to matched paired primary tumor. MMR testing of recurrent or metastasis should be considered for guiding immunotherapy if primary uterine tumor exhibits abnormal subclonal MMR loss.
Collapse
Affiliation(s)
- Robert M Ta
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States of America
| | - Jonathan L Hecht
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States of America
| | - Douglas I Lin
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States of America.
| |
Collapse
|
27
|
O’Brien O, Ryan É, Creavin B, Kelly ME, Mohan HM, Geraghty R, Winter DC, Sheahan K. Correlation of immunohistochemical mismatch repair protein status between colorectal carcinoma endoscopic biopsy and resection specimens. J Clin Pathol 2018; 71:631-636. [DOI: 10.1136/jclinpath-2017-204946] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/11/2018] [Accepted: 01/13/2018] [Indexed: 12/15/2022]
Abstract
BackgroundMicrosatellite instability is reflective of a deficient mismatch repair system (dMMR), which may be due to either sporadic or germline mutations in the relevant mismatch repair (MMR) gene. MMR status is frequently determined by immunohistochemistry (IHC) for mismatch repair proteins (MMRPs) on colorectal cancer (CRC) resection specimens. However, IHC testing performed on endoscopic biopsy may be as reliable as that performed on surgical resections.AimWe aimed to evaluate the reliability of MMR IHC staining on preoperative CRC endoscopic biopsies compared with matched-surgical resection specimens.MethodsA retrospective search of our institution’s histopathology electronic database was performed. Patients with CRC who had MMR IHC performed on both their preoperative endoscopic biopsy and subsequent resection from January 2010 to January 2016 were included. Concordance of MMR staining between biopsy and resection specimens was assessed.ResultsFrom 2000 to 2016, 53 patients had MMR IHC performed on both their preoperative colorectal endoscopic biopsy and resection specimens; 10 patients (18.87%) demonstrated loss of ≥1 MMRP on their initial endoscopic tumour biopsy. The remainder (81.13%) showed preservation of staining for all MMRPs. There was complete agreement in MMR IHC status between the preoperative endoscopic biopsies and corresponding resection specimens in all cases (κ=1.000, P<0.000) with a sensitivity of 100% (95% CI 69.15 to 100) and specificity of 100% (95% CI 91.78 to 100) for detection of dMMR.ConclusionEndoscopic biopsies are a suitable source of tissue for MMR IHC analysis. This may provide a number of advantages to both patients and clinicians in the management of CRC.
Collapse
|
28
|
Bartosch C, Clarke B, Bosse T. Gynaecological neoplasms in common familial syndromes (Lynch and HBOC). Pathology 2017; 50:222-237. [PMID: 29287922 DOI: 10.1016/j.pathol.2017.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 12/22/2022]
Abstract
Recognising hereditary predisposition in a cancer patient has implications both for the patient and the patient's kindred. For the latter, cascade germline testing can reassure those not-at-risk family members while carriers can be enrolled in cancer screening and prevention programs that are medically effective and economically sustainable for health care systems. Furthermore, in many of these syndromes, ramifications of molecular phenotypes are increasing, and it is now emerging that, in addition, they convey prognostic and predictive information. Although cancer predisposition syndromes are rare, these molecular phenotypes also occur as somatic events in sporadic cancer settings. The information obtained from these molecular phenotypes, regardless of germline or somatic origin, is being incorporated into clinical management in view of their manifold significance. Thus, increasingly, bespoke management of cancer patients involves testing for both germline and somatic mutations in tumours. Lynch syndrome and BRCA-1 and BRCA-2-associated hereditary breast and ovarian cancer are hereditary cancer syndromes frequently involving the gynaecological tract but tumours associated with similar molecular alterations may also occur sporadically. Thus, the molecular phenotype of mismatch repair deficiency, microsatellite instability or hypermutator phenotype may be attributable to germline or somatic events. Similarly, homologous recombination deficiency or 'BRCAness' in ovarian cancers may be syndromic or sporadic. While hereditary syndromes are well recognised, the prognostic and predictive implications of these molecular phenotypes have only recently been elucidated and these aspects will finally ensure that molecular screening may become standard of care. Thus, nowadays pathologists are asked to designate the molecular phenotype of these cancers and then determine whether it is due to hereditary or sporadic causes.
Collapse
Affiliation(s)
- Carla Bartosch
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Blaise Clarke
- Department of Laboratory Medicine and Pathobiology, University of Toronto, University Health Network, Toronto, Ontario, Canada
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, The Netherlands.
| |
Collapse
|
29
|
Markow M, Chen W, Frankel WL. Immunohistochemical Pitfalls: Common Mistakes in the Evaluation of Lynch Syndrome. Surg Pathol Clin 2017; 10:977-1007. [PMID: 29103543 DOI: 10.1016/j.path.2017.07.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
At least 15% of colorectal cancers diagnosed in the United States are deficient in mismatch repair mechanisms. Most of these are sporadic, but approximately 3% of colorectal cancers result from germline alterations in mismatch repair genes and represent Lynch syndrome. It is critical to identify patients with Lynch syndrome to institute appropriate screening and surveillance for patients and their families. Exclusion of Lynch syndrome in sporadic cases is equally important because it reduces anxiety for patients and prevents excessive spending on unnecessary surveillance. Immunohistochemistry is one of the most widely used screening tools for identifying patients with Lynch syndrome.
Collapse
Affiliation(s)
- Michael Markow
- Department of Pathology, The Ohio State University Wexner Medical Center, 129 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210, USA
| | - Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, 129 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210, USA
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University Wexner Medical Center, 129 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
30
|
Marginean EC, Melosky B. Is There a Role for Programmed Death Ligand-1 Testing and Immunotherapy in Colorectal Cancer With Microsatellite Instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol Lab Med 2017; 142:17-25. [PMID: 29144791 DOI: 10.5858/arpa.2017-0040-ra] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - Colorectal cancer (CRC) represents the third most-common cancer in developed countries and is a leading cause of cancer deaths worldwide. Two recognized pathways contribute to CRC development: a more-common chromosomal instability pathway and, in 15% of cases, a deficient mismatch repair or microsatellite instability-high (MSI-H) pathway. The MSI-H CRC can be associated with somatic or germline mutations. Microsatellite status has been recognized as a prognostic and predictive biomarker. OBJECTIVES - To summarize the molecular pathways of CRC, with an emphasis on the MSI (mismatch repair) pathway; the recommended MSI testing algorithms and interpretation; and the prognostic and predictive role of MSI-H status in personalized treatment, including adjuvant chemotherapy, targeted therapy, and immune checkpoint inhibitor therapy. DATA SOURCES - A PubMed (US National Library of Medicine, Bethesda, Maryland) review was performed for articles pertaining to CRC, MSI and mismatch repair systems, molecular classification, immune response, programmed death receptor-1/programmed death ligand-1, and immunotherapy. CONCLUSIONS - Although the TNM classification of malignant tumor stage remains the key determinant of CRC prognosis and treatment, there are considerable stage-independent, interindividual differences in clinical outcome and therapy response by patients. In addition, MSI-H status has an important role in CRC management and can be reliably detected by molecular and immunohistochemistry techniques and genetic testing. Efforts must be made to identify whether MSI-H CRC is germline or sporadic to ensure appropriate treatment, accurate prognosis, and risk assessment for relatives. Microsatellite status has been recognized as a good prognostic indicator and is predictive of a poor response to 5-fluorouracil-based chemotherapy and a good response to programmed death ligand-1 inhibitor pembrolizumab in metastatic/refractory MSI-H CRC.
Collapse
Affiliation(s)
- Esmeralda Celia Marginean
- From the Department of Pathology, University of Ottawa, Ottawa, Ontario, Canada (Dr Marginean); the Gastrointestinal Pathology Section, Ottawa Hospital, Ottawa (Dr Marginean); the Department of Medical Oncology, University of British Columbia, Vancouver, Canada (Dr Melosky); and the Department of Oncology, British Columbia Cancer Agency, Vancouver (Dr Melosky)
| | - Barbara Melosky
- From the Department of Pathology, University of Ottawa, Ottawa, Ontario, Canada (Dr Marginean); the Gastrointestinal Pathology Section, Ottawa Hospital, Ottawa (Dr Marginean); the Department of Medical Oncology, University of British Columbia, Vancouver, Canada (Dr Melosky); and the Department of Oncology, British Columbia Cancer Agency, Vancouver (Dr Melosky)
| |
Collapse
|
31
|
Chen W, Swanson BJ, Frankel WL. Molecular genetics of microsatellite-unstable colorectal cancer for pathologists. Diagn Pathol 2017; 12:24. [PMID: 28259170 PMCID: PMC5336657 DOI: 10.1186/s13000-017-0613-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/20/2017] [Indexed: 12/26/2022] Open
Abstract
Background Microsatellite-unstable colorectal cancers (CRC) that are due to deficient DNA mismatch repair (dMMR) represent approximately 15% of all CRCs in the United States. These microsatellite-unstable CRCs represent a heterogenous group of diseases with distinct oncogenesis pathways. There are overlapping clinicopathologic features between some of these groups, but many important differences are present. Therefore, determination of the etiology for the dMMR is vital for proper patient management and follow-up. Main body Epigenetic inactivation of MLH1 MMR gene (sporadic microsatellite-unstable CRC) and germline mutation in an MMR gene (Lynch syndrome, LS) are the two most common mechanisms in the pathogenesis of microsatellite instability in CRC. However, in a subset of dMMR CRC cases that are identified by screening tests, no known LS-associated genetic alterations are appreciated by current genetic analysis. When the etiology for dMMR is unclear, it leads to patient anxiety and creates challenges for clinical management. Conclusion It is critical to distinguish LS patients from other patients with tumors due to dMMR, so that the proper screening protocol can be employed for the patients and their families, with the goal to save lives while avoiding unnecessary anxiety and costs. This review summarizes the major pathogenesis pathways of dMMR CRCs, their clinicopathologic features, and practical screening suggestions. In addition, we include frequently asked questions for MMR immunohistochemistry interpretation.
Collapse
Affiliation(s)
- Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, S301 Rhodes Hall, 450 W. 10th Ave, Columbus, Ohio, 43210, USA.,Department of Pathology, The Ohio State University Wexner Medical Center, 129 Hamilton Hall, Columbus, Ohio, 43210, USA
| | - Benjamin J Swanson
- Department of Pathology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University Wexner Medical Center, S301 Rhodes Hall, 450 W. 10th Ave, Columbus, Ohio, 43210, USA. .,Department of Pathology, The Ohio State University Wexner Medical Center, 129 Hamilton Hall, Columbus, Ohio, 43210, USA.
| |
Collapse
|
32
|
Kuan SF, Ren B, Brand R, Dudley B, Pai RK. Neoadjuvant therapy in microsatellite-stable colorectal carcinoma induces concomitant loss of MSH6 and Ki-67 expression. Hum Pathol 2017; 63:33-39. [PMID: 28232158 DOI: 10.1016/j.humpath.2017.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/26/2017] [Accepted: 02/03/2017] [Indexed: 12/22/2022]
Abstract
Universal screening using immunohistochemistry for DNA mismatch-repair proteins (MLH1, MSH2, MSH6, and PMS2) is advocated by major professional medical organizations to identify Lynch syndrome-associated colorectal carcinoma. Loss of MSH6 expression independent of MSH2 expression has been reported in microsatellite-stable (MSS) colorectal carcinoma after neoadjuvant therapy. The mechanism remains unclear. We studied the immunohistochemical expression of MSH2, MSH6, and Ki-67 in MSS colorectal carcinoma with (n=50) or without (n=64) preoperative neoadjuvant therapy and Lynch syndrome-associated colorectal carcinoma with confirmed MSH6 germline mutation (n=3). Twelve of 50 MSS colorectal carcinoma postneoadjuvant resections demonstrated reduced MSH6 expression, with loss of expression ranging from 20% to 100% of tumor cells. Eight of 64 MSS colorectal carcinomas without neoadjuvant therapy also exhibited reduced MSH6 expression but to a lesser degree (10%-50% of tumor cells with loss of expression). In both subgroups, concomitant loss of MSH6 and Ki-67 expressions was demonstrated in the same tumor areas in consecutive tissue sections. However, 3 cases of Lynch syndrome-associated colorectal carcinoma due to germline MSH6 mutation revealed complete loss of MSH6 expression with discordant positive Ki-67 staining in the tumor cells. The MSH2-independent, Ki-67-related expression of MSH6 in colorectal carcinoma helps to explain the heterogeneous MSH6 staining in MSS colorectal carcinoma with or without neoadjuvant therapy.
Collapse
Affiliation(s)
- Shih-Fan Kuan
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213.
| | - Bing Ren
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Randall Brand
- Department of Internal Medicine, Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Beth Dudley
- Department of Internal Medicine, Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| |
Collapse
|
33
|
Vilkin A, Leibovici-Weissman Y, Halpern M, Morgenstern S, Brazovski E, Gingold-Belfer R, Wasserberg N, Brenner B, Niv Y, Sneh-Arbib O, Levi Z. Immunohistochemistry staining for mismatch repair proteins: the endoscopic biopsy material provides useful and coherent results. Hum Pathol 2015; 46:1705-1711. [PMID: 26359539 DOI: 10.1016/j.humpath.2015.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 07/07/2015] [Accepted: 07/15/2015] [Indexed: 01/07/2023]
Abstract
Immunohistochemistry (IHC) testing for mismatch repair proteins (MMRP) in patients with colorectal cancer can be performed on endoscopic biopsy material or the surgical resection material. Data are continuing to accumulate regarding the deleterious effect of neoadjuvant chemoradiation on MMRP expression. However, despite continuing rise in the use of endoscopic biopsies for IHC, most pathology departments still use mainly the surgical materials for IHC testing. In this study we compared the quality of stains among 96 colon cancer subjects with paired endoscopic and surgical material available for MLH1, MSH2, MSH6, and PMS2 stains (96 × 4, yielding 384 paired stains). Each slide received both a quantitative score (immunoreactivity [0-3] × percent positivity [0-4]) and a qualitative score (absent; weak and focal; strong). The quantitative scores of all MMRP were significantly higher among the endoscopic material (P<.001 for all). In 358 pairs (93.2%), both the endoscopic and operative material stained either strong (322, 83.9%) or absent (36, 9.4%). In 26 pairs (6.8%), the endoscopic material stained strong, whereas the operative material stained focal and weak. No endoscopic biopsy materials stained focal and weak. Our findings indicate that the biopsy material may provide more coherent results. Although these results may indicate that biopsy material provides coherent and useful results, it is yet to be determined if the demonstrated differences pose a real clinical problem in interpreting final results of IHC staining of such kind. Hence, we suggest that when available, the endoscopic material rather than the operative one should serve as the primary substrate for IHC staining.
Collapse
Affiliation(s)
- Alex Vilkin
- The Gastroenterology Department, Beilinson Hospital, Petah Tiqva, PO 49100, Israel
| | | | - Marisa Halpern
- The Pathology Department, Hasharon Hospital, Petah Tiqva, PO 49100, Israel
| | - Sara Morgenstern
- The Pathology Department, Beilinson Hospital, Petah Tiqva, PO 49100, Israel
| | - Eli Brazovski
- The Pathology Department, Tel Aviv Medical Center, PO 49100, Israel
| | | | - Nir Wasserberg
- The Surgical Division, Rabin Medical Center, Beilinson Hospital, Petah Tiqva, PO 49100, Israel
| | - Baruch Brenner
- The Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petah Tiqva, PO 49100, Israel
| | - Yaron Niv
- The Gastroenterology Department, Beilinson Hospital, Petah Tiqva, PO 49100, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, PO 69978 Israel
| | - Orly Sneh-Arbib
- The Gastroenterology Department, Beilinson Hospital, Petah Tiqva, PO 49100, Israel
| | - Zohar Levi
- The Gastroenterology Department, Beilinson Hospital, Petah Tiqva, PO 49100, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, PO 69978 Israel.
| |
Collapse
|