601
|
Lizardo DY, Kuang C, Hao S, Yu J, Huang Y, Zhang L. Immunotherapy efficacy on mismatch repair-deficient colorectal cancer: From bench to bedside. Biochim Biophys Acta Rev Cancer 2020; 1874:188447. [PMID: 33035640 PMCID: PMC7886024 DOI: 10.1016/j.bbcan.2020.188447] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancers (CRCs) with deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) often have sustained responses to immune checkpoint inhibitors (ICIs) including selective monoclonal antibodies against Program Death 1 (PD-1), Programmed Death Ligand 1(PD-L1), and cytotoxic T lymphocyte associated antigen 4 (CTLA-4). However, a substantial fraction of dMMR CRCs do not respond or ultimately develop resistance to immunotherapy. The majority (~85%) of CRCs are MMR proficient (pMMR) or microsatellite stable (MSS) and lack response to ICIs. Understanding the biology and mechanisms underlying dMMR-associated immunogenicity is urgently needed for improving the therapeutic efficacy of immunotherapy on CRC. Compared to pMMR/MSS CRCs, dMMR/MSI CRCs typically have increased tumor mutational burden (TMB), lower response rate to 5-fluorouracil-based chemotherapy, distinctive immunological features such as high tumor-infiltrating lymphocytes (TILs), and better prognosis. Here, we review the current understanding of the clinical relevance of dMMR/MSI in CRCs, the molecular basis and rationales for targeting dMMR CRC with immunotherapy, and clinical approaches using ICIs as single agents or in combination with other therapies for MSI-H CRCs. Furthermore, we address the potential strategies to sensitize pMMR/MSS CRC to immunotherapy by converting an immunologically "cold" microenvironment into a "hot" one.
Collapse
Affiliation(s)
- Darleny Y Lizardo
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoyuan Kuang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Suisui Hao
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yi Huang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
602
|
Kato M, Sasaki T, Inoue T. Current experimental human tissue-derived models for prostate cancer research. Int J Urol 2020; 28:150-162. [PMID: 33247498 DOI: 10.1111/iju.14441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022]
Abstract
Scientists engaged in prostate cancer research have been conducting experiments using two-dimensional cultures of prostate cancer cell lines for decades. However, these experiments fail to reproduce and reflect the clinical course of individual patients with prostate cancer, or the molecular and genetic characteristics of prostate cancer, the basic requirement for most of the preclinical studies on prostate cancer. The use of human prostate cancer tissues in experiments has enabled the collection and verification of clinically relevant data, including chemical reactions, changes in proteins, and specific gene expression. Tissue recombination models have been employed for studying prostate development, the initiation and progression of prostate cancer, and the tumor microenvironment. Notably, the epithelial-stromal interaction, which might play a critical role in prostate cancer pathogenesis, can be reproduced in this model. Patient-derived xenograft models have been developed as powerful avatars comprising patient-derived prostate cancer tissues implanted in immunocompromised mice and could serve as a precision medicine approach for each prostate cancer patient. Spheroid and organoid assays, representative of modern three-dimensional cultures, can replicate the conditions in human prostate tumors and the prostate organ itself as a miniature model. Although an intact immune system against the tumor is missing from the models aimed at investigating immuno-oncological reagents in various malignancies, all these experimental models can help researchers in developing new drugs and selecting appropriate treatment strategies for prostate cancer patients.
Collapse
Affiliation(s)
- Manabu Kato
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Takeshi Sasaki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Takahiro Inoue
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| |
Collapse
|
603
|
Diao Z, Han Y, Chen Y, Zhang R, Li J. The clinical utility of microsatellite instability in colorectal cancer. Crit Rev Oncol Hematol 2020; 157:103171. [PMID: 33290824 DOI: 10.1016/j.critrevonc.2020.103171] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/17/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Microsatellite instability (MSI) became the spotlight after the US FDA' s approval of MSI as an indication of immunotherapy for cancer patients. Immunohistochemical detection of loss of MMR proteins and PCR amplification of specific microsatellite repeats are widely used in clinical practice. Next-generation sequencing is a promising tool for identifying MSI patients. Circulating tumour DNA provides a convenient alternative when tumour tissue is unavailable. MSI detection is an effective tool to screen for Lynch syndrome. Early-stage CRC patients with MSI generally have a better prognosis and a reduced response to chemotherapy; instead, they are more likely to respond to immunotherapy. In this review, we aimed to assess the clinical utility of MSI as a biomarker in CRC. We will provide an overview of the available methods for evaluation of the analytical validity of MSI detection and elaborate the evidence on the clinical validity of MSI in the management of CRC patients.
Collapse
Affiliation(s)
- Zhenli Diao
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Peking University Fifth School of Clinical Medicine, Beijing Hospital, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, PR China
| | - Yanxi Han
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, PR China
| | - Yuqing Chen
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, PR China
| | - Rui Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, PR China.
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China.
| |
Collapse
|
604
|
Wu Y, Zhang L, He S, Guan B, He A, Yang K, Gong Y, Li X, Zhou L. Identification of immune-related LncRNA for predicting prognosis and immunotherapeutic response in bladder cancer. Aging (Albany NY) 2020; 12:23306-23325. [PMID: 33221763 PMCID: PMC7746369 DOI: 10.18632/aging.104115] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/09/2020] [Indexed: 12/24/2022]
Abstract
Long noncoding RNAs (lncRNAs) have multiple functions in the cancer immunity response and the tumor microenvironment. To investigate the immune-related lncRNA (IRlncRNA) signature for predicting prognosis and immunotherapeutic response in bladder cancer (BLCA), we extracted BLCA data from The Cancer Genome Atlas (TCGA) database. Finally, a total of 405 cases were enrolled and 8 prognostic IRlncRNAs (MIR181A2HG, AC114730.3, LINC00892, PTPRD-AS1, LINC01013, MRPL23-AS1,LINC01395, AC002454.1) were identified in the training set. Risk scores were calculated to divide patients into high-risk and low-risk groups, and the high-risk patients tended to have a poor overall survival (OS). Multivariate Cox regression analysis confirmed that the IRlncRNA signature could be an independent prognostic factor. The results were subsequently confirmed in the validating set. Additionally, this 8-IRlncRNA classifier was related to recurrence free survival (RFS) of BLCA. Functional characterization revealed this signature mediated immune-related phenotype. This signature was also associated with immune cell infiltration (i.e., macrophages M0, M2, Tregs, CD8 T cells, and neutrophils) and immune checkpoint inhibitors (ICIs) immunotherapy-related biomarkers [mismatch repair (MMR) genes, tumor mutation burden (TMB) and immune checkpoint genes]. The present study highlighted the value of the 8-IRlncRNA signature as a predictor of prognosis and immunotherapeutic response in BLCA.
Collapse
Affiliation(s)
- Yucai Wu
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Lei Zhang
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Bao Guan
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Anbang He
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Kunlin Yang
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| |
Collapse
|
605
|
Jung G, Benítez-Ribas D, Sánchez A, Balaguer F. Current Treatments of Metastatic Colorectal Cancer with Immune Checkpoint Inhibitors-2020 Update. J Clin Med 2020; 9:E3520. [PMID: 33142689 PMCID: PMC7694085 DOI: 10.3390/jcm9113520] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 12/29/2022] Open
Abstract
During the last 20 years, chemotherapy has improved survival rates of colorectal cancer (CRC). However, the majority of metastatic cases do not respond to or progress after first line conventional chemotherapy and contribute to the fatalities of patients with CRC. Insights into the immune contexture of the tumor microenvironment (TME) have enabled the development of new systemic treatments that boost the host immune system against the tumor-the immune checkpoint inhibitors (ICI). These promising drugs have already shown astonishing efficacies in other cancer types and have raised new hope for the treatment of metastatic CRC (mCRC). In this review, we will summarize the results of the clinical trials that led to their accelerated approval by the U.S. Food and Drug Administration (FDA) in 2017, as well as all relevant recent studies conducted since then-some of which are not published yet. We will focus on therapeutic efficacy, but also discuss the available data for drug safety and security, changes in quality of life indicators and predictive biomarkers for treatment response. The burgeoning evidence for a potential use of ICIs in other settings than mCRC will also be mentioned. For each trial, we have made a preliminary assessment of the quality of clinical trial design and of the "European Society of Medical Oncology (ESMO) magnitude of clinical benefit" (ESMO-MCBS) in order to provide the first evidence-based recommendation to the reader.
Collapse
Affiliation(s)
- Gerhard Jung
- Gastroenterology Department, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, 08036 Barcelona, Spain; (G.J.); (A.S.)
- Colorectal Cancer Research Laboratory, Institute for Research in Biomedicine, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Daniel Benítez-Ribas
- Immunology Department, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain;
| | - Ariadna Sánchez
- Gastroenterology Department, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, 08036 Barcelona, Spain; (G.J.); (A.S.)
| | - Francesc Balaguer
- Gastroenterology Department, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, 08036 Barcelona, Spain; (G.J.); (A.S.)
| |
Collapse
|
606
|
Luchini C, Paolino G, Mattiolo P, Piredda ML, Cavaliere A, Gaule M, Melisi D, Salvia R, Malleo G, Shin JI, Cargnin S, Terrazzino S, Lawlor RT, Milella M, Scarpa A. KRAS wild-type pancreatic ductal adenocarcinoma: molecular pathology and therapeutic opportunities. J Exp Clin Cancer Res 2020; 39:227. [PMID: 33115526 PMCID: PMC7594413 DOI: 10.1186/s13046-020-01732-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/13/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease, whose main molecular trait is the MAPK pathway activation due to KRAS mutation, which is present in 90% of cases.The genetic landscape of KRAS wild type PDAC can be divided into three categories. The first is represented by tumors with an activated MAPK pathway due to BRAF mutation that occur in up to 4% of cases. The second includes tumors with microsatellite instability (MSI) due to defective DNA mismatch repair (dMMR), which occurs in about 2% of cases, also featuring a high tumor mutational burden. The third category is represented by tumors with kinase fusion genes, which marks about 4% of cases. While therapeutic molecular targeting of KRAS is an unresolved challenge, KRAS-wild type PDACs have potential options for tailored treatments, including BRAF antagonists and MAPK inhibitors for the first group, immunotherapy with anti-PD-1/PD-L1 agents for the MSI/dMMR group, and kinase inhibitors for the third group.This calls for a complementation of the histological diagnosis of PDAC with a routine determination of KRAS followed by a comprehensive molecular profiling of KRAS-negative cases.
Collapse
Affiliation(s)
- Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134, Verona, Italy
| | - Gaetano Paolino
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134, Verona, Italy
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134, Verona, Italy
| | - Maria L Piredda
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Alessandro Cavaliere
- Section of Oncology, Department of Medicine, University and Hospital Trust of Verona, Piazzale L.A. Scuro 10, 37134, Verona, VR, Italy
| | - Marina Gaule
- Section of Oncology, Department of Medicine, University and Hospital Trust of Verona, Piazzale L.A. Scuro 10, 37134, Verona, VR, Italy
| | - Davide Melisi
- Section of Oncology, Department of Medicine, University and Hospital Trust of Verona, Piazzale L.A. Scuro 10, 37134, Verona, VR, Italy
| | - Roberto Salvia
- Department of Surgery, University of Verona, 37134, Verona, Italy
| | - Giuseppe Malleo
- Department of Surgery, University of Verona, 37134, Verona, Italy
| | - Jae Il Shin
- Yonsei University College of Medicine, 03722, Seoul, Republic of Korea
| | - Sarah Cargnin
- Department of Pharmaceutical Sciences and Interdepartmental Research Center of Pharmacogenetics and Pharmacogenomics (CRIFF), University of Piemonte Orientale, 28100, Novara, Italy
| | - Salvatore Terrazzino
- Department of Pharmaceutical Sciences and Interdepartmental Research Center of Pharmacogenetics and Pharmacogenomics (CRIFF), University of Piemonte Orientale, 28100, Novara, Italy
| | - Rita T Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Michele Milella
- Section of Oncology, Department of Medicine, University and Hospital Trust of Verona, Piazzale L.A. Scuro 10, 37134, Verona, VR, Italy.
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134, Verona, Italy
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134, Verona, Italy
| |
Collapse
|
607
|
Cilona M, Locatello LG, Novelli L, Gallo O. The Mismatch Repair System (MMR) in Head and Neck Carcinogenesis and Its Role in Modulating the Response to Immunotherapy: A Critical Review. Cancers (Basel) 2020; 12:cancers12103006. [PMID: 33081243 PMCID: PMC7602801 DOI: 10.3390/cancers12103006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The dysfunction of the mismatch repair system, an important mechanism for the detection and correction of DNA replication mistakes, may often lead to instability in the length of specific genetic sequences, known as microsatellites, and to the accumulation of mutations. Microsatellite instability is a well-known risk factor for the development of colorectal cancers and other types of tumors but is also considered a positive predictor of the immunotherapy response. Malignancies harboring such a specific genomic instability are very immunogenic because of the great number of aberrant antigens they produce. Therapies based on the blockade of specific immune checkpoints have shown to induce an effective immune response against microsatellite-unstable cancer. Many studies proved that microsatellite instability has a decisive role in the carcinogenesis and the malignant progression of head and neck cancer and, in the near future, it may become a useful tool in tailoring immunotherapy also in this field of precision oncology. Abstract The mismatch repair (MMR) system has a major role in the detection and correction of DNA replication errors, resulting from DNA polymerase slippage or nucleotides misincorporation. Specific inherited/acquired alterations or epigenetic inactivation of MMR genes are associated with microsatellite instability (MSI): the loss of crucial function in repairing DNA alterations can promote carcinogenesis by favoring the accumulation of thousands of mutations in a broad spectrum of different anatomic sites such as colon, stomach, prostate, esophagus, endometrium, lung and head and neck. Recent extensive data suggest that tumor mutational burden strongly correlates with a clinical response to immunotherapy using checkpoint inhibitors and this response is influenced by MMR deficiency in a wide range of human solid cancers. In this context, few data about this crucial point are available for head and neck cancer (HNC). In this review, we discuss the role of MMR alterations and the resulting MSI in HNC pathogenesis. Furthermore, by summarizing the clinical available data on how they influence the progression of precancerous lesions and the risk of recurrence or second primary tumors, we want to define the current role of MSI in the management of HNC. Finally, we analyze the complex interaction between cancer cells and the immune system addressing the data now available about a potential correlation between microsatellite instability and immunotherapy response in HNC.
Collapse
Affiliation(s)
- Maria Cilona
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3-50134 Florence, Italy; (M.C.); (L.G.L.)
| | - Luca Giovanni Locatello
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3-50134 Florence, Italy; (M.C.); (L.G.L.)
| | - Luca Novelli
- Department of Pathology, Careggi University Hospital, Largo Brambilla, 3-50134 Florence, Italy;
| | - Oreste Gallo
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3-50134 Florence, Italy; (M.C.); (L.G.L.)
- Correspondence: ; Tel.: +39-0557947989
| |
Collapse
|
608
|
Wang HC, Yeh TJ, Chan LP, Hsu CM, Cho SF. Exploration of Feasible Immune Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma Treatment in Real World Clinical Practice. Int J Mol Sci 2020; 21:E7621. [PMID: 33076306 PMCID: PMC7589088 DOI: 10.3390/ijms21207621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Recurrent locally advanced or metastatic head and neck squamous cell carcinoma (HNSCC) is associated with dismal prognosis because of its highly invasive behavior and resistance to conventional intensive chemotherapy. The combination of targeted therapy and conventional chemotherapy has significantly improved clinical outcomes. In recent years, the development of immunotherapies, such as immune checkpoint inhibitors (ICIs), has further increased treatment responses and prolonged survival. However, the limited response rate, risk of immunotherapy-related adverse effects and high cost of immunotherapy make the identification of predictive markers to optimize treatment efficacy a critical issue. Biomarkers are biological molecules that have been widely utilized to predict treatment response to certain treatments and clinical outcomes or to detect disease. An ideal biomarker should exhibit good predictive ability, which can guide healthcare professionals to achieve optimal treatment goals and bring clinical benefit to patients. In this review, we summarized the results of recent and important studies focused on HNSCC ICI immunotherapy and discussed potential biomarkers including their strengths and limitations, aiming to gain more insight into HNSCC immunotherapy in real world clinical practice.
Collapse
Affiliation(s)
- Hui-Ching Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (T.-J.Y.); (L.-P.C.)
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tsung-Jang Yeh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (T.-J.Y.); (L.-P.C.)
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Leong-Perng Chan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (T.-J.Y.); (L.-P.C.)
- Department of Otolaryngology-Head and Neck Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chin-Mu Hsu
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Feng Cho
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
609
|
Evaluation of 3 molecular-based assays for microsatellite instability detection in formalin-fixed tissues of patients with endometrial and colorectal cancers. Sci Rep 2020; 10:16386. [PMID: 33009475 PMCID: PMC7532161 DOI: 10.1038/s41598-020-73421-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022] Open
Abstract
Microsatellite instability (MSI) status is routinely assessed in patients with colorectal and endometrial cancers as it contributes to Lynch syndrome initial screening, tumour prognosis and selecting patients for immunotherapy. Currently, standard reference methods recommended for MSI/dMMR (deficient MisMatch Repair) testing consist of immunohistochemistry and pentaplex PCR-based assays, however, novel molecular-based techniques are emerging. Here, we aimed to evaluate the performance of a custom capture-based NGS method and the Bio-Rad ddPCR and Idylla approaches for the determination of MSI status for theranostic purposes in 30 formalin-fixed paraffin embedded (FFPE) tissue samples from patients with endometrial (n = 15) and colorectal (n = 15) cancers. All samples were previously characterised using IHC and Promega MSI Analysis System and these assays set as golden standard. Overall agreement, sensitivity and specificity of our custom-built NGS panel were 93.30%, 93.75% and 92.86% respectively. Overall agreement, sensitivity and specificity were 100% with the Idylla MSI system. The Bio-Rad ddPCR MSI assay showed a 100% concordance, sensitivity and specificity. The custom capture-based NGS, Bio-Rad ddPCR and Idylla approaches represent viable and complementary options to IHC and Promega MSI Analysis System for the detection of MSI. Bio-Rad ddPCR and Idylla MSI assays accounts for easy and fast screening assays while the NGS approach offers the advantages to simultaneously detect MSI and clinically relevant genomic alterations.
Collapse
|
610
|
Echle A, Grabsch HI, Quirke P, van den Brandt PA, West NP, Hutchins GGA, Heij LR, Tan X, Richman SD, Krause J, Alwers E, Jenniskens J, Offermans K, Gray R, Brenner H, Chang-Claude J, Trautwein C, Pearson AT, Boor P, Luedde T, Gaisa NT, Hoffmeister M, Kather JN. Clinical-Grade Detection of Microsatellite Instability in Colorectal Tumors by Deep Learning. Gastroenterology 2020; 159:1406-1416.e11. [PMID: 32562722 PMCID: PMC7578071 DOI: 10.1053/j.gastro.2020.06.021] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/22/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Microsatellite instability (MSI) and mismatch-repair deficiency (dMMR) in colorectal tumors are used to select treatment for patients. Deep learning can detect MSI and dMMR in tumor samples on routine histology slides faster and less expensively than molecular assays. However, clinical application of this technology requires high performance and multisite validation, which have not yet been performed. METHODS We collected H&E-stained slides and findings from molecular analyses for MSI and dMMR from 8836 colorectal tumors (of all stages) included in the MSIDETECT consortium study, from Germany, the Netherlands, the United Kingdom, and the United States. Specimens with dMMR were identified by immunohistochemistry analyses of tissue microarrays for loss of MLH1, MSH2, MSH6, and/or PMS2. Specimens with MSI were identified by genetic analyses. We trained a deep-learning detector to identify samples with MSI from these slides; performance was assessed by cross-validation (N = 6406 specimens) and validated in an external cohort (n = 771 specimens). Prespecified endpoints were area under the receiver operating characteristic (AUROC) curve and area under the precision-recall curve (AUPRC). RESULTS The deep-learning detector identified specimens with dMMR or MSI with a mean AUROC curve of 0.92 (lower bound, 0.91; upper bound, 0.93) and an AUPRC of 0.63 (range, 0.59-0.65), or 67% specificity and 95% sensitivity, in the cross-validation development cohort. In the validation cohort, the classifier identified samples with dMMR with an AUROC of 0.95 (range, 0.92-0.96) without image preprocessing and an AUROC of 0.96 (range, 0.93-0.98) after color normalization. CONCLUSIONS We developed a deep-learning system that detects colorectal cancer specimens with dMMR or MSI using H&E-stained slides; it detected tissues with dMMR with an AUROC of 0.96 in a large, international validation cohort. This system might be used for high-throughput, low-cost evaluation of colorectal tissue specimens.
Collapse
Affiliation(s)
- Amelie Echle
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Heike Irmgard Grabsch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands; Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Philip Quirke
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Piet A van den Brandt
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Nicholas P West
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Gordon G A Hutchins
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Lara R Heij
- Visceral and Transplant Surgery, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Xiuxiang Tan
- Visceral and Transplant Surgery, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Susan D Richman
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Jeremias Krause
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Elizabeth Alwers
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Josien Jenniskens
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Kelly Offermans
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Richard Gray
- Clinical Trial Service Unit, University of Oxford, Oxford, United Kingdom
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany; Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Tom Luedde
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany; Division of Gastroenterology, Hepatology, and Hepatobiliary Oncology, Aachen, Germany
| | | | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany; Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany; Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
611
|
Cao R, Yang F, Ma SC, Liu L, Zhao Y, Li Y, Wu DH, Wang T, Lu WJ, Cai WJ, Zhu HB, Guo XJ, Lu YW, Kuang JJ, Huan WJ, Tang WM, Huang K, Huang J, Yao J, Dong ZY. Development and interpretation of a pathomics-based model for the prediction of microsatellite instability in Colorectal Cancer. Theranostics 2020; 10:11080-11091. [PMID: 33042271 PMCID: PMC7532670 DOI: 10.7150/thno.49864] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022] Open
Abstract
Microsatellite instability (MSI) has been approved as a pan-cancer biomarker for immune checkpoint blockade (ICB) therapy. However, current MSI identification methods are not available for all patients. We proposed an ensemble multiple instance deep learning model to predict microsatellite status based on histopathology images, and interpreted the pathomics-based model with multi-omics correlation. Methods: Two cohorts of patients were collected, including 429 from The Cancer Genome Atlas (TCGA-COAD) and 785 from an Asian colorectal cancer (CRC) cohort (Asian-CRC). We established the pathomics model, named Ensembled Patch Likelihood Aggregation (EPLA), based on two consecutive stages: patch-level prediction and WSI-level prediction. The initial model was developed and validated in TCGA-COAD, and then generalized in Asian-CRC through transfer learning. The pathological signatures extracted from the model were analyzed with genomic and transcriptomic profiles for model interpretation. Results: The EPLA model achieved an area-under-the-curve (AUC) of 0.8848 (95% CI: 0.8185-0.9512) in the TCGA-COAD test set and an AUC of 0.8504 (95% CI: 0.7591-0.9323) in the external validation set Asian-CRC after transfer learning. Notably, EPLA captured the relationship between pathological phenotype of poor differentiation and MSI (P < 0.001). Furthermore, the five pathological imaging signatures identified from the EPLA model were associated with mutation burden and DNA damage repair related genotype in the genomic profiles, and antitumor immunity activated pathway in the transcriptomic profiles. Conclusions: Our pathomics-based deep learning model can effectively predict MSI from histopathology images and is transferable to a new patient cohort. The interpretability of our model by association with pathological, genomic and transcriptomic phenotypes lays the foundation for prospective clinical trials of the application of this artificial intelligence (AI) platform in ICB therapy.
Collapse
Affiliation(s)
- Rui Cao
- Information Management and Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Yang
- AI Lab, Tencent, Shenzhen, China
| | - Si-Cong Ma
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Liu
- Information Management and Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Zhao
- AI Lab, Tencent, Shenzhen, China
- Department of Computer Science, Technical University of Munich, Munich, Germany
| | - Yan Li
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - De-Hua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tongxin Wang
- Indiana University Bloomington, Bloomington, USA
| | | | | | - Hong-Bo Zhu
- Information Management and Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue-Jun Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu-Wen Lu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun-Jie Kuang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | - Kun Huang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Regenstrief Institute, Indianapolis, IN, USA
| | | | | | - Zhong-Yi Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
612
|
Malapelle U, Parente P, Pepe F, De Luca C, Cerino P, Covelli C, Balestrieri M, Russo G, Bonfitto A, Pisapia P, Fiordelisi F, D’Armiento M, Bruzzese D, Loupakis F, Pietrantonio F, Triassi M, Fassan M, Troncone G, Graziano P. Impact of Pre-Analytical Factors on MSI Test Accuracy in Mucinous Colorectal Adenocarcinoma: A Multi-Assay Concordance Study. Cells 2020; 9:2019. [PMID: 32887373 PMCID: PMC7565496 DOI: 10.3390/cells9092019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
Immunohistochemistry (IHC) and polymerase chain reaction (PCR) and fragment separation by capillary electrophoresis represent the current clinical laboratory standard for the evaluation of microsatellite instability (MSI) status. The importance of reporting MSI status in colorectal cancer is based on its potential for guiding treatment and as a prognostic indicator. It is also used to identify patients for Lynch syndrome testing. Our aim was to evaluate pre-analytical factors, such as age of formalin-fixed and paraffin-embedded (FFPE) block, neoplastic cell percentage, mucinous component, and DNA integrity, that may influence the accuracy of MSI testing and assess the concordance between three different MSI evaluation approaches. We selected the mucinous colorectal cancer (CRC) histotype for this study as it may possibly represent an intrinsic diagnostic issue due to its low tumor cellularity. Seventy-five cases of mucinous CRC and corresponding normal colon tissue samples were retrospectively selected. MMR proteins were evaluated by IHC. After DNA quality and quantity evaluation, the Idylla™ and TapeStation 4200 platforms were adopted for the evaluation of MSI status. Seventy-three (97.3%) cases were successfully analyzed by the three methodologies. Overall, the Idylla™ platform showed a concordance rate with IHC of 98.0% for microsatellite stable (MSS)/proficient MMR (pMMR) cases and 81.8% for MSI/deficient MMR (dMMR) cases. The TapeStation 4200 system showed a concordance rate with IHC of 96.0% for MSS/pMMR cases and 45.4% for MSI/dMMR cases. The concordance rates of the TapeStation 4200 system with respect to the Idylla™ platform were 98.1% for MSS profile and 57.8% for MSI profile. Discordant cases were analyzed using the Titano MSI kit. Considering pre-analytical factors, no significant variation in concordance rate among IHC analyses and molecular systems was observed by considering the presence of an acellular mucus cut-off >50% of the tumor area, FFPE year preparation, and DNA concentration. Conversely, the Idylla™ platform showed a significant variation in concordance rate with the IHC approach by considering a neoplastic cell percentage >50% (p-value = 0.002), and the TapeStation 4200 system showed a significant variation in concordance rate with the IHC approach by considering a DNA integrity number (DIN) ≥4 as cut-off (p-value = 0.009). Our data pinpoint a central role of the pre-analytical phase in the diagnostic outcome of MSI testing in CRC.
Collapse
MESH Headings
- Adenocarcinoma, Mucinous/diagnosis
- Adenocarcinoma, Mucinous/genetics
- Adenocarcinoma, Mucinous/pathology
- Aged
- Case-Control Studies
- Colorectal Neoplasms/diagnosis
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis
- Colorectal Neoplasms, Hereditary Nonpolyposis/genetics
- Colorectal Neoplasms, Hereditary Nonpolyposis/pathology
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- Diagnosis, Differential
- Electrophoresis, Capillary/standards
- Female
- Humans
- Immunohistochemistry/standards
- Male
- Microsatellite Instability
- Middle Aged
- Polymerase Chain Reaction/standards
- Prognosis
- Retrospective Studies
- Tissue Embedding/methods
- Tissue Embedding/standards
- Tissue Fixation/methods
- Tissue Fixation/standards
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (A.B.); (F.F.); (P.G.)
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Caterina De Luca
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Pellegrino Cerino
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Claudia Covelli
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (A.B.); (F.F.); (P.G.)
| | - Mariangela Balestrieri
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.B.); (M.F.)
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Antonio Bonfitto
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (A.B.); (F.F.); (P.G.)
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Fabiola Fiordelisi
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (A.B.); (F.F.); (P.G.)
| | - Maria D’Armiento
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Dario Bruzzese
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Fotios Loupakis
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Istituto Oncologico Veneto (IRCSS), 35128 Padua, Italy;
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, 20133 Milano, Italy;
- Oncology and Hemato-Oncology Department, University of Milan, 20133 Milan, Italy
| | - Maria Triassi
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.B.); (M.F.)
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.C.); (G.R.); (P.P.); (M.D.); (D.B.); (M.T.)
| | - Paolo Graziano
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (A.B.); (F.F.); (P.G.)
| |
Collapse
|
613
|
Luchini C, Grillo F, Fassan M, Vanoli A, Capelli P, Paolino G, Ingravallo G, Renzulli G, Doglioni C, D’Amuri A, Mattiolo P, Pecori S, Parente P, Florena AM, Zamboni G, Scarpa A. Malignant epithelial/exocrine tumors of the pancreas. Pathologica 2020; 112:210-226. [PMID: 33179623 PMCID: PMC7931574 DOI: 10.32074/1591-951x-167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic malignant exocrine tumors represent the most important cause of cancer-related death for pancreatic neoplasms. The most common tumor type in this category is represented by pancreatic ductal adenocarcinoma (PDAC), an ill defined, stroma-rich, scirrhous neoplasm with glandular differentiation. Here we present the relevant characteristics of the most important PDAC variants, namely adenosquamous carcinoma, colloid carcinoma, undifferentiated carcinoma, undifferentiated carcinoma with osteoclast-like giant cells, signet ring carcinoma, medullary carcinoma and hepatoid carcinoma. The other categories of malignant exocrine tumors, characterized by fleshy, stroma-poor, circumscribed neoplasms, include acinar cell carcinoma (pure and mixed), pancreatoblastoma, and solid pseudopapillary neoplasms. The most important macroscopic, histologic, immunohistochemical and molecular hallmarks of all these tumors, highlighting their key diagnostic/pathological features are presented. Lastly, standardized indications regarding gross sampling and how to compile a formal pathology report for pancreatic malignant exocrine tumors will be provided.
Collapse
Affiliation(s)
- Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Federica Grillo
- Anatomic Pathology, San Martino IRCCS Hospital, Genova, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital, Italy
| | - Paola Capelli
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Gaetano Paolino
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Giuseppe Ingravallo
- Department of Emergency and Organ Transplantation, Section of Pathological Anatomy, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppina Renzulli
- Department of Emergency and Organ Transplantation, Section of Pathological Anatomy, University of Bari Aldo Moro, Bari, Italy
| | - Claudio Doglioni
- Vita e Salute University, Milan, Italy
- Pathology Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Sara Pecori
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Ada M. Florena
- Department of Sciences for Promotion of Health and Mother and Child Care, Anatomic Pathology, University of Palermo, Italy
| | - Giuseppe Zamboni
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- IRCSS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-NET Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
614
|
Fassan M, Scarpa A, Remo A, De Maglio G, Troncone G, Marchetti A, Doglioni C, Ingravallo G, Perrone G, Parente P, Luchini C, Mastracci L. Current prognostic and predictive biomarkers for gastrointestinal tumors in clinical practice. Pathologica 2020; 112:248-259. [PMID: 33179625 PMCID: PMC7931577 DOI: 10.32074/1591-951x-158] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
The pathologist emerged in the personalized medicine era as a central actor in the definition of the most adequate diagnostic and therapeutic algorithms. In the last decade, gastrointestinal oncology has seen a significantly increased clinical request for the integration of novel prognostic and predictive biomarkers in histopathological reports. This request couples with the significant contraction of invasive sampling of the disease, thus conferring to the pathologist the role of governor for both proper pathologic characterization and customized processing of the biospecimens. This overview will focus on the most commonly adopted immunohistochemical and molecular biomarkers in the routine clinical characterization of gastrointestinal neoplasms referring to the most recent published recommendations, guidelines and expert opinions.
Collapse
Affiliation(s)
- Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Italy
| | - Aldo Scarpa
- ARC-NET Research Centre, University of Verona, Italy
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Andrea Remo
- Pathology Unit, Service Department, ULSS9 “Scaligera”, Verona, Italy
| | | | - Giancarlo Troncone
- Department of Public Health, Federico II University Medical School Naples, Italy
| | - Antonio Marchetti
- Center of Predictive Molecular Medicine, Center for Excellence on Aging and Translational Medicine, University of Chieti-Pescara, Italy
| | - Claudio Doglioni
- Vita e Salute University, Milan, Italy
- Pathology Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Ingravallo
- Department of Emergency and Organ Transplantation, Section of Pathological Anatomy, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Perrone
- Department of Pathology, Campus Bio-Medico University, Rome, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Luca Mastracci
- Anatomic Pathology, San Martino IRCCS Hospital,, Genova, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
| |
Collapse
|
615
|
Lombardi G, Barresi V, Indraccolo S, Simbolo M, Fassan M, Mandruzzato S, Simonelli M, Caccese M, Pizzi M, Fassina A, Padovan M, Masetto E, Gardiman MP, Bonavina MG, Caffo M, Persico P, Chioffi F, Denaro L, Dei Tos AP, Scarpa A, Zagonel V. Pembrolizumab Activity in Recurrent High-Grade Gliomas with Partial or Complete Loss of Mismatch Repair Protein Expression: A Monocentric, Observational and Prospective Pilot Study. Cancers (Basel) 2020; 12:2283. [PMID: 32823925 PMCID: PMC7464918 DOI: 10.3390/cancers12082283] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Pembrolizumab demonstrated promising results in hypermutated tumors of diverse origin. Immunohistochemical loss of mismatch repair (MMR) proteins has been suggested as a surrogate of hypermutation in high-grade gliomas (HGG). We evaluated the efficacy and safety of pembrolizumab in relapsing HGGs with immunohistochemical loss of at least 1 MMR protein. Molecular biomarkers of pembrolizumab activity were also analyzed. METHODS Consecutive patients with recurrent HGG and partial or complete loss of MMR protein expression were prospectively enrolled; they received pembrolizumab 200 mg once every 3 weeks until disease progression. The primary endpoint was disease control rate (DCR). Post hoc exploratory analyses included next-generation sequencing to assess tumor mutational burden (TMB), and immunostaining for CD8+ T-cells and CD68+ macrophages. RESULTS Among 310 HGG patients screened, 13 cases with MMR loss were enrolled: eight glioblastoma, four anaplastic astrocytoma, and one anaplastic oligodendroglioma. Median age was 43 years. DCR was 31%: four patients had stable disease and no patient had complete or partial response. TMB ranged between 6.8 and 23.4 mutations/megabase. Neither TMB nor gene mutations, nor CD8+ T-cell and CD68+ macrophage content, were associated with pembrolizumab activity. CONCLUSIONS pembrolizumab showed no apparent benefit in these patients. No molecular biomarker was found to be associated with pembrolizumab activity.
Collapse
Affiliation(s)
- Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of oncology-IRCCS, 35128 Padua, Italy; (M.C.); (M.P.); (V.Z.)
| | - Valeria Barresi
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (V.B.); (M.S.); (A.S.)
| | - Stefano Indraccolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology—IRCCS, 35128 Padua, Italy; (S.I.); (A.F.); (E.M.)
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (V.B.); (M.S.); (A.S.)
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, 35128 Padua, Italy; (M.F.); (M.P.); (M.P.G.); (A.P.D.T.)
| | - Susanna Mandruzzato
- Department of Surgery, Oncology and Gastroenterology, University of Padova and IOV-IRCCS, 35128 Padova, Italy;
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, 20089 Milan, Italy; (M.S.); (P.P.)
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of oncology-IRCCS, 35128 Padua, Italy; (M.C.); (M.P.); (V.Z.)
| | - Marco Pizzi
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, 35128 Padua, Italy; (M.F.); (M.P.); (M.P.G.); (A.P.D.T.)
| | - Arianna Fassina
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology—IRCCS, 35128 Padua, Italy; (S.I.); (A.F.); (E.M.)
| | - Marta Padovan
- Department of Oncology, Oncology 1, Veneto Institute of oncology-IRCCS, 35128 Padua, Italy; (M.C.); (M.P.); (V.Z.)
| | - Elena Masetto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology—IRCCS, 35128 Padua, Italy; (S.I.); (A.F.); (E.M.)
| | - Marina Paola Gardiman
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, 35128 Padua, Italy; (M.F.); (M.P.); (M.P.G.); (A.P.D.T.)
| | | | - Maria Caffo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy;
| | - Pasquale Persico
- Department of Biomedical Sciences, Humanitas University, 20089 Milan, Italy; (M.S.); (P.P.)
| | - Franco Chioffi
- Neurosurgery Unit, Azienda Ospedaliera di Padova, 35128 Padua, Italy;
| | - Luca Denaro
- Academic Neurosurgery, Department of Neurosciences, University of Padua, 35128 Padua, Italy;
| | - Angelo Paolo Dei Tos
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, 35128 Padua, Italy; (M.F.); (M.P.); (M.P.G.); (A.P.D.T.)
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (V.B.); (M.S.); (A.S.)
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of oncology-IRCCS, 35128 Padua, Italy; (M.C.); (M.P.); (V.Z.)
| |
Collapse
|
616
|
Kryklyva V, ter Linden E, Kroeze LI, de Voer RM, van der Kolk BM, Stommel MW, Hermans JJ, Luchini C, Wood LD, Hruban RH, Nagtegaal ID, Ligtenberg MJ, Brosens LA. Medullary Pancreatic Carcinoma Due to Somatic POLE Mutation: A Distinctive Pancreatic Carcinoma With Marked Long-Term Survival. Pancreas 2020; 49:999-1003. [PMID: 32658072 PMCID: PMC7368848 DOI: 10.1097/mpa.0000000000001588] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 05/06/2020] [Indexed: 12/12/2022]
Abstract
Medullary pancreatic carcinoma (MPC) is a rare histological variant of pancreatic ductal adenocarcinoma (PDAC). Because of its rarity, data on the molecular background of MPC are limited. Previous studies have shown that a subset of MPCs is microsatellite instable due to mismatch repair deficiency. Here, we present a unique case of a female patient in her 60s who is a long-term survivor after surgery for pancreatic cancer. The patient had a microsatellite stable MPC with a somatic mutation of the polymerase epsilon gene (POLE). Both microsatellite instable and POLE-mutated cancers are usually associated with high tumor mutational burden and antigen load, resulting in a prominent antitumor immune response and overall better survival. The current case illustrates that, in addition to mismatch repair deficiency, MPC can develop because of a somatic POLE mutation, resulting in a tumor with a high tumor mutational burden and leading to a better prognosis compared with conventional PDAC. This new finding may have important implications in the management of patients with MPC and calls for further studies on the role of POLE in PDAC.
Collapse
Affiliation(s)
- Valentyna Kryklyva
- From the Department of Pathology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen
| | - Esther ter Linden
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht
| | - Leonie I. Kroeze
- From the Department of Pathology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen
| | | | | | | | - John J. Hermans
- Radiology and Nuclear Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences/Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Claudio Luchini
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Laura D. Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Iris D. Nagtegaal
- From the Department of Pathology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen
| | - Marjolijn J.L. Ligtenberg
- From the Department of Pathology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen
- Departments of Human Genetics
| | - Lodewijk A.A. Brosens
- From the Department of Pathology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht
| |
Collapse
|
617
|
Cai L, Sun Y, Wang K, Guan W, Yue J, Li J, Wang R, Wang L. The Better Survival of MSI Subtype Is Associated With the Oxidative Stress Related Pathways in Gastric Cancer. Front Oncol 2020; 10:1269. [PMID: 32850385 PMCID: PMC7399340 DOI: 10.3389/fonc.2020.01269] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/18/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Gastric cancer (GC) is the third leading fatal cancer in the world and its incidence ranked second among all malignant tumors in China. The molecular classification of GC, proposed by the The Cancer Genome Atlas (TCGA), was added to the updated edition (2019) of WHO classification for digestive system tumor. Although MSI and EBV subtypes appeared as ever-increasingly significant roles in immune checkpoint inhibitor therapy, the underlying mechanisms are still unclear. Methods: We systematically summarized the relationship between EBV, d-MMR/MSI-H subtypes and clinicopathological parameters in 271 GC cases. Furthermore, GSE62254/ACRG and TCGA-STAD datasets, originated from Gene Expression Omnibus (GEO) and TCGA respectively, were analyzed to figure out the prognosis related molecular characteristics by bioinformatics methods. Results: Patients with MSI subtype had better prognosis than the MSS subtype (P = 0.013) and considered as an independent biomarker by the univariate analysis (P = 0.017) and multivariate analysis (P = 0.050). While there was no significant difference between EBV positive and negative tissues (P = 0.533). The positive prognostic value conferred by MSI in different cohorts was revalidated via the clinical analysis of GSE62254/ACRG and TCGA-STAD datasets regardless of race. Then key gene module that tightly associated with better status and longer OS time for MSI cases was obtained from weighted gene co-expression network analysis(WGCNA). NUBP2 and ENDOG were screened from the gene cluster and oxidative phosphorylation, reactive oxygen species(ROS) and glutathione metabolism were analyzed to be the differential pathways in their highly expressed groups. Conclusions: Our results manifested the significant prognostic value of MSI in Chinese GC cohort and comparisons with other populations. More opportunities to induce apoptosis of cancer cells, led by the unbalance between antioxidant system and ROS accumulation, lay foundations for unveiling the better prognosis in MSI phenotype through the bioinformatics analysis.
Collapse
Affiliation(s)
- Lei Cai
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yeqi Sun
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kezhou Wang
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenbin Guan
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juanqing Yue
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junlei Li
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruifen Wang
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lifeng Wang
- Department of Pathology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
618
|
Abstract
Mismatch repair deficiency (MMRD) is involved in the initiation of both hereditary and sporadic tumors. MMRD has been extensively studied in colorectal cancer and endometrial cancer, but not so in other tumors, such as ovarian carcinoma. We have determined the expression of mismatch repair proteins in a large cohort of 502 early-stage epithelial ovarian carcinoma entailing all the 5 main subtypes: high-grade serous carcinoma, endometrioid ovarian carcinoma (EOC), clear cell carcinoma (CCC), mucinous carcinoma, and low-grade serous carcinoma. We studied the association of MMRD with clinicopathologic and immunohistochemical features, including tumor-infiltrating lymphocytes in EOC, the histologic type in which MMRD is most frequent. In addition, MLH1 promoter methylation status and massive parallel sequencing were used to evaluate the proportion of sporadic and Lynch syndrome-associated tumors, and the most frequently mutated genes in MMRD EOCs. MMRD occurred only in endometriosis-associated histologic types, and it was much more frequent in EOC (18%) than in CCC (2%). The most frequent immunohistochemical pattern was loss of MLH1/PMS2, and in this group, 80% of the cases were sporadic and secondary to MLH1 promoter hypermethylation. The presence of somatic mutations in mismatch repair genes was the other mechanism of MMRD in sporadic tumors. In this series, the minimum estimated frequency of Lynch syndrome was 35% and it was due to germline mutations in MLH1, MSH2, and MSH6. ARID1A, PTEN, KTM2B, and PIK3CA were the most common mutated genes in this series. Interestingly, possible actionable mutations in ERRB2 were found in 5 tumors, but no TP53 mutations were detected. MMRD was associated with younger age and increased tumor-infiltrating lymphocytes. Universal screening in EOC and mixed EOC/CCC is recommended for the high frequency of MMRD detected; however, for CCC, additional clinical and pathologic criteria should be evaluated to help select cases for analysis.
Collapse
|
619
|
Cohen R, Shi Q, André T. Immunotherapy for Early Stage Colorectal Cancer: A Glance into the Future. Cancers (Basel) 2020; 12:E1990. [PMID: 32708216 PMCID: PMC7409300 DOI: 10.3390/cancers12071990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) have reshaped therapeutic strategies for cancer patients. The development of ICI for early stage colorectal cancer is accompanied by specific challenges: (i) the selection of patients who are likely to benefit from these treatments, i.e., patients with tumors harboring predictive factors of efficacy of ICI, such as microsatellite instability and/or mismatch repair deficiency (MSI/dMMR), or other potential parameters (increased T cell infiltration using Immunoscore® or others, high tumor mutational burden, POLE mutation), (ii) the selection of patients at risk of disease recurrence (poor prognostic features), and (iii) the choice of an accurate clinical trial methodological framework. In this review, we will discuss the ins and outs of clinical research of ICI for early stage MSI/dMMR CC patients in adjuvant and neoadjuvant settings. We will then summarize data that might support the development of ICI in localized colorectal cancer beyond MSI/dMMR.
Collapse
Affiliation(s)
- Romain Cohen
- Department of Medical Oncology, Hôpital Saint-Antoine, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), F-75012 Paris, France;
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA;
| | - Qian Shi
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA;
| | - Thierry André
- Department of Medical Oncology, Hôpital Saint-Antoine, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), F-75012 Paris, France;
| |
Collapse
|
620
|
Obayashi M, Shibasaki Y, Koakutsu T, Hayashi Y, Shoji T, Hirayama K, Yamazaki M, Takayanagi Y, Shibata H, Nakamura M, Maruo H. Pancreatic undifferentiated carcinoma with osteoclast-like giant cells curatively resected after pembrolizumab therapy for lung metastases: a case report. BMC Gastroenterol 2020; 20:220. [PMID: 32652936 PMCID: PMC7353752 DOI: 10.1186/s12876-020-01362-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/02/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Therapy targeting programmed death-1 or programmed death-1 ligand-1 (PD-1/PD-L1) has been developed for various solid malignant tumors, such as melanoma and non-small-cell lung cancer (NSCLC), but this approach has little effect in the treatment of pancreatic cancer. Pancreatic undifferentiated carcinoma with osteoclast-like giant cells (UCOGC) is a rare pancreatic malignancy having unique morphology and is considered a variant of pancreatic ductal adenocarcinoma (PDAC). Although UCOGC has been reported to have better prognosis than conventional PDAC, the optimal treatment for UCOGC with distant metastases has not been determined. CASE PRESENTATION A 66-year-old man was initially diagnosed with NSCLC with multiple intrapulmonary metastases and abdominal lymph node metastasis in the tail of the pancreas, and bronchial biopsy and diagnostic imaging were performed. Pathologic examination of the lung showed poorly differentiated adenocarcinoma cells expressing epithelial marker and PD-L1. Therefore, pembrolizumab monotherapy for NSCLC was given. The pulmonary lesions shrank markedly and were in complete remission after 8 months of anti-PD-1 therapy, though no therapeutic effect was observed in the pancreatic site. Distal pancreatectomy was then performed, and histopathological examination showed that the tumor was UCOGC originating from the pancreas. The histologic findings of the resected specimen mimicked those of the lung biopsy specimen, leading to the final assessment that the lung tumors were metastatic foci that migrated from the UCOGC, and only the metastatic lesions benefited from pembrolizumab therapy. CONCLUSION Immune checkpoint inhibitors have limited therapeutic effects on primary lesions of pancreatic cancer, but they may exert antitumor effects on pulmonary metastases of UCOGC.
Collapse
Affiliation(s)
- Miku Obayashi
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Yasushi Shibasaki
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Toru Koakutsu
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Yoshiro Hayashi
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Tsuyoshi Shoji
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Kazuhisa Hirayama
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Masanori Yamazaki
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Yasuhiro Takayanagi
- Department of Gastroenterology, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Hiroshi Shibata
- Department of Respiratory Medicine, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Masato Nakamura
- Department of Pathology, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan
| | - Hirotoshi Maruo
- Department of Surgery, Shizuoka City Shimizu Hospital, 1231 Miyakami, Shimizu-ku, Shizuoka, 424-8636, Japan.
| |
Collapse
|
621
|
Yoshino T, Pentheroudakis G, Mishima S, Overman MJ, Yeh KH, Baba E, Naito Y, Calvo F, Saxena A, Chen LT, Takeda M, Cervantes A, Taniguchi H, Yoshida K, Kodera Y, Kitagawa Y, Tabernero J, Burris H, Douillard JY. JSCO-ESMO-ASCO-JSMO-TOS: international expert consensus recommendations for tumour-agnostic treatments in patients with solid tumours with microsatellite instability or NTRK fusions. Ann Oncol 2020; 31:861-872. [PMID: 32272210 DOI: 10.1016/j.annonc.2020.03.299] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/15/2020] [Indexed: 01/05/2023] Open
Abstract
A Japan Society of Clinical Oncology (JSCO)-hosted expert meeting was held in Japan on 27 October 2019, which comprised experts from the JSCO, the Japanese Society of Medical Oncology (JSMO), the European Society for Medical Oncology (ESMO), the American Society of Clinical Oncology (ASCO), and the Taiwan Oncology Society (TOS). The purpose of the meeting was to focus on what we have learnt from both microsatellite instability (MSI)/deficient mismatch repair (dMMR) biomarkers in predicting the efficacy of anti-programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) immunotherapy, and the neurotrophic tyrosine receptor kinase (NTRK) gene fusions in predicting the efficacy of inhibitors of the tropomyosin receptor kinase (TRK) proteins across a range of solid tumour types. The recent regulatory approvals of the anti-PD-1 antibody pembrolizumab and the TRK inhibitors larotrectinib and entrectinib, based on specific tumour biomarkers rather than specific tumour type, have heralded a paradigm shift in cancer treatment approaches. The purpose of the meeting was to develop international expert consensus recommendations on the use of such tumour-agnostic treatments in patients with solid tumours. The aim was to generate a reference document for clinical practice, for pharmaceutical companies in the design of clinical trials, for ethics committees in the approval of clinical trial protocols and for regulatory authorities in relation to drug approvals, with a particular emphasis on diagnostic testing and patient selection.
Collapse
Affiliation(s)
- T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - G Pentheroudakis
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece
| | - S Mishima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - M J Overman
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - K-H Yeh
- Department of Medical Oncology, National Taiwan University Cancer Center and Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - E Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Y Naito
- Department of Experimental Therapeutics/Breast and Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - F Calvo
- Department of Clinical Pharmacology, University of Paris and Institute Gustave Roussy, Villejuif, France
| | - A Saxena
- Department of Medicine, Division of Hematology & Medical Oncology, Thoracic Oncology Service, Weill Cornell Medicine, New York, USA
| | - L-T Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - M Takeda
- Department of Medical Oncology, Kindai University, Osaka, Japan
| | - A Cervantes
- CIBERONC, Department of Medical Oncology, Institute of Health Research, INCLIVIA, University of Valencia, Valencia, Spain
| | - H Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - K Yoshida
- Department of Surgical Oncology, Gifu University, Graduate School of Medicine, Gifu, Japan
| | - Y Kodera
- Department of Gastrointestinal Surgery, Nagoya University, Nagoya, Japan
| | - Y Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - J Tabernero
- Medical Oncology Department, Vall d' Hebron University Hospital, Vall d'Hebron Institute of Oncology (V.H.I.O.), Barcelona, Spain
| | - H Burris
- The Sarah Cannon Research Institute, Nashville, USA
| | | |
Collapse
|
622
|
Fassan M, Brignola S, Pennelli G, Alberti G, Angerilli V, Bressan A, Pellino A, Lanza C, Salmaso R, Lonardi S, Pucciarelli S, Spolverato G, Scarpa M, Realdon S, Farinati F, Luchini C, Rugge M, Loupakis F. PD-L1 expression in gastroesophageal dysplastic lesions. Virchows Arch 2020; 477:151-156. [PMID: 31724072 DOI: 10.1007/s00428-019-02693-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Abstract
Immunotherapy has been recently approved for gastric (GC) and gastroesophageal-junction adenocarcinomas (GEC), and PD-L1 immunohistochemical evaluation represents a promising predictive biomarker in this oncological setting. A series of 125 gastroesophageal dysplastic lesions (52 low-grade, 73 high-grade) was investigated for PD-L1 and DNA mismatch repair proteins status. PD-L1 was positive (combined positive score (CPS) ≥ 1) in 48 (31.0%) dysplastic lesions. A higher prevalence of PD-L1-positive cases was observed among esophageal specimens compared with gastric ones (p = 0.0003), in high-grade and adenocarcinoma samples in comparison with low-grade dysplasia (p < 0.0001), and in lesions with mismatch repair deficiency (p = 0.028). For 30 dysplastic samples, a synchronous matched invasive lesion (GC = 15, GEC = 15) was available and tested for PD-L1 expression; a discordant PD-L1 status was observed in 12/30 (40%) cases. A relatively high prevalence in PD-L1 positivity was observed among gastroesophageal dysplastic lesions and this should be taken into consideration for future therapeutic strategies based on this biomarker.
Collapse
Affiliation(s)
- Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy.
| | - Stefano Brignola
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy
| | - Gianmaria Pennelli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy
| | - Giulia Alberti
- Unit of Medical Oncology 1, Department of Oncology, Istituto Oncologico Veneto, IOV-IRCCS, Padua, PD, Italy
- Department of Oncology, Surgery and Gastroenterology (DISCOG), University of Padua, Padua, PD, Italy
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy
| | - Alessandra Bressan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy
| | - Antonio Pellino
- Unit of Medical Oncology 1, Department of Oncology, Istituto Oncologico Veneto, IOV-IRCCS, Padua, PD, Italy
| | - Cristiano Lanza
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy
| | - Roberta Salmaso
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy
| | - Sara Lonardi
- Unit of Medical Oncology 1, Department of Oncology, Istituto Oncologico Veneto, IOV-IRCCS, Padua, PD, Italy
| | - Salvatore Pucciarelli
- 1st Surgery Unit, Department of Surgical Oncology and Gastroenterology (DiSCOG), University of Padua, Padua, Italy
| | - Gaya Spolverato
- 1st Surgery Unit, Department of Surgical Oncology and Gastroenterology (DiSCOG), University of Padua, Padua, Italy
| | - Marco Scarpa
- Surgery Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padua, PD, Italy
| | - Stefano Realdon
- Gastroenterology Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padua, PD, Italy
| | - Fabio Farinati
- Gastroenterology Unit, Department of Surgical Oncology and Gastroenterology (DiSCOG), University of Padua, Padua, PD, Italy
| | - Claudio Luchini
- Section of Pathology, Department of Diagnostics and Public Health, University and Hospital Trust of Verona, Verona, Italy
| | - Massimo Rugge
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121, Padua, PD, Italy
- Veneto Cancer Registry, Padua, Italy
| | - Fotios Loupakis
- Unit of Medical Oncology 1, Department of Oncology, Istituto Oncologico Veneto, IOV-IRCCS, Padua, PD, Italy
| |
Collapse
|
623
|
Baraibar I, Ros J, Mulet N, Salvà F, Argilés G, Martini G, Cuadra JL, Sardo E, Ciardiello D, Tabernero J, Élez E. Incorporating traditional and emerging biomarkers in the clinical management of metastatic colorectal cancer: an update. Expert Rev Mol Diagn 2020; 20:653-664. [PMID: 32552041 DOI: 10.1080/14737159.2020.1782194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Molecular profiling has led to significantly longer survival in metastatic colorectal cancer (mCRC) patients. Clinical guidelines recommend testing for KRAS/NRAS, BRAF and MSI status, and new biomarkers such as HER2 amplification and NTRK fusions have emerged more recently in refractory CRC, supported by overwhelming clinical relevance. These biomarkers can guide treatment management to improve clinical outcomes in these patients. AREAS COVERED Preclinical and clinical data over the last decade were reviewed for known and novel biomarkers with clinical implications in refractory CRC. Molecular alterations are described for classic and novel biomarkers, and data for completed and ongoing studies with targeted and immunotherapies are presented. EXPERT OPINION Use of targeted therapies based on biomarker testing in CRC has enabled impressive improvements in clinical outcomes in refractory patients. BRAF, MSI, NRAS and KRAS should be tested upfront in all patients given their indisputable therapeutic implications. Other molecular alterations such as HER2 and NTRK are emerging. Testing for these alterations may further improve outcomes for refractory CRC patients. Nonetheless, many key aspects remain to be defined including the optimal timing and technique for testing, the most adequate panel, and whether all patients should be tested for all alterations.
Collapse
Affiliation(s)
- Iosune Baraibar
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Javier Ros
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Nuria Mulet
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain.,Department of Medical Oncology, Institut Català D' oncologia-IDIBELL , Barcelona, Spain
| | - Francesc Salvà
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Guillem Argilés
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Giulia Martini
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain.,Dipartimento di Medicina di Precisione, Università Degli Studi Della Campania Luigi Vanvitelli , Naples, Italy
| | | | - Emilia Sardo
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain
| | - Davide Ciardiello
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain.,Dipartimento di Medicina di Precisione, Università Degli Studi Della Campania Luigi Vanvitelli , Naples, Italy
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Elena Élez
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| |
Collapse
|
624
|
Kallenbach-Thieltges A, Großerueschkamp F, Jütte H, Kuepper C, Reinacher-Schick A, Tannapfel A, Gerwert K. Label-free, automated classification of microsatellite status in colorectal cancer by infrared imaging. Sci Rep 2020; 10:10161. [PMID: 32576892 PMCID: PMC7311536 DOI: 10.1038/s41598-020-67052-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Challenging histopathological diagnostics in cancer include microsatellite instability-high (MSI-H) colorectal cancer (CRC), which occurs in 15% of early-stage CRC and is caused by a deficiency in the mismatch repair system. The diagnosis of MSI-H cannot be reliably achieved by visual inspection of a hematoxylin and eosin stained thin section alone, but additionally requires subsequent molecular analysis. Time- and sample-intensive immunohistochemistry with subsequent fragment length analysis is used. The aim of the presented feasibility study is to test the ability of quantum cascade laser (QCL)-based infrared (IR) imaging as an alternative diagnostic tool for MSI-H in CRC. We analyzed samples from 100 patients with sporadic CRC UICC stage II and III. Forty samples were used to develop the random forest classifier and 60 samples to verify the results on an independent blinded dataset. Specifically, 100% sensitivity and 93% specificity were achieved based on the independent 30 MSI-H- and 30 microsatellite stable (MSS)-patient validation cohort. This showed that QCL-based IR imaging is able to distinguish between MSI-H and MSS for sporadic CRC - a question that goes beyond morphological features - based on the use of spatially resolved infrared spectra used as biomolecular fingerprints.
Collapse
Affiliation(s)
- Angela Kallenbach-Thieltges
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Frederik Großerueschkamp
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Hendrik Jütte
- Institute of Pathology, Ruhr University Bochum, Bochum, Germany
| | - Claus Kuepper
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Anke Reinacher-Schick
- Department of Hematology, Oncology and Palliative Care, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | | | - Klaus Gerwert
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany. .,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany.
| |
Collapse
|
625
|
Horimoto Y, Thinzar Hlaing M, Saeki H, Kitano S, Nakai K, Sasaki R, Kurisaki-Arakawa A, Arakawa A, Otsuji N, Matsuoka S, Tokuda E, Arai M, Saito M. Microsatellite instability and mismatch repair protein expressions in lymphocyte-predominant breast cancer. Cancer Sci 2020; 111:2647-2654. [PMID: 32449246 PMCID: PMC7385389 DOI: 10.1111/cas.14500] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
The frequency of microsatellite instability (MSI) is reportedly extremely low in breast cancer, despite widespread clinical expectations that many patients would be responsive to immune-checkpoint inhibitors (ICI). Considering that some triple-negative breast cancers (TNBC) responded well to ICI in a clinical trial and that a high density of tumor-infiltrating lymphocytes (TILs) is frequently observed in other cancers with high levels of microsatellite instability (MSI-H), we hypothesized that some TNBC with a high density of TILs would be MSI-H. Medullary carcinoma (MedCa) of the breast, a rare histological type, is characterized by a high density of TILs. Considering that MedCa of the colon is often MSI-H, we suspected that MedCa in breast cancer might also include MSI-H tumors. Therefore, we conducted MSI tests on such breast cancers with a high density of TILs. The MSI status of 63 TIL-high TNBC and 38 MedCa tumors, all from Asian women who had undergone curative surgery, were determined retrospectively. DNA mismatch repair (MMR) proteins and PD-L1 expression were also investigated immunohistochemically. All samples were microsatellite stable, being negative for all microsatellite markers. TIL-high TNBC with low MLH1 protein had higher levels of PD-L1 in stromal immune cells (P = .041). MedCa tumors showed significantly higher PD-L1 expression in immune cells than in TIL-high TNBC (<.001). We found that MSI-H tumors were absent in TIL-high breast cancers. Examination of MMR proteins, not a purpose of Lynch syndrome screening, may merit further studies to yield predictive information for identifying patients who are likely to benefit from ICI.
Collapse
Affiliation(s)
- Yoshiya Horimoto
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - May Thinzar Hlaing
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Harumi Saeki
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigehisa Kitano
- Cancer Immunotherapy Development, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Katsuya Nakai
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ritsuko Sasaki
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Atsushi Arakawa
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Naomi Otsuji
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shuji Matsuoka
- Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Emi Tokuda
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | - Masami Arai
- Diagnostics and Therapeutics of Intractable Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Clinical Genetics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mitsue Saito
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
626
|
Abstract
PURPOSE OF REVIEW Although all primary central nervous system (CNS) tumors are rare, certain tumor types each represent less than 2% of the total and an annual incidence of about 1000 patients or less. Most of them are disproportionally diagnosed in children and young adults, but older adults can also be affected and are rarely recruited to clinical trials. Recent new molecular techniques have led to reclassification of some of these tumors and discovery of actionable molecular alterations. RECENT FINDINGS We review recent progress in the molecular understanding and therapeutic options of selected rare CNS tumors, with a focus on select clinical trials (temozolomide and lapatinib for recurrent ependymoma; vemurafenib for BRAFV600E-mutated tumors), as well as tumor-agnostic approvals (pembrolizumab, larotrectinib) and their implications for rare CNS tumors. SUMMARY Although rare CNS tumors are a very small fraction of the total of cancers, they represent a formidable challenge. There is a need for dedicated clinical trials with strong correlative component in patients of all ages with rare CNS tumors. Critical research questions include relevance of the selected target for specific tumor types, persistence of the actionable biomarker at recurrence, blood-brain barrier penetration, and analysis of mechanisms of primary and acquired resistance.
Collapse
|
627
|
Pécriaux A, Favre L, Calderaro J, Charpy C, Derman J, Pujals A. Detection of microsatellite instability in a panel of solid tumours with the Idylla MSI Test using extracted DNA. J Clin Pathol 2020; 74:36-42. [PMID: 32513848 DOI: 10.1136/jclinpath-2020-206581] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022]
Abstract
AIM During the last few years, determination of microstatellite instability (MSI) status has become a routine part of clinical practice, essentially to detect Lynch syndrome. Recently, MSI testing has increased with the development of immunotherapy and has expanded to a large panel of solid tumours. The aim of our work was to evaluate a fully automated system developed by Biocartis, the Idylla MSI Test, which performs an MSI analysis within 150 min. METHODS A comparison between pentaplex PCR, immunohistochemistry and Idylla MSI Test was performed in 53 colorectal carcinoma samples, 7 small intestine adenocarcinomas, 15 duodenal and pancreatic adenocarcinomas, 16 gastric tumours, 15 endometrial adenocarcinomas, 5 ovarian carcinomas and 4 cases of urinary tract tumours using extracted DNA. Limit-of-detection (LOD) experiment was also done using a commercial DNA known to harbour MSI phenotype. RESULTS The overall sensitivity was 94% and the overall specificity was 100%. Two invalid and three false-negative results were observed. Our experiments showed that the amount of DNA loaded into the cartridge was decisive and should be superior to 25 ng. LOD comprised between 4% and 8%. CONCLUSION Overall, we have demonstrated that the Idylla MSI Test is a rapid and valid option to detect MSI phenotype which can be used in a large panel of solid tumours.
Collapse
Affiliation(s)
| | - Loetitia Favre
- Department of Pathology, CHU Henri Mondor, Créteil, France.,Université Paris-Est Créteil Val de Marne Faculté de médecine, Creteil, France
| | - Julien Calderaro
- Department of Pathology, CHU Henri Mondor, Créteil, France.,Université Paris-Est Créteil Val de Marne Faculté de médecine, Creteil, France
| | - Cécile Charpy
- Department of Pathology, CHU Henri Mondor, Créteil, France
| | | | - Anaïs Pujals
- Department of Pathology, CHU Henri Mondor, Créteil, France .,Université Paris-Est Créteil Val de Marne Faculté de médecine, Creteil, France
| |
Collapse
|
628
|
Riechelmann RP, Peixoto RD, Fernandes GDS, Weschenfelder RF, Prolla G, Filho DR, Andrade AC, Crosara M, Rego JFM, Gansl RC, Coimbra F, Aguiar S, Carvalho E, Hoff PM, Coutinho AK. Evidence-based recommendations for gastrointestinal cancers during the COVID-19 pandemic by the Brazilian Gastrointestinal Tumours Group. Ecancermedicalscience 2020; 14:1048. [PMID: 32565901 PMCID: PMC7289615 DOI: 10.3332/ecancer.2020.1048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE As of 2020, the world is facing the great challenge of the COVID-19 (Coronavirus disease 2019) pandemic, caused by the SARS-CoV-2 virus. While the overall mortality is low, the virus is highly virulent and may infect millions of people worldwide. This will consequently burden health systems, particularly by those individuals considered to be at high risk of severe complications from COVID-19. Such risk factors include advanced age, cardiovascular and pulmonary diseases, diabetes and cancer. However, few data on the outcomes of cancer patients infected by SARS CoV-2 exist. Therefore, there is a lack of guidance on how to manage cancer patients during the pandemic. We sought to propose specific recommendations about the management of patients with gastrointestinal malignancies. METHODS The Brazilian Gastrointestinal Tumours Group board of directors and members sought up-to-date scientific literature on each tumour type and discussed all recommendations by virtual meetings to provide evidence-based-and sometimes, expert opinion-recommendation statements. Our objectives were to recommend evidence-based approaches to both treat and minimise the risk of COVID-19 for cancer patients, and simultaneously propose how to decrease the use of hospital resources at a time these resources need to be available to treat COVID-19 patients. RESULTS Overall and tumour-specific recommendations were made by stage (including surgical, locoregional, radiotherapy, systemic treatments and follow-up strategies) for the most common gastrointestinal malignancies: esophagus, gastric, pancreas, bile duct, hepatocellular, colorectal, anal cancer and neuroendocrine tumours. CONCLUSIONS Our recommendations emphasise the importance of treating cancer patients, using the best evidence available, while simultaneously taking into consideration the world-wide health resource hyperutilisation to treat non-cancer COVID-19 patients.
Collapse
Affiliation(s)
| | | | | | | | - Gabriel Prolla
- Centro de Oncologia Hospital São Lucas da PUCRS, Porto Alegre, RS 90610-000, Brazil
| | | | | | | | | | - Rene C Gansl
- Hospital Israelita Albert Einstein, Sao Paulo, SP 05652-900, Brazil
| | - Felipe Coimbra
- AC Camargo Cancer Center, Sao Paulo, SP 01509-010, Brazil
| | - Samuel Aguiar
- AC Camargo Cancer Center, Sao Paulo, SP 01509-010, Brazil
| | | | - Paulo M Hoff
- Instituto do Cancer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP 01246-000, Brazil
| | - Anelisa K Coutinho
- Clinica AMO, Assistência Multidisciplinar em Oncologia, Salvador, BA 41950-640, Brazil
| |
Collapse
|
629
|
Post CCB, Westermann AM, Bosse T, Creutzberg CL, Kroep JR. PARP and PD-1/PD-L1 checkpoint inhibition in recurrent or metastatic endometrial cancer. Crit Rev Oncol Hematol 2020; 152:102973. [PMID: 32497971 DOI: 10.1016/j.critrevonc.2020.102973] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 12/28/2022] Open
Abstract
The prognosis of recurrent or metastatic endometrial cancer is poor, with five-year survival of only 10-20 %. First-line therapy consists of either platinum-based chemotherapy or hormonal therapy. No standard subsequent-line therapy has been identified. In recent years, significant progress has been made in the knowledge on underlying molecular biology of endometrial cancer and potential targets for therapy have been identified. Targeted therapies as poly (ADP-ribose) polymerase (PARP) inhibitors and immunotherapy as PD-1/PD-L1 checkpoint inhibitors have the potential to be effective against specific subtypes of endometrial cancer. Preclinical studies have shown that combining these agents may result in a synergistic effect. In this review, we focus on the molecular basis of checkpoint inhibition and targeted therapy as PARP inhibition in endometrial cancer and summarize available clinical data, and ongoing and planned clinical trials that investigate these agents as mono- or combination therapies in endometrial cancer and where relevant, other gynecological cancers.
Collapse
Affiliation(s)
- Cathalijne C B Post
- Departments of Medical Oncology, Leiden, the Netherlands; Departments of Radiation Oncology Leiden, the Netherlands.
| | - Anneke M Westermann
- Departments of Medical Oncology, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Tjalling Bosse
- Departments of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Judith R Kroep
- Departments of Medical Oncology, Leiden, the Netherlands
| |
Collapse
|
630
|
Teixido C, Reguart N. Using biomarkers to determine optimal combinations with immunotherapy (biomarker discovery perspective). Future Oncol 2020; 16:1677-1681. [PMID: 32407147 DOI: 10.2217/fon-2020-0171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Cristina Teixido
- Department of Pathology, Thoracic Oncology Unit, Hospital Clínic of Barcelona, IDIBAPS, Villarroel 170, Barcelona, 08036, Spain
| | - Noemi Reguart
- Department of Medical Oncology, Thoracic Oncology Unit, Hospital Clínic of Barcelona, IDIBAPS, Villarroel 170, Barcelona 08036, Spain
| |
Collapse
|
631
|
Steeghs EMP, Kroeze LI, Tops BBJ, van Kempen LC, Ter Elst A, Kastner-van Raaij AWM, Hendriks-Cornelissen SJB, Hermsen MJW, Jansen EAM, Nederlof PM, Schuuring E, Ligtenberg MJL, Eijkelenboom A. Comprehensive routine diagnostic screening to identify predictive mutations, gene amplifications, and microsatellite instability in FFPE tumor material. BMC Cancer 2020; 20:291. [PMID: 32264863 PMCID: PMC7137451 DOI: 10.1186/s12885-020-06785-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/25/2020] [Indexed: 02/08/2023] Open
Abstract
Background Sensitive and reliable molecular diagnostics is needed to guide therapeutic decisions for cancer patients. Although less material becomes available for testing, genetic markers are rapidly expanding. Simultaneous detection of predictive markers, including mutations, gene amplifications and MSI, will save valuable material, time and costs. Methods Using a single-molecule molecular inversion probe (smMIP)-based targeted next-generation sequencing (NGS) approach, we developed an NGS panel allowing detection of predictive mutations in 33 genes, gene amplifications of 13 genes and microsatellite instability (MSI) by the evaluation of 55 microsatellite markers. The panel was designed to target all clinically relevant single and multiple nucleotide mutations in routinely available lung cancer, colorectal cancer, melanoma, and gastro-intestinal stromal tumor samples, but is useful for a broader set of tumor types. Results The smMIP-based NGS panel was successfully validated and cut-off values were established for reliable gene amplification analysis (i.e. relative coverage ≥3) and MSI detection (≥30% unstable loci). After validation, 728 routine diagnostic tumor samples including a broad range of tumor types were sequenced with sufficient sensitivity (2.4% drop-out), including samples with low DNA input (< 10 ng; 88% successful), low tumor purity (5–10%; 77% successful), and cytological material (90% successful). 75% of these tumor samples showed ≥1 (likely) pathogenic mutation, including targetable mutations (e.g. EGFR, BRAF, MET, ERBB2, KIT, PDGFRA). Amplifications were observed in 5.5% of the samples, comprising clinically relevant amplifications (e.g. MET, ERBB2, FGFR1). 1.5% of the tumor samples were classified as MSI-high, including both MSI-prone and non-MSI-prone tumors. Conclusions We developed a comprehensive workflow for predictive analysis of diagnostic tumor samples. The smMIP-based NGS analysis was shown suitable for limited amounts of histological and cytological material. As smMIP technology allows easy adaptation of panels, this approach can comply with the rapidly expanding molecular markers.
Collapse
Affiliation(s)
- Elisabeth M P Steeghs
- Department of Pathology, Radboud university medical center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands
| | - Leonie I Kroeze
- Department of Pathology, Radboud university medical center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands
| | - Bastiaan B J Tops
- Department of Pathology, Radboud university medical center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands.,Department of Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Leon C van Kempen
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arja Ter Elst
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | - Mandy J W Hermsen
- Department of Pathology, Radboud university medical center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands
| | - Erik A M Jansen
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Petra M Nederlof
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ed Schuuring
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjolijn J L Ligtenberg
- Department of Pathology, Radboud university medical center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands.,Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Astrid Eijkelenboom
- Department of Pathology, Radboud university medical center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands.
| |
Collapse
|
632
|
Cohen R, Pudlarz T, Garcia-Larnicol ML, Vernerey D, Dray X, Clavel L, Jary M, Piessen G, Zaanan A, Aparicio T, Louvet C, Tournigand C, Chibaudel B, Tougeron D, Guimbaud R, Benouna J, Adenis A, Sokol H, Borg C, Duval A, Svrcek M, André T. [Localized MSI/dMMR gastric cancer patients, perioperative immunotherapy instead of chemotherapy: The GERCOR NEONIPIGA phase II study is opened to recruitment]. Bull Cancer 2020; 107:438-446. [PMID: 32057467 DOI: 10.1016/j.bulcan.2019.11.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/25/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Perioperative chemotherapy is the standard strategy for localized gastric cancers. Nevertheless, this strategy seems to be inefficient, if not deleterious, for patients with tumors harboring microsatellite instability (MSI) and/or mismatch repair deficiency (dMMR), a tumor phenotype predictive for the efficacy of immune checkpoint inhibitors (ICKi). AIM The GERCOR NEONIPIGA single-arm phase II study (NCT04006262; EUDRACT 2018-004712-22) aims at evaluating the efficacy of a peri-operative strategy with nivolumab and ipilimumab in neoadjuvant setting, then nivolumab alone after surgery for patients with resectable MSI/dMMR gastric cancer. MATERIAL AND METHODS Main inclusion criteria are: gastric and oesogastric junction adenocarcinoma (GOA), T2-T4, all N stage and M0, MSI/dMMR. Patients will be treated with nivolumab 240mg Q2W, 6 infusions, and ipilimumab 1mg/kg Q6W, 2 infusions in neoadjuvant setting. Following surgery, patients with TRG 1-2-3 (Mandard tumor regression grade), acceptable tolerance of neoadjuvant treatment and postoperative ECOG performance status 0-1, will be treated with adjuvant nivolumab 480mg Q4W, 9 infusions. RESULTS The primary endpoint is pathological complete response rate (pCR-R). Based on a Fleming design, with α=5% and β=20%, 27 patients have to be evaluated (H0=5%; H1=20%). Secondary endpoints include disease-free survival, overall survival and safety. CONCLUSION This study is planned to include 32 patients to evaluate the pCR-R with the combination of nivolumab and ipilimumab in neoadjuvant setting for MSI/dMMR localized GOA. The MSI/MMR status should be systematically assessed on diagnostic biopsies of all GOA. If it meets its primary endpoint, the GERCOR NEONIPIGA study might mark a turning point in the management of localized MSI/dMMR GOA patients.
Collapse
Affiliation(s)
- Romain Cohen
- AP-HP, Sorbonne Université, hôpital Saint-Antoine, department of medical oncology, 75012 Paris, France.
| | - Thomas Pudlarz
- AP-HP, hôpital Saint-Antoine, Department of medical oncology, 75012 Paris, France
| | | | - Dewi Vernerey
- University Hospital of Besançon, Methodology and Quality of Life Unit in Oncology, 25000 Besançon, France; University Bourgogne Franche-Comté, interactions Hôte-Greffon-Tumeur/Ingénierie cellulaire et génique, EFS BFC, UMR1098, Inserm, 25000 Besançon, France
| | - Xavier Dray
- AP-HP, Sorbonne Université, hôpital Saint-Antoine, department of endoscopy, 75012 Paris, France
| | - Léa Clavel
- Hôpital privé Jean-Mermoz, gastro-enterology and digestive oncology department, Lyon, France
| | - Marine Jary
- University Bourgogne Franche-Comté, interactions Hôte-Greffon-Tumeur/Ingénierie cellulaire et génique, EFS BFC, UMR1098, Inserm, 25000 Besançon, France; CHU de Jean-Minjoz, Medical oncology department, 3, boulevard Alexander-Fleming, 25000 Besançon, France
| | - Guillaume Piessen
- Hôpital Claude-Huriez, service de chirurgie digestive et oncologique, 59000 Lille, France
| | - Aziz Zaanan
- Paris Descartes University, Sorbonne Paris Cité, Georges-Pompidou European Hospital, Department of Digestive Oncology, Paris, France
| | - Thomas Aparicio
- AP-HP, université de Paris, Hôpital Saint-Louis, Gastro-enterology and digestive oncology department, Paris, France
| | - Christophe Louvet
- Institut mutualiste Montsouris, medical oncology department, 75014 Paris, France
| | | | - Benoist Chibaudel
- Hôpital Franco-Britannique, Fondation Cognacq-Jay, Medical Oncology, Levallois-Perret, France
| | - David Tougeron
- Poitiers University Hospital, Department of Gastroenterology, Poitiers, France
| | - Rosine Guimbaud
- CHU de Toulouse-IUCT Rangueil-Larrey, Digestive Medical Oncology department, Toulouse, France
| | - Jaafar Benouna
- University Hospital of Nantes, Digestive Oncology, Nantes, France
| | - Antoine Adenis
- ICM Val d'Aurelle, medical oncology department, Montpellier, France
| | - Harry Sokol
- AP-HP, Sorbonne Université, Centre de Recherche Saint-Antoine, CRSA, Hôpital Saint-Antoine, gastroenterology department, Inserm, 75012 Paris, France
| | - Christophe Borg
- University Bourgogne Franche-Comté, interactions Hôte-Greffon-Tumeur/Ingénierie cellulaire et génique, EFS BFC, UMR1098, Inserm, 25000 Besançon, France; Hôpital privé Jean-Mermoz, gastro-enterology and digestive oncology department, Lyon, France
| | - Alex Duval
- Sorbonne Université, centre de recherche Saint-Antoine, équipe Instabilité des microsatellites et cancer, équipe labellisée par la Ligue nationale contre le cancer et SIRIC CURAMUS, Inserm UMRS 938, 75012 Paris, France
| | - Magali Svrcek
- AP-HP, Sorbonne Université, hôpital Saint-Antoine, department of pathology, 75012 Paris, France
| | - Thierry André
- AP-HP, Sorbonne Université, hôpital Saint-Antoine, department of medical oncology, 75012 Paris, France
| |
Collapse
|
633
|
Jiao Q, Ren Y, Ariston Gabrie AN, Wang Q, Wang Y, Du L, Liu X, Wang C, Wang YS. Advances of immune checkpoints in colorectal cancer treatment. Biomed Pharmacother 2020; 123:109745. [DOI: 10.1016/j.biopha.2019.109745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/30/2022] Open
|
634
|
Schneeweiss A, Hartkopf AD, Müller V, Wöckel A, Lux MP, Janni W, Ettl J, Belleville E, Huober J, Thill M, Fasching PA, Kolberg HC, Pöschke P, Welslau M, Overkamp F, Tesch H, Fehm TN, Lüftner D, Schütz F. Update Breast Cancer 2020 Part 1 - Early Breast Cancer: Consolidation of Knowledge About Known Therapies. Geburtshilfe Frauenheilkd 2020; 80:277-287. [PMID: 32139917 PMCID: PMC7056401 DOI: 10.1055/a-1111-2431] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 02/08/2023] Open
Abstract
This review is intended to present the latest developments in the prevention and treatment of early breast cancer. The risk of breast cancer can be increasingly better characterised with large epidemiological studies on genetic and non-genetic risk factors. Through new analyses, the evidence for high-penetrance genes as well as for low-penetrance genes was able to be improved. New data on denosumab and atezolizumab are available in the neoadjuvant situation as is a pooled appraisal of numerous studies on capecitabine in the curative situation. There is also an update to the overall survival data of pertuzumab in the adjuvant situation with a longer follow-up observation period. Finally, digital medicine is steadily finding its way into science. A recently conducted study on automated breast cancer detection using artificial intelligence establishes the basis for a future review in clinical studies.
Collapse
Affiliation(s)
- Andreas Schneeweiss
- National Center for Tumor Diseases, Division Gynecologic Oncology, University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Andreas D. Hartkopf
- Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, Germany
| | - Volkmar Müller
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Johannes Ettl
- Department of Obstetrics and Gynecology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Jens Huober
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Marc Thill
- Agaplesion Markus Krankenhaus, Frauenklinik, Frankfurt, Germany
| | - Peter A. Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | - Patrik Pöschke
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | | | - Hans Tesch
- Oncology Practice at Bethanien Hospital Frankfurt, Frankfurt, Germany
| | - Tanja N. Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Diana Lüftner
- Charité University Hospital, Campus Benjamin Franklin, Department of Hematology, Oncology and Tumour Immunology, Berlin, Germany
| | - Florian Schütz
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
635
|
Huang Z, Zhang W, He D, Cui X, Tian S, Yin H, Song B. Development and validation of a radiomics model based on T2WI images for preoperative prediction of microsatellite instability status in rectal cancer: Study Protocol Clinical Trial (SPIRIT Compliant). Medicine (Baltimore) 2020; 99:e19428. [PMID: 32150094 PMCID: PMC7478495 DOI: 10.1097/md.0000000000019428] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Globally, colorectal cancer (CRC) is the third most commonly diagnosed cancer in males and the second in females. Rectal cancer (RC) accounts for about 28% of all newly diagnosed CRC cases. The treatment of choice for locally advanced RC is a combination of surgical resection and chemotherapy and/or radiotherapy. These patients can potentially be cured, but the clinical outcome depends on the tumor biology. Microsatellite instability (MSI) is an important biomarker in CRC, with crucial diagnostic, prognostic, and predictive implications. It is important to develop a noninvasive, repeatable, and reproducible method to reflect the microsatellite status. Magnetic resonance imaging (MRI) has been recommended as the preferred imaging examination for RC in clinical practice by both the National Comprehensive Cancer Network and the European Society for Medical Oncology guidelines. T2WI is the core sequence of MRI scanning protocol for RC. Radiomics, the high-throughput mining of quantitative image features from standard-of-care medical imaging that enables data to be extracted and applied within clinical-decision support systems to improve diagnostic, prognostic, and predictive accuracy, is gaining importance in cancer research.We proposed a hypothesis: A simple radiomics model based on only T2WI images can accurately evaluate the MSI status of RC preoperatively. OBJECTIVE To develop a radiomics model based on T2WI images for accurate preoperative diagnosis the MSI status of RC. METHOD All patients with RC were retrospectively enrolled. The dataset was randomly split into training cohort (70% of all patients) and testing cohort (30% of all patients). The radiomics features will be extracted from T2WI-MR images of the entire primary tumor region. Least absolute shrinkage and selection operator was used to select the most predictive radiomics features. Logistic regression models were constructed in the training/validation cohort to discriminate the MSI status using clinical factors, radiomics features, or their integration. The diagnostic performance of these 3 models was evaluated in the testing cohort based on their area under the curve, sensitivity, specificity, and accuracy. DISCUSSION This study will help us know whether radiomics model based on T2WI images to preoperative identify MSI status of RC.
Collapse
Affiliation(s)
- Zixing Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu
| | - Wei Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’ s Armed Police Forces, Leshan
| | - Du He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu
| | - Xing Cui
- Institute of Advanced Research, Infervision, Beijing, China
| | - Song Tian
- Institute of Advanced Research, Infervision, Beijing, China
| | - Hongkun Yin
- Institute of Advanced Research, Infervision, Beijing, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu
| |
Collapse
|
636
|
Rachiglio AM, Sacco A, Forgione L, Esposito C, Chicchinelli N, Normanno N. Colorectal cancer genomic biomarkers in the clinical management of patients with metastatic colorectal carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:53-70. [PMID: 36046264 PMCID: PMC9400741 DOI: 10.37349/etat.2020.00004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal carcinoma (CRC) is an heterogeneous disease in which different genetic alterations play a role in its pathogenesis and progression and offer potential for therapeutic intervention. The research on predictive biomarkers in metastatic CRC (mCRC) mainly focused on the identification of biomarkers of response or resistance to anti-epidermal growth factor receptor monoclonal antibodies. In this respect, international guidelines suggest testing mCRC patients only for KRAS, NRAS and BRAF mutations and for microsatellite instability. However, the use of novel testing methods is raising relevant issue related to these biomarkers, such as the presence of sub-clonal RAS mutations or the clinical interpretation of rare no-V600 BRAF variants. In addition, a number of novel biomarkers is emerging from recent studies including amplification of ERBB2, mutations in ERBB2, MAP2K1 and NF1 and rearrangements of ALK, ROS1, NTRK and RET. Mutations in POLE and the levels of tumor mutation burden also appear as possible biomarkers of response to immunotherapy in CRC. Finally, the consensus molecular subtypes classification of CRC based on gene expression profiling has prognostic and predictive implications. Integration of all these information will be likely necessary in the next future in order to improve precision/personalized medicine in mCRC patients.
Collapse
Affiliation(s)
- Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Alessandra Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Laura Forgione
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Claudia Esposito
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Nicoletta Chicchinelli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| |
Collapse
|
637
|
Jia M, Yao L, Yang Q, Chi T. Association of MSH2 Expression With Tumor Mutational Burden and the Immune Microenvironment in Lung Adenocarcinoma. Front Oncol 2020; 10:168. [PMID: 32154170 PMCID: PMC7046689 DOI: 10.3389/fonc.2020.00168] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/30/2020] [Indexed: 12/20/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapies that target programmed cell death 1 (PD1) and PD1 ligand 1 (PDL1) have demonstrated promising benefits in lung adenocarcinoma (LUAD), and tumor mutational burden (TMB) is the most robust biomarker associated with the efficacy of PD-1-PD-L1 axis blockade in LUAD, but the assessment of TMB by whole-exome sequencing (WES) is rather expensive and time-consuming. Although targeted panel sequencing has been developed and approved by the US Food and Drug Administration (FDA) to estimate TMB, we found that its predictive accuracy for ICB response was significantly lower than WES in LUAD. Given that previous studies were mainly focusing on genomic variations to explore surrogate biomarkers of TMB, we turned to examine the transcriptome-based correlation with TMB in this study. Combining three immunotherapeutic cohorts with two independent The Cancer Genome Atlas (TCGA) datasets, we revealed that the expression of mutS homolog 2 (MSH2), one of the most crucial genes involved in DNA mismatch repair (MMR) pathway, was the strongest feature associated with increased TMB in multivariate analysis. Furthermore, MSH2 expression also displayed a significantly positive correlation with smoking signature while an inverse association with MMR deficiency (MMRd) signature in LUAD. More importantly, high expression of MSH2 markedly correlated with increased PD-L1 expression and CD8+ T cell infiltration, both suggesting a prominent immunotherapy-responsive microenvironment in LUAD. Notably, detecting MSH2 expression is much easier, faster, and cheaper than TMB in clinical practice. Taken together, this study demonstrates the association of MSH2 expression with TMB and the immune microenvironment in LUAD. MSH2 expression may be developed as a potential surrogate biomarker of TMB to identify ICB responders in LUAD.
Collapse
Affiliation(s)
- Mingming Jia
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Linli Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tian Chi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,Department Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
638
|
Zhang Y, Wang L, Li R, Liu B. The emerging development of tumor mutational burden in patients with NSCLC. Future Oncol 2020; 16:469-481. [PMID: 32048882 DOI: 10.2217/fon-2019-0650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Immunocheckpoint inhibitors (ICIs) which target PD-1 and CTLA-4 have dramatically changed the history of non-small-cell lung cancer treatment. Multiple biomarkers especially tumor mutational burden (TMB) have been raised to be potential predictors of response to ICIs. However, great value of TMB has been observed in patients who receive ICIs monotherapy instead of ICIs combination therapy from latest exploratory studies. Thus, the innovative concept of TMB needs to be identified. This study uncovers specific aspects of TMB including signatures of TMB, factors related with variation, racial differences, heterogeneity between tissue TMB and blood-based TMB. Additionally, more and more factors are found valuable in clinical trials, suggesting that more markers should be further investigated as interesting candidates for response prediction beyond TMB.
Collapse
Affiliation(s)
- Yu Zhang
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Lifeng Wang
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Rutian Li
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| |
Collapse
|
639
|
Gazouli I, Zarkavelis G, Mauri D, Pentheroudakis G. Mismatch repair deficiency/microsatellite instability testing as predictive immunotherapy biomarkers-possible diagnostic missteps trusting a single method. ANNALS OF TRANSLATIONAL MEDICINE 2020; 7:S329. [PMID: 32016047 DOI: 10.21037/atm.2019.09.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ioanna Gazouli
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia, EMEKEN, Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia, EMEKEN, Ioannina, Greece
| | - Davide Mauri
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia, EMEKEN, Ioannina, Greece
| | - George Pentheroudakis
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia, EMEKEN, Ioannina, Greece
| |
Collapse
|
640
|
Michaelidou K, Agelaki S, Mavridis K. Molecular markers related to immunosurveillance as predictive and monitoring tools in non-small cell lung cancer: recent accomplishments and future promises. Expert Rev Mol Diagn 2020; 20:335-344. [PMID: 32000550 DOI: 10.1080/14737159.2020.1724785] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: The landscape of systemic treatment options for lung cancer has rapidly evolved with the emergence of immunomodulatory agents such as neutralizing antibodies targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1). Another major breakthrough was the introduction of biomarkers, such as PD-L1 expression and tumor mutational burden (TMB), predicting response to immunotherapy. However, markers for monitoring treatment response are still lacking.Areas covered: PD-L1 and TMB represent static pre-treatment evaluations. Dynamic biomarkers are required, along with static ones, to accurately predict and monitor immunotherapy response and to discriminate between responders and non-responders early in the course of treatment. The tumor immune contexture offers potential candidates that can be tested through the liquid biopsy approach, such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), exosomes, microRNAs (miRNAs), circular RNAs (circRNAs), RNA splice variants, and immune cell subsets.Expert opinion: A holistic approach combining information from tissue at the time of diagnosis and serial liquid biopsy data could lead to a novel combinatorial biomarker panel with enhanced treatment monitoring potential. Incorporating information from additional parts of the tumor-host ecosystem, such as metabolic markers and the microbiome is expected to provide added value to this strategy.
Collapse
Affiliation(s)
- Kleita Michaelidou
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece
| | - Sofia Agelaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece.,Department of Medical Oncology, University General Hospital, Heraklion, Greece
| | - Konstantinos Mavridis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| |
Collapse
|
641
|
Cheng AL, Hsu C, Chan SL, Choo SP, Kudo M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol 2020; 72:307-319. [PMID: 31954494 DOI: 10.1016/j.jhep.2019.09.025] [Citation(s) in RCA: 362] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/09/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitor (ICI) therapy targeting anti-programmed cell death-1 (anti-PD-1) or its ligand (anti-PD-L1) is the backbone of numerous combination regimens aimed at improving the objective response and survival of patients with hepatocellular carcinoma (HCC). Clinical trials of immuno-oncology regimens in other cancer types have shed light on issues of study design, including how to choose candidate regimens based on early-phase trial results, statistical considerations in trials with multiple primary endpoints, and the importance of predictive biomarkers. In this review, the updated data from early-phase trials of combination immunotherapy for HCC are summarised. Since the most extensively tested combination regimens for advanced HCC comprise anti-PD-1/anti-PD-L1 agents plus antiangiogenic agents, the relative benefit and antitumor mechanism of antiangiogenic multikinase inhibitors versus specific VEGF/VEGFR inhibitors are discussed. Other critical issues in the development of combination immunotherapy, including optimal management of immune-related adverse events and the value of ICI therapy in combination with locoregional treatment for HCC, are also explored.
Collapse
Affiliation(s)
- Ann-Lii Cheng
- National Taiwan University Cancer Center, Taipei, Taiwan; Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chiun Hsu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan; Center of Precision Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Stephen L Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Institute of Digestive Disease, The Chinese University of Hong Kong, China
| | | | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
642
|
Huyghe N, Baldin P, Van den Eynde M. Immunotherapy with immune checkpoint inhibitors in colorectal cancer: what is the future beyond deficient mismatch-repair tumours? Gastroenterol Rep (Oxf) 2020; 8:11-24. [PMID: 32104582 PMCID: PMC7034232 DOI: 10.1093/gastro/goz061] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/25/2019] [Accepted: 10/22/2019] [Indexed: 12/17/2022] Open
Abstract
Following initial success in melanoma and lung tumours, immune checkpoint inhibitors (ICIs) are now well recognized as a major immunotherapy treatment modality for multiple types of solid cancers. In colorectal cancer (CRC), the small subset that is mismatch-repair-deficient and microsatellite-instability-high (dMMR/MSI-H) derive benefit from immunotherapy; however, the vast majority of patients with proficient MMR (pMMR) or with microsatellite stable (MSS) CRC do not. Immunoscore and the consensus molecular subtype classifications are promising biomarkers in predicting therapeutic efficacy in selected CRC. In pMRR/MSS CRC, biomarkers are also needed to understand the molecular mechanisms governing immune reactivity and to predict their relationship to treatment. The continuous development of such biomarkers would offer new perspectives and more personalized treatments by targeting oncological options, including ICIs, which modify the tumour-immune microenvironment. In this review, we focus on CRC and discuss the current status of ICIs, the role of biomarkers to predict response to immunotherapy, and the approaches being explored to render pMMR/MSS CRC more immunogenic through the use of combined therapies.
Collapse
Affiliation(s)
- Nicolas Huyghe
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, Brussels, Belgium
| | - Paméla Baldin
- Department of Pathology, Cliniques Universitaires St-Luc, Institut Roi Albert II, Brussels, Belgium
| | - Marc Van den Eynde
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, Brussels, Belgium
- Department of Medical Oncology, Cliniques Universitaires St-Luc, Institut Roi Albert II, Brussels, Belgium
| |
Collapse
|
643
|
Banias L, Jung I, Bara T, Fulop Z, Simu P, Simu I, Satala C, Gurzu S. Immunohistochemical-based molecular subtyping of colorectal carcinoma using maspin and markers of epithelial-mesenchymal transition. Oncol Lett 2020; 19:1487-1495. [PMID: 31966075 PMCID: PMC6956164 DOI: 10.3892/ol.2019.11228] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023] Open
Abstract
The aim of the present study was to classify colorectal carcinoma (CRC) into molecular subtypes, based on immunohistochemical (IHC) assessments. A total of 112 CRC samples were molecularly classified based on the expression levels of epithelial-mesenchymal transition (EMT)-associated IHC markers. A total of three molecular subtypes were defined: Epithelial, membrane positivity for E-cadherin and β-catenin, negative for vimentin; mesenchymal, E-cadherin-negative, nuclear β-catenin- and vimentin-positive; and hybrid cases, epithelial tumor core and mesenchymal tumor buds. Most of the cases were diagnosed as moderately differentiated adenocarcinoma (n=89; 79.46%). The majority of cases (n=100; 89.28%) exhibited a mismatch repair proficient status (microsatellite stable CRCs). A predominance of epithelial-type (n=51; 45.54%) and hybrid CRCs (n=47; 41.96%) was observed, whereas a few cases (n=14; 12.50%) were classified as mesenchymal-type CRCs. This molecular classification was associated with pathological stage (P<0.01), pT stage (P=0.04), pN stage (P<0.01), the grade of tumor budding (P=0.04), and maspin expression in both the tumor core (P=0.04) and the invasion front (P<0.01). The mesenchymal-type cases predominantly exhibited lymph node metastases, high-grade budding and a tendency towards maspin nuclear predominance. All epithelial-type cases with maspin-only expression (n=18) were non-metastatic. Patients with CRC of the epithelial subtype and those with a lymph node ratio (LNR) ≤0.15 presented the best overall survival, followed by those with hybrid and mesenchymal subtypes. Nuclear maspin positivity was more frequent in cases with a high-budding degree compared with those with a low-budding degree (P=0.03). The EMT-associated molecular classification of CRCs may be used to identify the most aggressive CRCs, which show a mesenchymal phenotype, high-budding degree, maspin nuclear positivity and lymph node metastases. The pN stage, LNR and budding degree of patients, which can be evaluated with maspin expression, remain the most important prognostic factors.
Collapse
Affiliation(s)
- Laura Banias
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| | - Ioan Jung
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| | - Tivadar Bara
- Department of Surgery, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| | - Zsolt Fulop
- Department of Surgery, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| | - Patricia Simu
- Department of Radiology, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| | - Iunius Simu
- Department of Radiology, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| | - Catalin Satala
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| | - Simona Gurzu
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
- Research Center (CCAMF)-Microscopy Laboratory, University of Medicine, Pharmacy, Sciences and Technology, ‘George Emil Palade’, 540139 Targu-Mures, Romania
| |
Collapse
|
644
|
Fanelli GN, Dal Pozzo CA, Depetris I, Schirripa M, Brignola S, Biason P, Balistreri M, Dal Santo L, Lonardi S, Munari G, Loupakis F, Fassan M. The heterogeneous clinical and pathological landscapes of metastatic Braf-mutated colorectal cancer. Cancer Cell Int 2020; 20:30. [PMID: 32015690 PMCID: PMC6990491 DOI: 10.1186/s12935-020-1117-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/20/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a complex and molecularly heterogeneous disease representing one of the most frequent causes of cancer-related death worldwide. About 8-15% of CRCs harbor a mutation in BRAF gene, a proto-oncogene involved in cell proliferation, differentiation and survival through the MAPK signaling cascade. The acquisition of BRAF mutation is an early event in the "serrated" CRC carcinogenetic pathway and is associated with specific and aggressive clinico-pathological and molecular features. Despite that the presence of BRAF mutation is a well-recognized negative prognostic biomarker in metastatic CRC (mCRC), a great heterogeneity in survival outcome characterizes these patients, due to the complex, and still not completely fully elucidated, interactions between the clinical, genetic and epigenetic landscape of BRAF mutations. Because of the great aggressiveness of BRAF-mutated mCRCs, only 60% of patients can receive a second-line chemotherapy; so intensive combined and tailored first-line approach could be a potentially effective strategy, but to minimize the selective pressure of resistant clones and to reduce side effects, a better stratification of patients bearing BRAF mutations is needed.
Collapse
Affiliation(s)
- Giuseppe Nicolò Fanelli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121 Padua, Italy
| | - Carlo Alberto Dal Pozzo
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121 Padua, Italy
| | - Ilaria Depetris
- Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Marta Schirripa
- Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Stefano Brignola
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121 Padua, Italy
| | - Paola Biason
- Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Mariangela Balistreri
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121 Padua, Italy
| | - Luca Dal Santo
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121 Padua, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Giada Munari
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121 Padua, Italy
- Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Fotios Loupakis
- Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, via Gabelli 61, 35121 Padua, Italy
| |
Collapse
|
645
|
Bazzichetto C, Conciatori F, Luchini C, Simionato F, Santoro R, Vaccaro V, Corbo V, Falcone I, Ferretti G, Cognetti F, Melisi D, Scarpa A, Ciuffreda L, Milella M. From Genetic Alterations to Tumor Microenvironment: The Ariadne's String in Pancreatic Cancer. Cells 2020; 9:309. [PMID: 32012917 PMCID: PMC7072496 DOI: 10.3390/cells9020309] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/18/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
The threatening notoriety of pancreatic cancer mainly arises from its negligible early diagnosis, highly aggressive progression, failure of conventional therapeutic options and consequent very poor prognosis. The most important driver genes of pancreatic cancer are the oncogene KRAS and the tumor suppressors TP53, CDKN2A, and SMAD4. Although the presence of few drivers, several signaling pathways are involved in the oncogenesis of this cancer type, some of them with promising targets for precision oncology. Pancreatic cancer is recognized as one of immunosuppressive phenotype cancer: it is characterized by a fibrotic-desmoplastic stroma, in which there is an intensive cross-talk between several cellular (e.g., fibroblasts, myeloid cells, lymphocytes, endothelial, and myeloid cells) and acellular (collagen, fibronectin, and soluble factors) components. In this review; we aim to describe the current knowledge of the genetic/biological landscape of pancreatic cancer and the composition of its tumor microenvironment; in order to better direct in the intrinsic labyrinth of this complex tumor type. Indeed; disentangling the genetic and molecular characteristics of cancer cells and the environment in which they evolve may represent the crucial step towards more effective therapeutic strategies.
Collapse
Affiliation(s)
- Chiara Bazzichetto
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Fabiana Conciatori
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Francesca Simionato
- Division of Oncology, University of Verona, 37126 Verona, Italy; (F.S.); (M.M.)
| | - Raffaela Santoro
- Medicine-Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37126 Verona, Italy; (R.S.); (D.M.)
| | - Vanja Vaccaro
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Vincenzo Corbo
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37126 Verona, Italy; (V.C.); (A.S.)
| | - Italia Falcone
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Gianluigi Ferretti
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Francesco Cognetti
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Davide Melisi
- Medicine-Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37126 Verona, Italy; (R.S.); (D.M.)
| | - Aldo Scarpa
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37126 Verona, Italy; (V.C.); (A.S.)
| | - Ludovica Ciuffreda
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Michele Milella
- Division of Oncology, University of Verona, 37126 Verona, Italy; (F.S.); (M.M.)
| |
Collapse
|
646
|
Cedrés S, Ponce-Aix S, Iranzo P, Callejo A, Pardo N, Navarro A, Martinez-Marti A, Gómez-Abecia S, Zucchiatti AC, Sansano I, Enguita AB, Miquel JM, Viaplana C, Dienstmann R, Paz-Ares L, Felip E. Analysis of mismatch repair (MMR) proteins expression in a series of malignant pleural mesothelioma (MPM) patients. Clin Transl Oncol 2020; 22:1390-1398. [PMID: 31916017 DOI: 10.1007/s12094-019-02275-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/16/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Promising results have been reported with immune checkpoint inhibitors (ICI) in a small proportion of MPM patients. MMR deficiency (dMMR) has been well described in several malignancies and was approved as a biomarker for anti-PD-1 inhibitors. Next generation sequencing (NGS) data demonstrated that 2% of MPM harbor microsatellite instability. The aim of this study is to characterize MMR by immunohistochemistry (IHC) in a series of MPM including a subset of patients treated with immunotherapy. METHODS Tumors of 159 MPM p diagnosed between 2002 and 2017 were reviewed. Formalin-fixed, paraffin-embedded tissue was stained for MLH1, MSH2, MSH6 and PMS2 and tumors were classified as dMMR (MMR protein expression negative) and MMR intact (all MMR proteins positively expressed). We retrospectively collected clinical outcomes under standard chemotherapy and experimental immunotherapy in the entire cohort. RESULTS MMR protein expression was analyzed in 158 samples with enough tissue and was positive in all of the cases. Twenty two patients received ICI with anti-CTLA4 or anti-PD-1 blockade in clinical trials, 58% had a response or stable disease for more than 6 m, with median progression-free survival (PFS) of 5.7 m (2.1-26.1 m). The median overall survival (mOS) in all population was 15 months (m) (13.5-18.8 m). In a multivariable model factors associated to improved mOS were PS 0, neutrophil-lymphocyte ratio (NLR) < 5 and epithelioid histology (p < 0.001). CONCLUSIONS In our series we were unable to identify any MPM patient with dMMR by IHC. Further studies are needed to elucidate potential predictive biomarkers of ICI benefit in MPM.
Collapse
Affiliation(s)
- S Cedrés
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain.
- Servicio de Oncología, Hospital Vall D´Hebron, Paseo Vall d´Hebron 119-129, 08035, Barcelona, Spain.
| | - S Ponce-Aix
- CIBERONC, Madrid, Spain
- Medical Oncology Department, University Hospital Doce de Octubre, Madrid, Spain
| | - P Iranzo
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain
| | - A Callejo
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain
| | - N Pardo
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain
| | - A Navarro
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain
| | - A Martinez-Marti
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain
| | | | - A C Zucchiatti
- Pathology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - I Sansano
- Pathological Anatomy Department, University Hospital Doce de Octubre, Madrid, Spain
| | - A B Enguita
- Pathological Anatomy Department, University Hospital Doce de Octubre, Madrid, Spain
| | - J M Miquel
- Vall d´Hebron Institute of Oncology, Barcelona, Spain
| | - C Viaplana
- Oncology Data Science (ODysSey) Group, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - R Dienstmann
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain
- Oncology Data Science (ODysSey) Group, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - L Paz-Ares
- CIBERONC, Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Biomedical Research Foundation I+12, Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Medical School, Complutense University, Madrid, Spain
| | - E Felip
- Medical Oncology Department, Vall d´Hebron University Hospital and Institute of Oncology, Barcelona, Spain
| |
Collapse
|
647
|
Microsatellite instability in Japanese female patients with triple-negative breast cancer. Breast Cancer 2020; 27:490-498. [PMID: 31907878 PMCID: PMC7196096 DOI: 10.1007/s12282-019-01043-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/25/2019] [Indexed: 01/11/2023]
Abstract
Background It is important to identify biomarkers for triple-negative breast cancers (TNBCs). Recently, pembrolizumab, an immune checkpoint inhibitor (ICI) for programmed cell death 1 (PD-1), was approved as a treatment strategy for unresectable or metastatic tumor with high-frequency microsatellite instability (MSI-H) or mismatch repair deficiency, such as malignant melanoma, non-small cell lung cancer, renal cell cancer and urothelial cancer. In addition, results from clinical trials suggested that ICI was a promising treatment for TNBCs with accumulated mutations. However, the frequency of MSI in Japanese TNBCs still remains unclear. We aimed to analyze the presence of MSI-H in TNBCs as a biomarker for ICI therapy. Methods In this study, we retrospectively evaluated the MSI of 228 TNBCs using an innovative method, MSI Analysis System Version 1.2 (Promega), consisting of 5 microsatellite markers: BAT-26, NR-21, BAT-25, MONO-27 and NR-24 without a normal tissue control. Results Among 228 tumors, 222 (97.4%) were microsatellite stable, 4 (1.7%) low-frequency MSI and 2 (0.9%) MSI-H, respectively. Two MSI-H tumors were potentially aggressive pathologically as indicated by nuclear grade 3 and high Ki-67 (> 30%), and were classified as basal-like and non-BRCA-like, but were not consistent regarding tumor-infiltrating lymphocytes, CD8 and PD-L1 expression. Conclusions Although we found that MSI-H was uncommon (0.9%) in TNBCs, potential targets for ICIs exist in TNBCs. Therefore, MSI-H breast cancer patients should be picked up using not only conventional methods but also platforms for comprehensive genomic profiling.
Collapse
|
648
|
Tuo Z, Zheng X, Zong Y, Li J, Zou C, Lv Y, Liu J. HK3 is correlated with immune infiltrates and predicts response to immunotherapy in non-small cell lung cancer. Clin Transl Med 2020; 10:319-330. [PMID: 32508023 PMCID: PMC7240846 DOI: 10.1002/ctm2.6] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/01/2020] [Accepted: 03/01/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND With the knowledge of tumor immunobiology deepening among researchers, the breakthroughs in the field of tumor immunotherapy in recent years have provided new approaches for cancer therapy. While patients who receive treatment are all at risk of side effects, about one-fifth of them have sustained responses. It is crucial to figure out the underlying mechanism of how the immune system regulates the nonsmall cell lung cancer (NSCLC) microenvironment to improve the benefit of immunotherapy. Regarding glucose metabolism, the initial step is to generate glucose-6-phosphate by phosphorylating glucose with hexokinases-3 (HK3). According to a recent study, HK3 has a functional role in the treatment of acute promyelocytic leukemia and colorectal cancer. RESULTS Here, we studied the co-expression relationship between the glycolytic pathway gene and the immune checkpoint gene and found that the expression of HK3 in tumor tissues may be related to immune status. By analyzing The Cancer Genome Atlas (TCGA) data, we found that the expression of HK3 was closely related to the main clinical features as well as to molecular characteristics. We also predicted that cases with low expression of HK3 were usually malignant entities and were shown to be obvious genomic aberrations of driver oncogenes. At the same time, gene ontology analysis based on significantly related genes in HK3 expression showed that HK3 expression was linked to inflammatory activity and immune response. Additionally, HK3 showed a remarkable trend in predicting the efficacy of immunotherapy for patients receiving Keytruda (PD-1 monoclonal antibody) treatment. CONCLUSIONS This is the first comprehensive study to characterize HK3 expression in NSCLC from molecular and clinical aspects.
Collapse
Affiliation(s)
- Zhan Tuo
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xin Zheng
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yan Zong
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jie Li
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chunyan Zou
- The People's Hospital of Guangxi Zhuang Autonomous Region NanningGuangxi530021China
| | - Yi Lv
- Department of Pharmacy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Jun Liu
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
649
|
Kang J, Jeong JH, Hwang HS, Lee SS, Park DH, Oh DW, Song TJ, Kim KH, Hwang S, Hwang DW, Kim SC, Park JH, Hong SM, Kim KP, Ryoo BY, Yoo C. Efficacy and Safety of Pembrolizumab in Patients with Refractory Advanced Biliary Tract Cancer: Tumor Proportion Score as a Potential Biomarker for Response. Cancer Res Treat 2019; 52:594-603. [PMID: 32019287 PMCID: PMC7176957 DOI: 10.4143/crt.2019.493] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Purpose The current standard chemotherapy for advanced biliary tract cancer (BTC) has limited benefit, and novel therapies need to be investigated. Materials and Methods In this prospective cohort study, programmed death ligand-1 (PD-L1)–positive BTC patients who progressed on first-line gemcitabine plus cisplatin were enrolled. Pembrolizumab 200 mg was administered intravenously every 3 weeks. Results Between May 2018 and February 2019, 40 patients were enrolled. Pembrolizumab was given as second-line (47.5%) or ≥ third-line therapy (52.5%). The objective response rate was 10% and 12.5% by Response Evaluation Criteria in Solid Tumor (RECIST) v1.1 and immune- modified RECIST (imRECIST) and median duration of response was 6.3 months. Among patients with progressive disease as best response, one patient (1/20, 5.0%) achieved complete response subsequently. The median progression-free survival (PFS) and overall survival (OS) were 1.5 months (95% confidence interval [CI], 0.0 to 3.0) and 4.3 months (95% CI, 3.5 to 5.1), respectively, and objective response per imRECIST was significantly associated with PFS (p < 0.001) and OS (p=0.001). Tumor proportion score ≥ 50% was significantly associated with higher response rates including the response after pseudoprogression (vs. < 50%; 37.5% vs. 6.5%; p=0.049). Conclusion Pembrolizumab showed modest anti-tumor activity in heavily pretreated PD-L1–positive BTC patients. In patients who showed objective response, durable response could be achieved.
Collapse
Affiliation(s)
- Junho Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hee-Sang Hwang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Soo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Do Hyun Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong Wook Oh
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki-Hun Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shin Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Wook Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Song Cheol Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin-Hong Park
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
650
|
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: 1.3] [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
|