1
|
Boge F, Mosig A. Causality and scientific explanation of artificial intelligence systems in biomedicine. Pflugers Arch 2024:10.1007/s00424-024-03033-9. [PMID: 39470762 DOI: 10.1007/s00424-024-03033-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 11/01/2024]
Abstract
With rapid advances of deep neural networks over the past decade, artificial intelligence (AI) systems are now commonplace in many applications in biomedicine. These systems often achieve high predictive accuracy in clinical studies, and increasingly in clinical practice. Yet, despite their commonly high predictive accuracy, the trustworthiness of AI systems needs to be questioned when it comes to decision-making that affects the well-being of patients or the fairness towards patients or other stakeholders affected by AI-based decisions. To address this, the field of explainable artificial intelligence, or XAI for short, has emerged, seeking to provide means by which AI-based decisions can be explained to experts, users, or other stakeholders. While it is commonly claimed that explanations of artificial intelligence (AI) establish the trustworthiness of AI-based decisions, it remains unclear what traits of explanations cause them to foster trustworthiness. Building on historical cases of scientific explanation in medicine, we here propagate our perspective that, in order to foster trustworthiness, explanations in biomedical AI should meet the criteria of being scientific explanations. To further undermine our approach, we discuss its relation to the concepts of causality and randomized intervention. In our perspective, we combine aspects from the three disciplines of biomedicine, machine learning, and philosophy. From this interdisciplinary angle, we shed light on how the explanation and trustworthiness of artificial intelligence relate to the concepts of causality and robustness. To connect our perspective with AI research practice, we review recent cases of AI-based studies in pathology and, finally, provide guidelines on how to connect AI in biomedicine with scientific explanation.
Collapse
Affiliation(s)
- Florian Boge
- Institute for Philosophy and Political Science, Technical University Dortmund, Emil-Figge-Str. 50, 44227, Dortmund, Germany
| | - Axel Mosig
- Bioinformatics Group, Department for Biology and Biotechnology, Ruhr-University Bochum (RUB), Gesundheitscampus 4, 44801, Bochum, NRW, Germany.
- Center for Protein Diagnostics, Ruhr University Bochum, Gesundheitscampus 4, 44801, Bochum, Germany.
| |
Collapse
|
2
|
Mattis AJ, Chen JF, Gonzalez IA, Rais R, Dehner LP, Pfeifer J, He M. Immune checkpoint markers and tumour mutation burden in Wilms tumour: a study of 59 cases. Pathology 2024; 56:814-825. [PMID: 38879422 DOI: 10.1016/j.pathol.2024.03.005] [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: 09/19/2023] [Revised: 01/05/2024] [Accepted: 03/12/2024] [Indexed: 09/04/2024]
Abstract
Wilms tumour (WT) is the most common renal tumour in children, and studies of immune checkpoint inhibitors (ICIs) treatment and markers are limited in number. In this study we investigated the ICIs' related immune landscape by examining the expression of PD-L1, PD-1, CD8 and DNA mismatch repair (MMR) proteins by immunohistochemistry (IHC), tumour mutation burden (TMB), and correlations with histology and clinical outcome. Positive PD-L1 (SP263) expression was defined as modified combined positive score (CPS) ≥1. A total of 59 WTs (from 2000 to 2017), including eight (14.0%) with anaplasia, from 46 patients were analysed (45 primary and 14 metastatic). Thirteen WTs (13/59, 22%) were positive for PD-L1 (8 primary, 5 metastatic; CPS 1.11-3.42). Positive PD-L1 expression was associated with diffuse anaplasia (p<0.05) and significantly shorter progression-free survival (p<0.05) among WTs with favourable histology (n=39). CD8+ lymphocytes were present in all analysed WTs. A subset of CD8+ cells co-expressed PD-1, which was associated with favourable histology and treatment. MMR IHC stains identified two (2/18, 11%) WTs with isolated PMS2 loss. All six WTs analysed for TMB showed low mutation burden. We found CD8+ lymphocytes in all analysed WTs and identified a fraction of WT (17.8% of primary and 35.8% of metastatic) with positive PD-L1 CPS, suggesting potential response to ICIs in some patients.
Collapse
Affiliation(s)
- Aidas J Mattis
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Jie-Fu Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Ivan A Gonzalez
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Rehan Rais
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Louis P Dehner
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - John Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Mai He
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
3
|
Winkelmann R, Weißgerber M, Wild PJ, Bein J, Fleischmann M, Demes M, Balermpas P, Loth A, Bankov K, von der Grün J. Single Center Characterization of a Cohort of Salivary Gland Carcinomas. Life (Basel) 2024; 14:1089. [PMID: 39337873 PMCID: PMC11432769 DOI: 10.3390/life14091089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Salivary gland cancer (SGC) is a rare cancer that can present a diagnostic challenge to pathologists, with emerging, but still limited options for the treatment of recurrent/metastatic disease. We aimed to characterize the cohort of salivary gland cancers in our institute and generate a tissue microarray (TMA) with clinical data available for immunohistochemical analysis. We extracted the cases of salivary gland cancers in our institute and generated a TMA with 72 patients between 2002 and 2017 with sufficient paraffin block material. Follow-up data were present for all cases. The TMA was stained with three p53 antibodies as well as MSH2, MSH6, PMS2 and MLH1 antibodies. Additionally, we applied fragment analysis based on the Bethesda panel, and the IdyllaTM MSI test to cases with expression loss of any of the mismatch repair proteins (MMR-P) according to our immunohistochemistry (IHC). The investigated cohort shows that pT and pN stage are the only factors independently associated with survival, according to our multivariate analysis (p = 0.037 and p = 0.014). In univariate analysis, risk factors identified in our cohort were also age (p = 0.015), (lympho-) vascular invasion (p = 0.002 and p = 0.003) and risk stratification (p = 0.037). The p53 protein investigated by three antibodies showed no statistically significant association with survival or other tumor characteristics in the investigated cohort. According to MMR-P IHC, six cases of SGC showed an aberrant IHC phenotype. Additional IdyllaTM MSI test and fragment length analysis failed to confirm microsatellite instability. The pT and pN stage are the most important factors for survival in our cohort. In our cohort, antibodies directed against the protein p53 did not contribute to clinical decision-making and were not correlated with any known clinical characteristics. MSI appears to be insignificant in SGCs. Larger cohorts are needed for verification.
Collapse
Affiliation(s)
- Ria Winkelmann
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (M.W.); (P.J.W.); (J.B.); (M.D.); (K.B.)
| | - Maja Weißgerber
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (M.W.); (P.J.W.); (J.B.); (M.D.); (K.B.)
| | - Peter J. Wild
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (M.W.); (P.J.W.); (J.B.); (M.D.); (K.B.)
- Frankfurt Institute for Advanced Studies (FIAS), Ruth-Moufang-Straße 1, 60438 Frankfurt am Main, Germany
| | - Julia Bein
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (M.W.); (P.J.W.); (J.B.); (M.D.); (K.B.)
| | - Maximilian Fleischmann
- Department of Radiation Oncology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (M.F.); (P.B.); (J.v.d.G.)
| | - Melanie Demes
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (M.W.); (P.J.W.); (J.B.); (M.D.); (K.B.)
| | - Panagiotis Balermpas
- Department of Radiation Oncology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (M.F.); (P.B.); (J.v.d.G.)
- Department of Radiation Oncology, University Hospital Zürich, Rämistrasse 100, 8091 Zürich, Switzerland
| | - Andreas Loth
- Department of Otorhinolarygology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany;
| | - Katrin Bankov
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (M.W.); (P.J.W.); (J.B.); (M.D.); (K.B.)
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jens von der Grün
- Department of Radiation Oncology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany; (M.F.); (P.B.); (J.v.d.G.)
- Department of Radiation Oncology, University Hospital Zürich, Rämistrasse 100, 8091 Zürich, Switzerland
| |
Collapse
|
4
|
Heriyanto DS, Yoshuantari N, Akbariani G, Lau V, Hanini H, Hidayati Z, Arief MZ, Gunawan AN, Ridwanuloh AM, Kusharyoto W, Handaya AY, Ilyas M, Kurnianda J, Hutajulu SH, Susanti S. High Probability of Lynch Syndrome Among Colorectal Cancer Patients Is Associated With Higher Occurrence of KRAS and PIK3CA Mutations. World J Oncol 2024; 15:612-624. [PMID: 38993255 PMCID: PMC11236368 DOI: 10.14740/wjon1843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/25/2024] [Indexed: 07/13/2024] Open
Abstract
Background In Indonesia, early-onset colorectal cancer (EOCRC) rates are higher in patients < 50 years old compared to Western populations, possibly due to a higher frequency of Lynch syndrome (LS) in CRC patients. We aimed to examine the association of KRAS and PIK3CA mutations with LS. Methods In this retrospective cross-sectional single-center study, the PCR-HRM-based test was used for screening of microsatellite instability (MSI) mononucleotide markers (BAT25, BAT26, BCAT25, MYB, EWSR1), MLH1 promoter methylation, and oncogene mutations of BRAF (V600E), KRAS (exon 2 and 3), and PIK3CA (exon 9 and 20) in FFPE DNA samples. Results All the samples (n = 244) were from Dr. Sardjito General Hospital Yogyakarta, Indonesia. KRAS and PIK3CA mutations were found in 151/244 (61.88%) and 107/244 (43.85%) of samples, respectively. KRAS and PIK3CA mutations were significantly associated with MSI status in 32/42 (76.19%) and 25/42 (59.52%) of samples, respectively. KRAS mutation was significantly associated with LS status in 26/32 (81.25%) of samples. The PIK3CA mutation was present in a higher proportion in LS samples of 19/32 (59.38%), but not statistically significant. Clinicopathology showed that KRAS mutation was significantly associated with right-sided CRC and higher histology grade in 39/151 (25.83%) and 24/151 (16.44%) samples, respectively. PIK3CA mutation was significantly associated with female sex and lower levels of tumor-infiltrating lymphocytes in 62/107 (57.94%) and 26/107 (30.23%) samples, respectively. KRAS and PIK3CA mutations did not significantly affect overall survival (120 months) in LS and non-LS patients. Conclusions The high probability of LS in Indonesian CRC patients is associated with KRAS and PIK3CA mutations.
Collapse
Affiliation(s)
- Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr Sardjito General Hospital Yogyakarta, Indonesia
- Collaboration Research Center for Precision Oncology based Omics - PKR PrOmics, Yogyakarta, Indonesia
| | - Naomi Yoshuantari
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr Sardjito General Hospital Yogyakarta, Indonesia
| | - Gilang Akbariani
- Pathgen Diagnostik Teknologi, Ir. Soekarno Science and Technology Park, National Research and Innovation Agency Republic of Indonesia, Bogor, Indonesia
| | - Vincent Lau
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr Sardjito General Hospital Yogyakarta, Indonesia
| | - Hanifa Hanini
- Pathgen Diagnostik Teknologi, Ir. Soekarno Science and Technology Park, National Research and Innovation Agency Republic of Indonesia, Bogor, Indonesia
| | - Zulfa Hidayati
- Pathgen Diagnostik Teknologi, Ir. Soekarno Science and Technology Park, National Research and Innovation Agency Republic of Indonesia, Bogor, Indonesia
| | - Muhammad Zulfikar Arief
- Pathgen Diagnostik Teknologi, Ir. Soekarno Science and Technology Park, National Research and Innovation Agency Republic of Indonesia, Bogor, Indonesia
| | - Andrew Nobiantoro Gunawan
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr Sardjito General Hospital Yogyakarta, Indonesia
| | - Asep Muhamad Ridwanuloh
- Research Center for Genetic Engineering, Research Organization for Life Sciences and Environment, National Research and Innovation Agency Republic of Indonesia, Ir. Soekarno Science and Technology Park, Bogor, Indonesia
| | - Wien Kusharyoto
- Research Center for Genetic Engineering, Research Organization for Life Sciences and Environment, National Research and Innovation Agency Republic of Indonesia, Ir. Soekarno Science and Technology Park, Bogor, Indonesia
| | - Adeodatus Yuda Handaya
- Division of Digestive Surgeon, Department of Surgery, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr Sardjito General Hospital Yogyakarta, Indonesia
| | - Mohammad Ilyas
- Molecular Pathology Research Group, Academic Unit of Translational Medical Science, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Johan Kurnianda
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - Susanna Hilda Hutajulu
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - Susanti Susanti
- Pathgen Diagnostik Teknologi, Ir. Soekarno Science and Technology Park, National Research and Innovation Agency Republic of Indonesia, Bogor, Indonesia
- Molecular Pathology Research Group, Academic Unit of Translational Medical Science, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Muhammadiyah Purwokerto, Indonesia
| |
Collapse
|
5
|
Kiran N, Yashaswini C, Maheshwari R, Bhattacharya S, Prajapati BG. Advances in Precision Medicine Approaches for Colorectal Cancer: From Molecular Profiling to Targeted Therapies. ACS Pharmacol Transl Sci 2024; 7:967-990. [PMID: 38633600 PMCID: PMC11019743 DOI: 10.1021/acsptsci.4c00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
Precision medicine is transforming colorectal cancer treatment through the integration of advanced technologies and biomarkers, enhancing personalized and effective disease management. Identification of key driver mutations and molecular profiling have deepened our comprehension of the genetic alterations in colorectal cancer, facilitating targeted therapy and immunotherapy selection. Biomarkers such as microsatellite instability (MSI) and DNA mismatch repair deficiency (dMMR) guide treatment decisions, opening avenues for immunotherapy. Emerging technologies such as liquid biopsies, artificial intelligence, and machine learning promise to revolutionize early detection, monitoring, and treatment selection in precision medicine. Despite these advancements, ethical and regulatory challenges, including equitable access and data privacy, emphasize the importance of responsible implementation. The dynamic nature of colorectal cancer, with its tumor heterogeneity and clonal evolution, underscores the necessity for adaptive and personalized treatment strategies. The future of precision medicine in colorectal cancer lies in its potential to enhance patient care, clinical outcomes, and our understanding of this intricate disease, marked by ongoing evolution in the field. The current reviews focus on providing in-depth knowledge on the various and diverse approaches utilized for precision medicine against colorectal cancer, at both molecular and biochemical levels.
Collapse
Affiliation(s)
- Neelakanta
Sarvashiva Kiran
- Department
of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064, India
| | - Chandrashekar Yashaswini
- Department
of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064, India
| | - Rahul Maheshwari
- School
of Pharmacy and Technology Management, SVKM’s
Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Green Industrial Park, TSIIC,, Jadcherla, Hyderabad 509301, India
| | - Sankha Bhattacharya
- School
of Pharmacy and Technology Management, SVKM’S
NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Bhupendra G. Prajapati
- Shree.
S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat 384012, India
| |
Collapse
|
6
|
Tan X, Fang Y, Fan X, Deng W, Huang J, Cai Y, Zou J, Chen Z, Lin H, Xu L, Wang G, Zhan H, Huang S, Fu X. Testing region selection and prognostic analysis of MLH1 promoter methylation in colorectal cancer in China. Gastroenterol Rep (Oxf) 2024; 12:goae011. [PMID: 38566849 PMCID: PMC10985700 DOI: 10.1093/gastro/goae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/09/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
Background MLH1 promoter methylation analysis is recommended in screening for Lynch syndrome (LS) in patients with MLH1-deficient colorectal cancer (CRC). The study aims to identify specific methylation regions in the MLH1 promoter and to evaluate the clinicopathologic characteristics of and prognosis for patients with MLH1 methylation. Methods A total of 580 CRC cases were included. The DNA mismatch repair (MMR) protein expression was assessed by using immunohistochemistry (IHC). The methylation status of the Regions A, B, C, D, and E in the MLH1 promoter was tested by using bisulfite sequencing PCR. The specificities of the five regions were calculated. Associations between MLH1 methylation and clinicopathologic characteristics were evaluated. Kaplan-Meier analyses for overall survival (OS) were carried out. Results In 580 CRC cases, the specificities of the methylation test in Regions D and E were both 97.8%. In the MLH1-deficient CRCs, the frequencies of MLH1 methylation and BRAFV600E mutation were 52.6% and 14.6%, respectively; BRAFV600E mutation occurred in 27.7% of patients with MLH1-methylated CRC. In the MMR-deficient patients, compared with MLH1 unmethylation, MLH1 methylation was more common in patients who were aged ≥50 years, female, had no family history of LS-related tumors, and had tumors located at the right colon. In the MMR-deficient patients, the MLH1-methylated cases had lower OS rates than the unmethylated cases with a family history of LS-related tumors (P = 0.047). Conclusions Regions D and E in the MLH1 promoter are recommended for determining the MLH1 methylation status in screening for LS in MLH1-deficient CRC. In MMR-deficient patients, the MLH1-methylated cases had a worse OS than the unmethylated cases with a family history of LS-related cancer.
Collapse
Affiliation(s)
- Xiaoli Tan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yongzhen Fang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xinjuan Fan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Weihao Deng
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Jinglin Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yacheng Cai
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Jiaxin Zou
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Zhiting Chen
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Hanjie Lin
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Liang Xu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Guannan Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Huanmiao Zhan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Shuhui Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xinhui Fu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
7
|
Bratei AA, Stefan-van Staden RI. Pathological Features of Colorectal Adenocarcinoma Patients Related to MLH1. Cell Mol Bioeng 2024; 17:153-164. [PMID: 38737450 PMCID: PMC11082117 DOI: 10.1007/s12195-024-00797-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/30/2024] [Indexed: 05/14/2024] Open
Abstract
Background MLH1, one of the MMR proteins, is linked to DNA replication, its role being to repair the incorrect DNA sequences and to replace them with proper ones. The loss of the MLH1 gene expression is part of Lynch syndrome which can lead to a series of cancers like colorectal and endometrial ones. The aim of this paper is to correlate the levels of MLH1 in four different bio-logical fluids with clinicopathological features in colorectal cancer patients in order to predict them with high probability. Therefore, a mathematical model with given code in Matlab has been proposed to get the clinicopathological features with high probability by only introducing the values for MLH1 concentrations. All these data can be obtained in a very short time even before surgery which can be very helpful the surgeon and the oncologist. Methods Four types of samples (whole blood, saliva, urine and tissue) were analyzed using stochastic microsensors; concentrations of MLH1 were determined and compared with different macroscopic and micro-scopic pathological features to obtain mathematical models for early, non-invasive diagnostic of colorectal adenocarcinoma. Results There have been established criteria and mathematical models for tumor location, TNM grading system, depth of the tumor, lymphatic, vascular and perineural invasions and the presence of mucus in the tumoral mass. Conclusions By using whole blood, saliva and urine samples, the location can be approximated. The proposed mathematical models aimed to allow a minim/noninvasive characterization of the tumor and its location which can help the surgeon and the oncologist to choose faster the personalized treatment.
Collapse
Affiliation(s)
- Alexandru Adrian Bratei
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, Bucharest, Romania
- Laboratory of Electrochemistry and PATLAB, National Institute of Research for Electrochemistry and Condensed Matter, 060021 Bucharest-6, Romania
- Department of Pathology, Emergency University Hospital, Bucharest, Romania
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu-Mures, 540139 Targu Mures, Romania
| | - Raluca-Ioana Stefan-van Staden
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, Bucharest, Romania
- Laboratory of Electrochemistry and PATLAB, National Institute of Research for Electrochemistry and Condensed Matter, 060021 Bucharest-6, Romania
| |
Collapse
|
8
|
Soleimani A, Saeedi N, Al-Asady AM, Nazari E, Hanaie R, Khazaei M, Ghorbani E, Akbarzade H, Ryzhikov M, Avan A, Mehr SMH. Colorectal Cancer Stem Cell Biomarkers: Biological Traits and Prognostic Insights. Curr Pharm Des 2024; 30:1386-1397. [PMID: 38623972 DOI: 10.2174/0113816128291321240329050945] [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: 11/19/2023] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/17/2024]
Abstract
Due to self-renewal, differentiation, and limitless proliferation properties, Cancer Stem Cells (CSCs) increase the probability of tumor development. These cells are identified by using CSC markers, which are highly expressed proteins on the cell surface of CSCs. Recently, the therapeutic application of CSCs as novel biomarkers improved both the prognosis and diagnosis outcome of colorectal Cancer. In the present review, we focused on a specific panel of colorectal CSC markers, including LGR5, ALDH, CD166, CD133, and CD44, which offers a targeted and comprehensive analysis of their functions. The selection criteria for these markers cancer were based on their established significance in Colorectal Cancer (CRC) pathogenesis and clinical outcomes, providing novel insights into the CSC biology of CRC. Through this approach, we aim to elevate understanding and stimulate further research for developing effective diagnostic and therapeutic strategies in CRC.
Collapse
Affiliation(s)
- Atena Soleimani
- Department of Biochemistry, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Nikoo Saeedi
- Medical School, Islamic Azad University, Mashhad, Iran
| | | | - Elnaz Nazari
- Department of Physiology, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Reyhane Hanaie
- Department of Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Majid Khazaei
- Department of Physiology, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Elnaz Ghorbani
- Department of Microbiology, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Hamed Akbarzade
- Department of Biochemistry, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | - Mikhail Ryzhikov
- Department of Biochemistry, Saint Louis University, St. Louis, MO 63103, USA
| | - Amir Avan
- Department of Genetics, Mashhad University of Medical Sciences, Razavi Khorasan, Mashhad, Iran
| | | |
Collapse
|
9
|
Mandarano M, Pelliccia C, Tomasello L, Caselli E, Floridi C, Loreti E, Barberini F, Rulli A, Gili A, Potenza R, Puma F, Rosati E, Donini A, Petrina A, Baccari P, Del Sordo R, Colella R, Bellezza G, Sidoni A. A New Medium (HistoCold) for Surgical Specimens Preserving to Improve the Preanalytic Issues in Histopathological Samples Handling: Morphologic and Antigenic Analysis. Biopreserv Biobank 2023; 21:610-623. [PMID: 37192479 DOI: 10.1089/bio.2022.0128] [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] [Indexed: 05/18/2023] Open
Abstract
Introduction: The onset of precision medicine has led to the integration of traditional morphologic tissues evaluation with biochemical and molecular data for a more appropriate pathological diagnosis. The preanalytic phase and, particularly, timing of cold ischemia are crucial to guarantee high-quality biorepositories of formalin-fixed paraffin-embedded (FFPE) tissues for patients' needs and scientific research. However, delayed fixation using the gold-standard and carcinogenic fixative neutral-buffered formalin (NBF) can be a significant limitation to diagnosis and biopathological characterization. HistoCold (patented; Bio-Optica Milano S.p.A., Milano, Italy) is a nontoxic, stable, and refrigerated preservative solution for tissue handling. This study examined HistoCold's potential role in improving the preanalytic phase of the pathological diagnostic process. Materials and Methods: Breast, lung, or colorectal cancers (20, 25, and 10 cases, respectively) that were to be surgically resected were recruited between 2019 and 2021. Once specimens were surgically removed, three residual samples for each patient were first promptly immersed into HistoCold for 24, 48, and 72 hours and then FFPE. These were compared with routine specimens regarding morphologic features (hematoxylin and eosin) and tissue antigenicity (immunohistochemical stains). Results: Good concordance regarding both the morphologic characteristics of the neoplasms and their proteins expression between the routine and HistoCold handled tissues were found. The tissue handling with the solution never affected the histopathological diagnosis. Conclusions: The use of HistoCold for samples transporting is easy, allows for improving the management of cold ischemia time, and monitoring the fixation times in NBF, resulting in good quality tissue blocks for biobanking. Moreover, it could be a candidate to eliminate formalin from operating theaters. HistoCold looks very promising for the preanalytic phase of human tissues handling in the era of precision medicine, to provide the best service to patients, and to scientific research.
Collapse
Affiliation(s)
- Martina Mandarano
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cristina Pelliccia
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Laura Tomasello
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emanuele Caselli
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudia Floridi
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elisabetta Loreti
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Barberini
- Breast Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Antonio Rulli
- Breast Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessio Gili
- Section of Public Health, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rossella Potenza
- Thoracic Surgery Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Puma
- Thoracic Surgery Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emanuele Rosati
- Section of General and Emergency Surgery, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Annibale Donini
- Section of General and Emergency Surgery, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Adolfo Petrina
- General Surgery Unit, S.M. Misericordia Hospital, Perugia, Italy
| | - Paolo Baccari
- General Surgery Unit, S.M. Misericordia Hospital, Perugia, Italy
| | - Rachele Del Sordo
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Renato Colella
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Guido Bellezza
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Angelo Sidoni
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Grillo F, Ali M, Paudice M, Pigozzi S, Anselmi G, Scabini S, Sciallero S, Piol N, Mastracci L. Impact of formalin fixation on mismatch repair protein evaluation by immunohistochemistry. Virchows Arch 2023; 483:677-685. [PMID: 37773452 PMCID: PMC10673985 DOI: 10.1007/s00428-023-03661-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023]
Abstract
Mismatch repair/microsatellite instability (MMR/MSI) status in colorectal cancer (CRC) has become fundamental as a diagnostic, prognostic, and predictive factor. MMR immunohistochemistry (IHC) is considered a simple and reliable approach; however, its effectiveness depends on pre-analytic factors. Aim of this study was to investigate the impact of different fixation times/protocols on MMR protein IHC quality. Left over tissue from surgically resected CRC samples (cold ischemia time < 30 min) where fixed as follows: standard formalin fixation (24-48 h); hypo-fixation (<20 h); hyper-fixation (>90 h); cold (4°C) fixation (24-48 h); standard fixation for small sample size (0.5×0.5 cm). Samples for each group were collected from 30 resected CRC and the following parameters were evaluated on 600 immunohistochemical stains: intensity of expression; patchiness of staining; presence of central artefact. Forty-six immunoreactions were inadequate (score 0 intensity), the majority regarding MLH1 or PMS2 in the hypo-fixation group (47.8%), followed by the hyper-fixation group (28.1%); cold formalin fixation showed the least inadequate cases. Patchiness and central artefact were more frequent in hypo-fixation and standard fixation group compared to the others. MLH1 (closely followed by PMS2) performed worse with regard to immunostaining intensity (p=0.0002) in the standard and in the hypo-fixation group (p< 0.00001). Using a small sample size improved patchiness/central artefacts. This is the first study specifically created to evaluate the impact of fixation on MMR protein IHC, showing that both formalin hypo- and hyper-fixation can cause problems; 24-h formalin fixation as well as cold (4°C) formalin fixation are recommended for successful IHC MMR evaluation.
Collapse
Affiliation(s)
- Federica Grillo
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Murad Ali
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michele Paudice
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simona Pigozzi
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giorgia Anselmi
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefano Scabini
- Oncological Surgical Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefania Sciallero
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Nataniele Piol
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Mastracci
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
11
|
Grypari IM, Tzelepi V, Gyftopoulos K. DNA Damage Repair Pathways in Prostate Cancer: A Narrative Review of Molecular Mechanisms, Emerging Biomarkers and Therapeutic Targets in Precision Oncology. Int J Mol Sci 2023; 24:11418. [PMID: 37511177 PMCID: PMC10380086 DOI: 10.3390/ijms241411418] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer (PCa) has a distinct molecular signature, including characteristic chromosomal translocations, gene deletions and defective DNA damage repair mechanisms. One crucial pathway involved is homologous recombination deficiency (HRD) and it is found in almost 20% of metastatic castrate-resistant PCa (mCRPC). Inherited/germline mutations are associated with a hereditary predisposition to early PCa development and aggressive behavior. BRCA2, ATM and CHECK2 are the most frequently HRD-mutated genes. BRCA2-mutated tumors have unfavorable clinical and pathological characteristics, such as intraductal carcinoma. PARP inhibitors, due to the induction of synthetic lethality, have been therapeutically approved for mCRPC with HRD alterations. Mutations are detected in metastatic tissue, while a liquid biopsy is utilized during follow-up, recognizing acquired resistance mechanisms. The mismatch repair (MMR) pathway is another DNA repair mechanism implicated in carcinogenesis, although only 5% of metastatic PCa is affected. It is associated with aggressive disease. PD-1 inhibitors have been used in MMR-deficient tumors; thus, the MMR status should be tested in all metastatic PCa cases. A surrogate marker of defective DNA repair mechanisms is the tumor mutational burden. PDL-1 expression and intratumoral lymphocytes have ambivalent predictive value. Few experimental molecules have been so far proposed as potential biomarkers. Future research may further elucidate the role of DNA damage pathways in PCa, revealing new therapeutic targets and predictive biomarkers.
Collapse
Affiliation(s)
- Ioanna-Maria Grypari
- Cytology Department, Aretaieion University Hospital, National Kapodistrian University of Athens, 11528 Athens, Greece
| | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Kostis Gyftopoulos
- Department of Anatomy, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
12
|
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: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [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 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, 18 DS) and 20 endometrial specimens (9 LS, 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 two mixed carcinomas). Four blocks of normal mucosa away from tumor were selected from most cases, as available. MMR immunohistochemistry (IHC) specific to the primary tumor mutations were analyzed. dMMR crypts were found in 65% of LS and 0% of DS MMR mutated colonic adenocarcinomas (p < 0.001). Most dMMR crypts were detected in the colon (12 of 15) compared to ileum (3). dMMR crypts showed single and grouped loss of MMR IHC expression. dMMR glands were found in 67% of LS and 9% (1 of 11) of DS endometrial cases (p = 0.017). Most dMMR glands were found in the uterine wall, with 1 LS and 1 DS case exhibiting dMMR glands in the lower uterine segment. The majority of 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 rarer in those with DS MMR mutations.
Collapse
Affiliation(s)
- C Eric Freitag
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210
| | - Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210
| | - Rachel Pearlman
- Department of Internal Medicine, Clinical Cancer Genetics Program, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210
| | - Heather Hampel
- Division of Genetics and Genetic Counseling, City of Hope, Duarte, California, 91010
| | - Peter P Stanich
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210
| | - Casey M Cosgrove
- Division of Gynecologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, 43210
| | - Eric Q Konnick
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210.
| |
Collapse
|
13
|
Aminder S, Saveena J, Ankita S, Harpreet K, Kunal J, Vikram N, Sumit G, Bhavna G, Ramneek K. Histopathological Predictors of Microsatellite Instability in Colorectal Cancer-a Tertiary Care Center Experience. Indian J Surg Oncol 2023; 14:137-143. [PMID: 36891442 PMCID: PMC9986156 DOI: 10.1007/s13193-022-01633-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 08/23/2022] [Indexed: 10/14/2022] Open
Abstract
Microsatellite instability (MSI) has a therapeutic and prognostic implication in colorectal carcinomas (CRCs). It can be detected either by immunohistochemistry (IHC) or molecular studies. In developing countries, a significant proportion of the patients experience financial constraints limiting the utilization of healthcare facilities. We aimed to identify the possible clinicopathological variables which can be used as predictors of microsatellite instability in such patients. CRC cases received for MSI detection by IHC (for 1 and 1/2 years) were included. A panel of four IHC markers (anti-MLH1, anti-PMS2, anti-MSH2, and anti-MSH6) was used. Confirmation by the molecular study was recommended in all the IHC-proven MSI cases. Various clinicopathological parameters were evaluated as predictors of MSI. Microsatellite instability was detected in 40.6% (30/74) cases with MLH1 and PMS2 dual loss in 27% cases, MSH2 and MSH6 dual loss in 6.8%, loss of all four MMR proteins in 2.7%, and isolated PMS2 loss in 4.1%. MSI-H expression was shown by 36.5% cases with only 4.1% cases showing MSI-L expression. The age cut-off value to differentiate both the study groups (MSI vs MSS) was 63 years with a sensitivity of 47.7% and specificity of 86.7%. ROC curve showed an area under the curve of 0.65 (95% CI, 0.515-0.776; p-value = 0.03). On univariate analysis, age < 63 years, colon site, and absence of nodal metastasis were significantly higher in the MSI group. However, on multivariate analysis, only the age < 63 years was found to be significantly higher in the MSI group. Confirmation was molecular study could only be obtained in 12 cases and was completely concordant with MSI detection by IHC. MSI detection can be performed either by IHC or by molecular study. In this study, no histological parameter appeared to be the independent predictor of MSI status. The age < 63 years might predict the microsatellite instability, yet larger studies are needed for its validation. Thus, we recommend that IHC testing should be performed in all CRC cases.
Collapse
Affiliation(s)
- Singh Aminder
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Jindal Saveena
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Soni Ankita
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Kaur Harpreet
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Jain Kunal
- Department of Medical Oncology, Dayanand Medical College & Hospital, Ludhiana, Punjab India
| | - Narang Vikram
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Grover Sumit
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Garg Bhavna
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| | - Kaur Ramneek
- Department of Pathology, Dayanand Medical College & Hospital, Tagore Nagar, Ludhiana, Punjab 141001 India
| |
Collapse
|
14
|
Molnar A, Monroe H, Basri Aydin H, Arslan ME, Lightle A, Lee H, El Jabbour T. Tumors of the Digestive System: Comprehensive Review of Ancillary Testing and Biomarkers in the Era of Precision Medicine. Curr Oncol 2023; 30:2388-2404. [PMID: 36826143 PMCID: PMC9954843 DOI: 10.3390/curroncol30020182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Immunotherapy has remained at the vanguard of promising cancer therapeutic regimens due to its exceptionally high specificity for tumor cells and potential for significantly improved treatment-associated quality of life compared to other therapeutic approaches such as surgery and chemoradiation. This is especially true in the digestive system, where high rates of mutation give rise to a host of targetable tumor-specific antigens. Many patients, however, do not exhibit measurable improvements under immunotherapy due to intrinsic or acquired resistance, making predictive biomarkers necessary to determine which patients will benefit from this line of treatment. Many of these biomarkers are assessed empirically by pathologists according to nuanced scoring criteria and algorithms. This review serves to inform clinicians and pathologists of extant and promising upcoming biomarkers predictive of immunotherapeutic efficacy among digestive system malignancies and the ancillary testing required for interpretation by pathologists according to tumor site of origin.
Collapse
Affiliation(s)
- Attila Molnar
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| | - Hunter Monroe
- Department of Pathology, West Virginia University, Morgantown, WV 26506, USA
| | - Hasan Basri Aydin
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Mustafa Erdem Arslan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Lightle
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Hwajeong Lee
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Tony El Jabbour
- Department of Pathology, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
15
|
Williams HL, Dias Costa A, Zhang J, Raghavan S, Winter PS, Kapner KS, Ginebaugh SP, Väyrynen SA, Väyrynen JP, Yuan C, Navia AW, Wang J, Yang A, Bosse TL, Kalekar RL, Lowder KE, Lau MC, Elganainy D, Morales-Oyarvide V, Rubinson DA, Singh H, Perez K, Cleary JM, Clancy TE, Wang J, Mancias JD, Brais LK, Hill ER, Kozak MM, Linehan DC, Dunne RF, Chang DT, Koong AC, Hezel AF, Hahn WC, Shalek AK, Aguirre AJ, Nowak JA, Wolpin BM. Spatially Resolved Single-Cell Assessment of Pancreatic Cancer Expression Subtypes Reveals Co-expressor Phenotypes and Extensive Intratumoral Heterogeneity. Cancer Res 2023; 83:441-455. [PMID: 36459568 PMCID: PMC10548885 DOI: 10.1158/0008-5472.can-22-3050] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has been classified into classical and basal-like transcriptional subtypes by bulk RNA measurements. However, recent work has uncovered greater complexity to transcriptional subtypes than was initially appreciated using bulk RNA expression profiling. To provide a deeper understanding of PDAC subtypes, we developed a multiplex immunofluorescence (mIF) pipeline that quantifies protein expression of six PDAC subtype markers (CLDN18.2, TFF1, GATA6, KRT17, KRT5, and S100A2) and permits spatially resolved, single-cell interrogation of pancreatic tumors from resection specimens and core needle biopsies. Both primary and metastatic tumors displayed striking intratumoral subtype heterogeneity that was associated with patient outcomes, existed at the scale of individual glands, and was significantly reduced in patient-derived organoid cultures. Tumor cells co-expressing classical and basal markers were present in > 90% of tumors, existed on a basal-classical polarization continuum, and were enriched in tumors containing a greater admixture of basal and classical cell populations. Cell-cell neighbor analyses within tumor glands further suggested that co-expressor cells may represent an intermediate state between expression subtype poles. The extensive intratumoral heterogeneity identified through this clinically applicable mIF pipeline may inform prognosis and treatment selection for patients with PDAC. SIGNIFICANCE A high-throughput pipeline using multiplex immunofluorescence in pancreatic cancer reveals striking expression subtype intratumoral heterogeneity with implications for therapy selection and identifies co-expressor cells that may serve as intermediates during subtype switching.
Collapse
Affiliation(s)
- Hannah L. Williams
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jinming Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Srivatsan Raghavan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter S. Winter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kevin S. Kapner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Scott P. Ginebaugh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Sara A. Väyrynen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Juha P. Väyrynen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew W. Navia
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Junning Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Annan Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Radha L. Kalekar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kristen E. Lowder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mai Chan Lau
- Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Dalia Elganainy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Vicente Morales-Oyarvide
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Douglas A. Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Harshabad Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas E. Clancy
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jiping Wang
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Joseph D. Mancias
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Harvard Medical School; Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Lauren K. Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Emma R. Hill
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Margaret M. Kozak
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford, CA, USA
| | - David C. Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard F. Dunne
- Department of Medicine, Division of Hematology and Oncology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel T. Chang
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford, CA, USA
| | - Albert C. Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aram F. Hezel
- Department of Medicine, Division of Hematology and Oncology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - William C. Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alex K. Shalek
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew J. Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan A. Nowak
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Bösherz MS, Samarska IV, Gaisa NT. Scoring Systems for Immunohistochemistry in Urothelial Carcinoma. Methods Mol Biol 2023; 2684:3-25. [PMID: 37410225 DOI: 10.1007/978-1-0716-3291-8_1] [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] [Indexed: 07/07/2023]
Abstract
Immunohistochemistry is widely used in diagnostic and scientific analysis of urothelial carcinoma. Objective interpretation of staining results is mandatory for accuracy and comparability in diagnostic and therapeutic patient care as well as research.Herein we summarize and explain standardized microscopic evaluation and scoring approaches for immunohistochemical stainings. We focus on commonly used and generally feasible approaches for different cellular compartments and comment on their utility in diagnostics and research practice.
Collapse
Affiliation(s)
| | - Iryna V Samarska
- Department of Pathology, GROW - School for Oncology and Reproduction, Maastricht University, Medical Centre+, Maastricht, The Netherlands
| | - Nadine T Gaisa
- Institute of Pathology, University Hospital, RWTH Aachen University, Aachen, Germany
- German Study Group of Bladder Cancer (DFBK e.V.), Munich, Germany
| |
Collapse
|
17
|
Chan EM, Foster KJ, Bass AJ. WRN Is a Promising Synthetic Lethal Target for Cancers with Microsatellite Instability (MSI). Cancer Treat Res 2023; 186:313-328. [PMID: 37978143 DOI: 10.1007/978-3-031-30065-3_17] [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] [Indexed: 11/19/2023]
Abstract
Microsatellite instability (MSI), a type of genetic hypermutability arising from impaired DNA mismatch repair (MMR), is observed in approximately 3% of all cancers. Preclinical work has identified the RecQ helicase WRN as a promising synthetic lethal target for patients with MSI cancers. WRN depletion substantially impairs the viability of MSI, but not microsatellite stable (MSS), cells. Experimental evidence suggests that this synthetic lethal phenotype is driven by numerous TA dinucleotide repeats that undergo expansion mutations in the setting of long-standing MMR deficiency. The lengthening of TA repeats increases their propensity to form secondary DNA structures that require WRN to resolve. In the absence of WRN helicase activity, these unresolved DNA secondary structures stall DNA replication forks and induce catastrophic DNA damage.
Collapse
Affiliation(s)
- Edmond M Chan
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, USA.
- Broad Institute of MIT and Harvard, Cambridge, USA.
- New York Genome Center, New York, USA.
| | | | - Adam J Bass
- Novartis Institutes for BioMedical Research, Cambridge, USA
| |
Collapse
|
18
|
Vazzano J, Tomlinson J, Stanich PP, Pearlman R, Kalady MF, Chen W, Hampel H, Frankel WL. Universal tumor screening for lynch syndrome on colorectal cancer biopsies impacts surgical treatment decisions. Fam Cancer 2023; 22:71-76. [PMID: 35732921 PMCID: PMC9829580 DOI: 10.1007/s10689-022-00302-3] [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: 04/04/2022] [Accepted: 06/05/2022] [Indexed: 01/13/2023]
Abstract
Universal tumor screening (UTS) for Lynch syndrome (LS) on colorectal cancer (CRC) can be performed on biopsies or resection specimens. The advantage of biopsies is the chance to provide preoperative genetic counseling/testing (GC/T) so patients diagnosed with LS can make informed decisions regarding resection extent. We evaluated utilization of UTS on biopsies, percentage of patients with deficient mismatch repair (dMMR) who underwent GC/T preoperatively, and whether surgical/treatment decisions were impacted. We performed a retrospective review of medical records to assess CRC cases with dMMR immunohistochemical staining from 1/1/2017 to 2/26/2021. 1144 CRC patients had UTS using MMR immunohistochemistry; 559 biopsies (48.9%) and 585 resections (51.1%). The main reason UTS was not performed on biopsy was it occurred outside our health system. 58 (5%) of CRCs were dMMR and did not have MLH1 promoter hypermethylation (if MLH1 and PMS2 absent). 28/58 (48.3%) of dMMR cases were diagnosed on biopsy. Of those 28, 14 (50%) eventually underwent GC/T, and 7 (25%) had GT results prior to surgery. One of the 7 had incomplete documentation of results affecting their treatment plan. Of the remaining 6 with complete documentation, 5 underwent surgery and one was treated with immunotherapy only. Three patients elected a more extensive surgery. 6/28 (21.4%) dMMR patients identified on biopsy made an informed surgical/treatment decision based on their dMMR status/LS diagnosis. When applied, UTS on biopsy followed by genetic counseling and testing informs surgical decision-making. Process and implementation strategies are in place to overcome challenges to more broadly optimize this approach.
Collapse
Affiliation(s)
- Jennifer Vazzano
- Department of Pathology, The Ohio State University Wexner Medical Center, Optometry Clinic and Health Science Faculty Office Building, 1664 Neil Avenue, Suite 6100, Columbus, OH, 43210, USA.
| | - Jewel Tomlinson
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Peter P Stanich
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Rachel Pearlman
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Matthew F Kalady
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, Optometry Clinic and Health Science Faculty Office Building, 1664 Neil Avenue, Suite 6100, Columbus, OH, 43210, USA
| | - Heather Hampel
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University Wexner Medical Center, Optometry Clinic and Health Science Faculty Office Building, 1664 Neil Avenue, Suite 6100, Columbus, OH, 43210, USA
| |
Collapse
|
19
|
Salman FG, Kankaya D, Özakıncı H, Şahin Y, Kubilay E, Süer E, Hayme S, Baltacı S. Role of PD-1/PD-L1-mediated tumour immune escape mechanism and microsatellite instability in the BCG failure of high-grade urothelial carcinomas. Turk J Med Sci 2022; 52:1802-1813. [PMID: 36945968 PMCID: PMC10390201 DOI: 10.55730/1300-0144.5526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 10/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Intravesical BCG treatment fails inexplicably in 30%-45% of patients for high-grade nonmuscle-invasive bladder cancer (NMIBC). We aimed to investigate the role of PD-1/PD-L1 interaction on BCG failure of high-grade NMIBC and to identify biomarkers for predicting BCG responsive cases. METHODS Thirty BCG responsive and 29 nonresponsive NMIBCs were included in the study. Expressions of PDL1(SP-263), MSH2, MSH6, PMS2, and MLH1 were evaluated on pre- and post-BCG transurethral resection (TUR-B) specimens by immunohistochemistry. PD-L1(SP-263) expression was categorised as negative/low, high. DNA mismatch repair protein (MMR) expressions were classified as "reduced" if ≤30% of nuclei stained, "preserved" if >30% of nuclei stained. Microsatellite instability (MSI) testing was performed by PCR using five mononucleotide markers. RESULTS Reduced DNA MMR protein expression was found to be significantly higher in the pretreatment biopsies of BCG-responsive group than the BCG nonresponsive tumour group (p = 0.022). PD-L1 expression did not show any significant difference between the pre- and posttreatment TUR-B specimens of the BCG nonresponsive tumour group or between the pretreatment TUR-B specimens of BCG nonresponsive and the BCG responsive groups (p = 0.508, p = 0.708, respectively). DISCUSSION Immune escape of tumour cells by PD-1/PD-L1 interaction does not seem to have any role in BCG failure of NMIBCs. Reduced MMR expression may help to determine cases that will respond well to BCG therapy. A better antitumour activity of BCG in NMIBCs with reduced MMR expression may be related to the ongoing accumulation of cancer neoantigens in correlation with increased tumour mutation load as a result of DNA repair defects.
Collapse
Affiliation(s)
- Fadime Gül Salman
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Duygu Kankaya
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Hilal Özakıncı
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Yasemin Şahin
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Eralp Kubilay
- Department of Urology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Evren Süer
- Department of Urology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Serhat Hayme
- Department of Biostatistics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Sümer Baltacı
- Department of Urology, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
20
|
Reitsam NG, Märkl B, Dintner S, Waidhauser J, Vlasenko D, Grosser B. Concurrent loss of MLH1, PMS2 and MSH6 immunoexpression in digestive system cancers indicating a widespread dysregulation in DNA repair processes. Front Oncol 2022; 12:1019798. [PMID: 36387226 PMCID: PMC9643848 DOI: 10.3389/fonc.2022.1019798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
Immunohistochemical analysis of mismatch repair (MMR) protein expression is widely used to identify tumors with a deficient MMR (dMMR). MMR proteins (MLH1/PMS2 and MSH2/MSH6) work as functional heterodimers, which usually leads to the loss of expression in only one functional MMR heterodimer. Recently, there have been studies showing the simultaneous loss of immunoexpression in proteins of both heterodimers. Yet, this phenomenon has been rarely investigated. In this study, we retrospectively considered cases of different digestive system cancers (gastric cancer, ampullary cancer, small bowel cancer, colorectal cancer), which were immunohistochemically tested for dMMR within a 4-year period at our university hospital (n=352). Of the 103 cases showing dMMR, 5 cases (1.4% of all, 5.1% of dMMR cases) showed a concurrent loss of MLH1, PMS2 and MSH6 immunoexpression, whereas in the other 98 dMMR cases only one MMR heterodimer was affected. MLH1-/PMS2-/MSH6- cancer cases almost arose throughout the entire digestive tract: from the gastric antrum to the left colic flexur. To provide a comprehensive molecular characterization of this MLH1-/PMS2-/MSH6- immunophenotype, tumors were analyzed for microsatellite instability, MLH1 promotor hypermethylation and BRAF exon 15 status. Furthermore, we performed next-generation sequencing focusing on genes related to DNA repair. Here, we could detect pathogenic germline variants as well as multiple sporadic mutations in different genes involved in MMR and homologous recombination repair (HRR) respectively. The affected MMR/HRR-related genes were: ATM, BARD1, BRCA1, CDK12, CHEK1, CHEK2, FANCA, MLH1, MSH6, PALB2, TP53. Considering the biologic function of HRR/MMR proteins as potential drug targets and the low frequency of most of these mutations in digestive system cancers in general, their common occurrence in our MLH1-/PMS2-/MSH6- cases seems to be even more noteworthy, highlighting the need for recognition, awareness and further investigation of this unusual IHC staining pattern.
Collapse
Affiliation(s)
- Nic Gabriel Reitsam
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
- *Correspondence: Nic Gabriel Reitsam, ;
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Sebastian Dintner
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Johanna Waidhauser
- Department of Hematology and Oncology, University Medical Center Augsburg / University Hospital of Augsburg, Augsburg, Germany
| | - Dmytro Vlasenko
- General, Visceral and Transplantation Surgery, University Hospital of Augsburg, Augsburg, Germany
| | - Bianca Grosser
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
21
|
Senhaji N, Squalli Houssaini A, Lamrabet S, Louati S, Bennis S. Molecular and Circulating Biomarkers in Patients with Glioblastoma. Int J Mol Sci 2022; 23:7474. [PMID: 35806478 PMCID: PMC9267689 DOI: 10.3390/ijms23137474] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/28/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma is the most aggressive malignant tumor of the central nervous system with a low survival rate. The difficulty of obtaining this tumor material represents a major limitation, making the real-time monitoring of tumor progression difficult, especially in the events of recurrence or resistance to treatment. The identification of characteristic biomarkers is indispensable for an accurate diagnosis, the rigorous follow-up of patients, and the development of new personalized treatments. Liquid biopsy, as a minimally invasive procedure, holds promise in this regard. The purpose of this paper is to summarize the current literature regarding the identification of molecular and circulating glioblastoma biomarkers and the importance of their integration as a valuable tool to improve patient care.
Collapse
Affiliation(s)
- Nadia Senhaji
- Department of Biology, Faculty of Sciences, Moulay Ismail University, Meknes 50000, Morocco
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco; (A.S.H.); (S.L.); (S.B.)
| | - Asmae Squalli Houssaini
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco; (A.S.H.); (S.L.); (S.B.)
| | - Salma Lamrabet
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco; (A.S.H.); (S.L.); (S.B.)
| | - Sara Louati
- Medical Biotechnology Laboratory, Faculty of Medicine and Pharmacy of Rabat, Mohammed Vth University, Rabat 10000, Morocco;
| | - Sanae Bennis
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco; (A.S.H.); (S.L.); (S.B.)
| |
Collapse
|
22
|
Ceccon C, Angerilli V, Rasola C, Procaccio L, Sabbadin M, Bergamo F, Malapelle U, Lonardi S, Fassan M. Microsatellite Instable Colorectal Adenocarcinoma Diagnostics: The Advent of Liquid Biopsy Approaches. Front Oncol 2022; 12:930108. [PMID: 35837109 PMCID: PMC9273960 DOI: 10.3389/fonc.2022.930108] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/30/2022] [Indexed: 11/24/2022] Open
Abstract
The introduction of immunotherapy has revolutionized the oncological targeted therapy paradigm. Microsatellite instability (MSI) identifies a subgroup of colorectal cancers (CRCs) which respond to treatment with immune checkpoint inhibitors. Tissue biopsy is currently the gold standard for the assessment of MSI/Mismatch Repair deficiency (MMRd) by means immunohistochemistry or molecular assays. However, the application of liquid biopsy in the clinic may help to overcome several limitations of tissue analysis and may provide great benefit to the diagnostic scenario and therapeutic decision-making process. In the context of MSI/MMRd CRC, the use of liquid biopsy may allow to establish MSI/MMR status if tissue sampling cannot be performed or in case of discordant tissue biopsies. Liquid biopsy may also become a powerful tool to monitor treatment response and the onset resistance to immunotherapy over time and to stratify of MSI/MMRd patients according to their risk of relapse and metastases. The aim of this review is to summarize the main technical aspects and clinical applications, the benefits, and limitations of the use of liquid biopsy in MSI/MMRd colorectal cancer patients.
Collapse
Affiliation(s)
- Carlotta Ceccon
- Department of Medicine (DIMED), University of Padua, Padua, Italy
| | | | - Cosimo Rasola
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | | | | | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Sara Lonardi
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua, Italy
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
- *Correspondence: Matteo Fassan,
| |
Collapse
|
23
|
BRAF and MLH1 Analysis Algorithm for the Evaluation of Lynch Syndrome Risk in Colorectal Carcinoma Patients: Evidence-Based Data from the Analysis of 100 Consecutive Cases. JOURNAL OF MOLECULAR PATHOLOGY 2022. [DOI: 10.3390/jmp3030011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Several causes may lead to CRC, either extrinsic (sporadic forms) or genetic (hereditary forms), such as Lynch syndrome (LS). Most sporadic deficient mismatch repair (dMMR) CRC cases are characterized by the methylation of the MLH1 promoter gene and/or BRAF gene mutations. Usually, the first test performed is the mismatch repair deficiency analysis. If a tumor shows a dMMR, BRAF mutations and then the MLH1 promoter methylation status have to be assessed, according to the ACG/ASCO screening algorithm. In this study, 100 consecutive formalin-fixed and paraffin-embedded samples of dMMR CRC were analyzed for both BRAF mutations and MLH1 promoter methylation. A total of 47 (47%) samples were BRAF p.V600E mutated, while MLH1 promoter methylation was found in 77 cases (77.0%). The pipeline “BRAF-followed-by-MLH1-analysis” led to a total of 153 tests, while the sequence “MLH1-followed-by-BRAF-analysis” resulted in a total of 123 tests. This study highlights the importance of performing MLH1 analysis in LS screening of BRAF-WT specimens before addressing patients to genetic counseling. We show that MLH1 analysis performs better as a first-line test in the screening of patients with LS risk than first-line BRAF analysis. Our data indicate that analyzing MLH1 methylation as a first-line test is more cost-effective.
Collapse
|
24
|
Li JJX, Ip PPC. Endometrial Cancer: An Update on Prognostic Pathologic Features and Clinically Relevant Biomarkers. Surg Pathol Clin 2022; 15:277-299. [PMID: 35715162 DOI: 10.1016/j.path.2022.02.006] [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] [Indexed: 06/15/2023]
Abstract
The prognosis of endometrial cancers has historically been determined by the evaluation of histologic typing, grading, and staging. Recently, molecular classification, pioneered by the 4 prognostic categories from The Cancer Genome Atlas Research Network, has been shown to independently predict the outcome, correlate with biomarker expression, and predict response to adjuvant chemotherapy. In modern-day pathology practice, it has become necessary to integrate the time-honored prognostic pathologic features with molecular classification to optimize patient management. In this review, the significance of the molecular classification of endometrioid carcinomas, the application of practical diagnostic surrogate algorithms, and interpretation of test results will be addressed. Histologic features and theragnostic biomarkers will also be discussed in relation to the molecular subtypes of endometrial cancers.
Collapse
Affiliation(s)
- Joshua J X Li
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR
| | - Philip P C Ip
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, 102 Pok Fu Lam Road, Hong Kong SAR.
| |
Collapse
|
25
|
Sun F, Grenert JP, Tan L, Van Ziffle J, Joseph NM, Mulvey CK, Bergsland E. Checkpoint Inhibitor Immunotherapy to Treat Temozolomide-Associated Hypermutation in Advanced Atypical Carcinoid Tumor of the Lung. JCO Precis Oncol 2022; 6:e2200009. [PMID: 35737914 PMCID: PMC9249272 DOI: 10.1200/po.22.00009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/06/2022] [Accepted: 05/04/2022] [Indexed: 01/23/2023] Open
Affiliation(s)
- Fangdi Sun
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - James P. Grenert
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Lisa Tan
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Jessica Van Ziffle
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Nancy M. Joseph
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Claire K. Mulvey
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Emily Bergsland
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
26
|
Zito Marino F, Amato M, Ronchi A, Panarese I, Ferraraccio F, De Vita F, Tirino G, Martinelli E, Troiani T, Facchini G, Pirozzi F, Perrotta M, Incoronato P, Addeo R, Selvaggi F, Lucido FS, Caraglia M, Savarese G, Sirica R, Casillo M, Lieto E, Auricchio A, Cardella F, Docimo L, Galizia G, Franco R. Microsatellite Status Detection in Gastrointestinal Cancers: PCR/NGS Is Mandatory in Negative/Patchy MMR Immunohistochemistry. Cancers (Basel) 2022; 14:cancers14092204. [PMID: 35565332 PMCID: PMC9102010 DOI: 10.3390/cancers14092204] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Microsatellite instability (MSI) detection has a high impact on eligibility for immune checkpoint inhibitors in gastrointestinal cancers. The appropriate detection of MSI represents the major critical issue in clinical practice, thus a better understanding of the limits related to MSI testing is needed to avoid misinterpretations. This study addresses the discordance between IHC and PCR/NGS testing in a large retrospective series of colorectal and gastric cancers in order to improve diagnosis. Our findings show a disagreement between negative/patchy expression IHC and PCR/NGS results, suggesting that molecular testing is mandatory in this subset of tumors. Abstract Background: Microsatellite instability (MSI) is a predictive biomarker for immune checkpoint inhibitors. The main goal was to investigate the discordance between IHC and PCR/NGS for MSI testing in gastrointestinal cancers. Methods: Two series were analyzed through IHC for mismatch-repair-system proteins (MMRP) and PCR, with one series of 444 colorectal cancers (CRC) and the other of 176 gastric cancers (GC). All cases with discordant results between IHC and PCR were analyzed by NGS. IHC staining was evaluated as follows: proficient MMR (pMMR), with all MMR positive; deficient MMR (dMMR), with the loss of one heterodimer; and cases with the loss/patchy expression of one MMR (lo-paMMR). Cases with instability in at least two markers by PCR were MSI-high (MSI-H) and with instability in one marker, MSI-low (MSI-L). Cases without instability were evaluated as microsatellite-stable (MSS). Results: In the CRC cohort, 15 out of 444 cases were dMMR and 46 lo-paMMR. Among the 15 dMMR, 13 were MSI-H and 2 MSS. Among the 46 lo-paMMR, 13 were MSI-H and 33 were MSS. In the GC cohort, 13 out of 176 cases were dMMR and 6 cases lo-paMMR. Among the 13 dMMR, 12 were MSI-H and only 1 was MSS. All six lo-paMMR cases were MSS. All NGS results were in agreement with PCR. Conclusions: In clinical practice, MMR–IHC could be used as a screening test and additional molecular analysis is mandatory exclusively in cases carrying loss/patchy MMR-IHC.
Collapse
Affiliation(s)
- Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Martina Amato
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Iacopo Panarese
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Franca Ferraraccio
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Ferdinando De Vita
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Giuseppe Tirino
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Erika Martinelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Teresa Troiani
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Gaetano Facchini
- Medical Oncology Unit, SM delle Grazie Hospital, 80078 Pozzuoli, Naples, Italy;
| | - Felice Pirozzi
- General Surgery Unit, Santa Maria delle Grazie Hospital, 80078 Pozzuoli, Naples, Italy;
| | - Michele Perrotta
- Hepatology and Interventional Ultrasound Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Naples, Italy;
| | - Pasquale Incoronato
- Medical Oncology Unit, ASL Napoli 2 Nord Hospital, 80014 Giugliano, Naples, Italy;
| | - Raffaele Addeo
- Medical Oncology Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Naples, Italy;
| | - Francesco Selvaggi
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Francesco Saverio Lucido
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Roberto Sirica
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Marika Casillo
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Eva Lieto
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Annamaria Auricchio
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Francesca Cardella
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Ludovico Docimo
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Gennaro Galizia
- Department of Surgical Sciences, School of Medicine, Second University of Naples, Place Miraglia, 3th Building, West Side, 4th Floor, 80138 Naples, Italy;
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
- Correspondence: ; Tel.: +39-0815664000
| |
Collapse
|
27
|
Li H, Sun L, Zhuang Y, Tian C, Yan F, Zhang Z, Hu Y, Liu P. Molecular mechanisms and differences in lynch syndrome developing into colorectal cancer and endometrial cancer based on gene expression, methylation, and mutation analysis. Cancer Causes Control 2022; 33:489-501. [PMID: 35149954 PMCID: PMC8904372 DOI: 10.1007/s10552-021-01543-w] [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: 03/11/2021] [Accepted: 12/14/2021] [Indexed: 11/29/2022]
Abstract
Purpose The aim of this study was to screen biomarkers specific to Lynch syndrome (LS) with colorectal cancer (CRC) or endometrial cancer (EC) to explore the mechanisms by which LS develops into CRC and EC and their differences. Methods Differentially expressed or differentially methylated genes and differential mutations were identified in 10 LS, 50 CRC, and 50 EC patients from TCGA, and genes overlapping between LS and CRC or EC (named SGs-LCs and SGs-LEs, respectively) were identified. Afterward, we annotated the enriched GO terms and pathways and constructed a protein–protein interaction (PPI) network. Finally, samples from 10 clinical cases with MSI-H/MSS CRC and EC were collected to verify the mutations and their correlations with five LS pathogenic genes in the SGs-LCs and SGs-LEs. Results A total of 494 SGs-LCs and 104 SGs-LEs were identified and enriched in 106 and 14 GO terms, respectively. There were great differences in the gene count and enriched terms between SGs-LCs and SGs-LEs. In the PPI network, SST, GCG, SNAP25, and NPY had the highest degree of connection among the SGs-LCs, and KIF20A and NUF2 had the highest degree of connection among the SGs-LE. In the SGs-LCs and SGs-LEs, the genes whose expression levels affected the survival of LS, CRC or EC patients were quite different. Conclusions COL11A1 was found to be mutated in MSS CRC patients, similar to the mutations of MSH6. SST, GCG, SNAP25, and NPY may be biomarkers for the development of LS into CRC, and KIF20A and NUF2 may be markers of LS developing into EC. Supplementary Information The online version contains supplementary material available at 10.1007/s10552-021-01543-w.
Collapse
Affiliation(s)
- Hongfeng Li
- Department of Clinical Laboratory, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Liwei Sun
- Department of Interventional Oncology, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Yan Zhuang
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Department of Colorectal Oncology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Caijuan Tian
- Tianjin Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd, Tianjin, 300381, China
| | - Fang Yan
- Tianjin Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd, Tianjin, 300381, China
| | - Zhenzhen Zhang
- Tianjin Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd, Tianjin, 300381, China
| | - Yuanjing Hu
- Department of Gynecological Oncology, Tianjin Central Hospital of Gynecology & Obstetrics, No. 156 Nankaisan Road, Nankai District, Tianjin, 300100, China.
| | - Pengfei Liu
- Department of Oncology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, No. 354 Beima Road, Hongqiao District, Tianjin, 300120, China.
| |
Collapse
|
28
|
Kim H, Lim KY, Park JW, Kang J, Won JK, Lee K, Shim Y, Park CK, Kim SK, Choi SH, Kim TM, Yun H, Park SH. Sporadic and Lynch syndrome-associated mismatch repair-deficient brain tumors. J Transl Med 2022; 102:160-171. [PMID: 34848827 PMCID: PMC8784316 DOI: 10.1038/s41374-021-00694-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022] Open
Abstract
Mismatch repair-deficient (MMRD) brain tumors are rare among primary brain tumors and can be induced by germline or sporadic mutations. Here, we report 13 MMRD-associated (9 sporadic and 4 Lynch syndrome) primary brain tumors to determine clinicopathological and molecular characteristics and biological behavior. Our 13 MMRD brain tumors included glioblastoma (GBM) IDH-wildtype (n = 9) including 1 gliosarcoma, astrocytoma IDH-mutant WHO grade 4 (n = 2), diffuse midline glioma (DMG) H3 K27M-mutant (n = 1), and pleomorphic xanthoastrocytoma (PXA) (n = 1). Next-generation sequencing using a brain tumor-targeted gene panel, microsatellite instability (MSI) testing, Sanger sequencing for germline MMR gene mutation, immunohistochemistry of MMR proteins, and clinicopathological and survival analysis were performed. There were many accompanying mutations, suggesting a high tumor mutational burden (TMB) in 77%, but TMB was absent in one case of GBM, IDH-wildtype, DMG, and PXA, respectively. MSH2, MLH1, MSH6, and PMS2 mutations were found in 31%, 31%, 31% and 7% of patients, respectively. MSI-high and MSI-low were found in 50% and 8% of these gliomas, respectively and 34% was MSI-stable. All Lynch syndrome-associated GBMs had MSI-high. In addition, 77% (10/13) had histopathologically multinucleated giant cells. The progression-free survival tended to be poorer than the patients with no MMRD gliomas, but the number and follow-up duration of our patients were insufficient to get statistical significance. In the present study, we found that the most common MMRD primary brain tumor was GBM IDH-wildtype. The genetic profile of MMRD GBM was different from that of conventional GBM. MMRD gliomas with TMB and MSI-H may be sensitive to immunotherapy but resistant to temozolomide. Our findings can help develop better treatment options.
Collapse
Affiliation(s)
- Hyunhee Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ka Young Lim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeongwan Kang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Kyung Won
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwanghoon Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yumi Shim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ki Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Institute of Neuroscience, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Gonzalez RS, Streutker CJ, Torlakovic EE. Proficiency Testing to Improve Interobserver Agreement for Mismatch Repair Deficiency Immunohistochemistry: An Invitation to Join CBQA Readout. Appl Immunohistochem Mol Morphol 2022; 30:79-82. [PMID: 35175237 DOI: 10.1097/pai.0000000000000995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Raul S Gonzalez
- From the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Catherine J Streutker
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON
| | - Emina E Torlakovic
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatchewan, SK, Canada
| |
Collapse
|
30
|
Imyanitov E, Kuligina E. Molecular testing for colorectal cancer: Clinical applications. World J Gastrointest Oncol 2021; 13:1288-1301. [PMID: 34721767 PMCID: PMC8529925 DOI: 10.4251/wjgo.v13.i10.1288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/19/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Molecular genetic analysis is an integral part of colorectal cancer (CRC) management. The choice of systemic therapy for CRC is largely based on the results of tumor molecular testing. Evaluation of the KRAS and NRAS gene status is mandatory for consideration of anti-epidermal growth factor receptor (EGFR) therapy. Tumors with the BRAF V600E substitution are characterized by aggressive behaviour, may require intensified cytotoxic regimens and benefit from combined BRAF and EGFR inhibition. The inactivation of DNA mismatch repair (MMR), or MUTYH gene, or DNA polymerase epsilon results in excessive tumor mutational burden; these CRCs are highly antigenic and therefore sensitive to immune checkpoint inhibitors. Some CRCs are characterized by overexpression of the HER2 oncogene and respond to the appropriate targeted therapy. There are CRCs with clinical signs of hereditary predisposition to this disease, which require germline genetic testing. Liquid biopsy is an emerging technology that has the potential to assist CRC screening, control the efficacy of surgical intervention and guide disease monitoring. The landscape of CRC molecular diagnosis is currently undergoing profound changes due to the increasing use of next generation sequencing.
Collapse
Affiliation(s)
- Evgeny Imyanitov
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, Saint-Petersburg 194100, Russia
- Department of Oncology, I.I. Mechnikov North-Western Medical University, Saint-Petersburg 191015, Russia
| | - Ekaterina Kuligina
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
| |
Collapse
|
31
|
Rüschoff J, Baretton G, Bläker H, Dietmaier W, Dietel M, Hartmann A, Horn LC, Jöhrens K, Kirchner T, Knüchel R, Mayr D, Merkelbach-Bruse S, Schildhaus HU, Schirmacher P, Tiemann M, Tiemann K, Weichert W, Büttner R. MSI testing : What's new? What should be considered? DER PATHOLOGE 2021; 42:110-118. [PMID: 34477921 DOI: 10.1007/s00292-021-00948-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
Based on new trial data regarding immune checkpoint inhibitors (ICIs), the detection of high-grade microsatellite instability (MSI-H) or underlying deficient mismatch repair protein (dMMR) is now becoming increasingly important for predicting treatment response. For the first time, a PD‑1 ICI (pembrolizumab) has been approved by the European Medicines Agency (EMA) for first-line treatment of advanced (stage IV) dMMR/MSI‑H colorectal cancer (CRC). Further indications, such as dMMR/MSI‑H endometrial carcinoma (EC), have already succeeded (Dostarlimab, 2nd line treatment) and others are expected to follow before the end of 2021. The question of optimal testing in routine diagnostics should therefore be re-evaluated. Based on a consideration of the strengths and weaknesses of the widely available methods (immunohistochemistry and PCR), a test algorithm is proposed that allows quality assured, reliable, and cost-effective dMMR/MSI‑H testing. For CRC and EC, testing is therefore already possible at the primary diagnosis stage, in line with international recommendations (NICE, NCCN). The clinician is therefore enabled from the outset to consider not only the predictive but also the prognostic and predispositional implications of such a test when counseling patients and formulating treatment recommendations. As a basis for quality assurance, participation in interlaboratory comparisons and continuous documentation of results (e.g., QuIP Monitor) are strongly recommended.
Collapse
Affiliation(s)
- Josef Rüschoff
- Institute of Pathology, Nordhessen und Targos Molecular Pathology GmbH, Germaniastr. 7, 34119, Kassel, Germany.
| | - Gustavo Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus, Fetscherstr. 74, 01307, Dresden, Germany
| | - Hendrik Bläker
- Institute of Pathology, University Hospital Leipzig, Liebigstr. 26, Gebäude G, 04103, Leipzig, Germany
| | - Wolfgang Dietmaier
- Institute of Pathology, Center of Molecular Pathological Diagnostics, Universität Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Manfred Dietel
- Institute of Pathology, University Hospital Charité, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Arndt Hartmann
- Pathological Institute, University Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Lars-Christian Horn
- Institute of Pathology, University Hospital Leipzig, Liebigstr. 26, Gebäude G, 04103, Leipzig, Germany
| | - Korinna Jöhrens
- Institute of Pathology, University Hospital Carl Gustav Carus, Fetscherstr. 74, 01307, Dresden, Germany
| | - Thomas Kirchner
- Pathological Institute, Ludwig-Maximilians-University Munich, Thalkirchner Str. 36, 80337, München, Germany
| | - Ruth Knüchel
- Institute of Pathology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Doris Mayr
- Pathological Institute, Ludwig-Maximilians-University Munich, Thalkirchner Str. 36, 80337, München, Germany
| | | | - Hans-Ulrich Schildhaus
- Institute of Pathology, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Peter Schirmacher
- Pathological Institute, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Markus Tiemann
- Fangdieckstr. 75a, Institute of Hematopathology Hamburg, 22547, Hamburg, Germany
| | - Katharina Tiemann
- Fangdieckstr. 75a, Institute of Hematopathology Hamburg, 22547, Hamburg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Trogerstr. 18, 81675, München, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Köln, Germany
| |
Collapse
|
32
|
Insulinoma-associated Protein 1 (INSM1) Expression in Small Cell Neuroendocrine Carcinoma of the Urinary Tract. Appl Immunohistochem Mol Morphol 2021; 28:687-693. [PMID: 31876605 DOI: 10.1097/pai.0000000000000824] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Clinical guidelines state that neoadjuvant chemotherapy should be administered before surgery in muscle invasive urinary bladder small cell neuroendocrine carcinoma. Recently described marker insulinoma-associated protein 1 (INSM1) has been reported to be sensitive and specific for neuroendocrine differentiation, however, its efficacy in urinary tract small cell carcinoma is not well established. This study examines immunohistochemical expression of INSM1 on whole tissue sections of urinary tract small cell neuroendocrine carcinoma and compares INSM1 expression with established neuroendocrine markers. Immunohistochemical stains for CD56, INSM1, synaptophysin, and chromogranin were performed on 32 cases of small cell neuroendocrine carcinoma of the bladder. Staining was scored for intensity (0: no staining; 1: weak; 2: moderate; 3: strong) and proportion of cells stained (0: 0%; 1: >0% to ≤25%; 2: >25% to ≤50%; 3: >50% to ≤75%; 4: >75% to 100%). INSM1 was positive (intensity 1 to 3 or proportion 1 to 4) in 87% (28/32) of cases (20 with intensity 2 to 3, 17 with proportion 3 to 4). CD56, synaptophysin, and chromogranin were positive in 75% (24/32), 60% (19/32), and 44% (14/32) of cases, respectively. INSM1 was negative (n=4) or only showed weak intensity staining (n=7) in 34% (11/32) of cases. INSM1 is a sensitive marker of small cell neuroendocrine differentiation of the urinary tract. However, this study suggests that optimal utilization of INSM1 would be inclusion in a limited panel of stains rather than as a stand-alone screening marker given that it is negative or only shows weak intensity staining in a significant proportion of cases.
Collapse
|
33
|
Precision Medicine for the Treatment of Colorectal Cancer: the Evolution and Status of Molecular Profiling and Biomarkers. CURRENT COLORECTAL CANCER REPORTS 2021. [DOI: 10.1007/s11888-021-00466-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
34
|
Evaluating mismatch repair deficiency for solid tumor immunotherapy eligibility: immunohistochemistry versus microsatellite molecular testing. Hum Pathol 2021; 115:10-18. [PMID: 34052294 DOI: 10.1016/j.humpath.2021.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022]
Abstract
While many landmark solid tumor immunotherapy studies show clinical benefits for solid tumors with high microsatellite instability (MSI-H) and mismatch repair deficiency (dMMR), the methodologies focus only on confirmatory polymerase chain reaction (PCR) testing for MSI-H. Because some tumors are either dMMR or MSI-H but not the other, clinicians must choose between two testing methods for a broad patient population. We investigated the level of correlation between MMR protein immunohistochemistry (IHC) and microsatellite PCR testing results in 62 cancer patients. Thirty-five of the 62 cases (56.5%) were MSI-H by PCR, whereas 35 (56.5%) were dMMR by IHC. MMR IHC results correlated well with MSI PCR in 32 co-positive cases (91.4%) and 24 co-negative cases (88.9%). Six discrepant cases (9.7%) were identified, among which three were MSI-H and MMR intact, and three were dMMR and microsatellite stable. The results of this study highlight the implications of dMMR/MSI testing strategies on precision oncology. Co-testing with both MMR IHC and MSI PCR may be an effective screening strategy for evaluating immunotherapy eligibility status for solid tumors.
Collapse
|
35
|
Rüschoff J, Baretton G, Bläker H, Dietmaier W, Dietel M, Hartmann A, Horn LC, Jöhrens K, Kirchner T, Knüchel R, Mayr D, Merkelbach-Bruse S, Schildhaus HU, Schirmacher P, Tiemann M, Tiemann K, Weichert W, Büttner R. [MSI testing : What is new? What should be considered? German version]. DER PATHOLOGE 2021; 42:414-423. [PMID: 34043067 DOI: 10.1007/s00292-021-00944-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/21/2022]
Abstract
Based on new trial data regarding immune checkpoint inhibitors (ICIs), the detection of high-grade microsatellite instability (MSI-H) or underlying deficient mismatch repair protein (dMMR) is now becoming increasingly important for predicting treatment response. For the first time, a PD‑1 ICI (pembrolizumab) has been approved by the European Medicines Agency (EMA) for first-line treatment of advanced (stage IV) dMMR/MSI‑H colorectal cancer (CRC). Further indications, such as dMMR/MSI‑H endometrial carcinoma (EC), have already succeeded (Dostarlimab, 2nd line treatment) and others are expected to follow before the end of 2021. The question of optimal testing in routine diagnostics should therefore be re-evaluated. Based on a consideration of the strengths and weaknesses of the widely available methods (immunohistochemistry and PCR), a test algorithm is proposed that allows quality assured, reliable, and cost-effective dMMR/MSI‑H testing. For CRC and EC, testing is therefore already possible at the primary diagnosis stage, in line with international recommendations (NICE, NCCN). The clinician is therefore enabled from the outset to consider not only the predictive but also the prognostic and predispositional implications of such a test when counseling patients and formulating treatment recommendations. As a basis for quality assurance, participation in interlaboratory comparisons and continuous documentation of results (e.g., QuIP Monitor) are strongly recommended.
Collapse
Affiliation(s)
- Josef Rüschoff
- Institut für Pathologie Nordhessen, TARGOS Molecular Pathology GmbH, Germaniastr. 7, 34119, Kassel, Deutschland.
| | - Gustavo Baretton
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - Hendrik Bläker
- Institut für Pathologie, Universitätsklinikum Leipzig, Liebigstr. 26, Gebäude G, 04103, Leipzig, Deutschland
| | - Wolfgang Dietmaier
- Institut für Pathologie/Zentrum für molekularpathologische Diagnostik, Universität Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Deutschland
| | - Manfred Dietel
- Institut für Pathologie, Campus Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - Arndt Hartmann
- Pathologisches Institut, Universität Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Deutschland
| | - Lars-Christian Horn
- Institut für Pathologie, Universitätsklinikum Leipzig, Liebigstr. 26, Gebäude G, 04103, Leipzig, Deutschland
| | - Korinna Jöhrens
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - Thomas Kirchner
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
| | - Ruth Knüchel
- Institut für Pathologie, Universitätsklinikum RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland
| | - Doris Mayr
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
| | - Sabine Merkelbach-Bruse
- Institut für Pathologie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - Hans-Ulrich Schildhaus
- Institut für Pathologie, Universitätsklinikum Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Peter Schirmacher
- Pathologisches Institut, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Deutschland
| | - Markus Tiemann
- Institut für Hämatopathologie Hamburg, Fangdieckstr. 75a, 22547, Hamburg, Deutschland
| | - Katharina Tiemann
- Institut für Hämatopathologie Hamburg, Fangdieckstr. 75a, 22547, Hamburg, Deutschland
| | - Wilko Weichert
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
| | - Reinhard Büttner
- Institut für Pathologie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| |
Collapse
|
36
|
Guyot D'Asnières De Salins A, Tachon G, Cohen R, Karayan-Tapon L, Junca A, Frouin E, Godet J, Evrard C, Randrian V, Duval A, Svrcek M, Lascols O, Vignot S, Coulet F, André T, Fléjou JF, Cervera P, Tougeron D. Discordance between immunochemistry of mismatch repair proteins and molecular testing of microsatellite instability in colorectal cancer. ESMO Open 2021; 6:100120. [PMID: 33930657 PMCID: PMC8102173 DOI: 10.1016/j.esmoop.2021.100120] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Background DNA mismatch repair system deficiency (dMMR) is found in 15% of colorectal cancers (CRCs). Two methods are used to determine dMMR, immunohistochemistry (IHC) of MMR proteins and molecular testing of microsatellite instability (MSI). Only studies with a low number of patients have reported rates of discordance between these two methods, ranging from 1% to 10%. Materials and methods Overall, 3228 consecutive patients with CRCs from two centers were included. Molecular testing was carried out using the Pentaplex panel and IHC evaluated four (MLH1, MSH2, MSH6, and PMS2; cohort 1; n = 1085) or two MMR proteins (MLH1 and MSH2; cohort 2; n = 2143). The primary endpoint was the rate of discordance between MSI and MMR IHC tests. Results Fifty-one discordant cases (1.6%) were initially observed. Twenty-nine out of 51 discordant cases were related to IHC misclassifications. In cohort 1, after re-reading IHC and/or carrying out new IHC, 16 discordant cases were reclassified as nondiscordant. In cohort 2, after the addition of MSH6/PMS2 IHC and re-examination, 13 were reclassified as nondiscordant. In addition, 10 misclassifications of molecular tests were identified. Finally, only 12 discordant cases (0.4%) remained: 5 were proficient MMR/MSI and 7 were dMMR/microsatellite stable. Conclusions Our study confirmed the high degree of concordance between MSI and MMR IHC tests. Discordant cases must be reviewed, and if needed, tests must be repeated and analyzed by an expert team. Concordance between MMR IHC (four proteins) and MSI molecular testing (Pentaplex) is superior to 98% in CRC cases Most discordant cases are related to misinterpretation of the tests, which may lead to clinical management errors. Discordant cases must be reviewed and tests must be repeated because most cases will be reclassified as nondiscordant.
Collapse
Affiliation(s)
| | - G Tachon
- Faculty of Medicine, University of Poitiers, Poitiers, France; INSERM 1084, Experimental and Clinical Neurosciences Laboratory, University of Poitiers, Poitiers, France; Cancer Biology Department, Poitiers University Hospital, Poitiers, France
| | - R Cohen
- Sorbonne University, Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - L Karayan-Tapon
- Faculty of Medicine, University of Poitiers, Poitiers, France; INSERM 1084, Experimental and Clinical Neurosciences Laboratory, University of Poitiers, Poitiers, France; Cancer Biology Department, Poitiers University Hospital, Poitiers, France
| | - A Junca
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - E Frouin
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - J Godet
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - C Evrard
- Medical Oncology Department, Poitiers University Hospital, Poitiers, France
| | - V Randrian
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France; Faculty of Medicine, University of Poitiers, Poitiers, France
| | - A Duval
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - M Svrcek
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne University, Department of Pathology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - O Lascols
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - S Vignot
- Sorbonne University, Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - F Coulet
- Department of Genetics, Pitié Salpétrière Hospital, AP-HP and Sorbonne University, Paris, France
| | - T André
- Sorbonne University, Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, Paris, France; Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - J-F Fléjou
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne University, Department of Pathology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - P Cervera
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne University, Department of Pathology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - D Tougeron
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France; Faculty of Medicine, University of Poitiers, Poitiers, France; Medical Oncology Department, Poitiers University Hospital, Poitiers, France.
| |
Collapse
|
37
|
Leelatian N, Hong CS, Bindra RS. The Role of Mismatch Repair in Glioblastoma Multiforme Treatment Response and Resistance. Neurosurg Clin N Am 2021; 32:171-180. [PMID: 33781500 DOI: 10.1016/j.nec.2020.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mismatch repair (MMR) is a highly conserved DNA repair pathway that is critical for the maintenance of genomic integrity. This pathway targets base substitution and insertion-deletion mismatches, which primarily arise from replication errors that escape DNA polymerase proof-reading function. Here, the authors review key concepts in the molecular mechanisms of MMR in response to alkylation damage, approaches to detect MMR status in the clinic, and the clinical relevance of this pathway in glioblastoma multiforme treatment response and resistance.
Collapse
Affiliation(s)
- Nalin Leelatian
- Department of Pathology, Yale School of Medicine, 310 Cedar Street LH 108, New Haven, CT 06510, USA
| | - Christopher S Hong
- Department of Neurosurgery, Yale School of Medicine, 333 Cedar Street Tompkins 4, New Haven, CT 06510, USA
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, 333 Cedar Street Hunter 2, New Haven, CT 06510, USA.
| |
Collapse
|
38
|
Mismatch repair phenotype determines the implications of tumor grade and CDX2 expression in stage II-III colon cancer. Mod Pathol 2021; 34:161-170. [PMID: 32737450 DOI: 10.1038/s41379-020-0634-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/28/2022]
Abstract
Mismatch repair (MMR) deficiency is an indicator of good prognosis in localized colon cancer but also associated with lack of expression of caudal-type homeobox transcription factor 2 (CDX2) and high tumor grade; markers that in isolation indicate a poor prognosis. Our study aims to identify clinically relevant prognostic subgroups by combining information about tumor grade, MMR phenotype, and CDX2 expression. Immunohistochemistry for MMR proteins and CDX2 was performed in 544 patients with colon cancer stage II-III, including a cohort from a randomized trial. In patients with proficient MMR (pMMR) and CDX2 negativity, hazard ratio (HR) for cancer death was 2.93 (95% CI 1.23-6.99, p = 0.015). Cancer-specific survival for pMMR/CDX2-negative cases was 35.8 months (95% CI 23.4-48.3) versus 52.1-53.5 months (95% CI 45.6-58.6, p = 0.001) for the remaining cases (CDX2-positive tumors or deficient MMR (dMMR)/CDX2-negative tumors). In our randomized cohort, high tumor grade was predictive of response to adjuvant fluorouracil-levamisole in pMMR patients, with a significant interaction between tumor grade and treatment (p = 0.036). For pMMR patients, high tumor grade was a significant marker of poor prognosis in the surgery-only group (HR 4.60 (95% CI 1.68-12.61), p = 0.003) but not in the group receiving chemotherapy (HR 0.66 (95% CI 0.15-3.00), p = 0.587). To conclude, patients with pMMR and CDX2 negativity have a very poor prognosis. Patients with pMMR and high-graded tumors have a poor prognosis but respond well to adjuvant chemotherapy. CDX2 expression and tumor grade did not impact prognosis in patients with dMMR.
Collapse
|
39
|
Mismatch Repair System Genomic Scars in Gastroesophageal Cancers: Biology and Clinical Testing. GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2040031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Alterations in the mismatch repair (MMR) system result in genomic instability, neoantigen production, and immune response in cancer. There is evidence that gastroesophageal tumors with MMR deficiency may be susceptible to immune-checkpoint inhibitors treatment, especially in those presenting at advanced-stage disease. Although a number of biomarkers have been developed in histology-agnostic settings to assess MMR status, there is evidence that a tumor-specific testing approach would improve the selection of patients for immunotherapy. However, no testing methods have been developed specifically for gastroesophageal cancers so far. Here, we discuss the state of the art, current advances, and future perspectives of MMR-related biomarkers’ biologic and clinical role in gastroesophageal cancers.
Collapse
|
40
|
Gu L, Liu Y, Jiang C, Sun L, Zhou H. Identification and clinical validation of metastasis-associated biomarkers based on large-scale samples in colon-adenocarcinoma. Pharmacol Res 2020; 160:105087. [PMID: 32683036 DOI: 10.1016/j.phrs.2020.105087] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/14/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
AIM Distant metastasis is the main cause of death in patients with colon-adenocarcinoma(COAD). Due to the lack of effective molecular markers and treatment, the prognosis of patients with metastatic colon cancer is still rather poor. METHODS Metastatic related signature (MRS) of stage I and stage IV in colon cancer were identified from different cohorts. Univariate cox regression is used to analyze the relationship between MRS and the overall survival. L1000FWD and DGIdb databases are used to identify molecular drugs. Expression and functional experimental validation of the hub MRS were carried out. RESULTS 16 MRS were identified, of which 14 MRS was significantly correlated with overall survival. Further functional enrichment analysis showed that MRS was significantly involved with important biological functions such as cell migration, and apoptosis. As important metastatic related genes, GSR, FAS and CYP1B1 have significant interaction with drug molecules. Further studies have confirmed that the expression of FAS and GSR is low, and inhibition of its expression can promote the metastasis of COAD. CYP1B1 expression is highly expressed, and inhibition of its expression can attenuate the malignant biological behavior of colon cancer. CONCLUSION Our research could increase the understanding of the mechanism of colon cancer metastasis and provide theoretical basis for the treatment of metastatic colon cancer.
Collapse
Affiliation(s)
- Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Ye Liu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Chunhui Jiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Longci Sun
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Hong Zhou
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China.
| |
Collapse
|
41
|
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: 17] [Impact Index Per Article: 4.3] [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
|
42
|
Mettman D, Haer E, Olyaee M, Rastogi A, Madan R, O'Neil M, Kelting S, Dennis K, Fan F. The utility of immunohistochemical testing for mismatch repair proteins in fine needle aspiration specimens of pancreatic adenocarcinoma. Ann Diagn Pathol 2020; 47:151552. [PMID: 32570025 DOI: 10.1016/j.anndiagpath.2020.151552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Microsatellite instability (MSI) testing is recommended for all colonic and endometrial carcinomas to screen for Lynch syndrome. The role of MSI testing in pancreatic adenocarcinoma has not been well-established. Screening can be done via immunohistochemical (IHC) staining for mismatch repair (MMR) proteins (MLH1, MSH2, MSH6, PMS2). We report our experience and the clinical utility of MMR IHC on pancreatic adenocarcinomas in fine-needle aspiration (FNA) specimens. MATERIALS AND METHODS We performed a retrospective review to identify all patients diagnosed with pancreatic adenocarcinoma by FNA at our institution between December 2017 and September 2019. For cases with sufficient tumor cells for testing, the MMR results and morphology were summarized, as well as corresponding clinical information, including age, clinical stage, treatment, and concurrent other cancers. RESULTS From December 2017 to September 2019, there were a total of 184 pancreatic FNAs with a diagnosis of adenocarcinoma. Of these 184 FNAs, 65 (35%) contained sufficient material in the cell block to perform IHC for MMR. The cell block material was collected in either RPMI or CytoLyt. Poor technical quality precluded interpretation of PMS2 in 4 cases and MSH6 in 2 cases. All other cases showed intact expression of all four proteins. CONCLUSIONS IHC for MMR proteins can be done on specimens collected in RPMI or CytoLyt, but RPMI appears to be more reliable. None of the pancreatic adenocarcinomas in this study showed loss of MMR protein expression. Routine testing of MMR loss may not be indicated in pancreatic adenocarcinomas in the general patient population.
Collapse
Affiliation(s)
- Daniel Mettman
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Erin Haer
- Pathology and Laboratory Medicine Service, Kansas City VA Medical Center, Kansas City, KS, United States of America
| | - Mojtaba Olyaee
- Department of Internal Medicine, Gastrointestinal Division, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Amit Rastogi
- Department of Internal Medicine, Gastrointestinal Division, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Maura O'Neil
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Sarah Kelting
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Katie Dennis
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Fang Fan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America.
| |
Collapse
|
43
|
Aird JJ, Steel MJ, Chow C, Ho J, Wolber R, Gilks CB, Hoang LN, Schaeffer DF. Should you repeat mismatch repair testing in cases of tumour recurrence? An evaluation of repeat mismatch repair testing by the use of immunohistochemistry in recurrent tumours of the gastrointestinal and gynaecological tracts. Histopathology 2020; 76:521-530. [DOI: 10.1111/his.14026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/30/2019] [Indexed: 01/23/2023]
Affiliation(s)
- John J Aird
- Division of Anatomic Pathology Vancouver General Hospital Vancouver BC Canada
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver BC Canada
| | - Michael J Steel
- Division of Anatomic Pathology Vancouver General Hospital Vancouver BC Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre Jack Bell Research Centre Vancouver BC Canada
| | - Julie Ho
- Department of Molecular Oncology British Columbia Cancer Agency Vancouver BC Canada
| | - Robert Wolber
- Department of Laboratory Medicine and Pathology Lions Gate Hospital North Vancouver BC Canada
| | - C Blake Gilks
- Division of Anatomic Pathology Vancouver General Hospital Vancouver BC Canada
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver BC Canada
| | - Lynn N Hoang
- Division of Anatomic Pathology Vancouver General Hospital Vancouver BC Canada
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver BC Canada
| | - David F Schaeffer
- Division of Anatomic Pathology Vancouver General Hospital Vancouver BC Canada
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver BC Canada
| |
Collapse
|
44
|
Devall M, Jennelle LT, Bryant J, Bien S, Peters U, Powell S, Casey G. Modeling the effect of prolonged ethanol exposure on global gene expression and chromatin accessibility in normal 3D colon organoids. PLoS One 2020; 15:e0227116. [PMID: 31951625 PMCID: PMC6968849 DOI: 10.1371/journal.pone.0227116] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
In this study we aimed to explore the potential biological effect of ethanol exposure on healthy colon epithelial cells using normal human colon 3D organoid “mini-gut” cultures. In numerous published studies ethanol use has been shown to be an environmental risk factor for colorectal cancer (CRC) development; however, the influence of ethanol exposure on normal colon epithelial cell biology remains poorly understood. We investigated the potential molecular effects of ethanol exposure in normal colon 3D organoids in a small pilot study (n = 3) using RNA-seq and ATAC-seq. We identify 1965 differentially expressed genes and 2217 differentially accessible regions of chromatin in response to ethanol treatment. Further, by cross-referencing our results with previously published analysis in colorectal cancer cell lines, we have not only validated a number of reported differentially expressed genes, but also identified several novel candidates for future investigation. In summary, our data highlights the potential importance for the use of normal colon 3D organoid models as a novel tool for the investigation of the relationship between the effects of environmental risk factors associated with colorectal cancer and the molecular mechanisms through which they confer this risk.
Collapse
Affiliation(s)
- Matthew Devall
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lucas T. Jennelle
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jennifer Bryant
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Stephanie Bien
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Steven Powell
- Digestive Health Center, Gastroenterology and Heaptology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Graham Casey
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
45
|
Mann SA, Cheng L. Microsatellite instability and mismatch repair deficiency in the era of precision immuno-oncology. Expert Rev Anticancer Ther 2019; 20:1-4. [PMID: 31842633 DOI: 10.1080/14737140.2020.1705789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Steven A Mann
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|