201
|
Corti G, Bartolini A, Crisafulli G, Novara L, Rospo G, Montone M, Negrino C, Mussolin B, Buscarino M, Isella C, Barault L, Siravegna G, Siena S, Marsoni S, Di Nicolantonio F, Medico E, Bardelli A. A Genomic Analysis Workflow for Colorectal Cancer Precision Oncology. Clin Colorectal Cancer 2019; 18:91-101.e3. [DOI: 10.1016/j.clcc.2019.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/18/2022]
|
202
|
Vlachostergios PJ, Faltas BM. Treatment resistance in urothelial carcinoma: an evolutionary perspective. Nat Rev Clin Oncol 2019; 15:495-509. [PMID: 29720713 DOI: 10.1038/s41571-018-0026-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The emergence of treatment-resistant clones is a critical barrier to cure in patients with urothelial carcinoma. Setting the stage for the evolution of resistance, urothelial carcinoma is characterized by extensive mutational heterogeneity, which is detectable even in patients with early stage disease. Chemotherapy and immunotherapy both act as selective pressures that shape the evolutionary trajectory of urothelial carcinoma throughout the course of the disease. A detailed understanding of the dynamics of evolutionary drivers is required for the rational development of curative therapies. Herein, we describe the molecular basis of the clonal evolution of urothelial carcinomas and the use of genomic approaches to predict treatment responses. We discuss various mechanisms of resistance to chemotherapy with a focus on the mutagenic effects of the DNA dC->dU-editing enzymes APOBEC3 family of proteins. We also review the evolutionary mechanisms underlying resistance to immunotherapy, such as the loss of clonal tumour neoantigens. By dissecting treatment resistance through an evolutionary lens, the field will advance towards true precision medicine for urothelial carcinoma.
Collapse
Affiliation(s)
- Panagiotis J Vlachostergios
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Bishoy M Faltas
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center at Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
203
|
Ramón Y Cajal S, Hümmer S, Peg V, Guiu XM, De Torres I, Castellvi J, Martinez-Saez E, Hernandez-Losa J. Integrating clinical, molecular, proteomic and histopathological data within the tissue context: tissunomics. Histopathology 2019; 75:4-19. [PMID: 30667539 PMCID: PMC6851567 DOI: 10.1111/his.13828] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/19/2019] [Indexed: 12/14/2022]
Abstract
Malignant tumours show a marked degree of morphological, molecular and proteomic heterogeneity. This variability is closely related to microenvironmental factors and the location of the tumour. The activation of genetic alterations is very tissue‐dependent and only few tumours have distinct genetic alterations. Importantly, the activation state of proteins and signaling factors is heterogeneous in the primary tumour and in metastases and recurrences. The molecular diagnosis based only on genetic alterations can lead to treatments with unpredictable responses, depending on the tumour location, such as the tumour response in melanomas versus colon carcinomas with BRAF mutations. Therefore, we understand that the correct evaluation of tumours requires a system that integrates both morphological, molecular and protein information in a clinical and pathological context, where intratumoral heterogeneity can be assessed. Thus, we propose the term ‘tissunomics’, where the diagnosis will be contextualised in each tumour based on the complementation of the pathological, molecular, protein expression, environmental cells and clinical data.
Collapse
Affiliation(s)
- Santiago Ramón Y Cajal
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - Stefan Hümmer
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - Vicente Peg
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - Xavier M Guiu
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain.,Department of Pathology, Bellvitge University Hospital, Barcelona, Spain
| | - Inés De Torres
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Josep Castellvi
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - Elena Martinez-Saez
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Javier Hernandez-Losa
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| |
Collapse
|
204
|
McCoach CE, Bivona TG. Engineering Multidimensional Evolutionary Forces to Combat Cancer. Cancer Discov 2019; 9:587-604. [PMID: 30992280 PMCID: PMC6497542 DOI: 10.1158/2159-8290.cd-18-1196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/28/2018] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
With advances in technology and bioinformatics, we are now positioned to view and manage cancer through an evolutionary lens. This perspective is critical as our appreciation for the role of tumor heterogeneity, tumor immune compartment, and tumor microenvironment on cancer pathogenesis and evolution grows. Here, we explore recent knowledge on the evolutionary basis of cancer pathogenesis and progression, viewing tumors as multilineage, multicomponent organisms whose growth is regulated by subcomponent fitness relationships. We propose reconsidering some current tenets of the cancer management paradigm in order to take better advantage of crucial fitness relationships to improve outcomes of patients with cancer. SIGNIFICANCE: Tumor and tumor immune compartment and microenvironment heterogeneity, and their evolution, are critical disease features that affect treatment response. The impact and interplay of these components during treatment are viable targets to improve clinical response. In this article, we consider how tumor cells, the tumor immune compartment and microenvironment, and epigenetic factors interact and also evolve during treatment. We evaluate the convergence of these factors and suggest innovative treatment concepts that leverage evolutionary relationships to limit tumor growth and drug resistance.
Collapse
Affiliation(s)
- Caroline E McCoach
- Department of Medicine, University of California, San Francisco, California.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, California.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California
| |
Collapse
|
205
|
Circulating tumor DNA analyses predict progressive disease and indicate trastuzumab-resistant mechanism in advanced gastric cancer. EBioMedicine 2019; 43:261-269. [PMID: 31031019 PMCID: PMC6562020 DOI: 10.1016/j.ebiom.2019.04.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023] Open
Abstract
Background Circulating tumor DNA (ctDNA) isolated from plasma contains genetic mutations that can be representative of those found in primary tumor tissue DNA. These samples can provide insights into tumoral heterogeneity in patients with advanced gastric cancer (AGC). Although trastuzumab has been shown to be effective in first-line therapy for patients with metastatic gastric cancer with overexpression of human epidermal growth factor receptor 2 (HER2), the mechanism of AGC resistance is incompletely understood. Methods In this prospective study, we used targeted capture sequencing to analyze 173 serial ctDNA samples from 39 AGC patients. We analyzed cancer cell fractions with PyClone to understand the clonal population structure in cancer, and monitored serial samples during therapy. Serial monitoring of ctDNA using the molecular tumor burden index (mTBI), identified progressive disease before imaging results (mean: 18 weeks). Findings We reconstructed the clonal structure of ctDNA during anti-HER2 treatment, and identified 32 expanding mutations potentially related to trastuzumab resistance. Multiple pathways activating in the same patients revealed heterogeneity in trastuzumab resistance mechanisms in AGC. In patients who received chemotherapy, mTBI was validated for the prediction of progressive disease, with a sensitivity of 94% (15/16). A higher mTBI (≥1%) in pretreatment ctDNA was also a risk factor for progression-free survival. Conclusions Analysis of ctDNA clones based on sequencing is a promising approach to clinical management, and may lead to improved therapeutic strategies for AGC patients. Fund This work was supported by grants from the National International Cooperation Grant (to J.X.; Project No. 2014DFB33160).
Collapse
|
206
|
Abstract
Since the discovery that DNA alterations initiate tumorigenesis, scientists and clinicians have been exploring ways to counter these changes with targeted therapeutics. The sequencing of tumor DNA was initially limited to highly actionable hot spots-areas of the genome that are frequently altered and have an approved matched therapy in a specific tumor type. Large-scale genome sequencing programs quickly developed technological improvements that enabled the deployment of whole-exome and whole-genome sequencing technologies at scale for pristine sample materials in research environments. However, the turning point for precision medicine in oncology was the innovations in clinical laboratories that improved turnaround time, depth of coverage, and the ability to reliably sequence archived, clinically available samples. Today, tumor genome sequencing no longer suffers from significant technical or financial hurdles, and the next opportunity for improvement lies in the optimal utilization of the technologies and data for many different tumor types.
Collapse
Affiliation(s)
- Kenna R Mills Shaw
- Khalifa Bin Zayed Institute for Personalized Cancer Therapy and Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
| | - Anirban Maitra
- Khalifa Bin Zayed Institute for Personalized Cancer Therapy and Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
| |
Collapse
|
207
|
Sumalatha V, Rambabu A, Vamsikrishna N, Ganji N, Daravath S, Shivaraj. Synthesis, characterization, DNA binding propensity, nuclease efficacy, antioxidant and antimicrobial activities of Cu(II), Co(II) and Ni(II) complexes derived from 4-(trifluoromethoxy)aniline Schiff bases. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.cdc.2019.100213] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
208
|
Sato H, Soh J, Aoe K, Fujimoto N, Tanaka S, Namba K, Torigoe H, Shien K, Yamamoto H, Tomida S, Tao H, Okabe K, Kishimoto T, Toyooka S. Droplet digital PCR as a novel system for the detection of microRNA‑34b/c methylation in circulating DNA in malignant pleural mesothelioma. Int J Oncol 2019; 54:2139-2148. [PMID: 30942424 DOI: 10.3892/ijo.2019.4768] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/08/2019] [Indexed: 11/06/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare malignancy arising from the pleura that is difficult to diagnose, contributing to its dismal prognosis. Previously, we reported that the degree of microRNA (miR)‑34b/c methylation in circulating DNA is associated with the development of MPM. Herein, we present a newly developed droplet digital PCR (ddPCR)‑based assay for the detection of miR‑34b/c methylation in circulating DNA in patients with MPM. We originally prepared two probes within a short amplicon of 60 bp, designing one from the positive strand and the other from the complementary strand. The two probes functioned cooperatively, and our established assay detected DNA methylation accurately in the preliminary validation. We subsequently verified this assay using clinical samples. Serum samples from 35 cases of MPM, 29 cases of pleural plaque and 10 healthy volunteers were collected from 3 different institutions and used in this study. We divided the samples into 2 groups (group A, n=33; group B, n=41). A receiver‑operating characteristic curve analysis using the samples in group A determined the optimal cut‑off value for the diagnosis of MPM, with a sensitivity of 76.9% and a specificity of 90%. On the other hand, the use of the same criterion yielded a sensitivity of 59.1% and a specificity of 100% in group B, and corresponding values of 65.7 and 94.9% for the entire cohort, indicating a moderate sensitivity and a high specificity. In addition, when the analysis was focused on stage II or more advanced MPM, the sensitivity improved to 81.8%, suggesting the possibility that the methylated allele frequency in MPM may be associated with the stage of disease progression. On the whole, the findings of this study indicate that miR‑34b/c methylation in circulating DNA is a promising biomarker for the prediction of disease progression in patients with MPM.
Collapse
Affiliation(s)
- Hiroki Sato
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Junichi Soh
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Keisuke Aoe
- Department of Medical Oncology, National Hospital Organization, Yamaguchi‑Ube Medical Center, Ube, Yamaguchi 755‑0241, Japan
| | - Nobukazu Fujimoto
- Department of Respiratory Medicine, Okayama Rosai Hospital, Okayama 702‑8055, Japan
| | - Shin Tanaka
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Kei Namba
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Hidejiro Torigoe
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Kazuhiko Shien
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Hiromasa Yamamoto
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Shuta Tomida
- Department of Bioinformatics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Hiroyuki Tao
- Department of Clinical Research, National Hospital Organization, Yamaguchi‑Ube Medical Center, Ube, Yamaguchi 755‑0241, Japan
| | - Kazunori Okabe
- Department of Clinical Research, National Hospital Organization, Yamaguchi‑Ube Medical Center, Ube, Yamaguchi 755‑0241, Japan
| | - Takumi Kishimoto
- Department of Respiratory Medicine, Okayama Rosai Hospital, Okayama 702‑8055, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| |
Collapse
|
209
|
Schumacher D, Andrieux G, Boehnke K, Keil M, Silvestri A, Silvestrov M, Keilholz U, Haybaeck J, Erdmann G, Sachse C, Templin M, Hoffmann J, Boerries M, Schäfer R, Regenbrecht CRA. Heterogeneous pathway activation and drug response modelled in colorectal-tumor-derived 3D cultures. PLoS Genet 2019; 15:e1008076. [PMID: 30925167 PMCID: PMC6457557 DOI: 10.1371/journal.pgen.1008076] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 04/10/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022] Open
Abstract
Organoid cultures derived from colorectal cancer (CRC) samples are increasingly used as preclinical models for studying tumor biology and the effects of targeted therapies under conditions capturing in vitro the genetic make-up of heterogeneous and even individual neoplasms. While 3D cultures are initiated from surgical specimens comprising multiple cell populations, the impact of tumor heterogeneity on drug effects in organoid cultures has not been addressed systematically. Here we have used a cohort of well-characterized CRC organoids to study the influence of tumor heterogeneity on the activity of the KRAS/MAPK-signaling pathway and the consequences of treatment by inhibitors targeting EGFR and downstream effectors. MAPK signaling, analyzed by targeted proteomics, shows unexpected heterogeneity irrespective of RAS mutations and is associated with variable responses to EGFR inhibition. In addition, we obtained evidence for intratumoral heterogeneity in drug response among parallel “sibling” 3D cultures established from a single KRAS-mutant CRC. Our results imply that separate testing of drug effects in multiple subpopulations may help to elucidate molecular correlates of tumor heterogeneity and to improve therapy response prediction in patients. Commonly occurring genetic alterations and patient-specific genetic features are increasingly used to predict the possible action of targeted cancer therapies. Although several lines of evidence have suggested that preclinical and clinical responses concur, the heterogeneity of tumors remains a severe obstacle in routinely translating preclinical data to patient treatments. Here we present a rapid work flow that integrates drug testing of three-dimensional patient tumor-derived (organoid) cultures and assessment of their genetic make-up as well as that of their donor tumors by amplicon sequencing and targeted proteomics. While the organoid cultures largely recapitulated the genomic profiles of donor tumors, the overall treatment responses and inhibitor effects on the intracellular signaling system were quite variable. Notably, organoid cultures obtained by synchronous multi-regional sampling of the same colorectal tumor showed an up to 30-fold difference in drug response. A combinatorial drug treatment improved the response. These data were confirmed in matched mouse xenograft models from the same tumor. Our findings may help to refine preclinical testing of individual tumors by modelling heterogeneity in cultures, to better understand therapeutic failure in clinical settings and to find ways to overcome treatment resistance.
Collapse
Affiliation(s)
- Dirk Schumacher
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Geoffroy Andrieux
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Karsten Boehnke
- Eli Lilly and Company, Lilly Research Laboratories, Oncology Translational Research, New York, NY, United States of America
| | - Marlen Keil
- EPO Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin, Germany
| | | | | | | | - Johannes Haybaeck
- Department of Pathology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Department of Pathology, Neuropathology, and Molecular Pathology, Medical University of Innsbruck, Austria.,Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, Austria
| | - Gerrit Erdmann
- NMI TT Pharmaservices, Berlin, Germany.,ASC Oncology GmbH, Berlin, Germany
| | - Christoph Sachse
- NMI TT Pharmaservices, Berlin, Germany.,ASC Oncology GmbH, Berlin, Germany
| | - Markus Templin
- ASC Oncology GmbH, Berlin, Germany.,NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Jens Hoffmann
- EPO Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin, Germany
| | - Melanie Boerries
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Reinhold Schäfer
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Charité Comprehensive Cancer Center, Berlin, Germany
| | - Christian R A Regenbrecht
- cpo-Cellular Phenomics & Oncology Berlin-Buch GmbH, Berlin, Germany.,Department of Pathology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,ASC Oncology GmbH, Berlin, Germany
| |
Collapse
|
210
|
Zhang L, Liang Y, Li S, Zeng F, Meng Y, Chen Z, Liu S, Tao Y, Yu F. The interplay of circulating tumor DNA and chromatin modification, therapeutic resistance, and metastasis. Mol Cancer 2019; 18:36. [PMID: 30849971 PMCID: PMC6408771 DOI: 10.1186/s12943-019-0989-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Peripheral circulating free DNA (cfDNA) is DNA that is detected in plasma or serum fluid with a cell-free status. For cancer patients, cfDNA not only originates from apoptotic cells but also from necrotic tumor cells and disseminated tumor cells that have escaped into the blood during epithelial-mesenchymal transition. Additionally, cfDNA derived from tumors, also known as circulating tumor DNA (ctDNA), carries tumor-associated genetic and epigenetic changes in cancer patients, which makes ctDNA a potential biomarker for the early diagnosis of tumors, monitory and therapeutic evaluations, and prognostic assessments, among others, for various kinds of cancer. Moreover, analyses of cfDNA chromatin modifications can reflect the heterogeneity of tumors and have potential for predicting tumor drug resistance.
Collapse
Affiliation(s)
- Lei Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yiyi Liang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Shifu Li
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Fanyuan Zeng
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yongan Meng
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ziwei Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China.
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Fenglei Yu
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
211
|
Dzobo K, Rowe A, Senthebane DA, AlMazyadi MAM, Patten V, Parker MI. Three-Dimensional Organoids in Cancer Research: The Search for the Holy Grail of Preclinical Cancer Modeling. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:733-748. [PMID: 30571609 DOI: 10.1089/omi.2018.0172] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most solid tumors become therapy resistant and will relapse, with no durable treatment option available. One major impediment to our understanding of cancer biology and finding innovative approaches to cancer treatment stems from the lack of better preclinical tumor models that address and explain tumor heterogeneity and person-to-person differences in therapeutic and toxic responses. Past cancer research has been driven by inadequate in vitro assays utilizing two-dimensional monolayers of cancer cells and animal models. Additionally, animal models do not truly mimic the original human tumor, are time consuming, and usually costly. New preclinical models are needed for innovation in cancer translational research. Hence, it is time to welcome the three-dimensional (3D) organoids: self-organizing cells grown in 3D culture systems mimicking the parent tissues from which the primary cells originate. The 3D organoids offer deeper insights into the crucial cellular processes in tissue and organ formation and pathological processes. Generation of near-perfect physiological microenvironments allow 3D organoids to couple with gene editing tools, such as the clustered regularly interspersed short palindromic repeat (CRISPR)/CRISPR-associated 9 and the transcription activator-like effector nucleases to model human diseases, offering distinct advantages over current models. We explain in this expert review that through recapitulating patients' normal and tumor tissues, organoid technology can markedly advance personalized medicine and help reveal once hidden aspects of cancers. The use of defined tissue- or organ-specific matrices, among other factors, will likely allow organoid technology to realize its potential in innovating many fields of life sciences.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Center for Genetic Engineering and Biotechnology (ICGEB) , Cape Town Component, Cape Town, South Africa .,2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| | - Arielle Rowe
- 1 International Center for Genetic Engineering and Biotechnology (ICGEB) , Cape Town Component, Cape Town, South Africa
| | - Dimakatso A Senthebane
- 1 International Center for Genetic Engineering and Biotechnology (ICGEB) , Cape Town Component, Cape Town, South Africa .,2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| | - Mousa A M AlMazyadi
- 3 Al-Ahsa College of Medicine, King Faisal University , Al-Ahsa, Kingdom of Saudi Arabia
| | - Victoria Patten
- 2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| | - M Iqbal Parker
- 2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town , Cape Town, South Africa
| |
Collapse
|
212
|
Corcoran RB. Circulating Tumor DNA: Clinical Monitoring and Early Detection. ANNUAL REVIEW OF CANCER BIOLOGY 2019. [DOI: 10.1146/annurev-cancerbio-030518-055719] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Roughly 70 years after the presence of cell-free DNA (cfDNA) in circulating blood was discovered, cfDNA has emerged as a transformative technology in clinical oncology. The ability to assess the presence, level, and composition of tumor DNA from a routine, noninvasive blood draw has opened the door to a broad array of high-impact clinical applications. While cfDNA is rapidly gaining clinical favor as a means of tumor mutational profiling without the need for an invasive biopsy, emerging applications in the areas of clinical monitoring and early cancer detection hold tremendous promise. These developing applications of cfDNA are reviewed herein.
Collapse
Affiliation(s)
- Ryan B. Corcoran
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
213
|
A Study of Pre-Analytical Variables and Optimization of Extraction Method for Circulating Tumor DNA Measurements by Digital Droplet PCR. Cancer Epidemiol Biomarkers Prev 2019; 28:909-916. [DOI: 10.1158/1055-9965.epi-18-0586] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/13/2018] [Accepted: 02/25/2019] [Indexed: 11/16/2022] Open
|
214
|
Chatterjee N, Bivona TG. Polytherapy and Targeted Cancer Drug Resistance. Trends Cancer 2019; 5:170-182. [PMID: 30898264 PMCID: PMC6446041 DOI: 10.1016/j.trecan.2019.02.003] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 02/07/2023]
Abstract
A current challenge in cancer treatment is drug resistance. Even the most effective therapies often fail to produce a complete and durable tumor response and ultimately give rise to therapy resistance and tumor relapse. However, how resistance arises in cancer remains incompletely understood. While drug resistance in cancer is thought to be driven by irreversible genetic mutations, emerging evidence also implicates reversible proteomic and epigenetic mechanisms in the development of drug resistance. Tumor microenvironment-mediated mechanisms and tumor heterogeneity can significantly contribute to cancer treatment resistance. Here, we discuss the diverse and dynamic strategies that cancers use to evade drug response, the promise of upfront combination and intermittent therapies and therapy switching in forestalling resistance, and epigenetic reprogramming to combat resistance.
Collapse
Affiliation(s)
- Nilanjana Chatterjee
- Department of Medicine, University of California, San Francisco, 600 16(th) Street, Box 2140, Genentech Hall, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 600 16(th) Street, Box 2140, Genentech Hall, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 600 16(th) Street, Box 2140, Genentech Hall, San Francisco, CA 94158, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, 600 16(th) Street, Box 2140, Genentech Hall, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 600 16(th) Street, Box 2140, Genentech Hall, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 600 16(th) Street, Box 2140, Genentech Hall, San Francisco, CA 94158, USA.
| |
Collapse
|
215
|
Yaeger R, Corcoran RB. Targeting Alterations in the RAF-MEK Pathway. Cancer Discov 2019; 9:329-341. [PMID: 30770389 PMCID: PMC6397699 DOI: 10.1158/2159-8290.cd-18-1321] [Citation(s) in RCA: 301] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/26/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022]
Abstract
The MAPK pathway is one of the most commonly mutated oncogenic pathways in cancer. Although RAS mutations are the most frequent MAPK alterations, less frequent alterations in downstream components of the pathway, including the RAF and MEK genes, offer promising therapeutic opportunities. In addition to BRAFV600 mutations, for which several approved therapeutic regimens exist, other alterations in the RAF and MEK genes may provide more rare, but tractable, targets. However, recent studies have illustrated the complexity of MAPK signaling and highlighted that distinct alterations in these genes may have strikingly different properties. Understanding the unique functional characteristics of specific RAF and MEK alterations, reviewed herein, will be critical for developing effective therapeutic approaches for these targets. SIGNIFICANCE: Alterations in the RAF and MEK genes represent promising therapeutic targets in multiple cancer types. However, given the unique and complex signaling biology of the MAPK pathway, the diverse array of RAF and MEK alterations observed in cancer can possess distinct functional characteristics. As outlined in this review, understanding the key functional properties of different RAF and MEK alterations is fundamental to selecting the optimal therapeutic approach.
Collapse
Affiliation(s)
- Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Ryan B Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
216
|
Tumor Heterogeneity as a Predictor of Response to Neoadjuvant Chemotherapy in Locally Advanced Rectal Cancer. Clin Colorectal Cancer 2019; 18:102-109. [PMID: 30935775 DOI: 10.1016/j.clcc.2019.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/04/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (nCRT) is the standard of care for locally advanced adenocarcinoma of the rectum, but it is currently unknown which patients have disease that will respond. This study tested the correlation between response to nCRT and intratumoral heterogeneity using next-generation sequencing assays. PATIENTS AND METHODS DNA was extracted from formalin-fixed, paraffin-embedded biopsy samples from a cohort of patients with locally advanced rectal adenocarcinoma (T3/4 or N1/2 disease) who received nCRT. High read-depth sequencing of > 400 cancer-relevant genes was performed. Tumor mutations and variant allele frequencies were used to calculate mutant-allele tumor heterogeneity (MATH) scores as measures of intratumoral heterogeneity. Response to nCRT was pathologically scored after surgical resection. RESULTS Biopsy samples from 21 patient tumors were analyzed. Eight patients had disease noted to have complete response, 2 moderate, 4 minimal, and 7 poor. Higher MATH scores correlated with poorer response to treatment, demonstrating significantly increased tumor heterogeneity compared to complete response (P = .039). CONCLUSION The application of MATH scores as a measure of tumor heterogeneity may provide a useful biomarker for treatment response in locally advanced rectal cancer.
Collapse
|
217
|
Seoane J, De Mattos-Arruda L, Le Rhun E, Bardelli A, Weller M. Cerebrospinal fluid cell-free tumour DNA as a liquid biopsy for primary brain tumours and central nervous system metastases. Ann Oncol 2019; 30:211-218. [PMID: 30576421 DOI: 10.1093/annonc/mdy544] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Challenges in obtaining tissue specimens from patients with brain tumours limit the diagnosis and molecular characterisation and impair the development of better therapeutic approaches. The analysis of cell-free tumour DNA in plasma (considered a liquid biopsy) has facilitated the characterisation of extra-cranial tumours. However, cell-free tumour DNA in plasma is limited in quantity and may not reliably capture the landscape of genomic alterations of brain tumours. Here, we review recent work assessing the relevance of cell-free tumour DNA from cerebrospinal fluid in the characterisation of brain cancer. We focus on the advances in the use of the cerebrospinal fluid as a source of cell-free tumour DNA to facilitate diagnosis, reveal actionable genomic alterations, monitor responses to therapy, and capture tumour heterogeneity in patients with primary brain tumours and brain and leptomeningeal metastases. Profiling cerebrospinal fluid cell-free tumour DNA provides the opportunity to precisely acquire and monitor genomic information in real time and guide precision therapies.
Collapse
Affiliation(s)
- J Seoane
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona; CIBERONC, Barcelona; Universitat Autònoma de Barcelona, Cerdanyola del Vallès.
| | - L De Mattos-Arruda
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona
| | - E Le Rhun
- Lille University, Inserm U1192 PRISM, Villeneuve d'Ascq; Neuro-oncology, Department of Neurosurgery, University Hospital, Lille; Neuro-oncology, Breast Unit, Department of Medical Oncology, Oscar Lambret Center, Lille, France
| | - A Bardelli
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO); Department of Oncology, University of Torino, Candiolo (TO), Italy
| | - M Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
218
|
Kato S, Schwaederlé MC, Fanta PT, Okamura R, Leichman L, Lippman SM, Lanman RB, Raymond VM, Talasaz A, Kurzrock R. Genomic Assessment of Blood-Derived Circulating Tumor DNA in Patients With Colorectal Cancers: Correlation With Tissue Sequencing, Therapeutic Response, and Survival. JCO Precis Oncol 2019; 3:PO.18.00158. [PMID: 31032472 PMCID: PMC6484865 DOI: 10.1200/po.18.00158] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
PURPOSE Genomic alterations in blood-derived circulating tumor DNA (ctDNA) from patients with colorectal cancers were correlated with clinical outcomes. PATIENTS AND METHODS Next-generation sequencing of ctDNA (54- to 73-gene panel) was performed in 94 patients with colorectal cancer. RESULTS Most patients (96%) had metastatic or recurrent disease at the time of blood draw. The median number of nonsynonymous alterations per patient was three (range, zero to 30). The most frequently aberrant genes were TP53 (52.1% of patients), KRAS (34%), and APC (28.7%). Concordance between tissue and blood next-generation sequencing ranged from 63.2% (APC) to 85.5% (BRAF). Altogether, 74 patients (79%) had one or more nonsynonymous alterations, 69 (73%) had one or more potentially actionable alterations, and 61 (65%) had an alteration actionable by a drug approved by the US Food and Drug Administration (on or off label). Lung metastases correlated with improved survival from diagnosis in univariable analysis. ctDNA of 5% or more from blood tests as well as EGFR and ERBB2 (HER2) nonsynonymous alterations correlated with worse survival (but only ERBB2 remained significant in multivariable analysis). No two patients had identical molecular portfolios. Overall, 65% versus 31% of patients treated with matched (n = 17) versus unmatched therapy (n = 18) after ctDNA testing achieved stable disease for 6 months or more, partial response, or complete response (P = .045); progression-free survival, 6.1 versus 2.3 months (P = .08); and survival not reached versus 9.4 months (P = .146; all by multivariable analysis). CONCLUSION Patients with colorectal cancer have heterogeneous ctDNA profiles, and most harbor potentially actionable ctDNA alterations. Matched therapy yielded higher rates of stable disease for 6 months or more, partial response, or complete response. ctDNA assessment may have clinical utility and merits further investigation.
Collapse
Affiliation(s)
- Shumei Kato
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Maria C. Schwaederlé
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Paul T. Fanta
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Ryosuke Okamura
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Lawrence Leichman
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Scott M. Lippman
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Richard B. Lanman
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Victoria M. Raymond
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - AmirAli Talasaz
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| | - Razelle Kurzrock
- Shumei Kato, Maria C. Schwaederlé, Paul T. Fanta, Ryosuke Okamura, Lawrence Leichman, Scott M. Lippman, and Razelle Kurzrock, University of California San Diego Moores Cancer Center, La Jolla; and Richard B. Lanman, Victoria M. Raymond, and AmirAli Talasaz, Guardant Health, Redwood City, CA
| |
Collapse
|
219
|
Sabnis AJ, Bivona TG. Principles of Resistance to Targeted Cancer Therapy: Lessons from Basic and Translational Cancer Biology. Trends Mol Med 2019; 25:185-197. [PMID: 30686761 DOI: 10.1016/j.molmed.2018.12.009] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/23/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022]
Abstract
Identification of the genomic drivers of cancer has led to the clinical development of targeted therapies that strike at the heart of many malignancies. Nonetheless, many cancers outsmart such precision-medicine efforts, and thus therapeutic resistance contributes significantly to cancer mortality. Attempts to understand the basis for resistance in patient samples and laboratory models has yielded two major benefits: one, more effective chemical inhibitors and rational combination therapies are now employed to prevent or circumvent resistance pathways; and two, our understanding of how oncogenic mutations drive cancer cell survival and oncogene addiction is deeper and broader, highlighting downstream or parallel cellular programs that shape these phenotypes. This review discusses emerging principles of resistance to therapies targeted against key oncogenic drivers.
Collapse
Affiliation(s)
- Amit J Sabnis
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
220
|
Giordano G, Parcesepe P, D’Andrea MR, Coppola L, Di Raimo T, Remo A, Manfrin E, Fiorini C, Scarpa A, Amoreo CA, Conciatori F, Milella M, Caruso FP, Cerulo L, Porras A, Pancione M. JAK/Stat5-mediated subtype-specific lymphocyte antigen 6 complex, locus G6D (LY6G6D) expression drives mismatch repair proficient colorectal cancer. J Exp Clin Cancer Res 2019; 38:28. [PMID: 30670049 PMCID: PMC6343337 DOI: 10.1186/s13046-018-1019-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/26/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Human microsatellite-stable (MSS) colorectal cancers (CRCs) are immunologically "cold" tumour subtypes characterized by reduced immune cytotoxicity. The molecular linkages between immune-resistance and human MSS CRC is not clear. METHODS We used transcriptome profiling, in silico analysis, immunohistochemistry, western blot, RT-qPCR and immunofluorescence staining to characterize novel CRC immune biomarkers. The effects of selective antagonists were tested by in vitro assays of long term viability and analysis of kinase active forms using anti-phospho antibodies. RESULTS We identified the lymphocyte antigen 6 complex, locus G6D (LY6G6D) as significantly overexpressed (around 15-fold) in CRC when compared with its relatively low expression in other human solid tumours. LY6G6D up-regulation was predominant in MSS CRCs characterized by an enrichment of immune suppressive regulatory T-cells and a limited repertoire of PD-1/PD-L1 immune checkpoint receptors. Coexpression of LY6G6D and CD15 increases the risk of metastatic relapse in response to therapy. Both JAK-STAT5 and RAS-MEK-ERK cascades act in concert as key regulators of LY6G6D and Fucosyltransferase 4 (FUT4), which direct CD15-mediated immune-resistance. Momelotinib, an inhibitor of JAK1/JAK2, consistently abrogated the STAT5/LY6G6D axis in vitro, sensitizing MSS cancer cells with an intact JAK-STAT signaling, to efficiently respond to trametinib, a MEK inhibitor used in clinical setting. Notably, colon cancer cells can evade JAK2/JAK1-targeted therapy by a reversible shift of the RAS-MEK-ERK pathway activity, which explains the treatment failure of JAK1/2 inhibitors in refractory CRC. CONCLUSIONS Combined targeting of STAT5 and MAPK pathways has superior therapeutic effects on immune resistance. In addition, the new identified LY6G6D antigen is a promising molecular target for human MSS CRC.
Collapse
Affiliation(s)
- Guido Giordano
- Oncology Unit, Casa Sollievo della Sofferenza-IRCCS, San Giovanni Rotondo, Italy
- Medical Oncology and Anatomic Pathology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Pietro Parcesepe
- Department of Diagnostics and Public Health – Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | | | - Luigi Coppola
- Medical Oncology and Anatomic Pathology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Tania Di Raimo
- Medical Oncology and Anatomic Pathology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Andrea Remo
- Pathology Unit, “Mater Salutis” Hospital AULSS9, Legnago (Verona), Italy
| | - Erminia Manfrin
- Department of Diagnostics and Public Health – Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Claudia Fiorini
- Department of Diagnostics and Public Health – Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health – Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Carla Azzurra Amoreo
- Pathology, IRCCS Regina Elena National Cancer Institute, Rome Italy, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Fabiana Conciatori
- Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome Italy, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Michele Milella
- Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome Italy, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Francesca Pia Caruso
- Department of Sciences and Technologies, University of Sannio, Via Port’Arsa, 1182100 Benevento, Italy
- Bioinformatics Laboratory, BIOGEM scrl, Ariano Irpino, Avellino, Italy
| | - Luigi Cerulo
- Department of Sciences and Technologies, University of Sannio, Via Port’Arsa, 1182100 Benevento, Italy
- Bioinformatics Laboratory, BIOGEM scrl, Ariano Irpino, Avellino, Italy
| | - Almudena Porras
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University Madrid, Madrid, Spain
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, Via Port’Arsa, 1182100 Benevento, Italy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University Madrid, Madrid, Spain
| |
Collapse
|
221
|
Molecular Analysis of Plasma From Patients With ROS1-Positive NSCLC. J Thorac Oncol 2019; 14:816-824. [PMID: 30664990 DOI: 10.1016/j.jtho.2019.01.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Circulating tumor DNA analysis is an emerging genotyping strategy that can identify tumor-specific genetic alterations in plasma including mutations and rearrangements. Detection of ROS1 fusions in plasma requires genotyping approaches that cover multiple breakpoints and target a variety of fusion partners. Compared to other molecular subsets of NSCLC, experience with detecting ROS1 genetic alterations in plasma is limited. METHODS To describe the spectrum of ROS1 fusions in NSCLC and determine sensitivity for detecting ROS1 fusions in plasma, we queried the Guardant Health plasma dataset and an institutional tissue database and compared plasma findings to tissue results. In addition, we used the Guardant360 NGS assay to detect potential genetic mediators of resistance in plasma from patients with ROS1-positive NSCLC who were relapsing on crizotinib. RESULTS We detected seven distinct fusion partners in plasma, most of which (n = 6 of 7) were also represented in the tissue dataset. Fusions pairing CD74 with ROS1 predominated in both cohorts (plasma: n = 35 of 56, 63%; tissue: n = 26 of 52, 50%). There was 100% concordance between the specific tissue- and plasma-detected ROS1 fusion for seven patients genotyped with both methods. Sensitivity for detecting ROS1 fusions in plasma at relapse on ROS1-directed therapy was 50%. Six (33%) of 18 post-crizotinib plasma specimens harbored ROS1 kinase domain mutations, five of which were ROS1 G2032R. Two (11%) post-crizotinib plasma specimens had genetic alterations (n = 1 each BRAF V600E and PIK3CA E545K) potentially associated with ROS1-independent signaling. CONCLUSIONS Plasma genotyping captures the spectrum of ROS1 fusions observed in tissue. Plasma genotyping is a promising approach to detecting mutations that drive resistance to ROS1-directed therapies.
Collapse
|
222
|
Lieu CH, Corcoran RB, Overman MJ. Integrating Biomarkers and Targeted Therapy Into Colorectal Cancer Management. Am Soc Clin Oncol Educ Book 2019; 39:207-215. [PMID: 31099678 DOI: 10.1200/edbk_240839] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
There have been substantial advances in the treatment of metastatic colorectal cancer (mCRC) over the past 15 years. Molecular characteristics of mCRC and identification of specific mutations can serve as predictive and prognostic indicators of disease and response to targeted therapies. When incorporated into clinical decision-making, these biomarkers can serve as critical tools in personalizing therapy to ensure the best outcomes. Additional improvements in the survival of patients with mCRC will be made possible with the identification of new predictive molecular biomarkers and their evaluation using rational and innovative clinical trials.
Collapse
Affiliation(s)
| | | | - Michael J Overman
- 3 Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
223
|
Circulating tumor DNA applications in monitoring the treatment of metastatic colorectal cancer patients. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2019. [PMCID: PMC7009314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colorectal cancer is the third most common cancer worldwide. New cancer treatment strategies such as monoclonal antibodies against growth factor and angiogenesis receptors have improved the overall survival (OS) and progression-free survival (PFS) in metastatic colorectal cancer (mCRC) patients. However, acquired resistance could happen after these therapies. Circulating tumor DNA (ctDNA) is the DNA fraction derived from tumor cells which could be applied as a non-invasive method for detecting tumor mutations before, during, and after therapies. Here, we reviewed most of the studies examining ctDNA as treatment monitoring in mCRC patients who receive different target therapies. Also, we compared ctDNA with other existing cancer-treatment monitoring methods.
Collapse
|
224
|
Sierant MC, Choi J. Single-Cell Ssequencing in Cancer: Recent Applications to Immunogenomics and Multi-omics Tools. Genomics Inform 2018; 16:e17. [PMID: 30602078 PMCID: PMC6440661 DOI: 10.5808/gi.2018.16.4.e17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor heterogeneity, the cellular mosaic of multiple lineages arising from the process of clonal evolution, has continued to thwart multi-omics analyses using traditional bulk sequencing methods. The application of single-cell sequencing, in concert with existing genomics methods, has enabled high-resolution interrogation of the genome, transcriptome, epigenome, and proteome. Applied to cancers, these single-cell multi-omics methods bypass previous limitations on data resolution and have enabled a more nuanced understanding of the evolutionary dynamics of tumor progression, immune evasion, metastasis, and treatment resistance. This review details the growing number of novel single-cell multi-omics methods applied to tumors and further discusses recent discoveries emerging from these approaches, especially in regard to immunotherapy.
Collapse
Affiliation(s)
- Michael C Sierant
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jungmin Choi
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA.,Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
225
|
Wu Z, Fournel L, Stadler N, Liu J, Boullier A, Hoyeau N, Fléjou JF, Duchatelle V, Djebrani-Oussedik N, Agopiantz M, Ségal-Bendirdjian E, Gompel A, Alifano M, Melander O, Trédaniel J, Forgez P. Modulation of lung cancer cell plasticity and heterogeneity with the restoration of cisplatin sensitivity by neurotensin antibody. Cancer Lett 2018; 444:147-161. [PMID: 30583074 DOI: 10.1016/j.canlet.2018.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022]
Abstract
Overall survival of patients with metastatic non-small cell lung cancer (NSCLC) has significantly improved with platinum-based salt treatments and recently with targeted therapies and immunotherapies. However, treatment failure occurs due to acquired or emerging tumor resistance. We developed a monoclonal antibody against the proform of neurotensin (LF-NTS mAb) that alters the homeostasis of tumors overexpressing NTSR1. Neurotensin is frequently overexpressed along with its high affinity receptor (NTSR1) in tumors from epithelial origins. This ligand/receptor complex contributes to the progression of many tumor types by activation of the cellular effects involved in tumor progression (proliferation, survival, migration, and invasion). We demonstrate that LF-NTS mAb operates on the plasticity of tumor cells overexpressing NTSR1 and lowers their aggressiveness. The mAb enables the restoration of platinum-based therapies responsiveness, while also decreasing metastatic processes. Efficacy dosage with long-term treatment showed no obvious adverse events, while demonstrating improvement in the performance status. Our data suggests that LF-NTS mAb is an ideal candidate to be safely added to the conventional standard of care in order to improve its efficacy.
Collapse
Affiliation(s)
- Zherui Wu
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France
| | - Ludovic Fournel
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France; Department of Thoracic Surgery, Cochin Hospital of Paris, AP-HP, Paris Descartes University, Paris, France; Paris-Sud, Paris-Saclay University, Orsay, France
| | - Nicolas Stadler
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France
| | - Jin Liu
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France
| | - Agnès Boullier
- CBH Biochemistry Laboratory, CHU Amiens-Picardie, Amiens, France
| | - Nadia Hoyeau
- Department of Pathology, Saint-Antoine Hospital, AP-HP, UPMC, Paris, France
| | | | - Véronique Duchatelle
- Department of Pathology, Groupe Hospitalier Paris Saint Joseph, Paris Descartes University, Paris, France
| | | | - Mikaël Agopiantz
- Department of Pathology, CHRU Nancy, University of Lorraine, Nancy, France
| | | | - Anne Gompel
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France; Department of Gynecology Obstetrics II and Reproductive Medicine, Paris Descartes University, AP-HP, Paris, France
| | - Marco Alifano
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France; Department of Thoracic Surgery, Cochin Hospital of Paris, AP-HP, Paris Descartes University, Paris, France
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Skåne University Hospital, CRC, Malmö, Sweden
| | - Jean Trédaniel
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France; Unit of Thoracic Oncology, Groupe Hospitalier Paris Saint Joseph, Paris Descartes University, Paris, France.
| | - Patricia Forgez
- INSERM UMRS 1007, Paris Descartes University, 75270, Paris Cedex 06, France.
| |
Collapse
|
226
|
Receptor Tyrosine Kinase-Targeted Cancer Therapy. Int J Mol Sci 2018; 19:ijms19113491. [PMID: 30404198 PMCID: PMC6274851 DOI: 10.3390/ijms19113491] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 12/14/2022] Open
Abstract
In the past two decades, several molecular targeted inhibitors have been developed and evaluated clinically to improve the survival of patients with cancer. Molecular targeted inhibitors inhibit the activities of pathogenic tyrosine kinases. Particularly, aberrant receptor tyrosine kinase (RTK) activation is a potential therapeutic target. An increased understanding of genetics, cellular biology and structural biology has led to the development of numerous important therapeutics. Pathogenic RTK mutations, deletions, translocations and amplification/over-expressions have been identified and are currently being examined for their roles in cancers. Therapies targeting RTKs are categorized as small-molecule inhibitors and monoclonal antibodies. Studies are underway to explore abnormalities in 20 types of RTK subfamilies in patients with cancer or other diseases. In this review, we describe representative RTKs important for developing cancer therapeutics and predicting or evaluated resistance mechanisms.
Collapse
|
227
|
Affiliation(s)
- Ryan B Corcoran
- From the Massachusetts General Hospital Cancer Center and the Department of Medicine, Harvard Medical School, Boston
| | - Bruce A Chabner
- From the Massachusetts General Hospital Cancer Center and the Department of Medicine, Harvard Medical School, Boston
| |
Collapse
|
228
|
Nakamura Y, Yoshino T. Clinical Utility of Analyzing Circulating Tumor DNA in Patients with Metastatic Colorectal Cancer. Oncologist 2018; 23:1310-1318. [PMID: 29700206 PMCID: PMC6291328 DOI: 10.1634/theoncologist.2017-0621] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/14/2018] [Indexed: 12/28/2022] Open
Abstract
Multiple genomic changes caused by clonal evolution induced by therapeutic pressure and corresponding intratumoral heterogeneity have posed great challenges for personalized therapy against metastatic colorectal cancer (mCRC) in the past decade. Liquid biopsy has emerged as an excellent molecular diagnostic tool for assessing predominant spatial and temporal intratumoral heterogeneity with minimal invasiveness.Previous studies have revealed that genomic alterations in RAS, BRAF, ERBB2, and MET, as well as other cancer-related genes associated with resistance to anti-epidermal growth factor receptor (EGFR) therapy, can be analyzed with high diagnostic accuracy by circulating tumor DNA (ctDNA) analysis. Furthermore, by longitudinally monitoring ctDNAs during anti-EGFR therapy, the emergence of genomic alterations can be detected as acquired resistance mechanisms in specific genes, mainly those associated with the mitogen-activated protein kinase signaling pathway. Analysis of ctDNA can also identify predictive biomarkers to immune checkpoint inhibitors, such as mutations in mismatch repair genes, microsatellite instability-high phenotype, and tumor mutation burden. Some prospective clinical trials evaluating targeted agents for genomic alterations in ctDNA or exploring resistance biomarkers by monitoring of ctDNA are ongoing.To determine the value of ctDNA analysis for decision-making by more accurate molecular marker-based selection of patients and identification of resistance mechanisms to targeted therapies or sensitive biomarkers for immune checkpoint inhibitors, clinical trials must be refined to evaluate the efficacy of study treatment in patients with targetable genomic alterations confirmed by ctDNA analysis, and resistance biomarkers should be explored by monitoring ctDNA in large-scale clinical trials. In the near future, ctDNA analysis will play an important role in precision medicine for mCRC. IMPLICATIONS FOR PRACTICE: Treatment strategies for metastatic colorectal cancer (mCRC) are determined according to the molecular profile, which is confirmed by analyzing tumor tissue. Analysis of circulating tumor DNA (ctDNA) may overcome the limitations of tissue-based analysis by capturing spatial and temporal intratumoral heterogeneity of mCRC. Clinical trials must be refined to test the value of ctDNA analysis in patient selection and identification of biomarkers. This review describes ctDNA analysis, which will have an important role in precision medicine for mCRC.
Collapse
Affiliation(s)
- Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| |
Collapse
|
229
|
Wei XL, Wu QN, Chen DL, Zeng ZL, Lu JB, Liu ZX, Ju HQ, Ren C, Pan ZZ, Wang FH, Xu RH. The Clinical and Biomarker Association of Programmed Death Ligand 1 and its Spatial Heterogeneous Expression in Colorectal Cancer. J Cancer 2018; 9:4325-4333. [PMID: 30519336 PMCID: PMC6277645 DOI: 10.7150/jca.27735] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Background: Programmed death ligand 1 (PD-L1) expression has been shown to predict benefit from anti-PD-1 treatment in several cancers. However, its predictive value in colorectal cancer seems limited. This study was aimed to explore the clinical and biomarker association of programmed death ligand 1 and its spatial heterogeneous expression in colorectal cancer. Methods: Tissue microarrays of 422 primary colorectal cancers from our hospital were used for the interpretation of PD-L1 and programmed death 1 (PD-1) expression, cluster of differentiation 4 (CD4) and CD8 density and microsatellite instability (MSI) status by immunohistochemistry. To assess the spatial heterogeneity of PD-L1 expression, Tissue microarrays of 383 paired intra-primary-tumor tissues, and 105 paired lymph node metastatic tumors and 64 paired distant metastatic tumors were also used. Results: PD-L1 was positive in 188 (44.5%) primary colorectal cancers. PD-L1 expression was associated with less advanced N category (P<0.001), less advanced TNM stage (P<0.001) and less nervous invasion (P=0.04). Higher PD-L1 expression was associated with higher PD-1 expression (P<0.001), higher CD4 (P<0.001) and CD8 (P<0.001) density and DNA mismatch repair deficiency (P=0.01). PD-L1 expression was associated with better disease-free survival and overall survival, but it was only an independent prognostic factor for disease-free survival (hazard ratio and 95% confidence interval: 0.42 [0.25-0.72], P<0.001). The probability of inconsistent PD-L1 expression was respectively 17.8%, 31.4% and 39.1% within primary tumors, between primary tumors and lymph node metastatic tumors, and between primary tumors and distant metastatic tumors. All the three differences were statistically significant (P<0.001, P<0.001 and P=0.05, respectively). Conclusions: PD-L1 expression was a marker of pre-existing immune responses in colorectal cancer, however, it was heterogeneously expressed in colorectal cancer, especially between primary and metastatic tumors. This might partially explain the low-efficiency of its predictive value for benefit from anti-PD-1 treatment.
Collapse
Affiliation(s)
- Xiao-Li Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Qi-Nian Wu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Dong-Liang Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Zhao-Lei Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jia-Bin Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Chao Ren
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
230
|
Elazezy M, Joosse SA. Techniques of using circulating tumor DNA as a liquid biopsy component in cancer management. Comput Struct Biotechnol J 2018; 16:370-378. [PMID: 30364656 PMCID: PMC6197739 DOI: 10.1016/j.csbj.2018.10.002] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022] Open
Abstract
Precision medicine in the clinical management of cancer may be achieved through the diagnostic platform called “liquid biopsy”. This method utilizes the detection of biomarkers in blood for prognostic and predictive purposes. One of the latest blood born markers under investigation in the field of liquid biopsy in cancer patients is circulating tumor DNA (ctDNA). ctDNA is released by tumor cells through different mechanisms and can therefore provide information about the genomic make-up of the tumor currently present in the patient. Through longitudinal ctDNA-based liquid biopsies, tumor dynamics may be monitored to predict and assess drug response and/or resistance. However, because ctDNA is highly fragmented and because its concentration can be extremely low in a high background of normal circulating DNA, screening for clinical relevant mutations is challenging. Although significant progress has been made in advancing the detection and analysis of ctDNA in the last few years, the current challenges include standardization and increasing current techniques to single molecule sensitivity in combination with perfect specificity. This review focuses on the potential role of ctDNA in the clinical management of cancer patients, the current technologies that are being employed, and the hurdles that still need to be taken to achieve ctDNA-based liquid biopsy towards precision medicine.
Collapse
Affiliation(s)
- Maha Elazezy
- University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Simon A Joosse
- University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
231
|
Khan KH, Cunningham D, Werner B, Vlachogiannis G, Spiteri I, Heide T, Mateos JF, Vatsiou A, Lampis A, Damavandi MD, Lote H, Huntingford IS, Hedayat S, Chau I, Tunariu N, Mentrasti G, Trevisani F, Rao S, Anandappa G, Watkins D, Starling N, Thomas J, Peckitt C, Khan N, Rugge M, Begum R, Hezelova B, Bryant A, Jones T, Proszek P, Fassan M, Hahne JC, Hubank M, Braconi C, Sottoriva A, Valeri N. Longitudinal Liquid Biopsy and Mathematical Modeling of Clonal Evolution Forecast Time to Treatment Failure in the PROSPECT-C Phase II Colorectal Cancer Clinical Trial. Cancer Discov 2018; 8:1270-1285. [PMID: 30166348 PMCID: PMC6380469 DOI: 10.1158/2159-8290.cd-17-0891] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 05/01/2018] [Accepted: 07/05/2018] [Indexed: 12/14/2022]
Abstract
Sequential profiling of plasma cell-free DNA (cfDNA) holds immense promise for early detection of patient progression. However, how to exploit the predictive power of cfDNA as a liquid biopsy in the clinic remains unclear. RAS pathway aberrations can be tracked in cfDNA to monitor resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. In this prospective phase II clinical trial of single-agent cetuximab in RAS wild-type patients, we combine genomic profiling of serial cfDNA and matched sequential tissue biopsies with imaging and mathematical modeling of cancer evolution. We show that a significant proportion of patients defined as RAS wild-type based on diagnostic tissue analysis harbor aberrations in the RAS pathway in pretreatment cfDNA and, in fact, do not benefit from EGFR inhibition. We demonstrate that primary and acquired resistance to cetuximab are often of polyclonal nature, and these dynamics can be observed in tissue and plasma. Furthermore, evolutionary modeling combined with frequent serial sampling of cfDNA allows prediction of the expected time to treatment failure in individual patients. This study demonstrates how integrating frequently sampled longitudinal liquid biopsies with a mathematical framework of tumor evolution allows individualized quantitative forecasting of progression, providing novel opportunities for adaptive personalized therapies.Significance: Liquid biopsies capture spatial and temporal heterogeneity underpinning resistance to anti-EGFR monoclonal antibodies in colorectal cancer. Dense serial sampling is needed to predict the time to treatment failure and generate a window of opportunity for intervention. Cancer Discov; 8(10); 1270-85. ©2018 AACR. See related commentary by Siravegna and Corcoran, p. 1213 This article is highlighted in the In This Issue feature, p. 1195.
Collapse
Affiliation(s)
- Khurum H Khan
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Benjamin Werner
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Georgios Vlachogiannis
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Inmaculada Spiteri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Timon Heide
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Javier Fernandez Mateos
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Alexandra Vatsiou
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Mahnaz Darvish Damavandi
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Hazel Lote
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Ian Said Huntingford
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Somaieh Hedayat
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Nina Tunariu
- Department of Radiology, The Royal Marsden NHS Trust, Londonand Sutton, United Kingdom
| | - Giulia Mentrasti
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Francesco Trevisani
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Sheela Rao
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Gayathri Anandappa
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Janet Thomas
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Clare Peckitt
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Nasir Khan
- Department of Radiology, The Royal Marsden NHS Trust, Londonand Sutton, United Kingdom
| | - Massimo Rugge
- Department of Medicine and Surgical Pathology, University of Padua, Padua, Italy
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Blanka Hezelova
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Annette Bryant
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Thomas Jones
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Paula Proszek
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Matteo Fassan
- Department of Medicine and Surgical Pathology, University of Padua, Padua, Italy
| | - Jens C Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Michael Hubank
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Chiara Braconi
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom.
| | - Nicola Valeri
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom.
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| |
Collapse
|
232
|
Anandappa G, Chau I. Evolving Tissue and Circulating Biomarkers as Prognostic and Predictive Tools in Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0410-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
233
|
Jia J, Morse MA, Nagy RJ, Lanman RB, Strickler JH. Cell-Free DNA Profiling to Discover Mechanisms of Exceptional Response to Cabozantinib Plus Panitumumab in a Patient With Treatment Refractory Metastatic Colorectal Cancer. Front Oncol 2018; 8:305. [PMID: 30211110 PMCID: PMC6121109 DOI: 10.3389/fonc.2018.00305] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/19/2018] [Indexed: 01/06/2023] Open
Abstract
MET amplification is rare in treatment-naïve metastatic colorectal cancer (CRC) tumors, but can emerge as a mechanism of resistance to anti-EGFR therapies. Preclinical and clinical data suggest that patients with MET amplified tumors benefit from MET-targeted therapy. Cabozantinib is an inhibitor of multiple tyrosine kinases, included c-MET. Panitumumab is an inhibitor of EGFR. This report describes a patient with KRAS, NRAS, and BRAF wild-type metastatic CRC who experienced disease progression on all standard chemotherapy and anti-EGFR antibody therapy. The patient was enrolled in a clinical trial evaluating the combination of cabozantinib plus panitumumab. After only 6 weeks of treatment, the patient experienced a significant anti-tumor response. Although tumor tissue was negative for MET amplification, molecular profiling of cell-free DNA (cfDNA) revealed MET amplification. This case represents the first report showing the activity of cabozantinib in combination with panitumumab in a patient with metastatic CRC, and suggests that MET amplification in cfDNA may be a biomarker of response. A clinical trial targeting MET amplified metastatic CRC is currently underway.
Collapse
Affiliation(s)
- Jingquan Jia
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Michael A. Morse
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | | | | | - John H. Strickler
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
234
|
The liquid biopsy in the management of colorectal cancer patients: Current applications and future scenarios. Cancer Treat Rev 2018; 70:1-8. [PMID: 30053724 DOI: 10.1016/j.ctrv.2018.07.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/17/2018] [Indexed: 02/06/2023]
Abstract
The term liquid biopsy refers to the analysis of biomarkers in any body fluid, including blood, urine and cerebrospinal fluid. In cancer, liquid biopsy testing allows the analysis of tumor-derived DNA, RNA, miRNA and proteins that can be either cell-free or contained in circulating tumor cells (CTC), extracellular vesicles (EVs) or platelets. A number of studies suggest that liquid biopsy testing could have a relevant role in the management of colorectal cancer (CRC) patients at different stages of the disease. Analysis of cell-free DNA (cfDNA), CTC and/or miRNA can provide relevant information for the early diagnosis of CRC and the identification of minimal residual disease and, more generally, the evaluation of the risk of recurrence in early CRC patients. In addition, liquid biopsy testing might allow the assessment of prognostic and predictive biomarkers in metastatic CRC patients, and the monitoring of the response to treatment and of the clonal evolution of the disease. While a number of elegant studies have shown the potential of liquid biopsy in CRC, the possibility to use this approach in the daily clinical practice is still limited. The use of non-standardized methods, the small cohorts of patients analyzed, the lack of demonstration of a clear clinical benefit are the main limitations of the studies with liquid biopsy in CRC reported up to now. The potential of this approach and the steps that need still to be taken to translate these preliminary findings in the clinic are discussed in this review.
Collapse
|
235
|
Siravegna G, Lazzari L, Crisafulli G, Sartore-Bianchi A, Mussolin B, Cassingena A, Martino C, Lanman RB, Nagy RJ, Fairclough S, Rospo G, Corti G, Bartolini A, Arcella P, Montone M, Lodi F, Lorenzato A, Vanzati A, Valtorta E, Cappello G, Bertotti A, Lonardi S, Zagonel V, Leone F, Russo M, Balsamo A, Truini M, Di Nicolantonio F, Amatu A, Bonazzina E, Ghezzi S, Regge D, Vanzulli A, Trusolino L, Siena S, Marsoni S, Bardelli A. Radiologic and Genomic Evolution of Individual Metastases during HER2 Blockade in Colorectal Cancer. Cancer Cell 2018; 34:148-162.e7. [PMID: 29990497 DOI: 10.1016/j.ccell.2018.06.004] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/18/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023]
Abstract
Targeting HER2 is effective in 24% of ERBB2 amplified metastatic colorectal cancer; however, secondary resistance occurs in most of the cases. We studied the evolution of individual metastases during treatment to discover spatially resolved determinants of resistance. Circulating tumor DNA (ctDNA) analysis identified alterations associated with resistance in the majority of refractory patients. ctDNA profiles and lesion-specific radiographic reports revealed organ- or metastasis-private evolutionary patterns. When radiologic assessments documented progressive disease in target lesions, response to HER2 blockade was retained in other metastases. Genomic and functional analyses on samples and cell models from eight metastases of a patient co-recruited to a postmortem study unveiled lesion-specific evolutionary trees and pharmacologic vulnerabilities. Lesion size and contribution of distinct metastases to plasma ctDNA were correlated.
Collapse
Affiliation(s)
| | - Luca Lazzari
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; FIRC Institute of Molecular Oncology (IFOM), Milan, Italy; Department of Oncology, University of Torino, Candiolo, TO 10060, Italy
| | - Giovanni Crisafulli
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; Department of Oncology, University of Torino, Candiolo, TO 10060, Italy
| | | | | | - Andrea Cassingena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Cosimo Martino
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy
| | | | | | | | - Giuseppe Rospo
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy
| | - Giorgio Corti
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy
| | | | - Pamela Arcella
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy
| | - Monica Montone
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy
| | - Francesca Lodi
- Department of Oncology, University of Torino, Candiolo, TO 10060, Italy
| | | | - Alice Vanzati
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Emanuele Valtorta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | | | - Andrea Bertotti
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; Department of Oncology, University of Torino, Candiolo, TO 10060, Italy
| | - Sara Lonardi
- Istituto Oncologico Veneto - IRCCS, Oncologia Medica 1, Padova 35128, Italy
| | - Vittorina Zagonel
- Istituto Oncologico Veneto - IRCCS, Oncologia Medica 1, Padova 35128, Italy
| | - Francesco Leone
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; Department of Oncology, University of Torino, Candiolo, TO 10060, Italy
| | | | | | - Mauro Truini
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; Department of Oncology, University of Torino, Candiolo, TO 10060, Italy
| | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Erica Bonazzina
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Silvia Ghezzi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Daniele Regge
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy
| | - Angelo Vanzulli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan 20122, Italy
| | - Livio Trusolino
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; Department of Oncology, University of Torino, Candiolo, TO 10060, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan 20122, Italy
| | - Silvia Marsoni
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; FIRC Institute of Molecular Oncology (IFOM), Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, TO, Italy; Department of Oncology, University of Torino, Candiolo, TO 10060, Italy.
| |
Collapse
|
236
|
Labgaa I, Villacorta-Martin C, D'Avola D, Craig AJ, von Felden J, Martins-Filho SN, Sia D, Stueck A, Ward SC, Fiel MI, Mahajan M, Tabrizian P, Thung SN, Ang C, Friedman SL, Llovet JM, Schwartz M, Villanueva A. A pilot study of ultra-deep targeted sequencing of plasma DNA identifies driver mutations in hepatocellular carcinoma. Oncogene 2018; 37:3740-3752. [PMID: 29628508 PMCID: PMC6035113 DOI: 10.1038/s41388-018-0206-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 01/12/2018] [Accepted: 02/16/2018] [Indexed: 01/28/2023]
Abstract
Cellular components of solid tumors including DNA are released into the bloodstream, but data on circulating-free DNA (cfDNA) in hepatocellular carcinoma (HCC) are still scarce. This study aimed at analyzing mutations in cfDNA and their correlation with tissue mutations in patients with HCC. We included 8 HCC patients treated with surgical resection for whom we collected paired tissue and plasma/serum samples. We analyzed 45 specimens, including multiregional tumor tissue sampling (n = 24), peripheral blood mononuclear cells (PMBC, n = 8), plasma (n = 8) and serum (n = 5). Ultra-deep sequencing (5500× coverage) of all exons was performed in a targeted panel of 58 genes, including frequent HCC driver genes and druggable mutations. Mutations detected in plasma included known HCC oncogenes and tumor suppressors (e.g., TERT promoter, TP53, and NTRK3) as well as a candidate druggable mutation (JAK1). This approach increased the detection rates previously reported for mutations in plasma of HCC patients. A thorough characterization of cis mutations found in plasma confirmed their tumoral origin, which provides definitive evidence of the release of HCC-derived DNA fragments into the bloodstream. This study demonstrates that ultra-deep sequencing of cfDNA is feasible and can confidently detect somatic mutations found in tissue; these data reinforce the role of plasma DNA as a promising minimally invasive tool to interrogate HCC genetics.
Collapse
Affiliation(s)
- Ismail Labgaa
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Carlos Villacorta-Martin
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Delia D'Avola
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Unit and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Clinica Universidad de Navarra, Pamplona, Spain
| | - Amanda J Craig
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Johann von Felden
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastiao N Martins-Filho
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Department of Pathology and Laboratory Medicine, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Daniela Sia
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ashley Stueck
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Stephen C Ward
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - M Isabel Fiel
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Milind Mahajan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Parissa Tabrizian
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Swan N Thung
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Celina Ang
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Division of Hematology and Medical Oncology, New York, USA
| | - Scott L Friedman
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Josep M Llovet
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Translational Research Laboratory, BCLC Group, IDIBAPS, CIBEREHD, Hospital Clinic, Universitat de Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Myron Schwartz
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Augusto Villanueva
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Division of Hematology and Medical Oncology, New York, USA.
| |
Collapse
|
237
|
Aguirre AJ, Nowak JA, Camarda ND, Moffitt RA, Ghazani AA, Hazar-Rethinam M, Raghavan S, Kim J, Brais LK, Ragon D, Welch MW, Reilly E, McCabe D, Marini L, Anderka K, Helvie K, Oliver N, Babic A, Da Silva A, Nadres B, Van Seventer EE, Shahzade HA, St Pierre JP, Burke KP, Clancy T, Cleary JM, Doyle LA, Jajoo K, McCleary NJ, Meyerhardt JA, Murphy JE, Ng K, Patel AK, Perez K, Rosenthal MH, Rubinson DA, Ryou M, Shapiro GI, Sicinska E, Silverman SG, Nagy RJ, Lanman RB, Knoerzer D, Welsch DJ, Yurgelun MB, Fuchs CS, Garraway LA, Getz G, Hornick JL, Johnson BE, Kulke MH, Mayer RJ, Miller JW, Shyn PB, Tuveson DA, Wagle N, Yeh JJ, Hahn WC, Corcoran RB, Carter SL, Wolpin BM. Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine. Cancer Discov 2018; 8:1096-1111. [PMID: 29903880 DOI: 10.1158/2159-8290.cd-18-0275] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/17/2018] [Accepted: 06/13/2018] [Indexed: 12/28/2022]
Abstract
Clinically relevant subtypes exist for pancreatic ductal adenocarcinoma (PDAC), but molecular characterization is not yet standard in clinical care. We implemented a biopsy protocol to perform time-sensitive whole-exome sequencing and RNA sequencing for patients with advanced PDAC. Therapeutically relevant genomic alterations were identified in 48% (34/71) and pathogenic/likely pathogenic germline alterations in 18% (13/71) of patients. Overall, 30% (21/71) of enrolled patients experienced a change in clinical management as a result of genomic data. Twenty-six patients had germline and/or somatic alterations in DNA-damage repair genes, and 5 additional patients had mutational signatures of homologous recombination deficiency but no identified causal genomic alteration. Two patients had oncogenic in-frame BRAF deletions, and we report the first clinical evidence that this alteration confers sensitivity to MAPK pathway inhibition. Moreover, we identified tumor/stroma gene expression signatures with clinical relevance. Collectively, these data demonstrate the feasibility and value of real-time genomic characterization of advanced PDAC.Significance: Molecular analyses of metastatic PDAC tumors are challenging due to the heterogeneous cellular composition of biopsy specimens and rapid progression of the disease. Using an integrated multidisciplinary biopsy program, we demonstrate that real-time genomic characterization of advanced PDAC can identify clinically relevant alterations that inform management of this difficult disease. Cancer Discov; 8(9); 1096-111. ©2018 AACR.See related commentary by Collisson, p. 1062This article is highlighted in the In This Issue feature, p. 1047.
Collapse
Affiliation(s)
- Andrew J Aguirre
- Dana-Farber Cancer Institute, Boston, Massachusetts. .,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Jonathan A Nowak
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nicholas D Camarda
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts.,Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Richard A Moffitt
- Department of Biomedical Informatics, Department of Pathology, Stony Brook University, Stony Brook, New York
| | - Arezou A Ghazani
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Srivatsan Raghavan
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Jaegil Kim
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | | | | | - Emma Reilly
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Devin McCabe
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts.,Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Lori Marini
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Kristin Anderka
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Karla Helvie
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Nelly Oliver
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Ana Babic
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Annacarolina Da Silva
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Brandon Nadres
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | | | | | - Kelly P Burke
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Thomas Clancy
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - James M Cleary
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Leona A Doyle
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kunal Jajoo
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nadine J McCleary
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A Meyerhardt
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Janet E Murphy
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Kimmie Ng
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Anuj K Patel
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Kimberly Perez
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Michael H Rosenthal
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Douglas A Rubinson
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Marvin Ryou
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Geoffrey I Shapiro
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Ewa Sicinska
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Stuart G Silverman
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Rebecca J Nagy
- Department of Medical Affairs, Guardant Health, Inc., Redwood City, California
| | - Richard B Lanman
- Department of Medical Affairs, Guardant Health, Inc., Redwood City, California
| | | | | | - Matthew B Yurgelun
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Charles S Fuchs
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts.,Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Levi A Garraway
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Gad Getz
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Jason L Hornick
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Bruce E Johnson
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Matthew H Kulke
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Robert J Mayer
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Jeffrey W Miller
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Paul B Shyn
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Nikhil Wagle
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Jen Jen Yeh
- Departments of Surgery and Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - William C Hahn
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Ryan B Corcoran
- Harvard Medical School, Boston, Massachusetts.,Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Scott L Carter
- Dana-Farber Cancer Institute, Boston, Massachusetts. .,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts.,Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Brian M Wolpin
- Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
238
|
Russo M, Lamba S, Lorenzato A, Sogari A, Corti G, Rospo G, Mussolin B, Montone M, Lazzari L, Arena S, Oddo D, Linnebacher M, Sartore-Bianchi A, Pietrantonio F, Siena S, Di Nicolantonio F, Bardelli A. Reliance upon ancestral mutations is maintained in colorectal cancers that heterogeneously evolve during targeted therapies. Nat Commun 2018; 9:2287. [PMID: 29895949 PMCID: PMC5997733 DOI: 10.1038/s41467-018-04506-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Attempts at eradicating metastatic cancers with targeted therapies are limited by the emergence of resistant subclones bearing heterogeneous (epi)genetic changes. We used colorectal cancer (CRC) to test the hypothesis that interfering with an ancestral oncogenic event shared by all the malignant cells (such as WNT pathway alterations) could override heterogeneous mechanisms of acquired drug resistance. Here, we report that in CRC-resistant cell populations, phylogenetic analysis uncovers a complex subclonal architecture, indicating parallel evolution of multiple independent cellular lineages. Functional and pharmacological modulation of WNT signalling induces cell death in CRC preclinical models from patients that relapsed during the treatment, regardless of the drug type or resistance mechanisms. Concomitant blockade of WNT and MAPK signalling restrains the emergence of drug-resistant clones. Reliance upon the WNT-APC pathway is preserved throughout the branched genomic drift associated with emergence of treatment relapse, thus offering the possibility of a common therapeutic strategy to overcome secondary drug resistance.
Collapse
Affiliation(s)
- Mariangela Russo
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy.
| | - Simona Lamba
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy
| | - Annalisa Lorenzato
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, SP 142 km 3.95, 10060, Candiolo, Turin, Italy
| | - Alberto Sogari
- Department of Oncology, University of Torino, SP 142 km 3.95, 10060, Candiolo, Turin, Italy.,FIRC Institute of Molecular Oncology (IFOM), 20139, Milan, Italy
| | - Giorgio Corti
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy
| | - Giuseppe Rospo
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy
| | | | - Monica Montone
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy
| | - Luca Lazzari
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, SP 142 km 3.95, 10060, Candiolo, Turin, Italy
| | - Sabrina Arena
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, SP 142 km 3.95, 10060, Candiolo, Turin, Italy
| | - Daniele Oddo
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, SP 142 km 3.95, 10060, Candiolo, Turin, Italy
| | - Michael Linnebacher
- Department of General Surgery, University of Rostock, Rostock, D-18057, Germany
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, 20162, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, 20122, Italy
| | - Filippo Pietrantonio
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, 20122, Italy.,Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, 20133, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, 20162, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, 20122, Italy
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, SP 142 km 3.95, 10060, Candiolo, Turin, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute-FPO, IRCCS, 10060, Candiolo, Turin, Italy. .,Department of Oncology, University of Torino, SP 142 km 3.95, 10060, Candiolo, Turin, Italy.
| |
Collapse
|
239
|
Schirripa M, Cohen SA, Battaglin F, Lenz HJ. Biomarker-driven and molecular targeted therapies for colorectal cancers. Semin Oncol 2018; 45:124-132. [PMID: 30262397 PMCID: PMC7496213 DOI: 10.1053/j.seminoncol.2017.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/27/2017] [Indexed: 12/23/2022]
Abstract
Improved clinical selection and identification of new molecules and innovative strategies have widened treatment options and increased overall survival in metastatic colorectal cancer patients in recent years. Biomarker-driven therapies represent an emerging issue in this field and new targeted treatments are under investigation and probably will be soon adopted into daily clinical practice. In the present review, the role RAS, BRAF mutations, Her2 amplification, microsatellite instability, and CpG island methylator phenotype are discussed according to their possible roles as prognostic, predictive markers, as well as possible biomarker-driven treatment options.
Collapse
Affiliation(s)
- Marta Schirripa
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Stacey A Cohen
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Francesca Battaglin
- Division of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Heinz-Josef Lenz
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
240
|
Zill OA, Banks KC, Fairclough SR, Mortimer SA, Vowles JV, Mokhtari R, Gandara DR, Mack PC, Odegaard JI, Nagy RJ, Baca AM, Eltoukhy H, Chudova DI, Lanman RB, Talasaz A. The Landscape of Actionable Genomic Alterations in Cell-Free Circulating Tumor DNA from 21,807 Advanced Cancer Patients. Clin Cancer Res 2018; 24:3528-3538. [DOI: 10.1158/1078-0432.ccr-17-3837] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/19/2018] [Accepted: 05/11/2018] [Indexed: 11/16/2022]
|
241
|
Agarwal N, Pal SK, Hahn AW, Nussenzveig RH, Pond GR, Gupta SV, Wang J, Bilen MA, Naik G, Ghatalia P, Hoimes CJ, Gopalakrishnan D, Barata PC, Drakaki A, Faltas BM, Kiedrowski LA, Lanman RB, Nagy RJ, Vogelzang NJ, Boucher KM, Vaishampayan UN, Sonpavde G, Grivas P. Characterization of metastatic urothelial carcinoma via comprehensive genomic profiling of circulating tumor DNA. Cancer 2018; 124:2115-2124. [PMID: 29517810 PMCID: PMC6857169 DOI: 10.1002/cncr.31314] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/09/2018] [Accepted: 01/25/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND Biomarker-guided clinical trials are increasingly common in metastatic urothelial carcinoma (mUC), yet patients for whom contemporary tumor tissue is not available are not eligible. Technological advancements in sequencing have made cell-free circulating DNA (cfDNA) next-generation sequencing (NGS) readily available in the clinic. The objective of the current study was to determine whether the genomic profile of mUC detected by NGS of cfDNA is similar to historical tumor tissue NGS studies. A secondary objective was to determine whether the frequency of genomic alterations (GAs) differed between lower tract mUC (mLTUC) and upper tract mUC (mUTUC). METHODS Patients from 13 academic medical centers in the United States who had a diagnosis of mUC between 2014 and 2017 and for whom cfDNA NGS results were available were included. cfDNA profiling was performed using a commercially available platform (Guardant360) targeting 73 genes. RESULTS Of 369 patients with mUC, 294 were diagnosed with mLTUC and 75 with mUTUC. A total of 2130 GAs were identified in the overall mUC cohort: 1610 and 520, respectively, in the mLTUC and mUTUC cohorts. In the mLTUC cohort, frequently observed GAs were similar between cfDNA NGS and historical tumor tissue studies, including tumor protein p53 (TP53) (P = 1.000 and .115, respectively), AT-rich interaction domain 1A (ARID1A) (P = .058 and .058, respectively), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) (P = .058 and .067, respectively), erb-b2 receptor tyrosine kinase 2 (ERBB2) (P = .565 and .074, respectively), and fibroblast growth factor receptor 3 (FGFR3) (P = .164 and .014, respectively). No significant difference was observed with regard to the frequency of GAs between patients with mLTUC and mUTUC. CONCLUSIONS Among patients with mUC for whom no tumor tissue was available, cfDNA NGS was able to identify a similar profile of GAs for biomarker-driven clinical trials compared with tumor tissue. Despite the more aggressive clinical course, cases of mUTUC demonstrated a circulating tumor DNA genomic landscape that was similar to that of mLTUC. Cancer 2018;124:2115-24. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Sumanta K. Pal
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Andrew W. Hahn
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Roberto H. Nussenzveig
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Gregory R. Pond
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Sumati V. Gupta
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Jue Wang
- Genitourinary Oncology Section, University of Arizona Cancer Center at Dignity Health, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona
| | - Mehmet A. Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Gurudatta Naik
- Department of Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Pooja Ghatalia
- Department of Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Christopher J. Hoimes
- Division of Hematology/Oncology, Case Western Reserve University, Seidman Cancer Center, Cleveland, Ohio
| | | | - Pedro C. Barata
- Division of Oncology, Department of Medicine, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alexandra Drakaki
- Department of Hematology/Oncology, University of California at Los Angeles Medical Center, Los Angeles, California
| | - Bishoy M. Faltas
- Department of Medical Oncology, Weill-Cornell Medical College, New York City, New York
| | | | | | | | | | - Kenneth M. Boucher
- Division of Biostatistics, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | | | - Guru Sonpavde
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Petros Grivas
- Division of Oncology, Department of Medicine, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
242
|
Konieczkowski DJ, Johannessen CM, Garraway LA. A Convergence-Based Framework for Cancer Drug Resistance. Cancer Cell 2018; 33:801-815. [PMID: 29763622 PMCID: PMC5957297 DOI: 10.1016/j.ccell.2018.03.025] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 02/02/2018] [Accepted: 03/26/2018] [Indexed: 02/07/2023]
Abstract
Despite advances in cancer biology and therapeutics, drug resistance remains problematic. Resistance is often multifactorial, heterogeneous, and prone to undersampling. Nonetheless, many individual mechanisms of targeted therapy resistance may coalesce into a smaller number of convergences, including pathway reactivation (downstream re-engagement of original effectors), pathway bypass (recruitment of a parallel pathway converging on the same downstream output), and pathway indifference (development of a cellular state independent of the initial therapeutic target). Similar convergences may also underpin immunotherapy resistance. Such parsimonious, convergence-based frameworks may help explain resistance across tumor types and therapeutic categories and may also suggest strategies to overcome it.
Collapse
|
243
|
Takayama Y, Suzuki K, Muto Y, Ichida K, Fukui T, Kakizawa N, Ishikawa H, Watanabe F, Hasegawa F, Saito M, Tsujinaka S, Futsuhara K, Miyakura Y, Noda H, Konishi F, Rikiyama T. Monitoring circulating tumor DNA revealed dynamic changes in KRAS status in patients with metastatic colorectal cancer. Oncotarget 2018; 9:24398-24413. [PMID: 29849949 PMCID: PMC5966256 DOI: 10.18632/oncotarget.25309] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/13/2018] [Indexed: 12/15/2022] Open
Abstract
KRAS mutated circulating tumor DNA (MctDNA) can be monitored in the blood of patients with metastatic colorectal cancer (mCRC), but dynamic changes have not been determined. Four hundred and fifty-seven plasma samples were collected prospectively from 85 mCRC patients who underwent chemotherapy. MctDNA in plasma was detected by droplet digital PCR, and the percentage of MctDNA in total circulating cell-free DNA was calculated. KRAS assessment in tumor tissues showed 29 patients with the mutant-type (MT) and 56 patients with the wild-type (WT). Twenty-three of 29 MT patients (79.3%) and 28 of 56 WT patients (50.0%) showed MctDNA. Emergence of MctDNA was recognized during treatments with various drugs. Regardless of KRAS status in tumor tissues, patients with MctDNA in blood showed poor progression-free survival with first-line treatment. Median percentage of MctDNA accounted for 10.10% in MT patients and 0.22% in WT patients. These differences between MT and WT likely affected patterns of changes in MctDNA. KRAS monitoring identified dynamic changes in MctDNA, such as continuous, intermittent, and transient changes (quick elevation and disappearance). Emergence of MctDNA involved drug resistance, except for transient changes, which were seen in WT patients and likely corresponded with the drug response. Transient changes could be involved in recovery of sensitivity to anti-EGFR antibody in WT patients. Monitoring MctDNA during various treatments showed dynamic changes in KRAS status and could provide useful information for determining treatments for patients with mCRC.
Collapse
Affiliation(s)
- Yuji Takayama
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Koichi Suzuki
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Yuta Muto
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Kosuke Ichida
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Taro Fukui
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Nao Kakizawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Hideki Ishikawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Fumiaki Watanabe
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Fumi Hasegawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Masaaki Saito
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Shingo Tsujinaka
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Kazushige Futsuhara
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Yasuyuki Miyakura
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Hiroshi Noda
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Fumio Konishi
- Department of Surgery, Nerima-Hikarigaoka Hospital, Nerima-ku, Tokyo 179-0072, Japan
| | - Toshiki Rikiyama
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| |
Collapse
|
244
|
Sartore-Bianchi A, Amatu A, Bonazzina E, Stabile S, Giannetta L, Cerea G, Schiavetto I, Bencardino K, Funaioli C, Ricotta R, Cipani T, Schirru M, Gambi V, Palmeri L, Carlo-Stella G, Rusconi F, Di Bella S, Burrafato G, Cassingena A, Valtorta E, Lauricella C, Pazzi F, Gambaro A, Ghezzi S, Marrapese G, Tarenzi E, Veronese S, Truini M, Vanzulli A, Siena S. Pooled Analysis of Clinical Outcome of Patients with Chemorefractory Metastatic Colorectal Cancer Treated within Phase I/II Clinical Studies Based on Individual Biomarkers of Susceptibility: A Single-Institution Experience. Target Oncol 2018; 12:525-533. [PMID: 28669023 PMCID: PMC5524857 DOI: 10.1007/s11523-017-0505-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Patients with metastatic colorectal cancer (mCRC) refractory to standard therapies have a poor prognosis. In this setting, recruitment into clinical trials is warranted, and studies driven by selection according to individual tumor molecular characteristics are expected to provide added value. Objective We retrospectively analyzed data from patients with mCRC refractory to or following failure of standard therapies who were enrolled into phase I/II clinical studies at the Niguarda Cancer Center based on the presence of a specific molecular profile expected to represent the target of susceptibility to the experimental drug(s). Patients and Methods From June 2011 to May 2016, 2044 patients with mCRC underwent molecular screening. Eighty patients (3.9%) were enrolled in ad hoc studies; the median age was 60 years (range 36–86) and the median number of previous treatment lines was five (range 2–8). Molecular characteristics exploited within these studies were MGMT promoter hypermethylation (48.7%), HER2 amplification (28.8%), BRAFV600E mutation (20%), and novel gene fusions involving ALK or NTRK (2.5%). Results One patient (1%) had RECIST (Response Evaluation Criteria In Solid Tumors) complete response (CR), 13 patients (16.5%) experienced a partial response (PR), and 28 (35%) stable disease (SD). Median progression-free survival (PFS) was 2.8 months (range 2.63–3.83), with 24% of patients displaying PFS >5 months. Median growth modulation index (GMI) was 0.85 (range 0–15.61) and 32.5% of patients had GMI >1.33. KRAS exon 2 mutations were found in 38.5% of patients, and among the 78 patients with known KRAS status, those with wild-type tumors had longer PFS than those with mutated tumors (3.80 [95% CI 2.80–5.03] vs. 2.13 months [95% CI 1.77–2.87], respectively, p = 0.001). Median overall survival (OS) was 7.83 months (range 7.17–9.33) for all patients, and patients with KRAS wild-type tumors had longer OS than those with mutated tumors (7.83 [95% CI 7.33–10.80] vs. 7.18 months [95% CI 5.63–9.33], respectively, p = 0.06). Conclusions This single-institution retrospective study indicates that in a heavily pretreated population approximately 4% of mCRC tumors display a potential actionable molecular context suitable for therapeutic intervention. Application of molecular selection is challenging but improves clinical outcome even in later lines of treatment.![]()
Collapse
Affiliation(s)
- Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Erica Bonazzina
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Stefano Stabile
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Laura Giannetta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Giulio Cerea
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Ilaria Schiavetto
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Katia Bencardino
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Chiara Funaioli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Riccardo Ricotta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Tiziana Cipani
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Michele Schirru
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Valentina Gambi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Laura Palmeri
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Giulia Carlo-Stella
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Francesca Rusconi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Sara Di Bella
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Giovanni Burrafato
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Andrea Cassingena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Emanuele Valtorta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Calogero Lauricella
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Federica Pazzi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Alessandra Gambaro
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Silvia Ghezzi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Giovanna Marrapese
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Emiliana Tarenzi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Silvio Veronese
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Mauro Truini
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Angelo Vanzulli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy.,Dipartimento di Oncologia e Emato-Oncologia, Università degli Studi di Milano, Milan, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy. .,Dipartimento di Oncologia e Emato-Oncologia, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
245
|
García-Foncillas J, Alba E, Aranda E, Díaz-Rubio E, López-López R, Tabernero J, Vivancos A. Incorporating BEAMing technology as a liquid biopsy into clinical practice for the management of colorectal cancer patients: an expert taskforce review. Ann Oncol 2018; 28:2943-2949. [PMID: 28945877 PMCID: PMC5834030 DOI: 10.1093/annonc/mdx501] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of mutation identification for advanced colorectal cancer treatment with anti-epidermal growth factor receptor agents is well established. However, due to delays in turnaround time, low-quality tissue samples, and/or lack of standardization of testing methods a significant proportion of patients are being treated without the information that Kirsten rat sarcoma and neuroblastoma rat sarcoma (RAS) testing can provide. The detection of mutated circulating tumor DNA by BEAMing technology addresses this gap in care and allows these patients to receive international guideline-recommended expanded RAS testing with rapid turnaround times. Furthermore, the overall concordance between OncoBEAM RAS colorectal cancer testing and standard of care tissue testing is very high (93.3%). This article presents an overview of the clinical utility and potential applications of this minimally invasive method, such as early detection of emergent resistance to anti-epidermal growth factor receptor therapy. If appropriately implemented, BEAMing technology holds considerable promise to enhance the quality of patient care and improve clinical outcomes.
Collapse
Affiliation(s)
- J García-Foncillas
- Cancer Institute, University Hospital Fundacion Jimenez Diaz, Autonomous University, Madrid
| | - E Alba
- Medical Oncology Unit, Regional University Hospital Virgen de la Victoria, IBIMA, Málaga
| | - E Aranda
- Biomedical Research Institute (IMIBIC), Hospital Reina Sofía, Faculty of Medicine, Universidad de Córdoba, Cordoba, Spain (CIBERONC)
| | - E Díaz-Rubio
- Research Institute IdISSC, Hospital Clínico San Carlos, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain (CIBERONC)
| | - R López-López
- Medical Oncology Department and Translational Medical Oncology Group, University Clinical Hospital & Health Research Institute (IDIS); CIBERONC, Santiago de Compostela, University School of Medicine, Santiago de Compostela, Spain
| | - J Tabernero
- Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAM), Barcelona, Spain (CIBERONC)
| | - A Vivancos
- Vall d'Hebron Institute of Oncology (VHIO), Cancer Genomics Lab., Barcelona, Spain
| |
Collapse
|
246
|
Feng C, Zhang L, Sun Y, Li X, Zhan L, Lou Y, Wang Y, Liu L, Zhang Y. GDPD5, a target of miR-195-5p, is associated with metastasis and chemoresistance in colorectal cancer. Biomed Pharmacother 2018; 101:945-952. [DOI: 10.1016/j.biopha.2018.03.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
|
247
|
Romano G, Chen PL, Song P, McQuade JL, Liang RJ, Liu M, Roh W, Duose DY, Carapeto FCL, Li J, Teh JLF, Aplin AE, Chen M, Zhang J, Lazar AJ, Davies MA, Futreal PA, Amaria RN, Zhang DY, Wargo JA, Kwong LN. A Preexisting Rare PIK3CAE545K Subpopulation Confers Clinical Resistance to MEK plus CDK4/6 Inhibition in NRAS Melanoma and Is Dependent on S6K1 Signaling. Cancer Discov 2018; 8:556-567. [PMID: 29496665 PMCID: PMC5932238 DOI: 10.1158/2159-8290.cd-17-0745] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 01/22/2018] [Accepted: 02/23/2018] [Indexed: 12/14/2022]
Abstract
Combined MEK and CDK4/6 inhibition (MEKi + CDK4i) has shown promising clinical outcomes in patients with NRAS-mutant melanoma. Here, we interrogated longitudinal biopsies from a patient who initially responded to MEKi + CDK4i therapy but subsequently developed resistance. Whole-exome sequencing and functional validation identified an acquired PIK3CAE545K mutation as conferring drug resistance. We demonstrate that PIK3CAE545K preexisted in a rare subpopulation that was missed by both clinical and research testing, but was revealed upon multiregion sampling due to PIK3CAE545K being nonuniformly distributed. This resistant population rapidly expanded after the initiation of MEKi + CDK4i therapy and persisted in all successive samples even after immune checkpoint therapy and distant metastasis. Functional studies identified activated S6K1 as both a key marker and specific therapeutic vulnerability downstream of PIK3CAE545K-induced resistance. These results demonstrate that difficult-to-detect preexisting resistance mutations may exist more often than previously appreciated and also posit S6K1 as a common downstream therapeutic nexus for the MAPK, CDK4/6, and PI3K pathways.Significance: We report the first characterization of clinical acquired resistance to MEKi + CDK4i, identifying a rare preexisting PIK3CAE545K subpopulation that expands upon therapy and exhibits drug resistance. We suggest that single-region pretreatment biopsy is insufficient to detect rare, spatially segregated drug-resistant subclones. Inhibition of S6K1 is able to resensitize PIK3CAE545K-expressing NRAS-mutant melanoma cells to MEKi + CDK4i. Cancer Discov; 8(5); 556-67. ©2018 AACR.See related commentary by Sullivan, p. 532See related article by Teh et al., p. 568This article is highlighted in the In This Issue feature, p. 517.
Collapse
Affiliation(s)
- Gabriele Romano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pei-Ling Chen
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ping Song
- Department of Bioengineering, Rice University, Houston, Texas
| | - Jennifer L McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roger J Liang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mingguang Liu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Whijae Roh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dzifa Y Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fernando C L Carapeto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jun Li
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jessica L F Teh
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Cutaneous Biology and Dermatology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Merry Chen
- Department of Neurooncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J Lazar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Davies
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rodabe N Amaria
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Y Zhang
- Department of Bioengineering, Rice University, Houston, Texas
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Neurooncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lawrence N Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
248
|
Peng C, Gu L, Wang L, Huang Q, Wang B, Guo G, Fan Y, Gao Y, Ma X, Zhang X. Role of presurgical targeted molecular therapy in renal cell carcinoma with an inferior vena cava tumor thrombus. Onco Targets Ther 2018; 11:1997-2005. [PMID: 29670375 PMCID: PMC5898583 DOI: 10.2147/ott.s158114] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Purpose The clinical benefit of targeted molecular therapy (TMT) in renal cell carcinoma (RCC) with an inferior vena cava (IVC) tumor thrombus remains controversial. The aim of this study was to investigate the effects of presurgical TMT on the heights and levels of IVC thrombi, and to assess its impact on surgical strategy. Patients and methods We retrospectively reviewed data from 18 patients with RCC involving IVC tumor thrombi who were treated at our hospital with presurgical TMT followed by an IVC thrombectomy. The changes in heights and levels of the IVC thrombi were compared using computed tomography or magnetic resonance imaging. Clinicopathological factors were also evaluated to assess their association with TMT efficacy. Results The tumor thrombus levels before TMT were stage I in 1 patient (5.6%), II in 12 patients (66.7%), III in 4 patients (22.2%), and IV in 1 patient (5.6%). After a median of two treatment cycles (range: 1–3), the thrombus height decreased measurably in 11 patients (61.1%) with an average shrinkage of 17.7%. The thrombus height remained stable in five patients (27.8%) and was enlarged in two (11.1%). Downstaging of the thrombus level occurred in four patients (22.2%); the surgical strategy was modified in three patients (16.7%) to avoid cardiopulmonary bypass and complicated liver mobilization under robot-assisted laparoscopy. Furthermore, a higher neutrophil count tended to be associated with a worse clinical TMT-associated outcome (P=0.056). Conclusion Our data suggest a limited influence of presurgical TMT with a positive benefit in RCC patients with level III and IV thrombus. Thrombus-level regression may potentially alter the surgical strategy, especially robotic surgery. However, our findings require validation with additional prospective investigations.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Liangyou Gu
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Lei Wang
- Department of Urology, Chinese PLA 534 Hospital, Luoyang, People's Republic of China
| | - Qingbo Huang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Baojun Wang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Gang Guo
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Yang Fan
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Yu Gao
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Xin Ma
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| | - Xu Zhang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Medical Academy, Beijing, People's Republic of China
| |
Collapse
|
249
|
Boesch M, Spizzo G, Seeber A. Concise Review: Aggressive Colorectal Cancer: Role of Epithelial Cell Adhesion Molecule in Cancer Stem Cells and Epithelial-to-Mesenchymal Transition. Stem Cells Transl Med 2018; 7:495-501. [PMID: 29667344 PMCID: PMC5980125 DOI: 10.1002/sctm.17-0289] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/31/2018] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide. In spite of various attempts to ameliorate outcome by escalating treatment, significant improvement is lacking particularly in the adjuvant setting. It has been proposed that cancer stem cells (CSCs) and the epithelial‐to‐mesenchymal transition (EMT) are at least partially responsible for therapy resistance in CRC. The epithelial cell adhesion molecule (EpCAM) was one of the first CSC antigens to be described. Furthermore, an EpCAM‐specific antibody (edrecolomab) has the merit of having launched the era of monoclonal antibody treatment in oncology in the 1990s. However, despite great initial enthusiasm, monoclonal antibody treatment has not proven successful in the adjuvant treatment of CRC patients. In the meantime, new insights into the function of EpCAM in CRC have emerged and new drugs targeting various epitopes have been developed. In this review article, we provide an update on the role of EpCAM in CSCs and EMT, and emphasize the potential predictive selection criteria for novel treatment strategies and refined clinical trial design. stemcellstranslationalmedicine2018;7:495–501
Collapse
Affiliation(s)
- Maximilian Boesch
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
| | - Gilbert Spizzo
- Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
| | - Andreas Seeber
- Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria
| |
Collapse
|
250
|
Corcoran RB, André T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, Hollebecque A, McRee AJ, Siena S, Middleton G, Muro K, Gordon MS, Tabernero J, Yaeger R, O'Dwyer PJ, Humblet Y, De Vos F, Jung AS, Brase JC, Jaeger S, Bettinger S, Mookerjee B, Rangwala F, Van Cutsem E. Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAFV600E-Mutant Colorectal Cancer. Cancer Discov 2018; 8:428-443. [PMID: 29431699 PMCID: PMC5882509 DOI: 10.1158/2159-8290.cd-17-1226] [Citation(s) in RCA: 417] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/21/2018] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
Although BRAF inhibitor monotherapy yields response rates >50% in BRAFV600-mutant melanoma, only approximately 5% of patients with BRAFV600E colorectal cancer respond. Preclinical studies suggest that the lack of efficacy in BRAFV600E colorectal cancer is due to adaptive feedback reactivation of MAPK signaling, often mediated by EGFR. This clinical trial evaluated BRAF and EGFR inhibition with dabrafenib (D) + panitumumab (P) ± MEK inhibition with trametinib (T) to achieve greater MAPK suppression and improved efficacy in 142 patients with BRAFV600E colorectal cancer. Confirmed response rates for D+P, D+T+P, and T+P were 10%, 21%, and 0%, respectively. Pharmacodynamic analysis of paired pretreatment and on-treatment biopsies found that efficacy of D+T+P correlated with increased MAPK suppression. Serial cell-free DNA analysis revealed additional correlates of response and emergence of KRAS and NRAS mutations on disease progression. Thus, targeting adaptive feedback pathways in BRAFV600E colorectal cancer can improve efficacy, but MAPK reactivation remains an important primary and acquired resistance mechanism.Significance: This trial demonstrates that combined BRAF + EGFR + MEK inhibition is tolerable, with promising activity in patients with BRAFV600E colorectal cancer. Our findings highlight the MAPK pathway as a critical target in BRAFV600E colorectal cancer and the need to optimize strategies inhibiting this pathway to overcome both primary and acquired resistance. Cancer Discov; 8(4); 428-43. ©2018 AACR.See related commentary by Janku, p. 389See related article by Hazar-Rethinam et al., p. 417This article is highlighted in the In This Issue feature, p. 371.
Collapse
Affiliation(s)
- Ryan B Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Thierry André
- Hôpital Saint-Antoine, and Sorbonne Universités, Paris, France
| | | | | | | | - Johanna C Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | | | - Autumn J McRee
- University of North Carolina, Chapel Hill, North Carolina
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Osopedale Metropolitano Niguarda and Department of Oncology and Hemato-Oncollogy, Università degli Studi di Milano, Milan, Italy
| | - Gary Middleton
- University of Birmingham and University Hospital, Birmingham, United Kingdom
| | - Kei Muro
- Aichi Cancer Center Hospital, Nagoya, Japan
| | | | | | - Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter J O'Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Filip De Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | | | - Savina Jaeger
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | - Fatima Rangwala
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | | |
Collapse
|