1
|
Ciardiello D, Boscolo Bielo L, Napolitano S, Martinelli E, Troiani T, Nicastro A, Latiano TP, Parente P, Maiello E, Avallone A, Normanno N, Pisconti S, Nisi C, Bordonaro R, Russo AE, Tamburini E, Toma I, Lotesoriere C, Vallarelli S, Zampino MG, Fazio N, Curigliano G, De Vita F, Ciardiello F, Martini G. Comprehensive genomic profiling by liquid biopsy captures tumor heterogeneity and identifies cancer vulnerabilities in patients with RAS/BRAF V600E wild-type metastatic colorectal cancer in the CAPRI 2-GOIM trial. Ann Oncol 2024:S0923-7534(24)03914-0. [PMID: 39214459 DOI: 10.1016/j.annonc.2024.08.2334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Emerging evidence supports tumor tissue-based comprehensive genomic profiling (CGP) in metastatic colorectal cancer (mCRC). Data on liquid biopsy-based circulating tumor DNA (ctDNA) CGP are scarce and mainly retrospective. Prospective comparison between the two tests is not currently available. MATERIALS AND METHODS The CAPRI 2-GOIM trial investigates efficacy and safety of ctDNA-driven, cetuximab-based sequence of three treatment lines in patients with RAS/BRAFV600E wild-type (WT) mCRC, as determined by the local laboratory. Before first-line therapy, CGP is carried out with FoundationOne (F1) CDx and F1 Liquid (F1L) CDx (324 genes) on tumor tissue DNA and plasma ctDNA, respectively. RESULTS For 2/207 (0.96%) patients, no ctDNA was detected by F1L CDx. No patient displayed tumor fraction (TF) below 1%, whereas elevated ctDNA (TF ≥ 10%) was detected among 140/205 (68.3%) patients. One thousand and thirteen genomic variants were identified. F1L CDx found KRAS, NRAS, or BRAFV600E alterations in 19 patients, whose tumors were classified as RAS/BRAFV600E WT by the local laboratory. Both F1 CDx and F1L CDx were available for 164/205 (80%) patients. A concordance of 61.4% between the two tests was observed. The concordance increased to 72.7% for F1L CDx with TF ≥ 10%. Concordance for genes potentially involved in anti-epidermal growth factor receptor resistance was found in 137/164 (83%) patients, increasing to 91.5% for F1L CDx with TF ≥ 10%. A higher number of genomic alterations was detected by F1L CDx compared with F1 CDx, including six cases with KRAS and NRAS alterations. Overall, 109/205 (53.2%) patients displayed at least one actionable genomic alteration (I to IIIB), according to the European Society for Medical Oncology Scale for Clinical Actionability of Molecular Targets (ESCAT). CONCLUSION Baseline liquid biopsy-based CGP is feasible, has high concordance with tumor tissue-based CGP, could better recapitulate tumor heterogeneity, and is clinically informative by identifying additional actionable genomic alterations in approximately half of RAS/BRAFV600E WT mCRC patients.
Collapse
Affiliation(s)
- D Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan
| | - L Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milan, Milan
| | - S Napolitano
- Department of Precision Medicine, The University of Campania Luigi Vanvitelli, Naples
| | - E Martinelli
- Department of Precision Medicine, The University of Campania Luigi Vanvitelli, Naples
| | - T Troiani
- Department of Precision Medicine, The University of Campania Luigi Vanvitelli, Naples
| | - A Nicastro
- Department of Precision Medicine, The University of Campania Luigi Vanvitelli, Naples
| | - T P Latiano
- Medical Oncology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo
| | - P Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo
| | - E Maiello
- Medical Oncology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo
| | - A Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples
| | - N Normanno
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Mendola
| | - S Pisconti
- Medical Oncology Unit, San Giuseppe Moscati Hospital, Statte
| | - C Nisi
- Medical Oncology Unit, San Giuseppe Moscati Hospital, Statte
| | - R Bordonaro
- Medical Oncology Unit, Azienda Ospedaliera ARNAS Garibaldi, Catania
| | - A E Russo
- Medical Oncology Unit, Azienda Ospedaliera ARNAS Garibaldi, Catania
| | - E Tamburini
- Department of Oncology and Palliative Care, Cardinale G Panico, Tricase City Hospital, Tricase
| | - I Toma
- Department of Oncology and Palliative Care, Cardinale G Panico, Tricase City Hospital, Tricase
| | - C Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS de Bellis Research Hospital, Castellana Grotte, Italy
| | - S Vallarelli
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS de Bellis Research Hospital, Castellana Grotte, Italy
| | - M G Zampino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan
| | - N Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan
| | - G Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milan, Milan
| | - F De Vita
- Department of Precision Medicine, The University of Campania Luigi Vanvitelli, Naples
| | - F Ciardiello
- Department of Precision Medicine, The University of Campania Luigi Vanvitelli, Naples.
| | - G Martini
- Department of Precision Medicine, The University of Campania Luigi Vanvitelli, Naples
| |
Collapse
|
2
|
Duenas-Gonzalez A, Gonzalez-Fierro A, Bornstein-Quevedo L, Gutierrez-Delgado F, Kast RE, Chavez-Blanco A, Dominguez-Gomez G, Candelaria M, Romo-Pérez A, Correa-Basurto J, Lizano M, Perez-de la Cruz V, Robles-Bañuelos B, Nuñez-Corona D, Martinez-Perez E, Verastegui E. Multitargeted polypharmacotherapy for cancer treatment. theoretical concepts and proposals. Expert Rev Anticancer Ther 2024; 24:665-677. [PMID: 38913911 DOI: 10.1080/14737140.2024.2372336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION The pharmacological treatment of cancer has evolved from cytotoxic to molecular targeted therapy. The median survival gains of 124 drugs approved by the FDA from 2003 to 2021 is 2.8 months. Targeted therapy is based on the somatic mutation theory, which has some paradoxes and limitations. While efforts of targeted therapy must continue, we must study newer approaches that could advance therapy and affordability for patients. AREAS COVERED This work briefly overviews how cancer therapy has evolved from cytotoxic chemotherapy to current molecular-targeted therapy. The limitations of the one-target, one-drug approach considering cancer as a robust system and the basis for multitargeting approach with polypharmacotherapy using repurposing drugs. EXPERT OPINION Multitargeted polypharmacotherapy for cancer with repurposed drugs should be systematically investigated in preclinical and clinical studies. Remarkably, most of these proposed drugs already have a long history in the clinical setting, and their safety is known. In principle, the risk of their simultaneous administration should not be greater than that of a first-in-human phase I study as long as the protocol is developed with strict vigilance to detect early possible side effects from their potential interactions. Research on cancer therapy should go beyond the prevailing paradigm targeted therapy.
Collapse
Affiliation(s)
- Alfonso Duenas-Gonzalez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas UNAM, Mexico City, Mexico
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Aurora Gonzalez-Fierro
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Francisco Gutierrez-Delgado
- Centro de Estudios y Prevención del Cancer Tuxtla Gutiérrez, Chiapas, México; Latin American School of Oncology (ELO), México City, Mexico
| | - Richard E Kast
- Head of Faculty, Brain Study, IIAIG Study Center, Burlington, VT, USA
| | - Alma Chavez-Blanco
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Myrna Candelaria
- Departamento de Hematología, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Adriana Romo-Pérez
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jose Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, SEPI-ESM, Instituto Politécnico Nacional, México, Mexico City, Mexico
| | - Marcela Lizano
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas UNAM, Mexico City, Mexico
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Veronica Perez-de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City, Mexico
| | | | - David Nuñez-Corona
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Erandi Martinez-Perez
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Emma Verastegui
- Departamento de Cuidados Paliativos, Division de Cirugia, Instituto Nacional de Cancerologia, Mexico City, Mexico
| |
Collapse
|
3
|
Liu B, Li S, Cheng Y, Song P, Xu M, Li Z, Shao W, Xin J, Fu Z, Gu D, Du M, Zhang Z, Wang M. Distinctive multicellular immunosuppressive hubs confer different intervention strategies for left- and right-sided colon cancers. Cell Rep Med 2024; 5:101589. [PMID: 38806057 PMCID: PMC11228667 DOI: 10.1016/j.xcrm.2024.101589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/11/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024]
Abstract
Primary colon cancers arising from the left and right sides exhibit distinct clinical and molecular characteristics. Sidedness-associated heterogeneity relies intricately on the oncogenic properties of cancer cells and multicellular interactions in tumor microenvironments. Here, combining transcriptomic profiling of 426,863 single cells from 105 colon cancer patients and validation with spatial transcriptomics and large-scale histological analysis, we capture common transcriptional heterogeneity patterns between left- and right-sided malignant epithelia through delineating two side-specific expression meta-programs. The proliferation stemness meta-program is notably enriched in left-sided malignant epithelia that colocalize with Mph-PLTP cells, activated regulatory T cells (Tregs), and exhausted CD8-LAYN cells, constituting the glucose metabolism reprogramming niche. The immune secretory (IS) meta-program exhibits specific enrichment in right-sided malignant epithelia, especially in smoking patients with right-sided colon cancer. The IShigh malignant epithelia spatially localize in hypoxic regions and facilitate immune evasion through attenuating Mph-SPP1 cell antigen presentation and recruiting innate-like cytotoxicity-reduced CD8-CD161 cells.
Collapse
Affiliation(s)
- Bingxin Liu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shuwei Li
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yifei Cheng
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Peng Song
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Menghuan Xu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhengyi Li
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wei Shao
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Junyi Xin
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zan Fu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Dongying Gu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Mulong Du
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.
| |
Collapse
|
4
|
Li XJ, Nie P, Herdewijn P, Sun JG. Unlocking the synthetic approaches and clinical application of approved small-molecule drugs for gastrointestinal cancer treatment: A comprehensive exploration. Eur J Med Chem 2023; 262:115928. [PMID: 37944387 DOI: 10.1016/j.ejmech.2023.115928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Gastrointestinal (GI) cancers encompass a group of malignancies affecting the digestive system, including the stomach, esophagus, liver, colon, rectum and pancreas. These cancers represent a significant global health burden, necessitating effective treatment strategies. Small-molecule drugs have emerged as crucial therapeutic options in the fight against GI cancers due to their oral bioavailability, targeted mechanisms of action, and well-established safety profiles. The review then elucidates the clinical applications and synthetic methods of clinically approved small-molecule drugs for the treatment of GI cancer, shedding light on their mechanisms of action and their potential in mitigating GI cancer progression. The review also discusses future prospects and the evolving landscape of small-molecule drug development in GI oncology, highlighting the potential for personalized medicine. In summary, this review provides valuable insights into cutting-edge strategies for harnessing clinically approved small-molecule drugs to combat GI cancer effectively.
Collapse
Affiliation(s)
- Xiao-Jing Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Peng Nie
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Piet Herdewijn
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Jian-Gang Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
5
|
Ottaiano A, De Luca A, Santorsola M, Scognamiglio G, Di Mauro A, Chiodini P, Lambiase M, Sacco A, Petrillo A, Granata V, Fusco R, Mercadante E, Martucci N, De Luca G, Rocca AL, Celentano E, Crispo A, Di Gennaro P, Tatangelo F, Ferrara G, Izzo F, Belli A, Patrone R, Delrio P, Rega D, De Franciscis S, Muto P, Ravo V, Di Franco R, Borzillo V, Santagata S, Rea G, Castaldo D, Pace U, De Feo G, Scala S, Nasti G, Normanno N. Oligo-metastatic neoPlasms from the gastro-intestinal tract: iDentIfiCaTIon of cliNical and molecular drivers: the PREDICTION study. BMC Cancer 2023; 23:1010. [PMID: 37858132 PMCID: PMC10588113 DOI: 10.1186/s12885-023-11479-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Metastatic disease in tumors originating from the gastrointestinal tract can exhibit varying degrees of tumor burden at presentation. Some patients follow a less aggressive disease course, characterized by a limited number of metastatic sites, referred to as "oligo-metastatic disease" (OMD). The precise biological characteristics that define the oligometastatic behavior remain uncertain. In this study, we present a protocol designed to prospectively identify OMD, with the aim of proposing novel therapeutic approaches and monitoring strategies. METHODS The PREDICTION study is a monocentric, prospective, observational investigation. Enrolled patients will receive standard treatment, while translational activities will involve analysis of the tumor microenvironment and genomic profiling using immunohistochemistry and next-generation sequencing, respectively. The first primary objective (descriptive) is to determine the prevalence of biological characteristics in OMD derived from gastrointestinal tract neoplasms, including high genetic concordance between primary tumors and metastases, a significant infiltration of T lymphocytes, and the absence of clonal evolution favoring specific driver genes (KRAS and PIK3CA). The second co-primary objective (analytic) is to identify a prognostic score for true OMD, with a primary focus on metastatic colorectal cancer. The score will comprise genetic concordance (> 80%), high T-lymphocyte infiltration, and the absence of clonal evolution favoring driver genes. It is hypothesized that patients with true OMD (score 3+) will have a lower rate of progression/recurrence within one year (20%) compared to those with false OMD (80%). The endpoint of the co-primary objective is the rate of recurrence/progression at one year. Considering a reasonable probability (60%) of the three factors occurring simultaneously in true OMD (score 3+), using a significance level of α = 0.05 and a test power of 90%, the study requires a minimum enrollment of 32 patients. DISCUSSION Few studies have explored the precise genetic and biological features of OMD thus far. In clinical settings, the diagnosis of OMD is typically made retrospectively, as some patients who undergo intensive treatment for oligometastases develop polymetastatic diseases within a year, while others do not experience disease progression (true OMD). In the coming years, the identification of true OMD will allow us to employ more personalized and comprehensive strategies in cancer treatment. TRIAL REGISTRATION ClinicalTrials.gov ID NCT05806151.
Collapse
Grants
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
- L4/8 Italian Government, Ministry of Health, Ricerca Corrente 2022
Collapse
Affiliation(s)
| | | | | | | | | | - Paolo Chiodini
- Section of Statistics, Department of Mental Health and Public Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, 80138, Italy
| | - Matilde Lambiase
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Alessandra Sacco
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | | | - Vincenza Granata
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Roberta Fusco
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | | | - Nicola Martucci
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Giuseppe De Luca
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | | | - Egidio Celentano
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Anna Crispo
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | | | | | - Gerardo Ferrara
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Francesco Izzo
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Andrea Belli
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Renato Patrone
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Paolo Delrio
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Daniela Rega
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | | | - Paolo Muto
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Vincenzo Ravo
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | | | | | - Sara Santagata
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Giuseppina Rea
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Daniela Castaldo
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Ugo Pace
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | | | - Stefania Scala
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Guglielmo Nasti
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| | - Nicola Normanno
- Istituto Nazionale Tumori, IRCCS "G. Pascale", Napoli, 80131, Italy
| |
Collapse
|
6
|
Ham-Karim H, Negm O, Ahmad N, Ilyas M. Investigating genomic, proteomic, and post-transcriptional regulation profiles in colorectal cancer: a comparative study between primary tumors and associated metastases. Cancer Cell Int 2023; 23:192. [PMID: 37670299 PMCID: PMC10478430 DOI: 10.1186/s12935-023-03020-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/06/2023] [Indexed: 09/07/2023] Open
Abstract
INTRODUCTION Approximately 50% of patients with primary colorectal carcinoma develop liver metastases. This study investigates the possible molecular discrepancies between primary colorectal cancer (pCRC) and their respective metastases. METHODS A total of 22 pairs of pCRC and metastases were tested. Mutation profiling of 26 cancer-associated genes was undertaken in 22/22primary-metastasis tumour pairs using next-generation sequencing, whilst the expression of a panel of six microRNAs (miRNAs) was investigated using qPCRin 21/22 pairs and 22 protein biomarkers was tested using Reverse Phase Protein Array (RPPA)in 20/22 patients' tumour pairs. RESULTS Among the primary and metastatic tumours the mutation rates for the individual genes are as follows:TP53 (86%), APC (44%), KRAS (36%), PIK3CA (9%), SMAD4 (9%), NRAS (9%) and 4% for FBXW7, BRAF, GNAS and CDH1. The primary-metastasis tumour mutation status was identical in 54/60 (90%) loci. However, there was discordance in heterogeneity status in 40/58 genetic loci (z-score = 6.246, difference = 0.3793, P < 0.0001). Furthermore, there was loss of concordance in miRNA expression status between primary and metastatic tumours, and 57.14-80.95% of the primary-metastases tumour pairs showed altered primary-metastasis relative expression in all the miRNAs tested. Moreover, 16 of 20 (80%) tumour pairs showed alteration in at least 3 of 6 (50%) of the protein biomarker pathways analysed. CONCLUSION The molecular alterations of primary colorectal tumours differ significantly from those of their matched metastases. These differences have profound implications for patients' prognoses and response to therapy.
Collapse
Affiliation(s)
- Hersh Ham-Karim
- Department of Pharmacy, College of Medicine, Komar University of Science and Technology, Chaq-Chaq-Qualaraisi, Sulaimani, Iraq.
| | - Ola Negm
- Division of Medical Sciences and Graduate Entry Medicine, Faculty of Medicine and Health Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Narmeen Ahmad
- Kurdistan Institution for Strategic Studies and Scientific Research, Qirga, Sulaimani, KRG, Iraq
| | - Mohammad Ilyas
- Department of Pharmacy, College of Medicine, Komar University of Science and Technology, Chaq-Chaq-Qualaraisi, Sulaimani, Iraq
- Nottingham Molecular Pathology Node, University of Nottingham, Nottingham, UK
| |
Collapse
|
7
|
Rao W, Liu Y, Li Y, Guo L, Qiu T, Dong L, Ying J, Li W. Potential unreliability of ALK variant allele frequency in the efficacy prediction of targeted therapy in NSCLC. Front Med 2023; 17:493-502. [PMID: 37010729 DOI: 10.1007/s11684-022-0946-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/28/2022] [Indexed: 04/04/2023]
Abstract
Anaplastic lymphoma kinase (ALK) is the most common fusion gene involved in non-small cell lung cancer (NSCLC), and remarkable response has been achieved with the use of ALK tyrosine kinase inhibitors (ALK-TKIs). However, the clinical efficacy is highly variable. Pre-existing intratumoral heterogeneity (ITH) has been proven to contribute to the poor treatment response and the resistance to targeted therapies. In this work, we investigated whether the variant allele frequencies (VAFs) of ALK fusions can help assess ITH and predict targeted therapy efficacy. Through the application of next-generation sequencing (NGS), 7.2% (326/4548) of patients were detected to be ALK positive. On the basis of the adjusted VAF (adjVAF, VAF normalization for tumor purity) of four different threshold values (adjVAF < 50%, 40%, 30%, or 20%), the association of ALK subclonality with crizotinib efficacy was assessed. Nonetheless, no statistical association was observed between median progression-free survival (PFS) and ALK subclonality assessed by adjVAF, and a poor correlation of adjVAF with PFS was found among the 85 patients who received first-line crizotinib. Results suggest that the ALK VAF determined by hybrid capture-based NGS is probably unreliable for ITH assessment and targeted therapy efficacy prediction in NSCLC.
Collapse
Affiliation(s)
- Wei Rao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
Fleming AM, Deschner BW, Williard FW, Drake JA, Vanderwalde A, Xiu J, Somer BG, Yakoub D, Tsao MW, Glazer ES, Dickson PV, Shibata D, Philip PA, Hwang JJ, Shields AF, Marshall JL, Korn WM, Lenz HJ, Deneve JL. Peritoneal metastases from primary appendiceal and colorectal carcinomas demonstrate distinct molecular identities on comprehensive tumor analysis. J Surg Oncol 2023; 127:815-822. [PMID: 36629137 DOI: 10.1002/jso.27198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/08/2022] [Accepted: 01/01/2023] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Published data comparing peritoneal metastases from appendiceal cancers (pAC) and colorectal cancers (pCRC) remain sparse. We compared pAC and pCRC using comprehensive tumor profiling (CTP). METHODS CTP was performed, including next-generation sequencing and analysis of copy number variation (CNV), microsatellite instability (MSI) and tumor mutational burden (TMB). RESULTS One hundred thirty-six pAC and 348 pCRC samples underwent CTP. The cohorts' age and gender were similar. pCRC demonstrated increased pathogenic variants (PATHs) in APC (48% vs. 3%, p < 0.01), ARID1A (12% vs. 2%, p < 0.01), BRAF (12% vs. 2%, p < 0.01), FBXW7 (7% vs. 2%, p < 0.01), KRAS (52% vs. 41%, p < 0.05), PIK3CA (15% vs. 2%, p < 0.01), and TP53 (53% vs. 23%, p < 0.01), and decreased PATHs in GNAS (8% vs. 31%, p < 0.01). There was no difference in CNV, fusion rate, or MSI. Median TMB was higher in pCRC (5.8 vs. 5.0 mutations per megabase, p = 0.0007). Rates of TMB-high tumors were similar (pAC 2.1% vs. pCRC 9.0%, p = 0.1957). pCRC had significantly more TMB-high tumors at lower thresholds. CONCLUSIONS Despite a reduced overall TMB, pAC demonstrated mutations distinct from those seen in pCRC. These may serve as discrete biomarkers for future study.
Collapse
Affiliation(s)
- Andrew M Fleming
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Benjamin W Deschner
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Division of Surgical Oncology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Forrest W Williard
- College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Justin A Drake
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ari Vanderwalde
- West Cancer Center, Germantown, Tennessee, USA.,Caris Life Sciences, Irving, Texas, USA
| | | | | | - Danny Yakoub
- Department of Surgery, Mayo Clinic Health System, Eau Claire, Wisconsin, USA
| | - Miriam W Tsao
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Division of Surgical Oncology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Evan S Glazer
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Division of Surgical Oncology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Paxton V Dickson
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Division of Surgical Oncology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - David Shibata
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Division of Surgical Oncology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Jimmy J Hwang
- Division of Hematology/Oncology, Atrium Health Levine Cancer Institute, Charlotte, North Carolina, USA
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - John L Marshall
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - W Michael Korn
- Division of Hematology/Oncology, University of California, San Francisco, California, USA
| | - Heinz-Josef Lenz
- Keck School of Medicine, Norris Comprehensive Cancer Center, Division of Medical Oncology, University of Southern California, Los Angeles, California, USA
| | - Jeremiah L Deneve
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Division of Surgical Oncology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
9
|
Maag N, Arndt A, Steinestel K. [Laser microdissection for the analysis of molecular heterogeneity in colorectal cancer]. PATHOLOGIE (HEIDELBERG, GERMANY) 2022; 43:36-41. [PMID: 36255446 DOI: 10.1007/s00292-022-01131-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Oncogenic driver mutations in RAS/RAF oncogenes are frequent in colorectal cancer (CRC). The presence of different subclones within a single tumor can lead to treatment failure in anti-EGFR/epidermal growth factor receptor-directed antibody therapies. The identification of different subclones and their mutational profiles within a single tumor and the identification of morphologically distinct tumor areas might help to unravel novel aspects of tumor biology and therapy resistance. OBJECTIVES The aim of this study was to identify intratumoral heterogeneity in CRC by using laser microdissection (LMD) in comparison to the routinely used method. We hereby applied LMD to identify and investigate tumor heterogeneity in CRC. METHODS We established LMD and purified DNA from several morphologically distinct tumor areas (n = 13) in CRCs from 2 patients and compared the results from routine testing to our newly established LMD approach. LMD enabled the comparative analysis of small tumor areas by cutting histologically selected elements under microscopic control using a laser beam. RESULTS In some cases, potential low-level mutations (PLLM) could not be detected using the routine method since they were masked by high-level mutations (HLM). The application of LMD enabled the identification of concomitant PLLM in NRAS and BRAF genes in the identical patient sample. CONCLUSION LMD improved spatial resolution in the molecular analysis of CRC tumor tissue compared to routine methods. Our results confirmed the presence of molecular heterogeneity in CRC. This should be kept in mind when interpreting sequencing results, since low frequency mutations can have an impact on the effectiveness of targeted therapy.
Collapse
Affiliation(s)
- Nathalie Maag
- Institut für Pathologie und Molekularpathologie, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Deutschland.
| | - Annette Arndt
- Institut für Pathologie und Molekularpathologie, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Deutschland
| | - Konrad Steinestel
- Institut für Pathologie und Molekularpathologie, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Deutschland
| |
Collapse
|
10
|
van den Bosch T, Vermeulen L, Miedema DM. Quantitative models for the inference of intratumor heterogeneity. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2022. [DOI: 10.1002/cso2.1034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Tom van den Bosch
- Laboratory for Experimental Oncology and Radiobiology Center for Experimental and Molecular Medicine Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism Amsterdam University Medical Centers Amsterdam The Netherlands
- Oncode Institute Amsterdam The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology Center for Experimental and Molecular Medicine Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism Amsterdam University Medical Centers Amsterdam The Netherlands
- Oncode Institute Amsterdam The Netherlands
| | - Daniël M. Miedema
- Laboratory for Experimental Oncology and Radiobiology Center for Experimental and Molecular Medicine Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism Amsterdam University Medical Centers Amsterdam The Netherlands
- Oncode Institute Amsterdam The Netherlands
| |
Collapse
|
11
|
Boccaccino A, Borelli B, Intini R, Antista M, Bensi M, Rossini D, Passardi A, Tamberi S, Giampieri R, Antonuzzo L, Noto L, Roviello G, Zichi C, Salati M, Puccini A, Noto C, Parisi A, Rihawi K, Persano M, Crespi V, Libertini M, Giordano M, Moretto R, Lonardi S, Cremolini C. Encorafenib plus cetuximab with or without binimetinib in patients with BRAF V600E-mutated metastatic colorectal cancer: real-life data from an Italian multicenter experience. ESMO Open 2022; 7:100506. [PMID: 35696748 PMCID: PMC9271503 DOI: 10.1016/j.esmoop.2022.100506] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Encorafenib plus cetuximab with or without binimetinib showed increased objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) compared with chemotherapy plus anti-EGFR in previously treated patients with BRAF V600E-mutated (mut) metastatic colorectal cancer (mCRC). Although no formal comparison was planned, addition of binimetinib to encorafenib plus cetuximab did not provide significant efficacy advantage. PATIENTS AND METHODS This real-life study was aimed at evaluating safety, activity, and efficacy of encorafenib plus cetuximab with or without binimetinib in patients with BRAF V600E-mut mCRC treated at 21 Italian centers within a nominal use program launched in May 2019. RESULTS Out of 133 patients included, 97 (73%) received encorafenib plus cetuximab (targeted doublet) and 36 (27%) the same therapy plus binimetinib (targeted triplet). Most patients had Eastern Cooperative Group Performance Status (ECOG-PS) of 0 or 1 (86%), right-sided primary tumor (69%), and synchronous disease (66%). Twenty (15%) tumors were DNA mismatch repair deficiency (dMMR)/microsatellite instability (MSI)-high. As many as 44 (34%) patients had received two or more prior lines of therapy, 122 (92%) were previously exposed to oxaliplatin, and 109 (82%) to anti-vascular endothelial growth factor (anti-VEGF). Most frequent adverse events were asthenia (62%) and anti-EGFR-related skin rash (52%). Any grade nausea (P = 0.03), vomiting (P = 0.04), and diarrhea (P = 0.07) were more frequent with the triplet therapy, while melanocytic nevi were less common (P = 0.06). Overall, ORR and disease control rate (DCR) were 23% and 69%, respectively, with numerically higher rates in the triplet group (ORR 31% versus 17%, P = 0.12; DCR 78% versus 65%, P = 0.23). Median PFS and OS were 4.5 and 7.2 months, respectively. Worse ECOG-PS, peritoneal metastases, and more than one prior treatment were independent poor prognostic factors for PFS and OS. Clonality of BRAF mutation measured as adjusted mutant allele fraction in tumor tissue was not associated with clinical outcome. CONCLUSIONS Our real-life data are consistent with those from the BEACON trial in terms of safety, activity, and efficacy. Patients in good general condition and not heavily pretreated are those more likely to derive benefit from the targeted treatment.
Collapse
Affiliation(s)
- A Boccaccino
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - B Borelli
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - R Intini
- Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology IOV-IRCCSP, Padova, Italy
| | - M Antista
- Medical Oncology Department, ENETS Center of Excellence, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - M Bensi
- Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Università Cattolica del Sacro Cuore, Roma, Italy
| | - D Rossini
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - A Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - S Tamberi
- UOC Oncologia Ravenna, AUSL Romagna, Ravenna, Italy
| | - R Giampieri
- Clinica Oncologica, Dipartimento Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - L Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - L Noto
- UOC Oncologia Medica, Policlinico "G.Rodolico" AOU Policlinico - San Marco, Catania, Italy
| | - G Roviello
- Department of Health Sciences, University of Florence, Florence, Italy
| | - C Zichi
- Oncologia Medica, A.O. Ordine Mauriziano - Umberto I, Torino, Italy
| | - M Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy; PhD Program Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - A Puccini
- Università degli Studi di Genova, Ospedale Policlinico San Martino IRCCS, Genova, Italy
| | - C Noto
- Università degli Studi di Udine, Dipartimento di Area Medica, Udine, Italy; Azienda Sanitaria Universitaria Friuli Centrale, Dipartimento di Oncologia medica, Udine, Italy
| | - A Parisi
- Medical Oncology, St. Salvatore Hospital, L'Aquila, Italy; Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - K Rihawi
- IRCSS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - M Persano
- Medical Oncology, University of Cagliari, Cagliari, Italy
| | - V Crespi
- Department of Oncology, University of Turin, Torino, Italy
| | - M Libertini
- Oncology Unit, Poliambulanza Foundation, Brescia, Italy
| | - M Giordano
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - R Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - S Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| | - C Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| |
Collapse
|
12
|
Imai T, Naruse M, Ochiai M, Matsumoto K, Ikeda S, Kani M, Kato Y, Hirayama A, Soga T, Hori Y, Yokoi A, Ochiai A. Different types of reactions to E7386 among colorectal cancer patient‑derived organoids and corresponding CAFs. Oncol Lett 2022; 24:221. [PMID: 35707761 PMCID: PMC9178669 DOI: 10.3892/ol.2022.13342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/27/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Toshio Imai
- Central Animal Division, National Cancer Center Research Institute, Tokyo 104‑0045, Japan
| | - Mie Naruse
- Central Animal Division, National Cancer Center Research Institute, Tokyo 104‑0045, Japan
| | - Masako Ochiai
- Central Animal Division, National Cancer Center Research Institute, Tokyo 104‑0045, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157‑8535, Japan
| | - Satsuki Ikeda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997‑0035, Japan
| | - Manami Kani
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997‑0035, Japan
| | - Yuyu Kato
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997‑0035, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997‑0035, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997‑0035, Japan
| | - Yusaku Hori
- Oncology Business Group, Eisai Co., Ltd., Tokyo 112‑8088, Japan
| | - Akira Yokoi
- Oncology Business Group, Eisai Co., Ltd., Tokyo 112‑8088, Japan
| | - Atsushi Ochiai
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo 104‑0045, Japan
| |
Collapse
|
13
|
Strömberg C, Martinez de la Maza L, Fernández Moro C, Gerling M, Jorns C, Sparrelid E, Löhr J, Villard C. Prognostic impact of inter-metastatic heterogeneity of viable tumour cells in colorectal liver metastases. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2022; 48:1656-1663. [DOI: 10.1016/j.ejso.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022]
|
14
|
Rachiglio AM, Forgione L, Pasquale R, Barone CA, Maiello E, Antonuzzo L, Cassata A, Tonini G, Bordonaro R, Rosati G, Zaniboni A, Lonardi S, Ferrari D, Frassineti GL, Tamberi S, Pisconti S, Di Fabio F, Roma C, Orlandi A, Latiano T, Damato A, Tortora G, Pinto C, Normanno N. Dynamics of RAS/BRAF Mutations in cfDNA from Metastatic Colorectal Carcinoma Patients Treated with Polychemotherapy and Anti-EGFR Monoclonal Antibodies. Cancers (Basel) 2022; 14:1052. [PMID: 35205799 PMCID: PMC8870112 DOI: 10.3390/cancers14041052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 02/05/2023] Open
Abstract
Analysis of plasma-derived cell-free DNA (cfDNA) might allow for the early identification of resistance in metastatic colorectal carcinoma (mCRC) patients receiving anti-EGFR monoclonal antibodies. We tested plasma samples from the Erbitux Metastatic Colorectal Cancer Strategy (ERMES) phase III trial of FOLFIRI+Cetuximab in first-line treatment of RAS/BRAF wild-type mCRC. Samples were collected at baseline (n = 37), at 8 weeks of treatment (n = 32), progressive disease (PD; n = 36) and 3 months after PD (n = 21). cfDNA testing was performed using the Idylla™ ctKRAS and ctNRAS-BRAF tests and the Oncomine Pan-Cancer Cell-Free Assay. Analysis of basal samples revealed RAS/BRAF mutations in 6/37 cases. A transient RAS positivity not associated with PD was observed at 8 weeks in five cases that showed no mutations at baseline and PD. The frequency of mutant cases increased at PD (33.3%) and decreased again at 3 months after PD (9.5%). The median progression-free survival (mPFS) of patients RAS/BRAF mutant at PD was 7.13 months versus 7.71 months in wild-type patients (p = 0.3892). These data confirm that the occurrence of RAS/BRAF mutations in mCRC patients receiving anti-EGFR agents is relatively frequent. However, the cfDNA dynamics of RAS mutations in patients treated with anti-EGFR agents plus polychemotherapy are complex and might not be directly associated with resistance to treatment.
Collapse
Affiliation(s)
- Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”-IRCCS, 80131 Naples, Italy; (A.M.R.); (L.F.); (R.P.); (C.R.)
| | - Laura Forgione
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”-IRCCS, 80131 Naples, Italy; (A.M.R.); (L.F.); (R.P.); (C.R.)
| | - Raffaella Pasquale
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”-IRCCS, 80131 Naples, Italy; (A.M.R.); (L.F.); (R.P.); (C.R.)
| | - Carlo Antonio Barone
- Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy; (C.A.B.); (A.O.); (G.T.)
| | - Evaristo Maiello
- IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (E.M.); (T.L.)
| | - Lorenzo Antonuzzo
- Medical Oncology Unit, Azienda Ospedaliero Universitaria Careggi, 50134 Florence, Italy;
| | - Antonino Cassata
- Medical Oncology Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”-IRCCS, 80131 Naples, Italy;
| | - Giuseppe Tonini
- Medical Oncology Unit, Università Campus Bio-Medico, 00128 Rome, Italy;
| | | | - Gerardo Rosati
- Medical Oncology Unit, Ospedale San Carlo, 85100 Potenza, Italy;
| | | | | | | | - Giovanni Luca Frassineti
- Medical Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | | | - Salvatore Pisconti
- Medical Oncology Division, S. Giuseppe Moscati Hospital, 74010 Taranto, Italy;
| | - Francesca Di Fabio
- Medical Oncology Unit, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | - Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”-IRCCS, 80131 Naples, Italy; (A.M.R.); (L.F.); (R.P.); (C.R.)
| | - Armando Orlandi
- Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy; (C.A.B.); (A.O.); (G.T.)
| | - Tiziana Latiano
- IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (E.M.); (T.L.)
| | - Angela Damato
- Medical Oncology Unit, Clinical Cancer Center, AUSL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy; (A.D.); (C.P.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Giampaolo Tortora
- Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy; (C.A.B.); (A.O.); (G.T.)
| | - Carmine Pinto
- Medical Oncology Unit, Clinical Cancer Center, AUSL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy; (A.D.); (C.P.)
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”-IRCCS, 80131 Naples, Italy; (A.M.R.); (L.F.); (R.P.); (C.R.)
| |
Collapse
|
15
|
Ueda K, Yamada T, Ohta R, Matsuda A, Sonoda H, Kuriyama S, Takahashi G, Iwai T, Takeda K, Miyasaka T, Shinji S, Chika N, Ishida H, Yoshida H. BRAF V600E mutations in right-side colon cancer: Heterogeneity detected by liquid biopsy. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2022; 48:1375-1383. [DOI: 10.1016/j.ejso.2022.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/30/2021] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
|
16
|
Yang S, Zhan X, Tang X, Zhao S, Yu L, Gao M, Luo D, Wang Y, Chang K, Chen M. A multiplexed circulating tumor DNA detection platform engineered from 3D-coded interlocked DNA rings. Bioact Mater 2021; 10:68-78. [PMID: 34901530 PMCID: PMC8637011 DOI: 10.1016/j.bioactmat.2021.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/24/2021] [Accepted: 09/03/2021] [Indexed: 12/31/2022] Open
Abstract
Circulating tumor DNA (ctDNA) is a critical biomarker not only important for the early detection of tumors but also invaluable for personalized treatments. Currently ctDNA detection relies on sequencing. Here, a platform termed three-dimensional-coded interlocked DNA rings (3D-coded ID rings) was created for multiplexed ctDNA identification. The ID rings provide a ctDNA recognition ring that is physically interlocked with a reporter ring. The specific binding of ctDNA to the recognition ring initiates target-responsive cutting via a restriction endonuclease; the cutting then triggers rolling circle amplification on the reporter ring. The signals are further integrated with internal 3D codes for multiplexed readouts. ctDNAs from non-invasive clinical specimens including plasma, feces, and urine were detected and validated at a sensitivity much higher than those obtained through sequencing. This 3D-coded ID ring platform can detect any multiple DNA fragments simultaneously without sequencing. We envision that our platform will facilitate the implementation of future personalized/precision medicine. A platform termed 3D-coded ID rings was created for multiplexed ctDNA detection. This platform was integrated with two schemes: the ID ring scheme and the 3D-coded scheme. The platform could achieve multiplexed detection with detection limit of 500 copies per million in non-invasive specimens.
Collapse
Affiliation(s)
- Sha Yang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xinyu Zhan
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Shuang Zhao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Lianyu Yu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Mingxuan Gao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Dan Luo
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853-5701, USA
| | - Yunxia Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.,College of Pharmacy and Laboratory Medicine, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
17
|
Guo S, Ye Y, Liu X, Gong Y, Xu M, Song L, Liu H. Intra-Tumor Heterogeneity of Colorectal Cancer Necessitates the Multi-Regional Sequencing for Comprehensive Mutational Profiling. Cancer Manag Res 2021; 13:9209-9223. [PMID: 34949941 PMCID: PMC8689048 DOI: 10.2147/cmar.s327596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/01/2021] [Indexed: 11/23/2022] Open
Abstract
Background The panorama and details of quantitative intratumor heterogeneity have not been fully investigated in colorectal cancer (CRC) patients with solitary lesion without distal metastasis, and its influences on sequencing interpretation and therapeutic strategies have not been explored. Methods Cancer tissues and matched blood from 70 sporadic CRC patients were collected and were divided into two cohorts. Four individual tissue biopsies were obtained from each of the 47 patients (multi-sample cohort). One random cancer tissue biopsy was obtained from each of the rest 23 patients (single-sample cohort). A 10 mL of blood was collected from all patients and the circulating cell-free DNA (cfDNA) was extracted. A 605-gene panel was used for targeted sequencing with tissue and paired blood. Results Mutational landscape revealed significantly higher mutational frequency in APC, CARD11 and CSMD3 in multi-sample cohort than single-sample cohort (P<0.05). The number of mutations and the ratio of trunk, shared and branch mutations showed extensive heterogeneity in multi-sample cohort, and the percentage of trunk mutations in major driver genes, including APC, TP53 and KRAS, was higher than 70%. A total of 929 mutations were detected in tissue/blood in multi-sample group, with 921(99.1%) from tissue and 472(50.8%) from blood (464 common mutations,49.9%). In contrast, 394 mutations were detected in tissue/blood in single-sample group, with 231 (58.6%) from tissue and 219 (55.6%) from blood (56 common mutations, 11.9%). The number of mutations of major driver genes detected in tissue was higher than that in blood in the multi-sample cohort, while it was similar in the single-sample group. Quantification analysis revealed differential correlation between tissue and blood VAF in trunk, shared and branch mutations. Meanwhile, VAF of trunk mutations was significantly higher than shared mutations and branch mutations. VAF exhibited significant differences among distinct stages, locations, differentiation and sex status. Conclusion Characteristic extensive heterogeneity was revealed for solitary CRC without distal metastasis. Multi-regional biopsy was necessary for comprehensive mutation detection in CRC.
Collapse
Affiliation(s)
- Shaohua Guo
- Department of General Surgery, The First Medical Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
- Department of General Surgery, The Eighth Medical Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yumeng Ye
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, People’s Republic of China
| | - Xinyi Liu
- Department of Medical Division, HaploX Biotechnology, Shenzhen, People’s Republic of China
| | - Yuan Gong
- Department of Gastroenterology, The Second Medical Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Mingyan Xu
- Department of Medical Division, HaploX Biotechnology, Shenzhen, People’s Republic of China
| | - Lele Song
- Department of Medical Division, HaploX Biotechnology, Shenzhen, People’s Republic of China
- Department of Radiotherapy, The Eighth Medical Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
- Lele Song Department of Radiotherapy, The Eighth Medical Center of the Chinese PLA General Hospital, Beijing, People’s Republic of ChinaTel +86-13240149188 Email
| | - Hongyi Liu
- Department of General Surgery, The First Medical Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
- Correspondence: Hongyi Liu Department of General Surgery, The First Medical Center of the Chinese PLA General Hospital, Beijing, People’s Republic of ChinaTel +86-10-66937533 Email
| |
Collapse
|
18
|
Yeoh Y, Low TY, Abu N, Lee PY. Regulation of signal transduction pathways in colorectal cancer: implications for therapeutic resistance. PeerJ 2021; 9:e12338. [PMID: 34733591 PMCID: PMC8544255 DOI: 10.7717/peerj.12338] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Resistance to anti-cancer treatments is a critical and widespread health issue that has brought serious impacts on lives, the economy and public policies. Mounting research has suggested that a selected spectrum of patients with advanced colorectal cancer (CRC) tend to respond poorly to both chemotherapeutic and targeted therapeutic regimens. Drug resistance in tumours can occur in an intrinsic or acquired manner, rendering cancer cells insensitive to the treatment of anti-cancer therapies. Multiple factors have been associated with drug resistance. The most well-established factors are the emergence of cancer stem cell-like properties and overexpression of ABC transporters that mediate drug efflux. Besides, there is emerging evidence that signalling pathways that modulate cell survival and drug metabolism play major roles in the maintenance of multidrug resistance in CRC. This article reviews drug resistance in CRC as a result of alterations in the MAPK, PI3K/PKB, Wnt/β-catenin and Notch pathways.
Collapse
Affiliation(s)
- Yeelon Yeoh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Imyanitov E, Kuligina E. Molecular testing for colorectal cancer: Clinical applications. World J Gastrointest Oncol 2021; 13:1288-1301. [PMID: 34721767 PMCID: PMC8529925 DOI: 10.4251/wjgo.v13.i10.1288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/19/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Molecular genetic analysis is an integral part of colorectal cancer (CRC) management. The choice of systemic therapy for CRC is largely based on the results of tumor molecular testing. Evaluation of the KRAS and NRAS gene status is mandatory for consideration of anti-epidermal growth factor receptor (EGFR) therapy. Tumors with the BRAF V600E substitution are characterized by aggressive behaviour, may require intensified cytotoxic regimens and benefit from combined BRAF and EGFR inhibition. The inactivation of DNA mismatch repair (MMR), or MUTYH gene, or DNA polymerase epsilon results in excessive tumor mutational burden; these CRCs are highly antigenic and therefore sensitive to immune checkpoint inhibitors. Some CRCs are characterized by overexpression of the HER2 oncogene and respond to the appropriate targeted therapy. There are CRCs with clinical signs of hereditary predisposition to this disease, which require germline genetic testing. Liquid biopsy is an emerging technology that has the potential to assist CRC screening, control the efficacy of surgical intervention and guide disease monitoring. The landscape of CRC molecular diagnosis is currently undergoing profound changes due to the increasing use of next generation sequencing.
Collapse
Affiliation(s)
- Evgeny Imyanitov
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, Saint-Petersburg 194100, Russia
- Department of Oncology, I.I. Mechnikov North-Western Medical University, Saint-Petersburg 191015, Russia
| | - Ekaterina Kuligina
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
| |
Collapse
|
20
|
Xu H, Lan Q, Huang Y, Zhang Y, Zeng Y, Su P, Chu Z, Lai W, Chu Z. The mechanisms of colorectal cancer cell mesenchymal-epithelial transition induced by hepatocyte exosome-derived miR-203a-3p. BMC Cancer 2021; 21:718. [PMID: 34147083 PMCID: PMC8214778 DOI: 10.1186/s12885-021-08419-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Liver metastasis is the most common cause of death in patients with colorectal cancer (CRC). Phosphatase of regenerating liver-3 induces CRC metastasis by epithelial-to-mesenchymal transition, which promotes CRC cell liver metastasis. Mesenchymal-to-epithelial transition (MET), the opposite of epithelial-to-mesenchymal transition, has been proposed as a mechanism for the establishment of metastatic neoplasms. However, the molecular mechanism of MET remains unclear. Methods Using Immunohistochemistry, western blotting, invasion assays, real-time quantitative PCR, chromatin immunoprecipitation, luciferase reporter assays, human miRNA arrays, and xenograft mouse model, we determined the role of hepatocyte exosome-derived miR-203a-3p in CRC MET. Results In our study, we found that miR-203a-3p derived from hepatocyte exosomes increased colorectal cancer cells E-cadherin expression, inhibited Src expression, and reduced activity. In this way miR-203a-3p induced the decreased invasion rate of CRC cells. Coclusion MiR-203a-3p derived from hepatocyte exosomes plays an important role of CRC cells to colonize in liver.
Collapse
Affiliation(s)
- Heyang Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiusheng Lan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongliang Huang
- Department of General Surgery, Foshan Maternal and Child Health Hospital, Southern Medical University, Foshan, China
| | - Yang Zhang
- Guangzhou Blood Center, Guangzhou, Guangdong Province, China
| | - Yujie Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pengwei Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ziqiang Chu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zhonghua Chu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
21
|
Dotolo S, Marabotti A, Rachiglio AM, Esposito Abate R, Benedetto M, Ciardiello F, De Luca A, Normanno N, Facchiano A, Tagliaferri R. A multiple network-based bioinformatics pipeline for the study of molecular mechanisms in oncological diseases for personalized medicine. Brief Bioinform 2021; 22:6287337. [PMID: 34050359 PMCID: PMC8574709 DOI: 10.1093/bib/bbab180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/17/2021] [Accepted: 04/20/2021] [Indexed: 01/03/2023] Open
Abstract
Motivation Assessment of genetic mutations is an essential element in the modern era of personalized cancer treatment. Our strategy is focused on ‘multiple network analysis’ in which we try to improve cancer diagnostics by using biological networks. Genetic alterations in some important hubs or in driver genes such as BRAF and TP53 play a critical role in regulating many important molecular processes. Most of the studies are focused on the analysis of the effects of single mutations, while tumors often carry mutations of multiple driver genes. The aim of this work is to define an innovative bioinformatics pipeline focused on the design and analysis of networks (such as biomedical and molecular networks), in order to: (1) improve the disease diagnosis; (2) identify the patients that could better respond to a given drug treatment; and (3) predict what are the primary and secondary effects of gene mutations involved in human diseases. Results By using our pipeline based on a multiple network approach, it has been possible to demonstrate and validate what are the joint effects and changes of the molecular profile that occur in patients with metastatic colorectal carcinoma (mCRC) carrying mutations in multiple genes. In this way, we can identify the most suitable drugs for the therapy for the individual patient. This information is useful to improve precision medicine in cancer patients. As an application of our pipeline, the clinically significant case studies of a cohort of mCRC patients with the BRAF V600E-TP53 I195N missense combined mutation were considered. Availability The procedures used in this paper are part of the Cytoscape Core, available at (www.cytoscape.org). Data used here on mCRC patients have been published in [55]. Supplementary Information A supplementary file containing a more detailed discussion of this case study and other cases is available at the journal site as Supplementary Data.
Collapse
Affiliation(s)
- Serena Dotolo
- Dipartimento di Scienze Aziendali, Management & Innovation Systems, Università degli Studi di Salerno, Fisciano (SA), Italy
| | - Anna Marabotti
- Dipartimento di Chimica e Biologia "A. Zambelli", Università degli Studi di Salerno, Fisciano (SA), Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori -IRCCS - Fondazione G. Pascale, Naples, Italy
| | | | - Fortunato Ciardiello
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Angelo Facchiano
- Institute of Food Sciences, Italian National Research Council (CNR), Avellino, Italy
| | - Roberto Tagliaferri
- Dipartimento di Scienze Aziendali, Management & Innovation Systems, Università degli Studi di Salerno, Fisciano (SA), Italy
| |
Collapse
|
22
|
Chen B, Zheng D, Yu W, Huang C, Ye J, Han G, Zhuang J. Cetuximab versus bevacizumab maintenance following prior 8-cycle modified FOLFOXIRI plus cetuximab in Asian postmenopausal women with treatment-naive KRAS and BRAF wild-type metastatic colorectal cancer. J Int Med Res 2021; 48:300060520930440. [PMID: 32993393 PMCID: PMC7545770 DOI: 10.1177/0300060520930440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the efficacy and safety of cetuximab (CE) versus bevacizumab (BE) maintenance treatment after prior 8-cycle modified 5-fluorouracil, folinate, oxaliplatin, and irinotecan (FOLFOXIRI) plus CE induction therapy in treatment-naive KRAS and BRAF wild-type (wt) metastatic colorectal cancer (mCRC). METHODS From 2012 to 2017, prospectively maintained databases were reviewed to assess Asian postmenopausal women with treatment-naive KRAS and BRAF wt mCRC who underwent modified FOLFOXIRI plus CE induction therapy, followed by CE or BE maintenance until disease progression or death. Co-primary clinical endpoints were progression-free survival (PFS) and overall survival (OS). RESULTS A total of 222 women were included (CE n = 110 and BE n = 112). At a median follow-up of 27.0 months (interquartile range, 6.5-38.6 months), median PFS was 21.9 months (95% confidence interval [CI] 16.4-24.4) and 17.7 months (95% CI 11.3-19.0) for CE and BE groups, respectively (hazard ratio [HR] 0.31, 95% CI 0.15-0.46); median OS was 26.0 months (95% CI 23.4-28.7) and 22.7 months (95% CI 21.2-24.3) for CE and BE groups, respectively (HR 0.22, 95% CI 0.11-0.37). CONCLUSIONS CE maintenance treatment is more poorly tolerated but has a slightly more modest survival benefit compared with BE maintenance treatment in mCRC.
Collapse
Affiliation(s)
- Baomin Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Donghua Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weiguang Yu
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cuiping Huang
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junxing Ye
- Department of Orthopaedics, The Third People's Hospital of Wuxi, Jiangsu Province; The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Guowei Han
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jintao Zhuang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Behrenbruch C, Prabhakaran S, Udayasiri D, Hollande F, Michael M, Hayes I, Heriot A, Knowles B, Thomson B. Survival benefit of neoadjuvant chemotherapy and surgery versus surgery first for resectable colorectal liver metastases: a cohort study. ANZ J Surg 2021; 91:1196-1202. [PMID: 33543551 DOI: 10.1111/ans.16613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 01/03/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is continued debate about the survival benefit of neoadjuvant chemotherapy (neoCT) in patients with resectable colorectal liver metastases (CRLM). METHODS In this retrospective cohort study, we included 201 patients with metastatic colorectal cancer who underwent their first CRLM resection and achieved resection of all sites of disease. We compared the overall survival (OS) and progression-free survival (PFS) between patients who received neoCT prior to CRLM resection with those who underwent CRLM upfront. A multivariable Cox proportional hazard regression analysis was performed to adjust for potential confounders. RESULTS A total of 101 of 201 (51.2%) patients received chemotherapy prior to CRLM resection and 100 of 201 had surgery upfront. Multivariable Cox proportional hazard regression showed no statistically significant difference in the hazard of death for those given neoCT prior to resection of CRLM compared with surgery first for both OS and PFS (OS: hazard ratio 1.74, 95% confidence interval 0.85-3.55, P = 0.127, PFS: hazard ratio 1.42, 95% confidence interval 0.93-2.19, P = 0.107). CONCLUSION In our series of patients with metastatic colorectal cancer who achieved surgical resection of all sites of disease, neoCT prior to CRLM resection was not associated with any survival benefit.
Collapse
Affiliation(s)
- Corina Behrenbruch
- Sir Peter MacCallum Department of Oncology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, Victoria, Australia.,Department of General Surgical Specialties, The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, Victoria, Australia
| | - Sowmya Prabhakaran
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Dilshan Udayasiri
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Royal Melbourne Hospital Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia.,Colorectal Surgery Unit, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Frédéric Hollande
- Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, Victoria, Australia.,Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Michael
- Sir Peter MacCallum Department of Oncology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Ian Hayes
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Royal Melbourne Hospital Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia.,Colorectal Surgery Unit, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Alexander Heriot
- Sir Peter MacCallum Department of Oncology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia.,Department of Surgery, St Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Brett Knowles
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Benjamin Thomson
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Royal Melbourne Hospital Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Romei C, Ramone T, Mulè C, Prete A, Cappagli V, Lorusso L, Torregrossa L, Basolo F, Ciampi R, Elisei R. RET mutated C-cells proliferate more rapidly than non-mutated neoplastic cells. Endocr Connect 2021; 10:124-130. [PMID: 33475524 PMCID: PMC7983519 DOI: 10.1530/ec-20-0589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/15/2021] [Indexed: 12/26/2022]
Abstract
A statistically significant higher prevalence of the RET p.Met918Thr somatic mutation, identified by direct sequencing, was previously reported in MTC > 2 cm than in smaller tumors. Aim of this study was to correlate the full RET and RAS mutation profile, identified by a Next Generation Sequencing approach, with the growth rate, proliferation and tumor size of MTC. Data of 149 sporadic MTC patients were correlated with RET mutations and Ki67 positivity. Eighty-one cases had a somatic RET mutation, 40 had a RAS mutation and 28 were negative. A statistically significant higher prevalence of RET mutations was found in MTC > 2 cm. A higher prevalence of RET more aggressive mutations, higher allelic frequencies and, higher percentage of Ki67 positive cells were found in larger tumors which had also a worse outcome. Our study highlights the predominant role of RET somatic mutations in MTC tumorigenesis. We demonstrate that RET mutation prevalence and allelic frequency (AF) are significantly higher in larger tumors. Based on these results, we can conclude that RET mutated C-cells's growth and proliferation are more rapid than those of non-mutated cells and give origin to bigger and more aggressive MTC.
Collapse
Affiliation(s)
- Cristina Romei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Teresa Ramone
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Mulè
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessandro Prete
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Virginia Cappagli
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Loredana Lorusso
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Liborio Torregrossa
- Department of Surgical, Medical, Molecular Pathology, University of Pisa, Pisa, Italy
| | - Fulvio Basolo
- Department of Surgical, Medical, Molecular Pathology, University of Pisa, Pisa, Italy
| | - Raffaele Ciampi
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Correspondence should be addressed to R Elisei:
| |
Collapse
|
25
|
Implementing anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer: challenges and future perspectives. Ann Oncol 2021; 31:30-40. [PMID: 31912793 DOI: 10.1016/j.annonc.2019.10.007] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/04/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) inhibitors are valuable therapeutics in metastatic colorectal cancer (mCRC). Anti-EGFR monoclonal antibodies (MoAbs), such as cetuximab or panitumumab, in combination with chemotherapy are effective treatment options for patients with RAS and BRAF wild-type mCRC. Nevertheless, several issues are still open concerning the optimal use of anti-EGFR drugs in the continuum of care of mCRC. Novel approaches for increasing the efficacy of anti-EGFR therapies include better molecular selection of EGFR-dependent mCRC, intensification of chemotherapy, combination of anti-EGFR MoAbs and immune checkpoint inhibitors, and reintroduction of EGFR blockade or 'rechallenge' in selected patients who have previously responded to anti-EGFR MoAb therapy. An extensive translational research program was conducted in the Cetuximab After Progression in KRAS wIld-type colorectal cancer patients-Gruppo Oncologico dell' Italia Meridionale (CAPRI-GOIM) study with the aims of determining which subgroups of patients could benefit from the continuous inhibition of EGFR, from evaluating the role of liquid biopsy-based and its concordance with tissue-based molecular testing, and from investigating novel potential mechanisms of resistance to anti-EGFR therapies. In this review, we summarize the translational and clinical findings of the CAPRI-GOIM program in the context of the current knowledge of therapeutic strategies and of ongoing research on more appropriate uses of anti-EGFR therapies in RAS and BRAF wild-type mCRC patients.
Collapse
|
26
|
Liu Y, Li L, Tian Y, Zhu X, Sun A, Sun Y, Qiao Y, Qi X, Wang T. Analysis of KRAS mutations in circulating tumor DNA and colorectal cancer tissue. Biotech Histochem 2020; 96:376-383. [PMID: 32876508 DOI: 10.1080/10520295.2020.1810775] [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: 12/24/2022] Open
Abstract
The mutation status of KRAS is important for anti-EGFR therapy in colorectal cancer (CRC) patients; however, detection of KRAS mutations in circulating tumor DNA (ctDNA) is problematic. We investigated tissue and plasma assays for KRAS mutations in CRC patients. The KRAS status of 407 CRC patients was evaluated using integration of amplification refractory mutation system polymerase chain reaction (PCR), melting curves and wild type DNA blocking (IAMB) in tissue and plasma samples. Disparate cases were re-evaluated by Sanger sequencing of tissue samples. General characteristics and tumor biomarkers including CEA, CA19-9 and CA125 were characterized. The prevalence of KRAS mutations was 40.8% in plasma and 49.1% in tissue. The overall percent agreement, positive percent agreement and negative percent agreement were 82.3, 76.3 and 90.8%, respectively. Older patients and higher TNM stage exhibited increased sensitivity for detecting KRAS mutations in plasma. We found 54.1% of patients with KRAS mutations using parallel analysis of tissue and plasma; only 36.4% of patients were detected by series analysis. We found that plasma based KRAS detection with IAMB technology is an alternative to tissue based KRAS testing. KRAS mutations can be identified more easily when both assays are used together.
Collapse
Affiliation(s)
- Yankui Liu
- Department of Pathology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Longhai Li
- Department of Epidemiology and Biostatistics, Jiangnan University School of Medicine, Wuxi, China
| | - Yu Tian
- Department of Epidemiology and Biostatistics, Jiangnan University School of Medicine, Wuxi, China
| | - Xiao Zhu
- Department of Good Clinical Practice, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Aijuan Sun
- Key Laboratory of PreMed Precision Medicine, Soochow University, Suzhou, China
| | - Yulong Sun
- Key Laboratory of PreMed Precision Medicine, Soochow University, Suzhou, China
| | - Yan Qiao
- Key Laboratory of PreMed Precision Medicine, Soochow University, Suzhou, China
| | - Xiaowei Qi
- Department of Pathology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Tao Wang
- Key Laboratory of PreMed Precision Medicine, Soochow University, Suzhou, China
| |
Collapse
|
27
|
Zheng Z, Yu T, Zhao X, Gao X, Zhao Y, Liu G. Intratumor heterogeneity: A new perspective on colorectal cancer research. Cancer Med 2020; 9:7637-7645. [PMID: 32853464 PMCID: PMC7571807 DOI: 10.1002/cam4.3323] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/17/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancers generally consist of multiple subclones. These subclones have their own unique characteristics, resulting in intratumor heterogeneity (ITH). As the discussion of ITH has advanced, a model describing the relationship of ITH to the tumor has gradually emerged. ITH can be divided into two types of intraprimary tumor heterogeneity and intraindividual tumor heterogeneity, the former for further understanding of tumor composition, and the latter for providing more information about evolutionary patterns. With the rapid development of new methods, such as next‐generation, polyguanine region sequencing, and Image detection, researchers may unravel the secrets underlying ITH. The higher the ITH of the tumor, the richer the interaction between the subclones maybe, or the greater the chance of the tumor getting more powerful subclones may be, thus increasing the malignant potential of the tumor. Existing evidence suggests that ITH may increase the ability of tumors to resist treatment and can be used as an independent influence on the prognosis of colorectal cancer. We reviewed 80 recent studies to give researchers a new perspective on colorectal cancer. There is still a limited amount of research in this area. Further study of the relationship between ITH and clinical endpoints may lead to the development of new treatment strategies.
Collapse
Affiliation(s)
- Zicheng Zheng
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Tao Yu
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Xin Gao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Yao Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin General Surgery Institute, Tianjin, China
| |
Collapse
|
28
|
Study of Ras Mutations' Prognostic Value in Metastatic Colorectal Cancer: STORIA Analysis. Cancers (Basel) 2020; 12:cancers12071919. [PMID: 32708575 PMCID: PMC7409181 DOI: 10.3390/cancers12071919] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Colorectal cancer (CRC) is the second most common cause of cancer-specific death in both sexes in Western countries. KRAS mutations occur in about 50% of metastatic CRCs (mCRCs). The prognostic value of specific KRAS mutations still remains unexplored and unclear. Methods: Two hundred and forty KRAS wild-type and 206 KRAS/NRAS mutant consecutive unresectable mCRC patients with PS Eastern Cooperative Oncology Group (ECOG) 0 or 1, aged < 80 years, and with a life expectancy >3 months entered into this study. DNA was extracted from paraffin-embedded formalin-fixed tumour tissues, and it was sequenced with the Oncomine Solid Tumour DNA kit (Thermo Fisher Scientific, Waltham, MA, USA). Data were analysed using the Torrent Suite Software v5.0 (Thermo Fisher Scientific). The primary outcome was the analysis of the prognostic role of different KRAS mutations in terms of overall survival (OS). Results: There were no significant differences among the most prevalent mutations (p.G12D, p.G12V, p.G13D, p.G12A, p.G12C, and p.G12S) in terms of age (<65 vs. ≥65 years), gender (male vs. female), grading (G1/G2 vs. G3), side of primary tumour (left vs. right), pT, and pN. At the median follow-up of 25.6 months, there were 77 deaths in KRAS-mutated patients and 94 in wild-type patients. Three homogeneous prognostic groups were identified: wild-type patients (group A, median survival: 27.5 months), p.G13D/p.G12A/p.G12V/p.G12D mutants (group B, median survival: 17.3 months), and p.G12C/p.G12S mutants (group C, median survival: 5.0 months, p < 0.0001 according to Log Rank test). Upon multivariate analysis, metastatic involvement and p.G12C/p.G12S KRAS mutation group C (vs. other mutations) emerged as independent prognostic variables for survival. Conclusions: We show that mutant KRAS is a negative prognostic factor and that p.G12C/p.G12S variants present the worst clinical courses. This information suggests a clear difference among KRAS mutations, and it will be useful to test potentiated and/or innovative therapeutic strategies in p.G12C/p.G12S metastatic CRC patients.
Collapse
|
29
|
Chen X, Lu J, Wu Y, Jiang X, Gu Y, Li Y, Zhao H, Jin M. Genetic features and application value of next generation sequencing in the diagnosis of synchronous multifocal lung adenocarcinoma. Oncol Lett 2020; 20:2829-2839. [PMID: 32782601 PMCID: PMC7400153 DOI: 10.3892/ol.2020.11843] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to elucidate the genetic features of multiple lung cancer (MLC) and identify effective molecular markers for diagnosis using next generation sequencing (NGS). The present data may also inform patient treatment and prognosis. A total of 35 lesions were obtained from 17 patients with MLC. Based on lesion histology and NGS, 13 cases of multiple primary lung cancer (MPLC) were identified and 4 cases were classified as intrapulmonary metastasis (IPM). All 4 patients with IPM exhibited an epidermal growth factor receptor (EGFR) mutation and synchronous mutation of at least one tumor suppressor gene. The frequency and percentage of EGFR mutations, accompanied with tumor suppressor genes, were significantly higher in patients with IPM compared with MPLC. Furthermore, a high EGFR-heterogeneity score and male sex were risk factors of IPM occurrence. There were significant differences in mean EGFR mutation abundance alone, mutations of tumor suppressor genes and mutations of EGFR combined with tumor suppressor genes between patients with adenocarcinoma (ADC) and adenocarcinoma in situ (AIS). In conclusion, histological characteristics combined with genetic alterations may be an effective method for the diagnosis of MPLC and IPM, and NGS may serve as a useful diagnostic tool. MLC exhibited unique molecular characteristics, including higher rates of EGFR mutations, EGFR driver mutations accompanied with tumor suppressor gene mutations and the absence of anaplastic lymphoma kinase mutations, which may help distinguish between patients with MPLC or IPM. The present study hypothesized that the mean frequency of EGFR mutations, mutations of tumor suppressor genes and mutations of both EGFR and tumor suppressor genes may serve an important role in the development of AIS to ADC. The results of the present study highlight the potential underlying mechanisms of lung ADC development, which may assist with future elucidation of effective treatments to prevent the progression of lung cancer.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China.,Department of Pathology, Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia 014010, P.R. China.,Department of Pathology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia 014010, P.R. China
| | - Jun Lu
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Yingying Wu
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Xingran Jiang
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Yajuan Gu
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Yunlong Li
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Hongying Zhao
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Mulan Jin
- Department of Pathology, Capital Medical University, Affiliated Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| |
Collapse
|
30
|
Leick KM, Kazarian AG, Rajput M, Tomanek-Chalkley A, Miller A, Shrader HR, McCarthy A, Coleman KL, Kasi PM, Chan CHF. Peritoneal Cell-Free Tumor DNA as Biomarker for Peritoneal Surface Malignancies. Ann Surg Oncol 2020; 27:5065-5071. [PMID: 32648179 DOI: 10.1245/s10434-020-08832-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/27/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Disease burden in patients with peritoneal carcinomatosis (PC) is difficult to estimate. We evaluate whether peritoneal cell-free tumor DNA can be used as a measure of disease burden. PATIENTS AND METHODS Malignant ascites or peritoneal lavage fluids were collected from patients with PC under approved IRB protocol. Cell-free DNA was extracted from peritoneal fluid. Droplet digital PCR (ddPCR) was performed using a commercially available KRAS G12/G13 screening kit. Mutant allele frequency (MAF) was calculated based on the numbers of KRAS wild-type and mutant droplets. Clinicopathological, treatment and outcome data were abstracted and correlated with MAF of cell-free KRAS mutant DNA. RESULTS Cell-free KRAS mutant DNA was detected in 15/37 (40%) malignant peritoneal fluids with a MAF of 0.1% to 26.2%. While peritoneal cell-free KRAS mutant DNA was detected in all the patients with KRAS mutant tumors (N = 10), 3/16 (19%) patients with KRAS wild-type tumors also had peritoneal cell-free KRAS mutant DNA. We also found that 71% (5/7) of patients with disease amenable to cytoreductive surgery (CRS) had a MAF of < 1% (median: 0.5%, range: 0.1-4.7%), while 75% (6/8) of patients with unresectable disease had a MAF of > 1% (median: 4.4%, range: 0.1-26.2%). CONCLUSIONS This pilot proof-of-principle study suggests that peritoneal cell-free tumor DNA detected by ddPCR may enable prediction of disease burden and a measure of disease amenable to CRS in patients with PC.
Collapse
Affiliation(s)
- Katie M Leick
- Department of Surgery, University of Iowa, Iowa City, IA, USA
| | | | - Maheen Rajput
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | | | - Ann Miller
- Department of Surgery, University of Iowa, Iowa City, IA, USA
| | | | - Ashley McCarthy
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Kristen L Coleman
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Pashtoon M Kasi
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.,Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Carlos H F Chan
- Department of Surgery, University of Iowa, Iowa City, IA, USA. .,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
31
|
Porru M, Zizza P, Panera N, Alisi A, Biroccio A, Leonetti C. Harnessing Omics Approaches on Advanced Preclinical Models to Discovery Novel Therapeutic Targets for the Treatment of Metastatic Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12071830. [PMID: 32650388 PMCID: PMC7408740 DOI: 10.3390/cancers12071830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Metastatic colorectal cancer (mCRC) remains challenging because of the emergence of resistance mechanisms to anti-epidermal growth factor receptor (EGFR) therapeutics, so more effective strategies to improve the patients’ outcome are needed. During the last decade, the application of a multi-omics approach has contributed to a deeper understanding of the complex molecular landscape of human CRC, identifying a plethora of drug targets for precision medicine. Target validation relies on the use of experimental models that would retain the molecular and clinical features of human colorectal cancer, thus mirroring the clinical characteristics of patients. In particular, organoids and patient-derived-xenografts (PDXs), as well as genetically engineered mouse models (GEMMs) and patient-derived orthotopic xenografts (PDOXs), should be considered for translational purposes. Overall, omics and advanced mouse models of cancer represent a portfolio of sophisticated biological tools that, if optimized for use in concert with accurate data analysis, could accelerate the anticancer discovery process and provide new weapons against cancer. In this review, we highlight success reached following the integration of omics and experimental models; moreover, results produced by our group in the field of mCRC are also presented.
Collapse
Affiliation(s)
- Manuela Porru
- UOSD SAFU, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCSS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Pasquale Zizza
- UOSD Oncogenomic and Epigenetic Unit, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCSS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Nadia Panera
- Research Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (N.P.); (A.A.)
| | - Anna Alisi
- Research Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (N.P.); (A.A.)
| | - Annamaria Biroccio
- UOSD Oncogenomic and Epigenetic Unit, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCSS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
- Correspondence: (A.B.); (C.L.)
| | - Carlo Leonetti
- UOSD SAFU, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCSS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
- Correspondence: (A.B.); (C.L.)
| |
Collapse
|
32
|
González-González M, Gutiérrez ML, Sayagués JM, Muñoz-Bellvís L, Orfao A. Genomic profiling of sporadic liver metastatic colorectal cancer. Semin Cancer Biol 2020; 71:98-108. [PMID: 32485312 DOI: 10.1016/j.semcancer.2020.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Sporadic colorectal cancer (sCRC) is the third leading cause of cancer death in the Western world. Approximately, a quarter of sCRC patients present metastatic dissemination at the moment of diagnosis, the liver being the most frequently affected organ. Additionally, this group of CRC patients is characterized by a worse prognosis. In the last decades, significant technological developments for genome analysis have fostered the identification and characterization of genetic alterations involved in the pathogenesis of sCRC. However, genetic alterations involved in the metastatic process through which tumor cells are able to colonize other tissues with a different microenvironment, still remain to be fully identified. Here, we review current knowledge about the most relevant genomic alterations involved in the liver metastatic process of sCRC, including detailed information about the genetic profile of primary colorectal tumors vs. their paired liver metastases.
Collapse
Affiliation(s)
- María González-González
- Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain; Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Biomedical Research Networking Centre Consortium-CIBER-CIBERONC, Spain
| | - María Laura Gutiérrez
- Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain; Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Biomedical Research Networking Centre Consortium-CIBER-CIBERONC, Spain
| | - José María Sayagués
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain; Department of Pathology, Universidad de Salamanca, Salamanca, Spain
| | - Luis Muñoz-Bellvís
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Biomedical Research Networking Centre Consortium-CIBER-CIBERONC, Spain; Department of General and Gastrointestinal Surgery, University Hospital of Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain; Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Biomedical Research Networking Centre Consortium-CIBER-CIBERONC, Spain.
| |
Collapse
|
33
|
Li Y, Monzo M, Moreno I, Martinez-Rodenas F, Hernandez R, Castellano JJ, Canals J, Han B, Muñoz C, Navarro A. KRAS mutations by digital PCR in circulating tumor cells isolated from the mesenteric vein are associated with residual disease and overall survival in resected colorectal cancer patients. Int J Colorectal Dis 2020; 35:805-813. [PMID: 32088737 DOI: 10.1007/s00384-020-03538-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2020] [Indexed: 02/04/2023]
Abstract
PURPOSE In colorectal cancer (CRC), circulating tumor cells (CTCs) are released into the mesenteric veins (MV). We chose to determine whether KRAS mutations detected in CTCs from blood obtained at the time of surgery could be a marker of survival. METHODS From 52 surgically resected CRC patients who later relapsed, samples of tumor tissue, normal tissue, and blood from the peripheral vein (PV) and MV were obtained from each patient at the time of surgery. KRAS mutations were assessed by Sanger sequencing and digital PCR (DGPCR) in tissue samples and by DGPCR in CTCs. Mutant KRAS copy number was assessed in CTCs. Results were correlated with overall survival (OS). RESULTS Sanger sequencing detected KRAS mutations in ten tumor samples (19.2%), while DGPCR detected mutations in 30 (58%). Mutations were detected in CTCs in 21 MV samples (40.4%) and 18 PV samples (34.6%). Patients with G13D mutations in CTCs from the MV had shorter OS than those with G12D mutations (28.1 vs 54.6 months; p = 0.025). Patients with a high mutant KRAS copy number in CTCs had shorter OS than those with a low mutant KRAS copy number (MV: 20.5 vs 43.7 months; p = 0.002; PV: 15.1 vs 38.2 months; p = 0.027). CONCLUSION DGPCR is more efficient than Sanger sequencing for detecting KRAS mutations. KRAS G13D mutations and high mutant KRAS copy number are associated with shorter OS. The analysis of KRAS mutations in CTCs from blood obtained at the time of surgery can identify patients with a higher risk of relapse.
Collapse
Affiliation(s)
- Yan Li
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Mariano Monzo
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Isabel Moreno
- Department of Medical Oncology and Surgery, Hospital Municipal de Badalona, Badalona, Spain
| | - Francisco Martinez-Rodenas
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
- Department of Medical Oncology and Surgery, Hospital Municipal de Badalona, Badalona, Spain
| | - Raquel Hernandez
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
- Department of Medical Oncology and Surgery, Hospital Municipal de Badalona, Badalona, Spain
| | - Joan J Castellano
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Jordi Canals
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Bing Han
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Carmen Muñoz
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Alfons Navarro
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain.
- Faculty of Medicine and Health Sciences-Hospital Clinic, University of Barcelona, Casanova 143, ES-08036, Barcelona, Spain.
| |
Collapse
|
34
|
Martini G, Ciardiello D, Vitiello PP, Napolitano S, Cardone C, Cuomo A, Troiani T, Ciardiello F, Martinelli E. Resistance to anti-epidermal growth factor receptor in metastatic colorectal cancer: What does still need to be addressed? Cancer Treat Rev 2020; 86:102023. [PMID: 32474402 DOI: 10.1016/j.ctrv.2020.102023] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022]
Abstract
Colorectal cancer (CRC) represents a global health problem, being one of the most diagnosed and aggressive tumors. Cetuximab and panitumumab monoclonal antibodies (mAbs) in combination with chemotherapy are an effective strategy for patients with RAS Wild Type (WT) metastatic colorectal cancer (mCRC). However, tumors are often unresponsive or develop resistance. In the last years, molecular alterations in principal oncogenes (RAS, BRAF, PI3KCA, PTEN) in the downstream pathway of the epidermal growth factor receptor (EGFR) and in other receptors (HER2, MET) that converge on MAPK-ERK signalling have been identified as novel mechanisms of resistance to anti-EGFR strategies. However, further efforts are needed to better stratify CRCs and ensure more individualized treatments. Herein, we describe the consolidated molecular drivers of resistance and the therapeutic strategies available so far, with an overview on potential biomarkers of response that could be integrated in clinical practice.
Collapse
Affiliation(s)
- Giulia Martini
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Pietro Paolo Vitiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Stefania Napolitano
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Claudia Cardone
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Antonio Cuomo
- Gastroenterology Unit, Ospedale Umberto I, Nocera Inferiore, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
35
|
Max Ma X, Bendell JC, Hurwitz HI, Ju C, Lee JJ, Lovejoy A, Mancao C, Nicholas A, Price R, Sommer N, Tikoo N, Yao L, Yaung SJ, Palma JF. Disease Monitoring Using Post-induction Circulating Tumor DNA Analysis Following First-Line Therapy in Patients with Metastatic Colorectal Cancer. Clin Cancer Res 2020; 26:4010-4017. [PMID: 32220893 DOI: 10.1158/1078-0432.ccr-19-1209] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 11/22/2019] [Accepted: 03/23/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE We assessed plasma circulating tumor DNA (ctDNA) level as a prognostic marker for progression-free survival (PFS) following first-line metastatic colorectal cancer (mCRC) therapy. EXPERIMENTAL DESIGN The Sequencing Triplet With Avastin and Maintenance (STEAM) was a randomized, phase II trial investigating efficacy of bevacizumab (BEV) plus 5-fluorouracil/leucovorin/oxaliplatin (FOLFOX) and 5-fluorouracil/leucovorin/irinotecan (FOLFIRI), administered concurrently or sequentially, versus FOLFOX-BEV in first-line mCRC. Evaluation of biomarkers associated with treatment outcomes was an exploratory endpoint. Patients in the biomarker-evaluable population (BEP) had 1 tissue sample, 1 pre-induction plasma sample, and 1 post-induction plasma sample collected ≤60 days of induction from last drug date. RESULTS Among the 280 patients enrolled in STEAM, 183 had sequenced and evaluable tumor tissue, 118 had matched pre-induction plasma, and 54 (BEP) had ctDNA-evaluable sequencing data for pre- and post-induction plasma. The most common somatic variants in tumor tissue and pre-induction plasma were TP53, APC, and KRAS. Patients with lower-than-median versus higher-than-median post-induction mean allele fraction (mAF) levels had longer median PFS (17.7 vs. 7.5 months, HR, 0.33; 95% confidence interval, 0.17-0.63). Higher levels of post-induction mAF and post-induction mean mutant molecules per milliliter (mMMPM), and changes in ctDNA (stratified by a 10-fold or 100-fold reduction in mAF between pre- and post-induction plasma), were associated with shorter PFS. Post-induction mAF and mMMPM generally correlated with each other (ρ = 0.987, P < 0.0001). CONCLUSIONS ctDNA quantification in post-induction plasma may serve as a prognostic biomarker for mCRC post-treatment outcomes.
Collapse
Affiliation(s)
- Xiaoju Max Ma
- Medical Scientific Affairs, Roche Sequencing Solutions, Inc., Pleasanton, California.
| | - Johanna C Bendell
- Drug Development Unit, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | - Herbert I Hurwitz
- Project Development Oncology, Genentech, Inc., South San Francisco, California
| | - Christine Ju
- Clinical Operations and Biometrics, Roche Molecular Systems, Pleasanton, California
| | - John J Lee
- Medical Scientific Affairs, Roche Sequencing Solutions, Inc., Pleasanton, California
| | - Alex Lovejoy
- Assay Development, Roche Sequencing Solutions, Inc., Pleasanton, California
| | - Christoph Mancao
- Oncology Biomarker Development, Genentech, Inc., Basel, Switzerland
| | - Alan Nicholas
- US Medical Affairs and Biometrics, Genentech, Inc., South San Francisco, California
| | - Richard Price
- Oncology Biomarker Development, Genentech, Inc., Basel, Switzerland
| | - Nicolas Sommer
- Medical Affairs BioOncology, Genentech, Inc., South San Francisco, California
| | - Nalin Tikoo
- Clinical Operations and Biometrics, Roche Molecular Systems, Pleasanton, California
| | - Lijing Yao
- Bioinformatics Research and Early Development, Roche Sequencing Solutions, Inc., Pleasanton, California
| | - Stephanie J Yaung
- Medical and Scientific Affairs Bioinformatics, Roche Sequencing Solutions, Inc., Pleasanton, California
| | - John F Palma
- Medical Scientific Affairs, Roche Sequencing Solutions, Inc., Pleasanton, California.
| |
Collapse
|
36
|
Park KJ, Kim IS, Yang EJ, Lim YT, Cho SH. Mutation analysis in Korean patients with T-cell acute lymphoblastic leukemia. Pediatr Hematol Oncol 2020; 37:129-139. [PMID: 31852326 DOI: 10.1080/08880018.2019.1701160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Genomic studies have illuminated the alterations in pathways underlying T-cell acute lymphoblastic leukemia (T-ALL) pathogenesis, but detailed mutation data by next-generation sequencing have not been reported in Korean patients. We aimed to investigate mutation frequency, spectrum, and pattern in the Korean patients with T-ALL. We designed a multigene panel targeting 101 genes and validated it using 10 reference materials. The mutation analysis was done in a total of 10 patients with T-ALL. Clinical data and laboratory tests including immunophenotyping, cytogenetics, and molecular genetic tests were also investigated. All of the 10 patients harbored at least one mutation (range 1-6 per patient). A total of 34 clinically significant mutations including 15 novel mutations were identified in 23 genes. The median of variant allelic frequencies (VAFs) and blasts were counted upto 33% (range 5-91%) and 79% (range 38-90%), respectively. Recurrent mutations were involved in epigenetic regulators (60%), NOTCH1 signaling (40%), PI3K-AKT (40%), JAK-STAT (30%), and transcription factors (30%). We found that both NOTCH signaling and JAK-STAT signaling were positively associated with epigenetic regulators, while showed mutually exclusive patterns with PI3K-AKT pathway. This study showed that the frequency of mutations in epigenetic regulators in Korean patients was significantly higher than expected. Distribution of VAF as well as mutation spectrum is considerably heterogeneous in Korean patients with T-ALL. Although from a limited number of patients, this study provides the first detailed mutational portrait of T-ALL of Korean patients, and gives additional insight into molecular pathogenesis of the disease.
Collapse
Affiliation(s)
- Kyoung-Jin Park
- Department of Laboratory Medicine, Myongji Hospital, Goyang, Republic of Korea
| | - In-Suk Kim
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Eu Jeen Yang
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Young Tak Lim
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Su-Hee Cho
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| |
Collapse
|
37
|
Pasquale R, Forgione L, Roma C, Fenizia F, Bergantino F, Rachiglio AM, De Luca A, Gallo M, Maiello MR, Palumbo G, Morabito A, Azzaro R, Normanno N. Targeted sequencing analysis of cell-free DNA from metastatic non-small-cell lung cancer patients: clinical and biological implications. Transl Lung Cancer Res 2020; 9:61-70. [PMID: 32206554 PMCID: PMC7082281 DOI: 10.21037/tlcr.2020.01.01] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sequencing artifacts, clonal hematopoietic mutations of indeterminate potential (CHIP) and tumor heterogeneity have been hypothesized to contribute to the low concordance between tissue and cell-free DNA (cfDNA) molecular profiling with targeted sequencing. METHODS We analyzed by targeted sequencing cfDNA from 30 healthy individuals, and cfDNA and matched tumor samples from 30 EGFR-mutant and 77 EGFR wild-type metastatic non-small-cell lung cancer (mNSCLC) patients. Discordant cases were solved by droplet digital PCR (ddPCR). RESULTS By testing cfDNA from healthy donors, we developed an algorithm to recognize sequencing artifacts. Applying this method to cfDNA from mNSCLC patients, EGFR mutations were detected with a good sensitivity (76.7%) and specificity (97.4%). In contrast, sensitivity and specificity for KRAS variants were 61.5% and 93.8%, respectively. All EGFR and KRAS variants detected in plasma but not in tissue were confirmed by ddPCR, thus excluding sequencing artifacts. In a fraction of cases, KRAS mutations found in plasma samples were confirmed in tumor tissue suggesting tumor heterogeneity. KRAS variants were found to be more likely sub-clonal as compared with EGFR mutations, and a correlation between clonal origin and frequency of detection in plasma was found. In a case with both EGFR and KRAS variants in cfDNA, we could demonstrate the presence of the KRAS variant in tumor tissue associated with lack of response to tyrosine kinase inhibitors (TKIs). CONCLUSIONS Although sequencing artifacts can be identified in targeted sequencing of cfDNA, tumor heterogeneity and CHIP are likely to influence the concordance between plasma and tissue testing.
Collapse
Affiliation(s)
- Raffaella Pasquale
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Laura Forgione
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Cristin Roma
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Francesca Fenizia
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Francesca Bergantino
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Anna M. Rachiglio
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Marianna Gallo
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Monica R. Maiello
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Giuliano Palumbo
- Medical Oncology, Ospedale Santa Maria della Pietà, Casoria, Italy
| | - Alessandro Morabito
- Thoracic Medical Oncology, Department of Hemathology, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Rosa Azzaro
- Transfusion Service, Department of Hemathology, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| |
Collapse
|
38
|
Xie B, Bai B, Xu Y, Liu Y, Lv Y, Gao X, Wu F, Fang Z, Lou Y, Pan H, Han W. Tumor-suppressive function and mechanism of HOXB13 in right-sided colon cancer. Signal Transduct Target Ther 2019; 4:51. [PMID: 31815008 PMCID: PMC6882800 DOI: 10.1038/s41392-019-0086-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 01/05/2023] Open
Abstract
Right-sided colon cancer (RCC) and left-sided colon cancer (LCC) differ in their clinical and molecular features. An investigation of differentially expressed genes (DEGs) between RCC and LCC could contribute to targeted therapy for colon cancer, especially RCC, which has a poor prognosis. Here, we identified HOXB13, which was significantly less expressed in RCC than in LCC and associated with prognosis in RCC, by using 5 datasets from the Gene Expression Omnibus (GEO). Tissue sample analysis showed that HOXB13 was differentially expressed between normal and only RCC tumor tissues. HOXB13 inhibited colon cancer cell proliferation and induced apoptosis both in vitro and in vivo. Furthermore, we found that HOXB13 might be regulated by DNMT3B and suppress C-myc expression to exert antitumor effects via β-catenin/TCF4 signals in RCC. In conclusion, the current study is the first to demonstrate that HOXB13 has a tumor-suppressive effect in RCC. High expression levels of HOXB13 are associated with prolonged overall survival in patients with RCC. The DNMT3B-HOXB13-C-myc signaling axis might be a molecular target for the treatment of RCC.
Collapse
Affiliation(s)
- Binbin Xie
- Department of Medical Oncology; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| | - Bingjun Bai
- Department of Colorectal Surgery; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| | - Yuzi Xu
- Department of Stomatology; Stomatology Hospital; School of Medicine, Zhejiang University, Hangzhou, 310000 PR China
| | - Yunlong Liu
- Department of Medical Oncology; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| | - Yiming Lv
- Department of Colorectal Surgery; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| | - Xing Gao
- Department of Medical Oncology; The Second Affiliated Hospital of Suzhou University; School of Medicine, Suzhou University, Suzhou, 215000 PR China
| | - Fei Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001 PR China
| | - Zhipeng Fang
- Department of Medical Oncology; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| | - Ying Lou
- Department of Medical Oncology; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| | - Hongming Pan
- Department of Medical Oncology; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| | - Weidong Han
- Department of Medical Oncology; Sir Run Run Shaw Hospital; School of Medicine, Zhejiang University, Hangzhou, 310016 PR China
| |
Collapse
|
39
|
Kwak Y, Seo AN, Lee HE, Lee HS. Tumor immune response and immunotherapy in gastric cancer. J Pathol Transl Med 2019; 54:20-33. [PMID: 31674166 PMCID: PMC6986974 DOI: 10.4132/jptm.2019.10.08] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
Remarkable developments in immuno-oncology have changed the landscape of gastric cancer (GC) treatment. Because immunotherapy intervenes with tumor immune response rather than directly targeting tumor cells, it is important to develop a greater understanding of tumor immunity. This review paper summarizes the tumor immune reaction and immune escape mechanisms while focusing on the role of T cells and their co-inhibitory signals, such as the immune checkpoint molecules programmed death-1 and programmed deathligand 1 (PD-L1). This paper also describes past clinical trials of immunotherapy for patients with GC and details their clinical implications. Strong predictive markers are essential to improve response to immunotherapy. Microsatellite instability, Epstein-Barr virus, PD-L1 expression, and tumor mutational burden are now regarded as potent predictive markers for immunotherapy in patients with GC. Novel immunotherapy and combination therapy targeting new immune checkpoint molecules such as lymphocyte-activation gene 3, T cell immunoglobulin, and mucin domain containing-3, and indoleamine 2,3-dioxygenase have been suggested, and trials are ongoing to evaluate their safety and efficacy. Immunotherapy is an important treatment option for patients with GC and has great potential for improving patient outcome, and further research in immuno-oncology should be carried out.
Collapse
Affiliation(s)
- Yoonjin Kwak
- Department of Pathology, Seoul National University Hospital, Seoul, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Hee Eun Lee
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - Hye Seung Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea.,Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
40
|
van Helden EJ, Angus L, Menke-van der Houven van Oordt CW, Heideman DAM, Boon E, van Es SC, Radema SA, van Herpen CML, de Groot DJA, de Vries EGE, Jansen MPHM, Sleijfer S, Verheul HMW. RAS and BRAF mutations in cell-free DNA are predictive for outcome of cetuximab monotherapy in patients with tissue-tested RAS wild-type advanced colorectal cancer. Mol Oncol 2019; 13:2361-2374. [PMID: 31350822 PMCID: PMC6822250 DOI: 10.1002/1878-0261.12550] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/09/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022] Open
Abstract
In metastatic colorectal cancer, RAS and BRAF mutations cause resistance to anti‐EGFR therapies, such as cetuximab. Heterogeneity in RAS and BRAF mutations might explain nonresponse in a subset of patients receiving cetuximab. Analyzing mutations in plasma‐derived circulating tumor DNA (ctDNA) could provide a more comprehensive overview of the mutational landscape as compared to analyses of primary and/or metastatic tumor tissue. Therefore, this prospective multicenter study followed 34 patients with metastatic colorectal cancer who were tissue‐tested as RAS wild‐type (exons 2–4) during routine work‐up and received third‐line cetuximab monotherapy. BRAF mutation status was also tested but did not exclude patients from therapy. At baseline and upon disease progression, cell‐free DNA (cfDNA) was isolated for targeted next‐generation sequencing (NGS). At 8 weeks, we determined that patients had benefited from treatment. NGS of cfDNA identified three patients with RAS mutations not detected in tumor tissue during routine work‐up. Another six patients had a BRAF or rare RAS mutation in ctDNA and/or tumor tissue. Relative to patients without mutations in RAS/BRAF, patients with mutations at baseline had shorter progression‐free survival [1.8 versus 4.9 months (P < 0.001)] and overall survival [3.1 versus 9.4 months (P = 0.001)]. In patients with clinical benefit (progressive disease after 8 weeks), ctDNA testing revealed previously undetected mutations in RAS/BRAF (71%) and EGFR (47%), which often emerged polyclonally. Our results indicate that baseline NGS of ctDNA can identify additional RAS mutation carriers, which could improve patient selection for anti‐EGFR therapies. Acquired resistance, in patients with initial treatment benefit, is mainly explained by polyclonal emergence of RAS,BRAF, and EGFR mutations in ctDNA.
Collapse
Affiliation(s)
- Erik J van Helden
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Lindsay Angus
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Daniëlle A M Heideman
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Eline Boon
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Suzanne C van Es
- Department of Medical Oncology, University Medical Center Groningen, The Netherlands
| | - Sandra A Radema
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Derk Jan A de Groot
- Department of Medical Oncology, University Medical Center Groningen, The Netherlands
| | | | - Maurice P H M Jansen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| |
Collapse
|
41
|
Onidani K, Shoji H, Kakizaki T, Yoshimoto S, Okaya S, Miura N, Sekikawa S, Furuta K, Lim CT, Shibahara T, Boku N, Kato K, Honda K. Monitoring of cancer patients via next-generation sequencing of patient-derived circulating tumor cells and tumor DNA. Cancer Sci 2019; 110:2590-2599. [PMID: 31169336 PMCID: PMC6676129 DOI: 10.1111/cas.14092] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/21/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Liquid biopsy of circulating tumor cells (CTC) and circulating tumor DNA (ctDNA) is gaining attention as a method for real-time monitoring in cancer patients. Conventional methods based upon epithelial cell adhesion molecule (EpCAM) expression have a risk of missing the most aggressive CTC subpopulations due to epithelial-mesenchymal transition and may, thus, underestimate the total number of actual CTC present in the bloodstream. Techniques utilizing a label-free inertial microfluidics approach (LFIMA) enable efficient capture of CTC without the need for EpCAM expression. In this study, we optimized a method for analyzing genetic alterations using next-generation sequencing (NGS) of extracted ctDNA and CTC enriched using an LFIMA as a first-phase examination of 30 patients with head and neck cancer, esophageal cancer, gastric cancer and colorectal cancer (CRC). Seven patients with advanced CRC were enrolled in the second-phase examination to monitor the emergence of alterations occurring during treatment with epidermal growth factor receptor (EGFR)-specific antibodies. Using LFIMA, we effectively captured CTC (median number of CTC, 14.5 cells/mL) from several types of cancer and detected missense mutations via NGS of CTC and ctDNA. We also detected time-dependent genetic alterations that appeared during anti-EGFR therapy in CTC and ctDNA from CRC patients. The results of NGS analyses indicated that alterations in the genomic profile revealed by the liquid biopsy could be expanded by using a combination of assays with CTC and ctDNA. The study was registered with the University Hospital Medical Information Network Clinical Trials Registry (ID: UMIN000014095).
Collapse
Affiliation(s)
- Kaoru Onidani
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan.,Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo, Japan
| | - Hirokazu Shoji
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan.,Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiko Kakizaki
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Seiichi Yoshimoto
- Department of Head and Neck Surgery, National Cancer Center Central Hospital, Tokyo, Japan
| | - Shinobu Okaya
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Nami Miura
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Shoichi Sekikawa
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo, Japan
| | - Koh Furuta
- Division of Clinical Laboratory, Kanagawa Cancer Center, Kanagawa, Japan
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore.,Biomedical Institute of Global Health Research and Technology, National University of Singapore, Singapore.,Mechanbiology Institute, National University of Singapore, Singapore, Singapore
| | - Takahiko Shibahara
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo, Japan
| | - Narikazu Boku
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Kazufumi Honda
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan.,Japan Agency for Medical Research and Development (AMED) CREST, Tokyo, Japan
| |
Collapse
|
42
|
Necessity of Genetic Evaluation of Metachronous Metastases of Colorectal Cancer: Quantitative Analysis of Genetic Discordance Between Metachronous Metastases and Radically Resected Primary Colorectal Cancers Using Next-Generation Sequencing. Dis Colon Rectum 2019; 62:832-839. [PMID: 31188184 DOI: 10.1097/dcr.0000000000001386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Mutation analyses provide the basis of selecting an appropriate target agent for the treatment of metastatic colorectal cancer. However, metachronous metastases developed after the treatment of primary tumor could create significant opportunities for different genetic profiles relative to the primary tumors. OBJECTIVE The purpose of this study was to assess the necessity of genetic evaluation of metachronous metastases; we performed a quantitative analysis of genetic discordance between metachronous metastases and radically resected primary colorectal cancers using next-generation sequencing. DESIGN This was a retrospective study. SETTINGS Patients from a single-institution tertiary care center were studied. PATIENTS We enrolled 33 patients who underwent resection of metachronous metastases between January 2014 and December 2016, ≥6 months after radical resection of primary colorectal cancer and whose tissue was available for analysis. MAIN OUTCOME MEASURES Tumor samples were analyzed by next-generation sequencing. The mutant allele frequency was analyzed to evaluate the proportion of mutations in the tumor tissue. RESULTS The mutant allele frequency of KRAS in metachronous metastases was higher in 6 cases (mean difference =% 25.5% (range, 9.5%-58.0%)) and lower in 3 cases (mean difference = 9.3% (range, 8.0-10.0%) compared with each of their primary tumors. In 1 case, the KRAS mutant-type (mutant allele frequency = 22.6%) metachronous metastasis had developed from the KRAS wild-type primary tumor. LIMITATIONS Tumor sample may not represent perfectly the whole tumor of the patient because of heterogeneity. CONCLUSIONS Genetic discordance can exist between metachronous metastases and radically resected primary colorectal cancers. For appropriate target therapy, genetic evaluation of metachronous metastases needs to be considered when possible. See Video Abstract at http://links.lww.com/DCR/A932.
Collapse
|
43
|
Guo TA, Wu YC, Tan C, Jin YT, Sheng WQ, Cai SJ, Liu FQ, Xu Y. Clinicopathologic features and prognostic value of KRAS, NRAS and BRAF mutations and DNA mismatch repair status: A single-center retrospective study of 1,834 Chinese patients with Stage I-IV colorectal cancer. Int J Cancer 2019; 145:1625-1634. [PMID: 31162857 PMCID: PMC6771586 DOI: 10.1002/ijc.32489] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
Abstract
Mutations of KRAS, NRAS, BRAF and DNA mismatch repair (MMR) status have become an important part of the assessment of patients with colorectal cancer (CRC), while respective clinicopathologic features and prognostic significance in specific stages and related detection strategies remain unclear. We retrospectively analyzed clinicopathologic features and prognosis of 1,834 patients with Stage I–IV colorectal adenocarcinoma. Mutations in KRAS, NRAS and BRAF and DNA MMR status were determined. The mutation rates of KRAS, NRAS and BRAF were 46.4, 3.2 and 3.5%, respectively, and the mismatch repair gene deletion (dMMR) rate was 5.6%. In a multivariate analysis, female, advanced age, tumor type histology, mucinous carcinoma and positive tumor deposits were associated with a high KRAS mutation rate. A high BRAF mutation rate was associated with female, poor differentiation, lymphovascular invasion and positive tumor deposits. Factors associated with high dMMR rates included low age, large tumor size, poor differentiation, Stages I–III. Tumor site was independently associated with KRAS mutation, BRAF mutation and dMMR. KRAS and BRAF mutations were independent risk factors for shorter overall survival (OS) in Stage IV tumors but not in Stage I–III tumors. NRAS mutation was an independent risk factor for shorter OS in Stage I–II tumors. dMMR was independently associated with longer OS in Stage III tumors. What's new? Mutations in KRAS, NRAS, BRAF and DNA mismatch repair (MMR) status are important biomarkers in the assessment of patients with colorectal cancer (CRC). However, the clinicopathologic features associated with these mutations—and their impact on prognosis—are unclear, especially at earlier stages of CRC. In this large Chinese study, the authors analyzed variables such as gender, age, tumor histology, lymphovascular invasion, etc., that were associated with particular oncogene mutations and overall survival. These results should provide guidance for improved clinical strategies and enhance the usefulness of these biomarkers.
Collapse
Affiliation(s)
- Tian-An Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Chen Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cong Tan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu-Tong Jin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA
| | - Wei-Qi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - San-Jun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fang-Qi Liu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
44
|
Rachiglio AM, Lambiase M, Fenizia F, Roma C, Cardone C, Iannaccone A, De Luca A, Carotenuto M, Frezzetti D, Martinelli E, Maiello E, Ciardiello F, Normanno N. Genomic Profiling of KRAS/NRAS/BRAF/PIK3CA Wild-Type Metastatic Colorectal Cancer Patients Reveals Novel Mutations in Genes Potentially Associated with Resistance to Anti-EGFR Agents. Cancers (Basel) 2019; 11:E859. [PMID: 31226844 PMCID: PMC6627713 DOI: 10.3390/cancers11060859] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
Previous findings suggest that metastatic colorectal carcinoma (mCRC) patients with KRAS/NRAS/BRAF/PIK3CA wild-type (quadruple-wt) tumors are highly sensitive to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (MoAbs). However, additional molecular alterations might be involved in the de novo resistance to these drugs. We performed a comprehensive molecular profiling of 21 quadruple-wt tumors from mCRC patients enrolled in the "Cetuximab After Progression in KRAS wild-type colorectal cancer patients" (CAPRI-GOIM) trial of first line FOLFIRI plus cetuximab. Tumor samples were analyzed with a targeted sequencing panel covering single nucleotide variants (SNVs), insertions/deletions (Indels), copy number variations (CNVs), and gene fusions in 143 cancer-related genes. The analysis revealed in all 21 patients the presence of at least one SNV/Indel and in 10/21 cases (48%) the presence of at least one CNV. Furthermore, 17/21 (81%) patients had co-existing SNVs/Indels in different genes. Quadruple-wt mCRC from patients with the shorter progression free survival (PFS) were enriched with peculiar genetic alterations in KRAS, FBXW7, MAP2K1, and NF1 genes as compared with patients with longer PFS. These data suggest that a wide genetic profiling of quadruple-wt mCRC patients might help to identify novel markers of de novo resistance to anti-EGFR MoAbs.
Collapse
Affiliation(s)
- Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Matilde Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Francesca Fenizia
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Claudia Cardone
- Department of Precision Medicine, Università degli Studi della Campania L Vanvitelli, 80131 Naples, Italy.
| | - Alessia Iannaccone
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
- Department of food and feed control, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici (NA), Italy.
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Marianeve Carotenuto
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Daniela Frezzetti
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Erika Martinelli
- Department of Precision Medicine, Università degli Studi della Campania L Vanvitelli, 80131 Naples, Italy.
| | - Evaristo Maiello
- Department of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy.
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania L Vanvitelli, 80131 Naples, Italy.
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| |
Collapse
|
45
|
Santos C, Azuara D, Viéitez JM, Páez D, Falcó E, Élez E, López-López C, Valladares M, Robles-Díaz L, García-Alfonso P, Bugés C, Durán G, Salud A, Navarro V, Capellá G, Aranda E, Salazar R. Phase II study of high-sensitivity genotyping of KRAS, NRAS, BRAF and PIK3CA to ultra-select metastatic colorectal cancer patients for panitumumab plus FOLFIRI: the ULTRA trial. Ann Oncol 2019; 30:796-803. [PMID: 30840064 DOI: 10.1093/annonc/mdz082] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Several studies show the importance of accurately quantifying not only KRAS and other low-abundant mutations because benefits of anti-EGFR therapies may depend on certain sensitivity thresholds. We assessed whether ultra-selection of patients using a high-sensitive digital PCR (dPCR) to determine KRAS, NRAS, BRAF and PIK3CA status can improve clinical outcomes of panitumumab plus FOLFIRI. PATIENTS AND METHODS This was a single-arm phase II trial that analysed 38 KRAS, NRAS, BRAF and PIK3CA hotspots in tumour tissues of irinotecan-resistant metastatic colorectal cancer patients who received panitumumab plus FOLFIRI until disease progression or early withdrawal. Mutation profiles were identified by nanofluidic dPCR and correlated with clinical outcomes (ORR, overall response rate; PFS, progression-free survival; OS, overall survival) using cut-offs from 0% to 5%. A quantitative PCR (qPCR) analysis was also performed. RESULTS Seventy-two evaluable patients were enrolled. RAS (KRAS/NRAS) mutations were detected in 23 (32%) patients and RAS/BRAF mutations in 25 (35%) by dPCR, while they were detected in 7 (10%) and 11 (15%) patients, respectively, by qPCR. PIK3CA mutations were not considered in the analyses as they were only detected in 2 (3%) patients by dPCR and in 1 (1%) patient by qPCR. The use of different dPCR cut-offs for RAS (KRAS/NRAS) and RAS/BRAF analyses translated into differential clinical outcomes. The highest ORR, PFS and OS in wild-type patients with their lowest values in patients with mutations were achieved with a 5% cut-off. We observed similar outcomes in RAS/BRAF wild-type and mutant patients defined by qPCR. CONCLUSIONS High-sensitive dPCR accurately identified patients with KRAS, NRAS, BRAF and PIK3CA mutations. The optimal RAS/BRAF mutational cut-off for outcome prediction is 5%, which explains that the predictive performance of qPCR was not improved by dPCR. The biological and clinical implications of low-frequent mutated alleles warrant further investigations. CLINICALTRIALS.GOV NUMBER NCT01704703. EUDRACT NUMBER 2012-001955-38.
Collapse
Affiliation(s)
- C Santos
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat; Department of Medical Oncology, Institut Català d'Oncologia Oncobell Program-IDIBELL, CIBERONC, L'Hospitalet de Llobregat
| | - D Azuara
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat
| | - J M Viéitez
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo
| | - D Páez
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - E Falcó
- Department of Medical Oncology, Hospital Son Llàtzer, Palma de Mallorca
| | - E Élez
- Department of Medical Oncology, Hospital Vall d'Hebrón, Barcelona
| | - C López-López
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander
| | - M Valladares
- Department of Medical Oncology, Hospital Universitario de A Coruña, A Coruña
| | - L Robles-Díaz
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid
| | - P García-Alfonso
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid
| | - C Bugés
- Department of Medical Oncology, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Badalona, Institut Català d'Oncologia-Hospital Germans Trias i Pujol
| | - G Durán
- Department of Medical Oncology, Hospital Universitario Virgen de la Victoria, Málaga
| | - A Salud
- Department of Medical Oncology, Hospital Universitari Arnau de Vilanova, Lleida
| | - V Navarro
- Clinical Research Unit, Institut Català d'Oncologia, L'Hospitalet de Llobregat
| | - G Capellá
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat
| | - E Aranda
- Department of Medical Oncology, IMIBIC, Hospital Universitario Reina Sofía, Universidad de Córdoba, CIBERONC, Córdoba, Spain
| | - R Salazar
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat; Department of Medical Oncology, Institut Català d'Oncologia Oncobell Program-IDIBELL, CIBERONC, L'Hospitalet de Llobregat.
| |
Collapse
|
46
|
Deans ZC, Butler R, Cheetham M, Dequeker EMC, Fairley JA, Fenizia F, Hall JA, Keppens C, Normanno N, Schuuring E, Patton SJ. IQN path ASBL report from the first European cfDNA consensus meeting: expert opinion on the minimal requirements for clinical ctDNA testing. Virchows Arch 2019; 474:681-689. [PMID: 31028539 PMCID: PMC6581928 DOI: 10.1007/s00428-019-02571-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 02/08/2019] [Accepted: 04/02/2019] [Indexed: 12/18/2022]
Abstract
Liquid biopsy testing is a new laboratory-based method that detects tumour mutations in circulating free DNA (cfDNA) derived from minimally invasive blood sampling techniques. Recognising the significance for clinical testing, in 2017, IQN Path provided external quality assessment for liquid biopsy testing. Representatives of those participating laboratories were invited to attend a workshop to discuss the findings and how to achieve quality implementation of cfDNA testing in the clinical setting, the discussion and outcomes of this consensus meeting are described below. Predictive molecular profiling using tumour tissue in order to select cancer patients eligible for targeted therapy is now routine in diagnostic pathology. If insufficient tumour tissue material is available, in some circumstances, recent European Medicines Agency (EMA) guidance recommends mutation testing with plasma cfDNA. Clinical applications of cfDNA include treatment selection based on clinically relevant mutations derived from pre-treatment samples and the detection of resistant mutations upon progression of the disease. In order to identify tumour-related mutations in amongst other nucleic acid material found in plasma samples, highly sensitive laboratory methods are needed. In the workshop, we discussed the variable approaches taken with regard to cfDNA extraction methods, the tests, and considered the impact of false-negative test results. We explored the lack of standardisation of complex testing procedures ranging from plasma collection, transport, processing and storage, cfDNA extraction, and mutation analysis, to interpretation and reporting of results. We will also address the current status of clinical validation and clinical utility, and its use in current diagnosis. This workshop revealed a need for guidelines on with standardised procedures for clinical cfDNA testing and reporting, and a requirement for cfDNA-based external quality assessment programs.
Collapse
Affiliation(s)
- Zandra C Deans
- UK NEQAS for Molecular Genetics, Department of Laboratory Medicine, Royal Infirmary of Edinburgh, Little France Crescent, Edinburgh, EH16 4SA, UK.
| | - Rachel Butler
- All Wales Genetic Laboratory, Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff, CF14 4XW, UK
| | - Melanie Cheetham
- European Molecular Genetics Quality Network, Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester, M13 9WL, UK
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium.,University Hospital of Leuven, Leuven, Belgium
| | - Jennifer A Fairley
- UK NEQAS for Molecular Genetics, Department of Laboratory Medicine, Royal Infirmary of Edinburgh, Little France Crescent, Edinburgh, EH16 4SA, UK
| | - Francesca Fenizia
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Jacqueline A Hall
- International Quality Network for Pathology (IQN Path ASBL), 3A Sentier de l'Esperance, 1474, Luxembourg, Luxembourg
| | - Cleo Keppens
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center of Groningen, Groningen, The Netherlands
| | - Simon J Patton
- European Molecular Genetics Quality Network, Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester, M13 9WL, UK
| |
Collapse
|
47
|
Ashktorab H, Azimi H, Varma S, Lee EL, Laiyemo AO, Nickerson ML, Brim H. Driver genes exome sequencing reveals distinct variants in African Americans with colorectal neoplasia. Oncotarget 2019; 10:2607-2624. [PMID: 31080553 PMCID: PMC6498998 DOI: 10.18632/oncotarget.26721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in the United States. African Americans are disproportionately affected by CRC. Our hypothesis is that driver genes with known and novel mutations have an impact on CRC outcome in this population. Therefore, we investigated the variants' profiles in a panel of 15 CRC genes. PATIENTS & METHODS Colorectal specimens (n=140) were analyzed by targeted exome sequencing using an Ion Torrent platform. Detected variants were validated in 36 samples by Illumina sequencing. The novel status of the validated variants was determined by comparison to publicly available databases. Annotated using ANNOVAR and in-silico functional analysis of these variants were performed to determine likely pathogenic variants. RESULTS Overall, 121 known and novel variants were validated: APC (27%), AMER1 (3%), ARID1 (7%), MSH3 (12%), MSH6 (10%), BRAF (4%), KRAS (6%), FBXW7 (4%), PIK3CA (6%), SMAD4 (5%), SOX9 (2%), TCF7L2 (2%), TGFBR2 (5%), TP53 (7%). From these validated variants, 12% were novel in 8 genes (AMER1, APC, ARID1A, BRAF, MSH6, PIK3CA, SMAD4, and TCF7L2). Of the validated variants, 23% were non-synonymous, 14% were stopgains, 24% were synonymous and 39% were intronic variants. CONCLUSION We here report the specifics of variants' profiles of African Americans with colorectal lesions. Validated variants showed that Tumor Suppressor Genes (TSGs) APC and ARID1 and DNA Mismatch repair (MMR) genes MSH3 and MSH6 are the genes with the highest numbers of validated variants. Oncogenes KRAS and PIK3CA are also altered and likely participate in the increased proliferative potential of the mutated colonic epithelial cells in this population.
Collapse
Affiliation(s)
- Hassan Ashktorab
- Department of Medicine, Cancer Center, Howard University, Washington, DC, USA
| | - Hamed Azimi
- Department of Medicine, Cancer Center, Howard University, Washington, DC, USA
| | | | - Edward L. Lee
- Department of Pathology, Howard University College of Medicine, Washington, DC, USA
| | - Adeyinka O. Laiyemo
- Department of Medicine, Cancer Center, Howard University, Washington, DC, USA
| | - Michael L. Nickerson
- Laboratory of Translational Genomics, National Cancer Institute, Bethesda, MD, USA
| | - Hassan Brim
- Department of Pathology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
48
|
Normanno N, Esposito Abate R, Lambiase M, Forgione L, Cardone C, Iannaccone A, Sacco A, Rachiglio AM, Martinelli E, Rizzi D, Pisconti S, Biglietto M, Bordonaro R, Troiani T, Latiano TP, Giuliani F, Leo S, Rinaldi A, Maiello E, Ciardiello F. RAS testing of liquid biopsy correlates with the outcome of metastatic colorectal cancer patients treated with first-line FOLFIRI plus cetuximab in the CAPRI-GOIM trial. Ann Oncol 2019; 29:112-118. [PMID: 28950295 DOI: 10.1093/annonc/mdx417] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Liquid biopsy is an alternative to tissue for RAS testing in metastatic colorectal carcinoma (mCRC) patients. Little information is available on the predictive role of liquid biopsy RAS testing in patients treated with first-line anti-EGFR monoclonal antibody-based therapy. Patients and methods In the CAPRI-GOIM trial, 340 KRAS exon-2 wild-type mCRC patients received first-line cetuximab plus FOLFIRI. Tumor samples were retrospectively assessed by next generation sequencing (NGS). Baseline plasma samples were analyzed for KRAS and NRAS mutations using beads, emulsion, amplification, and magnetics digital PCR (BEAMing). Discordant cases were solved by droplet digital PCR (ddPCR) or deep-sequencing. Results A subgroup of 92 patients with available both NGS data on tumor samples and baseline plasma samples were included in this study. Both NGS analysis of tumor tissue and plasma testing with BEAMing identified RAS mutations in 33/92 patients (35.9%). However, 10 cases were RAS tissue mutant and plasma wild-type, and additional 10 cases were tissue wild-type and plasma mutant, resulting in a concordance rate of 78.3%. Analysis of plasma samples with ddPCR detected RAS mutations in 2/10 tissue mutant, plasma wild-type patients. In contrast, in all tissue wild-type and plasma mutant cases, ddPCR or deep-sequencing analysis of tumor tissue confirmed the presence of RAS mutations at allelic frequencies ranging between 0.15% and 1.15%. The median progression-free survival of RAS mutant and wild-type patients according to tissue (7.9 versus 12.6 months; P = 0.004) and liquid biopsy testing (7.8 versus 13.8 moths; P < 0.001) were comparable. Similar findings were observed for the median overall survival of RAS mutant and wild-type patients based on tissue (22.1 versus 35.8 months; P = 0.016) and plasma (19.9 versus 35.8 months; P = 0.013) analysis. Conclusion This study indicates that RAS testing of liquid biopsy results in a similar outcome when compared with tissue testing in mCRC patients receiving first-line anti-EGFR monoclonal antibodies.
Collapse
Affiliation(s)
- N Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - R Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - M Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - L Forgione
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - C Cardone
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | - A Iannaccone
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - A Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - A M Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - E Martinelli
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | - D Rizzi
- Gruppo Oncologico dell'Italia Meridionale (GOIM), Bari, Italy
| | - S Pisconti
- Medical Oncology, Hospital SS. Annunziata, Taranto, Italy
| | - M Biglietto
- Medical Oncology, Hospital "A. Cardarelli," Napoli, Italy
| | - R Bordonaro
- Medical Oncology, Hospital Garibaldi-Nesima, Catania, Italy
| | - T Troiani
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | - T P Latiano
- Medical Oncology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - F Giuliani
- Medical Oncology, National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - S Leo
- Medical Oncology, Hospital Vito Fazzi, Lecce, Italy
| | - A Rinaldi
- Medical Oncology, Presidio Ospedaliero Polo Occidentale, Castellaneta, Bari, Italy
| | - E Maiello
- Medical Oncology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - F Ciardiello
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | | |
Collapse
|
49
|
Rachiglio AM, Fenizia F, Piccirillo MC, Galetta D, Crinò L, Vincenzi B, Barletta E, Pinto C, Ferraù F, Lambiase M, Montanino A, Roma C, Ludovini V, Montagna ES, De Luca A, Rocco G, Botti G, Perrone F, Morabito A, Normanno N. The Presence of Concomitant Mutations Affects the Activity of EGFR Tyrosine Kinase Inhibitors in EGFR-Mutant Non-Small Cell Lung Cancer (NSCLC) Patients. Cancers (Basel) 2019; 11:E341. [PMID: 30857358 PMCID: PMC6468673 DOI: 10.3390/cancers11030341] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023] Open
Abstract
Recent findings suggest that a fraction of EGFR-mutant non-small-cell lung cancers (NSCLC) carry additional driver mutations that could potentially affect the activity of EGFR tyrosine kinase inhibitors (TKIs). We investigated the role of concomitant KRAS, NRAS, BRAF, PIK3CA, MET and ERBB2 mutations (other mutations) on the outcome of 133 EGFR mutant patients, who received first-line therapy with EGFR TKIs between June 2008 and December 2014. Analysis of genomic DNA by Next Generation Sequencing (NGS) revealed the presence of hotspot mutations in genes other than the EGFR, including KRAS, NRAS, BRAF, ERBB2, PIK3CA, or MET, in 29/133 cases (21.8%). A p.T790M mutation was found in 9/133 tumour samples (6.8%). The progression free survival (PFS) of patients without other mutations was 11.3 months vs. 7 months in patients with other mutations (log-rank test univariate: p = 0.047). In a multivariate Cox regression model including the presence of other mutations, age, performance status, smoking status, and the presence of p.T790M mutations, the presence of other mutations was the only factor significantly associated with PFS (Hazard Ratio 1.63, 95% CI 1.04⁻2.58; p = 0.035). In contrast, no correlation was found between TP53 mutations and patients' outcome. These data suggest that a subgroup of EGFR mutant tumours have concomitant driver mutations that might affect the activity of first-line EGFR TKIs.
Collapse
Affiliation(s)
- Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Francesca Fenizia
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Maria Carmela Piccirillo
- Clinical Trials Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Domenico Galetta
- Medical Oncology, National Cancer Research Center "Giovanni Paolo II", 70126 Bari, Italy.
| | - Lucio Crinò
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Bruno Vincenzi
- Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy.
| | | | - Carmine Pinto
- Medical Oncology, S. Maria Nuova Hospital-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy.
| | - Francesco Ferraù
- Medical Oncology, "S. Vincenzo" Hospital, 98039 Taormina (ME), Italy.
| | - Matilde Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Agnese Montanino
- Medical Oncology, Thoraco-Pulmonary Department, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Vienna Ludovini
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy.
| | | | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Gaetano Rocco
- Thoracic Surgery, Thoraco-Pulmonary Department, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Gerardo Botti
- Surgical Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy.
| | - Alessandro Morabito
- Medical Oncology, Thoraco-Pulmonary Department, 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
|
50
|
Martini G, Cardone C, Vitiello PP, Belli V, Napolitano S, Troiani T, Ciardiello D, Della Corte CM, Morgillo F, Matrone N, Sforza V, Papaccio G, Desiderio V, Paul MC, Moreno-Viedma V, Normanno N, Rachiglio AM, Tirino V, Maiello E, Latiano TP, Rizzi D, Signoriello G, Sibilia M, Ciardiello F, Martinelli E. EPHA2 Is a Predictive Biomarker of Resistance and a Potential Therapeutic Target for Improving Antiepidermal Growth Factor Receptor Therapy in Colorectal Cancer. Mol Cancer Ther 2019; 18:845-855. [PMID: 30824612 DOI: 10.1158/1535-7163.mct-18-0539] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/28/2018] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
The EPHA2 tyrosine kinase receptor is implicated in tumor progression and targeted therapies resistance. We evaluated EPHA2 as a potential resistance marker to the antiepidermal growth factor receptor (EGFR) monoclonal antibody cetuximab in colorectal cancer. We studied activation of EPHA2 in a panel of human colorectal cancer cell lines sensitive or resistant to anti-EGFR drugs. The in vitro and in vivo effects of ALW-II-41-27 (an EPHA2 inhibitor) and/or cetuximab treatment were tested. Formalin-fixed paraffin-embedded tumor specimens from 82 RAS wild-type (WT) metastatic colorectal cancer patients treated with FOLFIRI + cetuximab as first-line therapy in the CAPRI-GOIM trial were assessed for EPHA2 expression by immunohistochemistry and correlated with treatment efficacy. EPHA2 was differentially activated in colorectal cancer cell lines. Combined treatment with ALW-II-41-27 plus cetuximab reverted primary and acquired resistance to cetuximab, causing cell growth inhibition, inducing apoptosis and cell-cycle G1-G2 arrest. In tumor xenograft models, upon progression to cetuximab, ALW-II-41-27 addition significantly inhibited tumor growth. EPHA2 protein expression was detected in 55 of 82 tumor samples, frequently expressed in less-differentiated and left-sided tumors. High levels of EPHA2 significantly correlated with worse progression-free survival [8.6 months; confidence interval (CI) 95%, 6.4-10.8; vs. 12.3 months; CI 95%, 10.4-14.2; P = 0.03] and with increased progression rate (29% vs. 9%, P = 0.02). A specific EPHA2 inhibitor reverts in vitro and in vivo primary and acquired resistance to anti-EGFR therapy. EPHA2 levels are significantly associated with worse outcome in patients treated with FOLFIRI + cetuximab. These results highlight EPHA2 as a potential therapeutic target in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Giulia Martini
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Claudia Cardone
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Pietro Paolo Vitiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Valentina Belli
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Stefania Napolitano
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Teresa Troiani
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Davide Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Carminia Maria Della Corte
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Floriana Morgillo
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Nunzia Matrone
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Vincenzo Sforza
- Department of Clinical Experimental Thoracic Oncology, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli Napoli, IT, Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli Napoli, IT, Naples, Italy
| | - Mariel C Paul
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, Vienna, Austria
| | - Veronica Moreno-Viedma
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, Vienna, Austria
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Naples, Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli Napoli, IT, Naples, Italy
| | - Evaristo Maiello
- Medical Oncology, Hospital Casa Sollievo Della Sofferenza-San Giovanni Rotondo (Foggia), San Giovanni Rotondo, Italy
| | - Tiziana Pia Latiano
- Medical Oncology, Hospital Casa Sollievo Della Sofferenza-San Giovanni Rotondo (Foggia), San Giovanni Rotondo, Italy
| | - Daniele Rizzi
- Medical Oncology, Hospital Casa Sollievo Della Sofferenza-San Giovanni Rotondo (Foggia), San Giovanni Rotondo, Italy
| | - Giuseppe Signoriello
- Biostatistics, Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Maria Sibilia
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, Vienna, Austria
| | - Fortunato Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy.
| | - Erika Martinelli
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania L. Vanvitelli, Naples, Italy.
| |
Collapse
|