101
|
Abstract
The activating BRAF mutation V600E and related mutations in this codon are most important for the activation of the RAS/RAF/MEK/ERK mitogen-activated protein kinase (MAPK) signalling pathway in melanoma. BRAF V600E mutations have been detected in ~40% of melanoma patients and BRAF V600K mutations in ~5% of melanoma patients. Activation of the MAPK pathway results in continuous stimulation of cell proliferation and inhibits programmed cell death. Vemurafenib (PLX4032) was developed as a low-molecular-weight molecule for the inhibition of the mutated serine-threonine kinase BRAF, and it selectively binds to the ATP-binding site of BRAF V600E kinase and inhibits its activity. The biochemical affinity of vemurafenib for mutated BRAF translates to potent inhibition of ERK phosphorylation and of cell proliferation exclusively in BRAF-mutant cell lines. In animal model experiments, it was demonstrated that vemurafenib achieved tumour regressions in cells harbouring the BRAF V600E mutation. The clinical trials with vemurafenib in unresectable metastatic melanoma in phases I, II and III for patients harbouring BRAF V600E mutations demonstrated all unexpected high objective response rates ranging between 50 and 80%. Median progression-free survival was prolonged from 2 months with dacarbazine to 7 months with vemurafenib, and median overall survival was, respectively, prolonged from 9 to 14 months. A major problem remains in the development of resistance to vemurafenib treatment after several months in the majority of patients, and multiple resistance mechanisms have already been described. Under vemurafenib treatment, about 25% of patients developed cutaneous squamous cell carcinomas of the keratoacanthoma type with low invasive potential and without the occurrence of metastasis. The overall tolerability of the drug was quite good, and many patients remained on treatment for long times. As other solid tumours like papillary thyroid cancer, colorectal cancer, non-small-cell lung cancer and ovarian cancer likewise harbour BRAF mutation, vemurafenib is also tested in these entities. In future, combinations of vemurafenib with other kinase inhibitors and with immunotherapies will improve its therapeutic potential.
Collapse
Affiliation(s)
- Claus Garbe
- Division of Dermatooncology, Department of Dermatology, University Medical Centre, Liebermeisterstr. 25, 72074, Tuebingen, Germany.
| | - Thomas K Eigentler
- Division of Dermatooncology, Department of Dermatology, University Medical Centre, Liebermeisterstr. 25, 72074, Tuebingen, Germany
| |
Collapse
|
102
|
In 't Veld SGJG, Duong KN, Snel M, Witteveen A, Beumer IJ, Delahaye LJMJ, Wehkamp D, Bernards R, Glas AM, Tian S. A Computational Workflow Translates a 58-Gene Signature to a Formalin-Fixed, Paraffin-Embedded Sample-Based Companion Diagnostic for Personalized Treatment of the BRAF-Mutation-Like Subtype of Colorectal Cancers. High Throughput 2017; 6:ht6040016. [PMID: 29479053 PMCID: PMC5748595 DOI: 10.3390/ht6040016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer patients with the BRAF(p.V600E) mutation have poor prognosis in metastatic setting. Personalized treatment options and companion diagnostics are needed to better treat these patients. Previously, we developed a 58-gene signature to characterize the distinct gene expression pattern of BRAF-mutation-like subtype (accuracy 91.1%). Further experiments repurposed drug Vinorelbine as specifically lethal to this BRAF-mutation-like subtype. The aim of this study is to translate this 58-gene signature from a research setting to a robust companion diagnostic that can use formalin-fixed, paraffin-embedded (FFPE) samples to select patients with the BRAF-mutation-like subtype. BRAF mutation and gene expression data of 302 FFPE samples were measured (mutants = 57, wild-type = 245). The performance of the 58-gene signature in FFPE samples showed a high sensitivity of 89.5%. In the identified BRAF-mutation-like subtype group, 50% of tumours were known BRAF mutants, and 50% were BRAF wild-type. The stability of the 58-gene signature in FFPE samples was evaluated by two control samples over 40 independent experiments. The standard deviations (SD) were within the predefined criteria (control 1: SD = 0.091, SD/Range = 3.0%; control 2: SD = 0.169, SD/Range = 5.5%). The fresh frozen version and translated FFPE version of this 58-gene signature were compared using 170 paired fresh frozen and FFPE samples and the result showed high consistency (agreement = 99.3%). In conclusion, we translated this 58-gene signature to a robust companion diagnostic that can use FFPE samples.
Collapse
Affiliation(s)
- Sjors G J G In 't Veld
- Department of Neurosurgery, VU University Medical Center, 1081HV Amsterdam, The Netherlands.
| | - Kim N Duong
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
| | - Mireille Snel
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
| | - Anke Witteveen
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
| | - Inès J Beumer
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
| | | | - Diederik Wehkamp
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
| | - René Bernards
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands.
| | - Annuska M Glas
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
| | - Sun Tian
- Agendia NV, Science Park 406, 1098XH Amsterdam, The Netherlands.
| |
Collapse
|
103
|
Sanz-Garcia E, Argiles G, Elez E, Tabernero J. BRAF mutant colorectal cancer: prognosis, treatment, and new perspectives. Ann Oncol 2017; 28:2648-2657. [PMID: 29045527 DOI: 10.1093/annonc/mdx401] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The MAPK cascade plays a crucial role in tumor cell proliferation and survival. Accumulating evidence suggests that mutations in the BRAF oncogene are not only associated with poor prognosis but also linked with less benefit when treated with anti-epidermal growth factor receptor antibodies in metastatic colorectal cancer (mCRC). Targeting this molecular aberration has thus become a matter of particular interest in mCRC drug development. In contrast to other malignances such as BRAF mutant melanoma, efficacy observed with BRAF inhibitors in monotherapy in mCRC is poor. Several mechanisms of resistance have been identified leading to the development of different treatment strategies that have shown promising activity in early clinical trials. Hence, rational combination of targeted therapies is expected to further increase the efficacy of selective BRAF inhibitors. Herein, we discuss the main clinical and molecular characteristics of BRAF mutant colorectal cancer and its translation into the clinic, with a focus on developmental therapeutics and combination strategies. In addition, we contextualize the available data with potential future approaches that include the extended access to next-generation sequencing platforms and gene expression strategies for molecular subtyping. These approaches will facilitate the identification of certain patient profiles providing more therapeutic possibilities.
Collapse
Affiliation(s)
- E Sanz-Garcia
- Medical Oncology Department, Vall D'Hebron University Hospital, Barcelona;; Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - G Argiles
- Medical Oncology Department, Vall D'Hebron University Hospital, Barcelona;; Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - E Elez
- Medical Oncology Department, Vall D'Hebron University Hospital, Barcelona;; Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - J Tabernero
- Medical Oncology Department, Vall D'Hebron University Hospital, Barcelona;; Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
104
|
Salem ME, Weinberg BA, Xiu J, El-Deiry WS, Hwang JJ, Gatalica Z, Philip PA, Shields AF, Lenz HJ, Marshall JL. Comparative molecular analyses of left-sided colon, right-sided colon, and rectal cancers. Oncotarget 2017; 8:86356-86368. [PMID: 29156800 PMCID: PMC5689690 DOI: 10.18632/oncotarget.21169] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/15/2017] [Indexed: 12/12/2022] Open
Abstract
Tumor sidedness has emerged as an important prognostic and predictive factor in the treatment of colorectal cancer. Recent studies demonstrate that patients with advanced right-sided colon cancers have a worse prognosis than those with left-sided colon or rectal cancers, and these patient subgroups respond differently to biological therapies. Historically, management of patients with metastatic colon and rectal cancers has been similar, and colon and rectal cancer patients have been grouped together in large clinical trials. Clearly, the differences in molecular biology among right-sided colon, left-sided colon, and rectal cancers should be further studied in order to account for disparities in clinical outcomes. We profiled 10,570 colorectal tumors (of which 2,413 were identified as arising from the left colon, right colon, or rectum) using next-generation sequencing, immunohistochemistry, chromogenic in-situ hybridization, and fragment analysis (Caris Life Sciences, Phoenix, AZ). Right-sided colon cancers had higher rates of microsatellite instability, more frequent aberrant activation of the EGFR pathway including higher BRAF and PIK3CA mutation rates, and increased mutational burden compared to left-sided colon and rectal cancers. Rectal cancers had higher rates of TOPO1 expression and Her2/neu amplification compared to both left- and right-sided colon cancers. Molecular variations among right-sided colon, left-sided colon, and rectal tumors may contribute to differences in clinical behavior. The site of tumor origin (left colon, right colon, or rectum) should certainly be considered when selecting treatment regimens and stratifying patients for future clinical trials.
Collapse
Affiliation(s)
- Mohamed E. Salem
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Benjamin A. Weinberg
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | | | - Jimmy J. Hwang
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | | | - Philip A. Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Anthony F. Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Heinz-Josef Lenz
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - John L. Marshall
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
105
|
Wang X, Wei Q, Gao J, Li J, Li J, Gong J, Li Y, Shen L. Clinicopathologic features and treatment efficacy of Chinese patients with BRAF-mutated metastatic colorectal cancer: a retrospective observational study. CHINESE JOURNAL OF CANCER 2017; 36:81. [PMID: 29037218 PMCID: PMC5644136 DOI: 10.1186/s40880-017-0247-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
Background The prognostic role of the V600E mutation of v-raf murine sarcoma viral oncogene homolog B1 (BRAF) in metastatic colorectal cancer (mCRC) is well established, but the therapeutic regimen targeting this disease is lacking. This study aimed to analyze the clinicopathologic features of and treatment efficacy of commonly used regimens on BRAF-mutated mCRCs. Methods We collected and reviewed the medical records of mCRC patients treated at Peking University Cancer Hospital & Institute (Beijing, China) between July 2011 and July 2016. Kirsten rat sarcoma viral oncogene homolog (KRAS), neuroblastoma RAS viral oncogene homolog (NRAS), and BRAF mutational status was assayed using direct sequencing. The details of clinicopathologic characteristics of patients and their responses to FOLFOXIRI regimen or standard therapy were obtained by reviewing the medical records. The progression-free survival (PFS) and overall survival (OS) were assessed using Kaplan–Meier analysis and compared using the log-rank test. Results Of 1694 patients studied, 75 had BRAF exon 15 mutations. Of these 75 patients, 71 had V600E mutation, 1 had D594G mutation, 2 had K601E mutation, and 1 had a novel T599_V600insAGA alteration. No patients had KRAS or NRAS mutations. Of 63 patients with BRAF V600E-mutated mCRC and sufficient clinical data, 27 (42.9%) had right-sided colon tumors, 19 (30.2%) had left-sided colon tumors, and 17 (26.9%) had rectal tumors; 26 (41.3%) had peritoneal metastases, and 50 (79.4%) had distant lymph node metastases. The patients with BRAF K601E- and T599_V600insAGA-mutated tumors had similar clinicopathologic features to those with BRAF V600E-mutated tumors. Patients with the BRAF V600E mutation benefited more from FOLFOXIRI regimen compared with patients who underwent standard therapy (overall response rate 83.3% vs. 14.0%; median PFS 6.4 months vs. 2.8 months, P = 0.220; median OS 11.0 months vs. 6.9 months, P = 0.048). Conclusions BRAF V600E mutations were commonly identified in right-sided tumors and showed a high incidence of peritoneal and distant lymph nodes metastases. This subtype of mCRC was characterized by short OS and unique patterns of metastasis. Compared with standard treatment regimens, the FOLFOXIRI regimen had acceptable and manageable toxicities and favorable efficacy on patients with BRAF-mutated mCRC.
Collapse
Affiliation(s)
- Xicheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Qing Wei
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Jing Gao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Jie Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China.
| |
Collapse
|
106
|
Roma C, Rachiglio AM, Pasquale R, Fenizia F, Iannaccone A, Tatangelo F, Antinolfi G, Parrella P, Graziano P, Sabatino L, Colantuoni V, Botti G, Maiello E, Normanno N. BRAF V600E mutation in metastatic colorectal cancer: Methods of detection and correlation with clinical and pathologic features. Cancer Biol Ther 2017; 17:840-8. [PMID: 27261210 DOI: 10.1080/15384047.2016.1195048] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The screening for BRAF V600E mutation is employed in clinical practice for its prognostic and potentially predictive role in patients with metastatic colorectal carcinoma (mCRC). Little information is available on the sensitivity and specificity of the testing methods to detect this mutation in CRC. By using serial dilution of BRAF mutant DNA with wild type DNA, we found that the sensitivity of allelic discrimination-Real Time PCR was higher than PCR-Sequencing (10% vs 20%). In agreement, the Real Time PCR assay displayed increased analytical sensitivity in detecting the BRAF V600E mutation as compared with PCR-Sequencing in a cohort of 510 consecutive CRCs (21 vs 16 cases). Targeted resequencing demonstrated that all cases negative by PCR-Sequencing had an allelic frequency of the BRAF mutation <20%, thus suggesting tumor heterogeneity. The association of BRAF mutations with clinical and pathological features was assessed next in a cohort of 840 KRAS exon 2 wild type CRC patients screened with the Real Time PCR assay. The BRAF V600E mutation frequency in this cohort was 7.8% that increased to 33.4% in females over 70 y of age with right-sided tumor location. BRAF mutations were also detected in 4.4% of male patients with left-sided tumors and aged <70 y. Fourteen of 61 (22.9%) BRAF V600E mutation bearing patients exhibited microsatellite instability (MSI) as assessed by T17 mononucleotide sequence within intron 8 of HSP110. Our study indicates that Real Time PCR-based assays are more sensitive than PCR-Sequencing to detect the BRAF V600E mutation in CRC and that BRAF mutations screening should not be restricted to selected patients on the basis of the clinical-pathological characteristics.
Collapse
Affiliation(s)
- Cristin Roma
- a Laboratory of Pharmacogenomics , Centro di Ricerche Oncologiche di Mercogliano (CROM)-Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| | - Anna Maria Rachiglio
- a Laboratory of Pharmacogenomics , Centro di Ricerche Oncologiche di Mercogliano (CROM)-Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| | - Raffaella Pasquale
- a Laboratory of Pharmacogenomics , Centro di Ricerche Oncologiche di Mercogliano (CROM)-Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| | - Francesca Fenizia
- a Laboratory of Pharmacogenomics , Centro di Ricerche Oncologiche di Mercogliano (CROM)-Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| | - Alessia Iannaccone
- a Laboratory of Pharmacogenomics , Centro di Ricerche Oncologiche di Mercogliano (CROM)-Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| | - Fabiana Tatangelo
- b Pathology Unit , Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| | - Giuseppe Antinolfi
- c Surgical Pathology Unit , Azienda Ospedaliera dei Colli , Naples , Italy
| | - Paola Parrella
- d Laboratory of Oncology , Hospital "Casa Sollievo Della Sofferenza," San Giovanni Rotondo , FG , Italy
| | - Paolo Graziano
- e Surgical Pathology Unit , Hospital "Casa Sollievo Della Sofferenza," San Giovanni Rotondo , FG , Italy
| | - Lina Sabatino
- f Department of Science and Technology , University of Sannio , Benevento , Italy
| | - Vittorio Colantuoni
- f Department of Science and Technology , University of Sannio , Benevento , Italy
| | - Gerardo Botti
- b Pathology Unit , Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| | - Evaristo Maiello
- g Department of Oncology , Hospital "Casa Sollievo Della Sofferenza," San Giovanni Rotondo , FG , Italy
| | - Nicola Normanno
- h Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS , Naples , Italy
| |
Collapse
|
107
|
Strickler JH, Wu C, Bekaii-Saab T. Targeting BRAF in metastatic colorectal cancer: Maximizing molecular approaches. Cancer Treat Rev 2017; 60:109-119. [PMID: 28946014 DOI: 10.1016/j.ctrv.2017.08.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/08/2017] [Accepted: 08/11/2017] [Indexed: 02/07/2023]
Abstract
Oncogenic mutations in B-type Raf kinase (BRAF) occur in 7-10% of metastatic colorectal cancers (mCRC). Despite recent improvements in survival in the general population of patients with mCRC, patients with BRAF-mutant mCRC continue to have poor response to most systemic therapies, and prognosis remains poor. There is a substantial unmet need for novel therapeutic strategies to treat patients with BRAF-mutant mCRC. This review outlines the epidemiology, molecular pathogenesis, prognosis, and mechanisms of treatment resistance of BRAF-mutated CRC. Additionally, this review highlights novel therapeutic strategies aimed at enhancing response and improving outcomes.
Collapse
Affiliation(s)
- John H Strickler
- Duke University School of Medicine, 20 Duke Medicine Circle, Durham, NC 27710, USA
| | - Christina Wu
- Emory University, 1365-C Clifton Rd NE, Atlanta, GA 30322, USA
| | | |
Collapse
|
108
|
Cremolini C, Pietrantonio F, Tomasello G, Dadduzio V, Moretto R, Morano F, Schirripa M, Antoniotti C, Fucà G, Bergamo F, Rossini D, Nichetti F, Ziampiri S, Ghidini M, Marmorino F, Prisciandaro M, Falcone A, De Braud F, Loupakis F, Lonardi S. Vinorelbine in BRAF V600E mutated metastatic colorectal cancer: a prospective multicentre phase II clinical study. ESMO Open 2017; 2:e000241. [PMID: 29209533 PMCID: PMC5703381 DOI: 10.1136/esmoopen-2017-000241] [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] [Received: 06/29/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND BRAF V600E mutation defines a specific colorectal cancer (CRC) subgroup with poor prognosis. Promising preclinical data showed synthetically lethal activity of mitotic spindle poisons on BRAF-mutated and BRAF-like CRC models. We designed a phase II trial to test the activity of vinorelbine in patients with BRAF V600E mutated metastatic CRC (mCRC). PATIENTS AND METHODS Patients progressed to or not deemed eligible for standard treatments received oral (60 mg/sqm) or intravenous (25 mg/sqm) vinorelbine, on days 1 and 8 every 21 days. Primary endpoint was objective response rate (ORR). RESULTS Twenty patients were enrolled; 75% of them were highly pretreated. No responses were observed (0%); only one patient had a confirmed disease stabilisation (5%). Median progression-free survival was 1 month (95% CI 0.8 to 1.8), median overall survival was 2.1 months (95% CI 1.6 to 3.7). No serious adverse events were observed. CONCLUSIONS Despite encouraging preclinical data, our study did not show signs of clinical activity for vinorelbine in this patients' population. Further investigations on molecular heterogeneity and dynamic evolution of BRAF V600E mutated mCRC are needed.
Collapse
Affiliation(s)
- Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Vincenzo Dadduzio
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto IRCCS, Padua, Italy
- Department of Medical Oncology Fondazione Policlinico, Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marta Schirripa
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto IRCCS, Padua, Italy
| | - Carlotta Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanni Fucà
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Bergamo
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto IRCCS, Padua, Italy
| | - Daniele Rossini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Stamatia Ziampiri
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto IRCCS, Padua, Italy
| | - Michele Ghidini
- Department of Oncology, ASST Ospedale di Cremona, Cremona, Italy
| | - Federica Marmorino
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Michele Prisciandaro
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alfredo Falcone
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Filippo De Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Fotios Loupakis
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto IRCCS, Padua, Italy
| | - Sara Lonardi
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto IRCCS, Padua, Italy
| |
Collapse
|
109
|
Ishida H, Yamaguchi T, Chiba K, Iijima T, Horiguchi SI. A case report of ascending colon adenosquamous carcinoma with BRAF V600E mutation. Int Cancer Conf J 2017; 6:93-97. [PMID: 31149479 DOI: 10.1007/s13691-017-0283-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/06/2017] [Indexed: 12/23/2022] Open
Abstract
A 78-year-old Japanese male was referred to our hospital with a 3-month history of anorexia and right abdominal pain. A colonoscopy showed circumferential narrowing of the ascending colon with deep ulceration. Biopsy was performed, and examination of the specimen demonstrated squamous cell carcinoma. Abdominal computed tomography demonstrated thickening of the wall of the ascending colon, multiple lymph node metastases, multiple liver metastases, and peritoneal dissemination. After right hemicolectomy, histological examination demonstrated adenosquamous carcinoma. Biomarker analysis showed a microsatellite stable, wild-type KRAS gene in exon 2 and BRAF V600E mutation. Despite undergoing intensive chemotherapy, the patient died 5 months postoperatively.
Collapse
Affiliation(s)
- Hiroyuki Ishida
- 1Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22, Honkomagome, Bunkyo-ku, Tokyo, 113-8677 Japan
| | - Tatsuro Yamaguchi
- 1Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22, Honkomagome, Bunkyo-ku, Tokyo, 113-8677 Japan.,2Hereditary Tumor Research Project, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Kazuro Chiba
- 3Department of Gastroenterology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Takeru Iijima
- 2Hereditary Tumor Research Project, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Shin-Ichiro Horiguchi
- 4Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| |
Collapse
|
110
|
Li D, Fu Q, Li M, Li J, Yin C, Zhao J, Li F. Primary tumor site and anti-EGFR monoclonal antibody benefit in metastatic colorectal cancer: a meta-analysis. Future Oncol 2017; 13:1115-1127. [PMID: 28110551 DOI: 10.2217/fon-2016-0468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM This meta-analysis aimed to document the impact of primary tumor site on anti-EGFR monoclonal antibody (mAb) benefit in metastatic colorectal cancer. MATERIALS & METHODS Tumors with metastatic left-sided colorectal cancer (LCC) were compared with tumors with metastatic right-sided colon cancer (RCC) with respect to anti-EGFR mAb objective response rate (ORR), overall survival (OS) and progression-free survival (PFS) benefit. RESULTS Comparing LCC with RCC, LCC was found to have significantly superior anti-EGFR mAb ORR (p < 0.00001), OS (p < 0.00001) and PFS (p < 0.00001) benefit. Additionally, anti-EGFR mAb therapy significantly improved both OS and PFS for LCC compared with no anti-EGFR mAb therapy, but not for RCC. The test of interaction was also apparent for OS (p = 0.0002) and PFS (p = 0.0002). CONCLUSION This meta-analysis demonstrated that LCC had markedly superior anti-EGFR mAb treatment benefit compared with RCC.
Collapse
Affiliation(s)
- Dandan Li
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.,Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Qiang Fu
- Department of Hepatobiliary Pancreatic Surgery, People's Hospital of Zhengzhou University, Zhengzhou University School of Medicine, Zhengzhou, Henan, China
| | - Man Li
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.,Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jun Li
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.,Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Can Yin
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.,Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jin Zhao
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.,Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Feng Li
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.,Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China.,Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.,Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
111
|
Abstract
In the last 20 years, improvements in metastatic colorectal cancer treatment lead to a radical raise of outcomes with median survival reaching now more than 30 months. Despite that, the identification of predictive and/or prognostic biomarker still represents a challenging issue, and until today, although clinician and researchers might face with a deeper knowledge of biological mechanisms related to colorectal cancer, many pieces of evidence underline the heterogeneity and the dynamism of such disease. In the present review, we describe the road leading to the discovery of RAS mutations, BRAF V600E mutation, and microsatellite instability role in colorectal cancer; second, we discuss some of the possible major pitfalls of biomarker research, and lastly, we give new suggestions for future research in this field.
Collapse
|
112
|
Nakayama I, Shinozaki E, Matsushima T, Wakatsuki T, Ogura M, Ichimura T, Ozaka M, Takahari D, Suenaga M, Chin K, Mizunuma N, Yamaguchi K. Retrospective study of RAS/PIK3CA/BRAF tumor mutations as predictors of response to first-line chemotherapy with bevacizumab in metastatic colorectal cancer patients. BMC Cancer 2017; 17:38. [PMID: 28068936 PMCID: PMC5223326 DOI: 10.1186/s12885-016-2994-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 12/13/2016] [Indexed: 12/22/2022] Open
Abstract
Background After analysis of minor RAS mutations (KRAS exon 3, 4/NRAS) in the FIRE-3 and PRIME studies, an expanded range of RAS mutations were established as a negative predictive marker for the efficacy of anti-EGFR antibody treatment. BRAF and PIK3CA mutations may be candidate biomarkers for anti-EGFR targeted therapies. However, it remains unknown whether RAS/PIK3CA/BRAF tumor mutations can predict the efficacy of bevacizumab in metastatic colorectal cancer. We assessed whether selection according to RAS/PIK3CA/BRAF mutational status could be beneficial for patients treated with bevacizumab as first-line treatment for metastatic colorectal cancer. Methods Of the 1001 consecutive colorectal cancer patients examined for RAS, PIK3CA, and BRAF tumor mutations using a multiplex kit (Luminex®), we studied 90 patients who received combination chemotherapy with bevacizumab as first-line treatment for metastatic colorectal cancer. The objective response rate (ORR) and progression-free survival (PFS) were evaluated according to mutational status. Results The ORR was higher among patients with wild-type tumors (64.3%) compared to those with tumors that were only wild type with respect to KRAS exon 2 (54.8%), and the differences in ORR between patients with wild-type and mutant-type tumors were greater when considering only KRAS exon 2 mutations (6.8%) rather than RAS/PIK3CA/BRAF mutations (18.4%). There were no statistically significant differences in ORR or PFS between all wild-type tumors and tumors carrying any of the mutations. Multivariate analysis revealed that liver metastasis and RAS and BRAF mutations were independent negative factors for disease progression after first-line treatment with bevacizumab. Conclusions Patient selection according to RAS/PIK3CA/BRAF mutations could help select patients who will achieve a better response to bevacizumab treatment. We found no clinical benefit of restricting combination therapy with bevacizumab for metastatic colorectal cancer patients with EGFR-wild type tumors. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2994-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Izuma Nakayama
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Eiji Shinozaki
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Tomohiro Matsushima
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takeru Wakatsuki
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Mariko Ogura
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takashi Ichimura
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Masato Ozaka
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Daisuke Takahari
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Mitsukuni Suenaga
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Keisho Chin
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Nobuyuki Mizunuma
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| |
Collapse
|
113
|
Li Y, Li W. BRAF mutation is associated with poor clinicopathological outcomes in colorectal cancer: A meta-analysis. Saudi J Gastroenterol 2017; 23:144-149. [PMID: 28611337 PMCID: PMC5470373 DOI: 10.4103/1319-3767.207712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/AIMS The clinical relevance of the BRAF mutation in colorectal carcinoma (CRC) remains controversial. We performed a comprehensive meta-analysis to evaluate the precise relationship of BRAF mutation to clinicopathological features. MATERIALS AND METHODS A systematic search of the electronic databases, including PubMed, the Web of Knowledge, and the China Journal Net was performed between January 2005 and December 2015. Outcomes of interest included gender, tumor site, tumor differentiation, node involvement, tumor size, and AJCC stage. We calculated the pooled odds ratios (ORs) or risk ratios with 95% confidence intervals (CIs) for each study using a random or fixed-effect model. RESULTS Twenty-five studies with a total of 13208 patients were included. BRAF mutation-positive CRC patients were 1464 (11.1%). Our meta-analysis revealed that, in patients with CRC, the BRAF mutation was associated with female, proximal site, poor differentiation, >5 cm size, and advanced AJCC stage. CONCLUSIONS This meta-analysis demonstrated that BRAF mutation was closely related to adverse pathological features and poor outcome of CRC.
Collapse
Affiliation(s)
- Yujie Li
- Department of Surgical Oncology, Ningbo NO. 2 Hospital, Hangzhou, Zhejiang, China
| | - Weier Li
- Department of Colorectal Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,Address for correspondence: Dr. Weier Li, Department of Colorectal Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China. E-mail:
| |
Collapse
|
114
|
Immunohistochemistry with the anti-BRAF V600E (VE1) antibody: impact of pre-analytical conditions and concordance with DNA sequencing in colorectal and papillary thyroid carcinoma. Pathology 2016; 46:509-17. [PMID: 25014730 PMCID: PMC4233678 DOI: 10.1097/pat.0000000000000119] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Summary The most common of all activating BRAF mutations (T1799A) leads to a substitution of valine (V) to glutamic acid (E) at the position 600 of the amino acid sequence. The major goal of this study was to compare detection of the BRAF V600E mutation by DNA sequencing with immunohistochemistry (IHC) using the anti-BRAF V600E (VE1) antibody. Archival formalin fixed, paraffin embedded tissues from 352 patients with colon adenocarcinoma (n = 279) and papillary thyroid carcinoma (n = 73) were evaluated for the BRAF V600E mutation by sequencing and IHC. The discordant cases were re-evaluated by repeat IHC, SNaPshot and next-generation sequencing (NGS). Furthermore, the effect of pre-analytical variables on the utility of this antibody was evaluated in two xenograft mouse models. After resolving 15 initially discordant cases, 212 cases were negative for the BRAF V600E mutation by IHC. Of these, 210 cases (99.1%) were also negative by sequencing and two cases (0.9%) remained discordant. Of the 140 cases that were IHC positive for BRAF V600E, 138 cases were confirmed by sequencing (98.6%) and two cases remained discordant (1.4%). Overall, the negative predictive value was 99.1%, positive predictive value 98.6%, sensitivity 98.6%, specificity 99.1% and overall percentage agreement 98.9% (348/352 cases). Tissue fixation studies indicated that tissues should be fixed for 12–24 h within 2 h of tissue collection with 10% neutral buffered formalin.
Collapse
|
115
|
Chemotherapeutic drug selectivity between wild-type and mutant BRaf kinases in colon cancer. J Mol Model 2016; 23:1. [DOI: 10.1007/s00894-016-3177-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/22/2016] [Indexed: 01/19/2023]
|
116
|
Atreya CE, Greene C, McWhirter RM, Ikram NS, Allen IE, Van Loon K, Venook AP, Yeh BM, Behr SC. Differential Radiographic Appearance of BRAF V600E-Mutant Metastatic Colorectal Cancer in Patients Matched by Primary Tumor Location. J Natl Compr Canc Netw 2016; 14:1536-1543. [PMID: 27956538 PMCID: PMC5551390 DOI: 10.6004/jnccn.2016.0165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND BRAF-mutant metastatic colorectal cancers (mCRCs) share many clinicopathologic features with right-sided colon tumors, including frequent peritoneal involvement. Because of the poorer outcomes associated with BRAF mutations, early enrollment in clinical trials has been encouraged. However, the use of standard eligibility and assessment criteria, such as measurable disease, has anecdotally impeded patient accrual and restricted appraisal of treatment response. We investigated whether the presence of a BRAF V600E mutation is differentially associated with sites and appearance of metastatic disease in patients matched by primary tumor location. METHODS A total of 40 patients with BRAF-mutant mCRC were matched to 80 patients with BRAF wild-type mCRC by location of primary tumor (right or left colon; rectum), sex, and age. Associations between BRAF mutation status and clinicopathologic characteristics and metastatic sites were analyzed using proportion tests. Survival was summarized with Kaplan-Meier and Cox regression methods. RESULTS The distribution of primary tumor locations was: 60% right colon, 30% left colon, and 10% rectum. Compared with BRAF wild-type tumors, BRAF-mutant tumors more commonly associated with peritoneal metastases (50% vs 31%; P=.045) and ascites (50% vs 24%; P=.0038). In patients with left colon primaries, BRAF mutations were associated with more frequent ascites (58% vs 12%; P=.0038) and less frequent liver metastases (42% vs 79%; P=.024). Among patients with right colon primaries, no significant difference in sites of disease by BRAF mutation status was observed. Disease was not measurable by RECIST 1.1 in 24% of patients with right-sided primary tumors, irrespective of BRAF mutation status. In the BRAF-mutated cohort, ascites correlated unfavorably with survival (hazard ratio, 2.35; 95% CI, 1.14, 4.83; P=.02). CONCLUSIONS Greater frequency of ascites and peritoneal metastases, which pose challenges for RECIST 1.1 interpretation of therapeutic outcomes, are seen with BRAF-mutant mCRC, even when patients are matched for primary tumor location.
Collapse
Affiliation(s)
- Chloe E. Atreya
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center
| | - Claire Greene
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center
| | - Ryan M. McWhirter
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center
| | | | - I. Elaine Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Katherine Van Loon
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center
| | - Alan P. Venook
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center
| | | | | |
Collapse
|
117
|
Park T, Choi CJ, Choi Y, Suh DC. Cost-effectiveness of cetuximab for colorectal cancer. Expert Rev Pharmacoecon Outcomes Res 2016; 16:667-677. [DOI: 10.1080/14737167.2016.1245618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Taehwan Park
- Department of Pharmacy Administration, St Louis College of Pharmacy, St. Louis, MO, USA
| | - Chang-ju Choi
- Department of Pharmacy, Chung-Ang University College of Pharmacy, Seoul, South Korea
| | - Yeera Choi
- Department of Pharmacy, Chung-Ang University College of Pharmacy, Seoul, South Korea
| | - Dong-Churl Suh
- Department of Pharmacy, Chung-Ang University College of Pharmacy, Seoul, South Korea
| |
Collapse
|
118
|
Kawakami H, Huang S, Pal K, Dutta SK, Mukhopadhyay D, Sinicrope FA. Mutant BRAF Upregulates MCL-1 to Confer Apoptosis Resistance that Is Reversed by MCL-1 Antagonism and Cobimetinib in Colorectal Cancer. Mol Cancer Ther 2016; 15:3015-3027. [PMID: 27765849 DOI: 10.1158/1535-7163.mct-16-0017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 08/26/2016] [Accepted: 09/27/2016] [Indexed: 01/19/2023]
Abstract
Oncogenic BRAFV600E mutations activate MAPK signaling and are associated with treatment resistance and poor prognosis in patients with colorectal cancer. In BRAFV600E-mutant colorectal cancers, treatment failure may be related to BRAFV600E-mediated apoptosis resistance that occurs by an as yet undefined mechanism. We found that BRAFV600E can upregulate anti-apoptotic MCL-1 in a gene dose-dependent manner using colorectal cancer cell lines isogenic for BRAF BRAFV600E-induced MCL-1 upregulation was confirmed by ectopic BRAFV600E expression that activated MEK/ERK signaling to phosphorylate (MCL-1Thr163) and stabilize MCL-1. Upregulation of MCL-1 was mediated by MEK/ERK shown by the ability of ERK siRNA to suppress MCL-1. Stabilization of MCL-1 by phosphorylation was shown by a phosphorylation-mimicking mutant and an unphosphorylated MCL-1 mutant that decreased or increased MCL-1 protein turnover, respectively. MEK/ERK inhibition by cobimetinib suppressed MCL-1 expression/phosphorylation and induced proapoptotic BIM to a greater extent than did vemurafenib in BRAFV600E cell lines. MCL-1 knockdown versus control shRNA significantly enhanced cobimetinib-induced apoptosis in vitro and in HT29 colon cancer xenografts. The small-molecule MCL-1 inhibitor, A-1210477, also enhanced cobimetinib-induced apoptosis in vitro that was due to disruption of the interaction of MCL-1 with proapoptotic BAK and BIM. Knockdown of BIM attenuated BAX, but not BAK, activation by cobimetinib plus A-1210477. In summary, BRAFV600E-mediated MEK/ERK activation can upregulate MCL-1 by phosphorylation/stabilization to confer apoptosis resistance that can be reversed by MCL-1 antagonism combined with cobimetinib, suggesting a novel therapeutic strategy against BRAFV600E-mutant CRCs. Mol Cancer Ther; 15(12); 3015-27. ©2016 AACR.
Collapse
Affiliation(s)
- Hisato Kawakami
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Shengbing Huang
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida
| | | | - Frank A Sinicrope
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
119
|
A Vulnerability of a Subset of Colon Cancers with Potential Clinical Utility. Cell 2016; 165:317-30. [PMID: 27058664 DOI: 10.1016/j.cell.2016.02.059] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 12/20/2015] [Accepted: 02/22/2016] [Indexed: 01/11/2023]
Abstract
BRAF(V600E) mutant colon cancers (CCs) have a characteristic gene expression signature that is also found in some tumors lacking this mutation. Collectively, they are referred to as "BRAF-like" tumors and represent some 20% of CCs. We used a shRNA-based genetic screen focused on genes upregulated in BRAF(V600E) CCs to identify vulnerabilities of this tumor subtype that might be exploited therapeutically. Here, we identify RANBP2 (also known as NUP358) as essential for survival of BRAF-like, but not for non-BRAF-like, CC cells. Suppression of RANBP2 results in mitotic defects only in BRAF-like CC cells, leading to cell death. Mechanistically, RANBP2 silencing reduces microtubule outgrowth from the kinetochores, thereby inducing spindle perturbations, providing an explanation for the observed mitotic defects. We find that BRAF-like CCs display far greater sensitivity to the microtubule poison vinorelbine both in vitro and in vivo, suggesting that vinorelbine is a potential tailored treatment for BRAF-like CCs.
Collapse
|
120
|
Matos P, Gonçalves V, Jordan P. Targeting the serrated pathway of colorectal cancer with mutation in BRAF. Biochim Biophys Acta Rev Cancer 2016; 1866:51-63. [DOI: 10.1016/j.bbcan.2016.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/15/2016] [Accepted: 06/19/2016] [Indexed: 12/19/2022]
|
121
|
Ahn DH, Ciombor KK, Mikhail S, Bekaii-Saab T. Genomic diversity of colorectal cancer: Changing landscape and emerging targets. World J Gastroenterol 2016; 22:5668-5677. [PMID: 27433082 PMCID: PMC4932204 DOI: 10.3748/wjg.v22.i25.5668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/20/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023] Open
Abstract
Improvements in screening and preventive measures have led to an increased detection of early stage colorectal cancers (CRC) where patients undergo treatment with a curative intent. Despite these efforts, a high proportion of patients are diagnosed with advanced stage disease that is associated with poor outcomes, as CRC remains one of the leading causes of cancer-related deaths in the world. The development of next generation sequencing and collaborative multi-institutional efforts to characterize the cancer genome has afforded us with a comprehensive assessment of the genomic makeup present in CRC. This knowledge has translated into understanding the prognostic role of various tumor somatic variants in this disease. Additionally, the awareness of the genomic alterations present in CRC has resulted in an improvement in patient outcomes, largely due to better selection of personalized therapies based on an individual’s tumor genomic makeup. The benefit of various treatments is often limited, where recent studies assessing the genomic diversity in CRC have identified the development of secondary tumor somatic variants that likely contribute to acquired treatment resistance. These studies have begun to alter the landscape of treatment for CRC that include investigating novel targeted therapies, assessing the role of immunotherapy and prospective, dynamic assessment of changes in tumor genomic alterations that occur during the treatment of CRC.
Collapse
|
122
|
Oddo D, Sennott EM, Barault L, Valtorta E, Arena S, Cassingena A, Filiciotto G, Marzolla G, Elez E, van Geel RMJM, Bartolini A, Crisafulli G, Boscaro V, Godfrey JT, Buscarino M, Cancelliere C, Linnebacher M, Corti G, Truini M, Siravegna G, Grasselli J, Gallicchio M, Bernards R, Schellens JHM, Tabernero J, Engelman JA, Sartore-Bianchi A, Bardelli A, Siena S, Corcoran RB, Di Nicolantonio F. Molecular Landscape of Acquired Resistance to Targeted Therapy Combinations in BRAF-Mutant Colorectal Cancer. Cancer Res 2016; 76:4504-15. [PMID: 27312529 DOI: 10.1158/0008-5472.can-16-0396] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/24/2016] [Indexed: 12/16/2022]
Abstract
Although recent clinical trials of BRAF inhibitor combinations have demonstrated improved efficacy in BRAF-mutant colorectal cancer, emergence of acquired resistance limits clinical benefit. Here, we undertook a comprehensive effort to define mechanisms underlying drug resistance with the goal of guiding development of therapeutic strategies to overcome this limitation. We generated a broad panel of BRAF-mutant resistant cell line models across seven different clinically relevant drug combinations. Combinatorial drug treatments were able to abrogate ERK1/2 phosphorylation in parental-sensitive cells, but not in their resistant counterparts, indicating that resistant cells escaped drug treatments through one or more mechanisms leading to biochemical reactivation of the MAPK signaling pathway. Genotyping of resistant cells identified gene amplification of EGFR, KRAS, and mutant BRAF, as well as acquired mutations in KRAS, EGFR, and MAP2K1 These mechanisms were clinically relevant, as we identified emergence of a KRAS G12C mutation and increase of mutant BRAF V600E allele frequency in the circulating tumor DNA of a patient at relapse from combined treatment with BRAF and MEK inhibitors. To identify therapeutic combinations capable of overcoming drug resistance, we performed a systematic assessment of candidate therapies across the panel of resistant cell lines. Independent of the molecular alteration acquired upon drug pressure, most resistant cells retained sensitivity to vertical MAPK pathway suppression when combinations of ERK, BRAF, and EGFR inhibitors were applied. These therapeutic combinations represent promising strategies for future clinical trials in BRAF-mutant colorectal cancer. Cancer Res; 76(15); 4504-15. ©2016 AACR.
Collapse
Affiliation(s)
- Daniele Oddo
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | - Erin M Sennott
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Ludovic Barault
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | - Emanuele Valtorta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Sabrina Arena
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | - Andrea Cassingena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Genny Filiciotto
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | - Giulia Marzolla
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | - Elena Elez
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Alice Bartolini
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | | | - Valentina Boscaro
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Jason T Godfrey
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | | | - Michael Linnebacher
- Department of General Surgery, Division of Molecular Oncology and Immunotherapy, University of Rostock, Rostock, Germany
| | - Giorgio Corti
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | - Mauro Truini
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giulia Siravegna
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy. FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Julieta Grasselli
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - René Bernards
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | | | - Alberto Bardelli
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy. Department of Oncology, Università degli Studi di Milano, Milan, Italy
| | - Ryan B Corcoran
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Torino, Italy.
| |
Collapse
|
123
|
Abstract
Substantial progress has been made in the treatment of colorectal cancer, where more effective therapies have led to improved outcomes in patients with advanced disease. However, the 5-year overall survival rate remains poor. Genomic sequencing has allowed us to understand that colorectal cancer is a heterogeneous disease, where tumor-specific variants affect the prognosis and outcomes in patients. This has shaped the future directions of treatment and the development of clinical trials, including the incorporation of novel targeted therapies and investigations into the role of immunotherapy in colorectal cancer.
Collapse
Affiliation(s)
- Daniel H Ahn
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 460 W 10th Ave., Columbus, OH 43210, USA
| | - Richard M Goldberg
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 460 W 10th Ave., Columbus, OH 43210, USA
| |
Collapse
|
124
|
Loupakis F, Moretto R, Aprile G, Muntoni M, Cremolini C, Iacono D, Casagrande M, Ferrari L, Salvatore L, Schirripa M, Rossini D, De Maglio G, Fasola G, Calvetti L, Pilotto S, Carbognin L, Fontanini G, Tortora G, Falcone A, Sperduti I, Bria E. Clinico-pathological nomogram for predicting BRAF mutational status of metastatic colorectal cancer. Br J Cancer 2016; 114:30-6. [PMID: 26575603 PMCID: PMC4716533 DOI: 10.1038/bjc.2015.399] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/15/2015] [Accepted: 10/20/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In metastatic colorectal cancer (mCRC), BRAFV600E mutation has been variously associated to specific clinico-pathological features. METHODS Two large retrospective series of mCRC patients from two Italian Institutions were used as training-set (TS) and validation-set (VS) for developing a nomogram predictive of BRAFV600E status. The model was internally and externally validated. RESULTS In the TS, data from 596 mCRC patients were gathered (RAS wild-type (wt) 281 (47.1%); BRAFV600E mutated 54 (9.1%)); RAS and BRAFV600E mutations were mutually exclusive. In the RAS-wt population, right-sided primary (odds ratio (OR): 7.80, 95% confidence interval (CI) 3.05-19.92), female gender (OR: 2.90, 95% CI 1.14-7.37) and mucinous histology (OR: 4.95, 95% CI 1.90-12.90) were independent predictors of BRAFV600E mutation, with high replication at internal validation (100%, 93% and 98%, respectively). A predictive nomogram was calculated: patients with the highest score (right-sided primary, female and mucinous) had a 81% chance to bear a BRAFV600E-mutant tumour; accuracy measures: AUC=0.812, SE:0.034, sensitivity:81.2%; specificity:72.1%. In the VS (508 pts, RAS wt: 262 (51.6%), BRAFV600E mutated: 49 (9.6%)), right-sided primary, female gender and mucinous histology were confirmed as independent predictors of BRAFV600E mutation with high accuracy. CONCLUSIONS Three simple and easy-to-collect characteristics define a useful nomogram for predicting BRAF status in mCRC with high specificity and sensitivity.
Collapse
Affiliation(s)
- Fotios Loupakis
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Roberto Moretto
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Giuseppe Aprile
- Dipartimento di Oncologia, Azienda Ospedaliero-Universitaria ‘Santa Maria della Misericordia', Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy
| | - Marta Muntoni
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Chiara Cremolini
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Donatella Iacono
- Dipartimento di Oncologia, Azienda Ospedaliero-Universitaria ‘Santa Maria della Misericordia', Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy
| | - Mariaelena Casagrande
- Dipartimento di Oncologia, Azienda Ospedaliero-Universitaria ‘Santa Maria della Misericordia', Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy
| | - Laura Ferrari
- Dipartimento di Oncologia, Azienda Ospedaliero-Universitaria ‘Santa Maria della Misericordia', Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy
| | - Lisa Salvatore
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Marta Schirripa
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Daniele Rossini
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Giovanna De Maglio
- Dipartimento di Medicina di Laboratorio, Azienda Ospedaliero-Universitaria ‘Santa Maria della Misericordia', Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy
| | - Gianpiero Fasola
- Dipartimento di Oncologia, Azienda Ospedaliero-Universitaria ‘Santa Maria della Misericordia', Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy
| | - Lorenzo Calvetti
- Dipartimento di Medicina, Oncologia Medica, University of Verona, Azienda Ospedaliera Universitaria Integrata, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| | - Sara Pilotto
- Dipartimento di Medicina, Oncologia Medica, University of Verona, Azienda Ospedaliera Universitaria Integrata, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| | - Luisa Carbognin
- Dipartimento di Medicina, Oncologia Medica, University of Verona, Azienda Ospedaliera Universitaria Integrata, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| | - Gabriella Fontanini
- Divisione di Patologia, Dipartimento di Chirurgia, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Giampaolo Tortora
- Dipartimento di Medicina, Oncologia Medica, University of Verona, Azienda Ospedaliera Universitaria Integrata, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| | - Alfredo Falcone
- Oncologia Medica 2 Universitaria, Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - Isabella Sperduti
- Direzione Scientifica, Biostatistics, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Emilio Bria
- Dipartimento di Medicina, Oncologia Medica, University of Verona, Azienda Ospedaliera Universitaria Integrata, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| |
Collapse
|
125
|
Abstract
Oncogenic BRAF mutations are found in ~10% of colorectal cancers (CRCs) and predict poor prognosis. Although BRAF inhibitors have demonstrated striking efficacy in BRAF mutant melanomas, BRAF inhibitor monotherapy is ineffective in BRAF mutant CRC. Over the past few years, studies have begun to define the molecular mechanisms underlying the relative resistance of BRAF mutant CRC to BRAF inhibitors, leading to the development of novel therapeutic strategies that are showing promising clinical activity in initial clinical trials. Our current understanding of the mechanisms of BRAF inhibitor resistance in BRAF mutant CRC and the therapeutic approaches currently in clinical trials for BRAF mutant CRC are reviewed herein.
Collapse
Affiliation(s)
- Ryan B Corcoran
- 1 Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA ; 2 Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
126
|
Clarke CN, Kopetz ES. BRAF mutant colorectal cancer as a distinct subset of colorectal cancer: clinical characteristics, clinical behavior, and response to targeted therapies. J Gastrointest Oncol 2015; 6:660-7. [PMID: 26697199 DOI: 10.3978/j.issn.2078-6891.2015.077] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Despite new and more effective cytotoxic chemotherapy, limitations to conventional agents have been reached in a subset of patients with advanced colorectal cancer (CRC). The identification of novel prognostic and predictive biomarkers to guide individualized treatment plans is critical to overcoming therapeutic resistance. Mutation of the BRAF proto-oncogene is linked to a variety of cancers and is increasingly being used as a prognostic tool and therapeutic target. This paper is a comprehensive review of the literature that summarizes the clinical, pathologic, and molecular features of BRAF mutated CRC that support the hypothesis that BRAF mutant cancers represent a distinct subset of CRC with its own clinical implications with regard to prognosis, treatments and emerging therapeutic strategies.
Collapse
Affiliation(s)
- Callisia N Clarke
- 1 Department of Surgical Oncology, 2 Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - E Scott Kopetz
- 1 Department of Surgical Oncology, 2 Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
127
|
BRAF Mutations in Non-Metastatic Colorectal Cancer: Current Relevance and Future Implications. CURRENT COLORECTAL CANCER REPORTS 2015. [DOI: 10.1007/s11888-015-0295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
128
|
Kopetz S, Desai J, Chan E, Hecht JR, O'Dwyer PJ, Maru D, Morris V, Janku F, Dasari A, Chung W, Issa JPJ, Gibbs P, James B, Powis G, Nolop KB, Bhattacharya S, Saltz L. Phase II Pilot Study of Vemurafenib in Patients With Metastatic BRAF-Mutated Colorectal Cancer. J Clin Oncol 2015; 33:4032-8. [PMID: 26460303 PMCID: PMC4669589 DOI: 10.1200/jco.2015.63.2497] [Citation(s) in RCA: 520] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose BRAF V600E mutation is seen in 5% to 8% of patients with metastatic colorectal cancer (CRC) and is associated with poor prognosis. Vemurafenib, an oral BRAF V600 inhibitor, has pronounced activity in patients with metastatic melanoma, but its activity in patients with BRAF V600E–positive metastatic CRC was unknown. Patients and Methods In this multi-institutional, open-label study, patients with metastatic CRC with BRAF V600 mutations were recruited to an expansion cohort at the previously determined maximum-tolerated dose of 960 mg orally twice a day. Results Twenty-one patients were enrolled, of whom 20 had received at least one prior metastatic chemotherapy regimen. Grade 3 toxicities included keratoacanthomas, rash, fatigue, and arthralgia. Of the 21 patients treated, one patient had a confirmed partial response (5%; 95% CI, 1% to 24%) and seven other patients had stable disease by RECIST criteria. Median progression-free survival was 2.1 months. Patterns of concurrent mutations, microsatellite instability status, CpG island methylation status, PTEN loss, EGFR expression, and copy number alterations were not associated with clinical benefit. In contrast to prior expectations, concurrent KRAS and NRAS mutations were detected at low allele frequency in a subset of the patients' tumors (median, 0.21% allele frequency) and were apparent mechanisms of acquired resistance in vemurafenib-sensitive patient-derived xenograft models. Conclusion In marked contrast to the results seen in patients with BRAF V600E–mutant melanoma, single-agent vemurafenib did not show meaningful clinical activity in patients with BRAF V600E mutant CRC. Combination strategies are now under development and may be informed by the presence of intratumor heterogeneity of KRAS and NRAS mutations.
Collapse
Affiliation(s)
- Scott Kopetz
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia.
| | - Jayesh Desai
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Emily Chan
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Joel Randolph Hecht
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Peter J O'Dwyer
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Dipen Maru
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Van Morris
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Filip Janku
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Arvind Dasari
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Woonbook Chung
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Jean-Pierre J Issa
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Peter Gibbs
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Brian James
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Garth Powis
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Keith B Nolop
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Suman Bhattacharya
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Leonard Saltz
- Scott Kopetz, Dipen Maru, Van Morris, Filip Janku, and Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Emily Chan, Vanderbilt-Ingram Cancer Center, Nashville, TN; Joel Randolph Hecht, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles; Brian James and Garth Powis, Sanford Burnham Institute, La Jolla; Keith B. Nolop, Plexxikon, Berkeley; Suman Bhattacharya, Genentech, South San Francisco, CA; Peter J. O'Dwyer, Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA; Woonbook Chung and Jean-Pierre J. Issa, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA; Leonard Saltz, Memorial Sloan-Kettering Cancer Center, New York, NY; and Jayesh Desai and Peter Gibbs, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
129
|
Geramizadeh B. Molecular Biomarkers of Colorectal Cancer: A Review of Published Articles From Iran. ACTA ACUST UNITED AC 2015. [DOI: 10.17795/acr-30100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
130
|
Corcoran RB, Atreya CE, Falchook GS, Kwak EL, Ryan DP, Bendell JC, Hamid O, Messersmith WA, Daud A, Kurzrock R, Pierobon M, Sun P, Cunningham E, Little S, Orford K, Motwani M, Bai Y, Patel K, Venook AP, Kopetz S. Combined BRAF and MEK Inhibition With Dabrafenib and Trametinib in BRAF V600-Mutant Colorectal Cancer. J Clin Oncol 2015; 33:4023-31. [PMID: 26392102 DOI: 10.1200/jco.2015.63.2471] [Citation(s) in RCA: 389] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To evaluate dabrafenib, a selective BRAF inhibitor, combined with trametinib, a selective MEK inhibitor, in patients with BRAF V600-mutant metastatic colorectal cancer (mCRC). PATIENTS AND METHODS A total of 43 patients with BRAF V600-mutant mCRC were treated with dabrafenib (150 mg twice daily) plus trametinib (2 mg daily), 17 of whom were enrolled onto a pharmacodynamic cohort undergoing mandatory biopsies before and during treatment. Archival tissues were analyzed for microsatellite instability, PTEN status, and 487-gene sequencing. Patient-derived xenografts were established from core biopsy samples. RESULTS Of 43 patients, five (12%) achieved a partial response or better, including one (2%) complete response, with duration of response > 36 months; 24 patients (56%) achieved stable disease as best confirmed response. Ten patients (23%) remained in the study > 6 months. All nine evaluable during-treatment biopsies had reduced levels of phosphorylated ERK relative to pretreatment biopsies (average decrease ± standard deviation, 47% ± 24%). Mutational analysis revealed that the patient achieving a complete response and two of three evaluable patients achieving a partial response had PIK3CA mutations. Neither PTEN loss nor microsatellite instability correlated with efficacy. Responses to dabrafenib plus trametinib were comparable in patient-derived xenograft-bearing mice and the biopsied lesions from each corresponding patient. CONCLUSION The combination of dabrafenib plus trametinib has activity in a subset of patients with BRAF V600-mutant mCRC. Mitogen-activated protein kinase signaling was inhibited in all patients evaluated, but to a lesser degree than observed in BRAF-mutant melanoma with dabrafenib alone. PIK3CA mutations were identified in responding patients and thus do not preclude response to this regimen. Additional studies targeting the mitogen-activated protein kinase pathway in this disease are warranted.
Collapse
Affiliation(s)
- Ryan B Corcoran
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE.
| | - Chloe E Atreya
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Gerald S Falchook
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Eunice L Kwak
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - David P Ryan
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Johanna C Bendell
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Omid Hamid
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Wells A Messersmith
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Adil Daud
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Razelle Kurzrock
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Mariaelena Pierobon
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Peng Sun
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Elizabeth Cunningham
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Shonda Little
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Keith Orford
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Monica Motwani
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Yuchen Bai
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Kiran Patel
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Alan P Venook
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| | - Scott Kopetz
- Ryan B. Corcoran, Eunice L. Kwak, and David P. Ryan, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; Chloe E. Atreya, Adil Daud, and Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco; Omid Hamid, Angeles Clinic and Research Institute, Los Angeles, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver; Wells A. Messersmith, University of Colorado Cancer Center and University of Colorado, Aurora, CO; Johanna C. Bendell, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Razelle Kurzrock and Scott Kopetz, University of Texas, MD Anderson Cancer Center, Houston, TX; Mariaelena Pierobon, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA; Peng Sun, Elizabeth Cunningham, Shonda Little, Keith Orford, Monica Motwani, and Yuchen Bai, GlaxoSmithKline, Philadelphia, PA; and Kiran Patel, Incyte, Wilmington, DE
| |
Collapse
|
131
|
Coulson R. Molecular Profiling in Resectable Colorectal Liver Metastases: The Role of KRAS Mutation Status in Assessing Prognosis in the Preoperative Setting. J Adv Pract Oncol 2015; 6:470-4. [PMID: 27069739 PMCID: PMC4803464 DOI: 10.6004/jadpro.2015.6.5.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
132
|
Cremolini C, Loupakis F, Antoniotti C, Lupi C, Sensi E, Lonardi S, Mezi S, Tomasello G, Ronzoni M, Zaniboni A, Tonini G, Carlomagno C, Allegrini G, Chiara S, D'Amico M, Granetto C, Cazzaniga M, Boni L, Fontanini G, Falcone A. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol 2015; 16:1306-15. [PMID: 26338525 DOI: 10.1016/s1470-2045(15)00122-9] [Citation(s) in RCA: 732] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 06/22/2015] [Accepted: 06/24/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND In the TRIBE study, FOLFOXIRI (fluorouracil, leucovorin, oxaliplatin, and irinotecan) plus bevacizumab significantly improved progression-free survival of patients with metastatic colorectal cancer compared with FOLFIRI (fluorouracil, leucovorin, and irinotecan) plus bevacizumab. In this updated analysis, we aimed to provide mature results for overall survival-a secondary endpoint-and report treatment efficacy in RAS and BRAF molecular subgroups. METHODS TRIBE was an open-label, multicentre, phase 3 randomised study of patients (aged 18-70 years with Eastern Cooperative Oncology Group [ECOG] performance status of 2 or less and aged 71-75 years with an ECOG performance status of 0) with unresectable metastatic colorectal cancer who were recruited from 34 Italian oncology units. Patients were randomly assigned (1:1) via a web-based procedure to receive FOLFIRI plus bevacizumab or FOLFOXIRI plus bevacizumab. Bevacizumab was given as a 5 mg/kg intravenous dose. FOLFIRI consisted of a 180 mg/m(2) intravenous infusion of irinotecan for 60 min followed by a 200 mg/m(2) intravenous infusion of leucovorin for 120 min, a 400 mg/m(2) intravenous bolus of fluorouracil, and a 2400 mg/m(2) continuous infusion of fluorouracil for 46 h. FOLFOXIRI consisted of a 165 mg/m(2) intravenous infusion of irinotecan for 60 min, followed by an 85 mg/m(2) intravenous infusion of oxaliplatin given concurrently with 200 mg/m(2) leucovorin for 120 min, followed by a 3200 mg/m(2) continuous infusion of fluorouracil for 48 h. Tissue samples for RAS and BRAF mutational status analyses were centrally collected. In this updated analysis, we assessed the secondary endpoint of overall survival in the main cohort and treatment efficacy in RAS and BRAF molecular subgroups. All analyses were by intention to treat. TRIBE was concluded on Nov 30, 2014. The trial is registered with ClinicalTrials.gov, number NCT00719797. FINDINGS Between July 17, 2008, and May 31, 2011, 508 patients were randomly assigned. At a median follow-up of 48·1 months (IQR 41·7-55·6), median overall survival was 29·8 months (95% CI 26·0-34·3) in the FOLFOXIRI plus bevacizumab group compared with 25·8 months (22·5-29·1) in the FOLFIRI plus bevacizumab group (hazard ratio [HR] 0·80, 95% CI 0·65-0·98; p=0·03). Median overall survival was 37·1 months (95% CI 29·7-42·7) in the RAS and BRAF wild-type subgroup compared with 25·6 months (22·4-28·6) in the RAS-mutation-positive subgroup (HR 1·49, 95% CI 1·11-1·99) and 13·4 months (8·2-24·1) in the BRAF-mutation-positive subgroup (HR 2·79, 95% CI 1·75-4·46; likelihood-ratio test p<0·0001). Treatment effect was not significantly different across molecular subgroups (pinteraction=0·52). INTERPRETATION FOLFOXIRI plus bevacizumab is a feasible treatment option for those patients who meet the inclusion criteria of the present study, irrespective of baseline clinical characteristics and RAS or BRAF mutational status.
Collapse
Affiliation(s)
- Chiara Cremolini
- Department of Oncology, University Hospital of Pisa, Pisa, Italy; Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Fotios Loupakis
- Department of Oncology, University Hospital of Pisa, Pisa, Italy; Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Carlotta Antoniotti
- Department of Oncology, University Hospital of Pisa, Pisa, Italy; Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Cristiana Lupi
- Department of Surgical Pathology, Medical, Molecular, and Critical Area, University of Pisa, Pisa, Italy
| | - Elisa Sensi
- Department of Surgical Pathology, Medical, Molecular, and Critical Area, University of Pisa, Pisa, Italy
| | - Sara Lonardi
- Medical Oncology 1, Oncology Institute IRCCS Veneto, Padua, Italy
| | - Silvia Mezi
- Day Hospital of Oncology, Policlinico Umberto I, Rome, Italy
| | - Gianluca Tomasello
- Division of Medicine and Medical Oncology, Hospital of Cremona, Cremona, Italy
| | - Monica Ronzoni
- Department of Oncology, Hospital San Raffaele IRCSS, Milan, Italy
| | - Alberto Zaniboni
- Department of Medical Oncology, Poliambulanza Foundation, Brescia, Italy
| | | | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, University Hospital Federico II, Naples, Italy
| | | | - Silvana Chiara
- Medical Oncology 2, IRCCS University Hospital San Martino, Genoa, Italy
| | - Mauro D'Amico
- Unit of Medical Oncology, Galliera Hospital, Genoa, Italy
| | - Cristina Granetto
- Unit of Medical Oncology, Medical Centre Hospital Santa Croce and Carle, Cuneo, Italy
| | | | - Luca Boni
- Clinical Trials Coordinating Center, Toscano Cancer Institute, University Hospital Careggi, Florence, Italy
| | - Gabriella Fontanini
- Department of Surgical Pathology, Medical, Molecular, and Critical Area, University of Pisa, Pisa, Italy
| | - Alfredo Falcone
- Department of Oncology, University Hospital of Pisa, Pisa, Italy; Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
133
|
Wild-type APC predicts poor prognosis in microsatellite-stable proximal colon cancer. Br J Cancer 2015; 113:979-88. [PMID: 26305864 PMCID: PMC4578087 DOI: 10.1038/bjc.2015.296] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/08/2015] [Accepted: 07/20/2015] [Indexed: 12/14/2022] Open
Abstract
Background: APC mutations (APC-mt) occur in ∼70% of colorectal cancers (CRCs), but their relationship to prognosis is unclear. Methods: APC prognostic value was evaluated in 746 stage I–IV CRC patients, stratifying for tumour location and microsatellite instability (MSI). Microarrays were used to identify a gene signature that could classify APC mutation status, and classifier ability to predict prognosis was examined in an independent cohort. Results: Wild-type APC microsatellite stable (APC-wt/MSS) tumours from the proximal colon showed poorer overall and recurrence-free survival (OS, RFS) than APC-mt/MSS proximal, APC-wt/MSS distal and APC-mt/MSS distal tumours (OS HR⩾1.79, P⩽0.015; RFS HR⩾1.88, P⩽0.026). APC was a stronger prognostic indicator than BRAF, KRAS, PIK3CA, TP53, CpG island methylator phenotype or chromosomal instability status (P⩽0.036). Microarray analysis similarly revealed poorer survival in MSS proximal cancers with an APC-wt-like signature (P=0.019). APC status did not affect outcomes in MSI tumours. In a validation on 206 patients with proximal colon cancer, APC-wt-like signature MSS cases showed poorer survival than APC-mt-like signature MSS or MSI cases (OS HR⩾2.50, P⩽0.010; RFS HR⩾2.14, P⩽0.025). Poor prognosis APC-wt/MSS proximal tumours exhibited features of the sessile serrated neoplasia pathway (P⩽0.016). Conclusions: APC-wt status is a marker of poor prognosis in MSS proximal colon cancer.
Collapse
|
134
|
Karapetis CS, Maru D, Waring P, Tie J, Michael MZ. Incorporating traditional and emerging biomarkers in the clinical management of metastatic colorectal cancer. Expert Rev Mol Diagn 2015; 15:1033-48. [PMID: 26166616 DOI: 10.1586/14737159.2015.1052797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The role of biomarker assessment in determining the best therapeutic options for patients with metastatic colorectal cancer has become increasingly complex and important. Biomarkers that predict the efficacy and/or toxicity of such treatments can affect medical decision making, leading to decreased harm and/or costs associated with treatment and improvements in therapeutic outcomes for patients. This review discusses traditional and emerging biomarkers of potential clinical utility for patients with metastatic colorectal cancer, current assays and methods used in clinical practice, technologies that have allowed the identification of new biomarkers and key considerations for oncologists and pathologists when determining appropriate biomarker evaluations to be undertaken for their patients.
Collapse
Affiliation(s)
- Christos S Karapetis
- Flinders Medical Centre, Sturt Road, Bedford Park, South Australia 5042, Australia
| | | | | | | | | |
Collapse
|
135
|
Cremolini C, Di Bartolomeo M, Amatu A, Antoniotti C, Moretto R, Berenato R, Perrone F, Tamborini E, Aprile G, Lonardi S, Sartore-Bianchi A, Fontanini G, Milione M, Lauricella C, Siena S, Falcone A, de Braud F, Loupakis F, Pietrantonio F. BRAF codons 594 and 596 mutations identify a new molecular subtype of metastatic colorectal cancer at favorable prognosis. Ann Oncol 2015; 26:2092-7. [PMID: 26153495 DOI: 10.1093/annonc/mdv290] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/01/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND While the negative prognostic role of BRAF V600E mutation in metastatic colorectal cancer (mCRC) is well established, the impact of BRAF codons 594 and 596 mutations, occurring in <1% of CRCs, is completely unknown. The present work aims to describe clinical, pathological and molecular features and prognosis of BRAF codons 594 and 596 mutant mCRCs, compared with BRAF V600E mutant and wild-type ones. PATIENTS AND METHODS Patients treated for mCRC at three Italian Institutions between October 2006 and October 2014, with available KRAS and NRAS codon 12, 13, 59, 61, 117 and 146 and BRAF codon 594, 596 and 600 mutational status, as detected by means of direct sequencing or matrix assisted laser desorption ionization time-of-flight MassArray, were included. RESULTS Ten patients bearing BRAF codons 594 or 596 mutated tumors were identified and compared with 77 and 542 patients bearing BRAF V600E mutated and BRAF wild-type tumors, respectively. While BRAF V600E mutated tumors were more frequently right-sided, mucinous and with peritoneal spread, BRAF 594 or 596 mutated were more frequently rectal, nonmucinous and with no peritoneal spread. All BRAF 594 or 596 mutated tumors were microsatellite stable. Patients with BRAF codons 594 or 596 mutated tumors had markedly longer overall survival (OS) when compared with BRAF V600E mutated [median OS: 62.0 versus 12.6 months; hazard ratio: 0.36 (95% confidence interval 0.20-0.64), P = 0.002], both at univariate and multivariate analyses. CONCLUSIONS BRAF codon 594 or 596 mutated mCRCs are different from BRAF V600E ones in terms of molecular features, pathological characteristics and clinical outcome. This is consistent with preclinical evidences of a kinase inactivating effect of these mutations. The role of CRAF in transducing the intracellular signal downstream BRAF 594 or 596 mutated proteins opens the way to further preclinical investigation.
Collapse
Affiliation(s)
- C Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa
| | - M Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - A Amatu
- S. C. Oncologia Falck, Niguarda Cancer Center, Ospedale Niguarda Ca' Granda, Milano
| | - C Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa
| | - R Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa
| | - R Berenato
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - F Perrone
- Department of Pathology and Molecular Biology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - E Tamborini
- Department of Pathology and Molecular Biology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - G Aprile
- Department of Medical Oncology, Azienda Ospedaliero-Universitaria, Udine
| | - S Lonardi
- UOC Oncologia Medica 1, Istituto Oncologico Veneto-IRCCS, Padova
| | - A Sartore-Bianchi
- S. C. Oncologia Falck, Niguarda Cancer Center, Ospedale Niguarda Ca' Granda, Milano
| | - G Fontanini
- Department of Surgery, Division of Pathology, University of Pisa, Pisa
| | - M Milione
- Department of Pathology and Molecular Biology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | | | - S Siena
- S. C. Oncologia Falck, Niguarda Cancer Center, Ospedale Niguarda Ca' Granda, Milano University of Milan, Milan, Italy
| | - A Falcone
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa
| | - F de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - F Loupakis
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa
| | - F Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| |
Collapse
|
136
|
Sorbye H, Dragomir A, Sundström M, Pfeiffer P, Thunberg U, Bergfors M, Aasebø K, Eide GE, Ponten F, Qvortrup C, Glimelius B. High BRAF Mutation Frequency and Marked Survival Differences in Subgroups According to KRAS/BRAF Mutation Status and Tumor Tissue Availability in a Prospective Population-Based Metastatic Colorectal Cancer Cohort. PLoS One 2015; 10:e0131046. [PMID: 26121270 PMCID: PMC4484806 DOI: 10.1371/journal.pone.0131046] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/28/2015] [Indexed: 01/23/2023] Open
Abstract
RAS and BRAF mutations impact treatment and prognosis of metastatic colorectal cancer patients (mCRC), but the knowledge is based on trial patients usually not representative for the general cancer population. Patient characteristics, treatment and efficacy according to KRAS, BRAF and MSI status were analyzed in a prospectively collected unselected population-based cohort of 798 non-resectable mCRC patients. The cohort contained many patients with poor performance status (39% PS 2-4) and elderly (37% age>75), groups usually not included in clinical trials. Patients without available tissue micro array (TMA) (42%) had worse prognostic factors and inferior survival (all patients; 7m vs 11m, chemotherapy-treated;12m vs 17m). The 92 patients (21%) with BRAF mutation had a poor prognosis regardless of microsatellite instability, but receipt of 1-2nd chemotherapy was similar to wildtype BRAF patients. Median survival in this cohort varied from 1 month in BRAF mutated patients not given chemotherapy to 26 months in wildtype KRAS/BRAF patients <75 years in good PS. TMA availability, BRAF mutation and KRAS mutation were all independent prognostic factors for survival. The observed 21% BRAF mutation incidence is higher than the previously and repeatedly reported incidence of 5-12% in mCRC. Screening for BRAF mutations before selection of treatment is relevant for many patients, especially outside clinical trials. A BRAF mutation only partly explained the very poor prognosis of many mCRC patients. Survival in unselected metastatic colorectal cancer patients is extremely variable and subgroups have an extremely short survival compared to trial patients. Patients without available TMA had worse prognostic factors and shorter survival, which questions the total generalizability of present TMA studies and implies that we lack information on the biologically worst mCRC cases. Lack of available tissue is an important underexposed issue which introduces sample bias, and this should be recognized more clearly when conclusions are made from translational mCRC studies.
Collapse
Affiliation(s)
- Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| | - Anca Dragomir
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Magnus Sundström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Ulf Thunberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Monica Bergfors
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Kristine Aasebø
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Geir Egil Eide
- Department of Global Public Health and Primary Care, Lifestyle Epidemiology Group, University of Bergen, Bergen, Norway
| | - Fredrik Ponten
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Camilla Qvortrup
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
137
|
Wang F, Bai L, Liu TS, Yu YY, He MM, Liu KY, Luo HY, Zhang DS, Jin Y, Wang FH, Wang ZQ, Wang DS, Qiu MZ, Ren C, Li YH, Xu RH. Right-sided colon cancer and left-sided colorectal cancers respond differently to cetuximab. CHINESE JOURNAL OF CANCER 2015; 34:384-93. [PMID: 26111811 PMCID: PMC4593341 DOI: 10.1186/s40880-015-0022-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 04/08/2015] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Right-sided colon cancer (RSCC) and left-sided colorectal cancer (LSCRC) differ with respect to their biology and genomic patterns. This study aimed to examine whether the primary tumor location is associated with the response to cetuximab in patients with metastatic colorectal cancer (mCRC). METHODS Patients with mCRC treated with cetuximab and standard chemotherapy as first- or second-line treatments were compared with randomly chosen patients who were treated with chemotherapy alone between 2005 and 2013. The main outcome measures were the overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). The differences in the outcome were analyzed by using the chi-squared test, Student's t test, and Kaplan-Meier method. RESULTS The treatment results of 206 patients with mCRC treated with cetuximab and standard chemotherapy as first- or second-line treatments were compared with those of 210 patients who were treated with chemotherapy alone. As a first-line treatment, cetuximab with chemotherapy was associated with a significantly higher ORR (49.4 % vs. 28.6 %, P = 0.005) as well as longer PFS (9.1 vs. 6.2 months, P = 0.002) and OS (28.9 vs. 20.1 months, P = 0.036) than chemotherapy alone in patients with LSCRC. However, cetuximab neither improved the ORR (36.4 % vs. 26.2 %, P = 0.349) nor prolonged PFS (5.6 vs. 5.7 months, P = 0.904) or OS (25.1 vs. 19.8 months, P = 0.553) in patients with RSCC. As a second-line treatment, cetuximab exhibited a tendency to improve the ORR (23.5 % vs. 10.2 %, P = 0.087) and prolong PFS (4.9 vs. 3.5 months, P = 0.064), and it significantly prolonged OS (17.1 vs. 12.4 months, P = 0.047) compared with chemotherapy alone in the patients with LSCRC. In contrast, as a second-line treatment, cetuximab neither improved the ORR (7.1 % vs. 11.4 %, P = 0.698) nor prolonged PFS (3.3 vs. 4.2 months, P = 0.761) or OS (13.4 vs. 13.0 months, P = 0.652) in patients with RSCC. CONCLUSIONS The addition of cetuximab to chemotherapy in both first- and second-line treatments of mCRC may only benefit patients with primary LSCRC.
Collapse
Affiliation(s)
- Feng 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, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Long Bai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| | - Yi-Yi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| | - Ming-Ming He
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Kai-Yan Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Hui-Yan Luo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Dong-Sheng Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Yin Jin
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. 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, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Zhi-Qiang 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, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - De-Shen 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, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. 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, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Yu-Hong Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center: State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. 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, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| |
Collapse
|
138
|
Scartozzi M, Giampieri R, Aprile G, Iacono D, Santini D, dell’Aquila E, Silvestris N, Gnoni A, Bonotto M, Puzzoni M, Demurtas L, Cascinu S. The distinctive molecular, pathological and clinical characteristics ofBRAF-mutant colorectal tumors. Expert Rev Mol Diagn 2015; 15:979-87. [DOI: 10.1586/14737159.2015.1047346] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
139
|
Ye JX, Liu Y, Qin Y, Zhong HH, Yi WN, Shi XY. KRAS and BRAF gene mutations and DNA mismatch repair status in Chinese colorectal carcinoma patients. World J Gastroenterol 2015; 21:1595-1605. [PMID: 25663779 PMCID: PMC4316102 DOI: 10.3748/wjg.v21.i5.1595] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/28/2014] [Accepted: 12/08/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate gene mutations and DNA mismatch repair (MMR) protein abnormality in Chinese colorectal carcinoma (CRC) patients and their correlations with clinicopathologic features.
METHODS: Clinical and pathological information for 535 patients including 538 tumors was reviewed and recorded. Mutation analyses for exon 2 of KRAS gene and exon 15 of BRAF gene were performed by Sanger sequencing except that in 9 tumors amplification refractory mutation system PCR was used. Expression of MMR proteins including MHL1, MSH2, MSH6 and PMS2 was evaluated by immunohistochemistry. Correlations of KRAS and BRAF mutation status and the expression status of MMR proteins with age, gender, cancer stage, location, and histology were analyzed. Correlations between KRAS or BRAF mutations and MMR protein expression were also explored.
RESULTS: The overall frequencies of KRAS and BRAF mutations were 37.9% and 4.4%, respectively. KRAS mutations were more common in patients ≥ 50 years old (39.8% vs 22% in patients < 50 years old, P < 0.05). The frequencies of BRAF mutants were higher in tumors from females (6.6% vs males 2.8%, P < 0.05), located in the right colon (9.6% vs 2.1% in the left colon, 1.8% in the rectum, P < 0.01), with mucinous differentiation (9.8% vs 2.8% without mucinous differentiation, P < 0.01), or being poorly differentiated (9.5% vs 3.4% well/moderately differentiated, P < 0.05). MMR deficiency was strongly associated with proximal location (20.5% in the right colon vs 9.2% in the left colon and 5.1% in the rectum, P < 0.001), early cancer stage (15.0% in stages I-II vs 7.7% in stages III-IV, P < 0.05), and mucinous differentiation (20.2% vs 9.2% without mucin, P < 0.01). A higher frequency of MLH1/PMS2 loss was found in females (9.2% vs 4.4% in males, P < 0.05), and MSH2/MSH6 loss tended to be seen in younger (<50 years old) patients (12.0% vs 4.0% ≥ 50 years old, P < 0.05). MMR deficient tumors were less likely to have KRAS mutations (18.8% vs 41.7% in MMR proficient tumors, P < 0.05) and tumors with abnormal MLH1/PMS2 tended to harbor BRAF mutations (15.4% vs 4.2% in MMR proficient tumors, P < 0.05).
CONCLUSION: The frequency of sporadic CRCs having BRAF mutation, MLH1 deficiency and MSI in Chinese population may be lower than that in the Western population.
Collapse
|
140
|
Herr R, Köhler M, Andrlová H, Weinberg F, Möller Y, Halbach S, Lutz L, Mastroianni J, Klose M, Bittermann N, Kowar S, Zeiser R, Olayioye MA, Lassmann S, Busch H, Boerries M, Brummer T. B-Raf inhibitors induce epithelial differentiation in BRAF-mutant colorectal cancer cells. Cancer Res 2014; 75:216-29. [PMID: 25381152 DOI: 10.1158/0008-5472.can-13-3686] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BRAF mutations are associated with aggressive, less-differentiated and therapy-resistant colorectal carcinoma. However, the underlying mechanisms for these correlations remain unknown. To understand how oncogenic B-Raf contributes to carcinogenesis, in particular to aspects other than cellular proliferation and survival, we generated three isogenic human colorectal carcinoma cell line models in which we can dynamically modulate the expression of the B-Raf(V600E) oncoprotein. Doxycyclin-inducible knockdown of endogenous B-Raf(V600E) decreases cellular motility and invasion in conventional and three-dimensional (3D) culture, whereas it promotes cell-cell contacts and induces various hallmarks of differentiated epithelia. Importantly, all these effects are recapitulated by B-Raf (PLX4720, vemurafenib, and dabrafenib) or MEK inhibitors (trametinib). Surprisingly, loss of B-Raf(V600E) in HT29 xenografts does not only stall tumor growth, but also induces glandular structures with marked expression of CDX2, a tumor-suppressor and master transcription factor of intestinal differentiation. By performing the first transcriptome profiles of PLX4720-treated 3D cultures of HT29 and Colo-205 cells, we identify several upregulated genes linked to epithelial differentiation and effector functions, such as claudin-1, a Cdx-2 target gene encoding a critical tight junction component. Thereby, we provide a mechanism for the clinically observed correlation between mutant BRAF and the loss of Cdx-2 and claudin-1. PLX4720 also suppressed several metastasis-associated transcripts that have not been implicated as targets, effectors or potential biomarkers of oncogenic B-Raf signaling so far. Together, we identify a novel facet of clinically applied B-Raf or MEK inhibitors by showing that they promote cellular adhesion and differentiation of colorectal carcinoma cells.
Collapse
Affiliation(s)
- Ricarda Herr
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany
| | - Martin Köhler
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany. Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Hana Andrlová
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany
| | - Florian Weinberg
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany
| | - Yvonne Möller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sebastian Halbach
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany. Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Lisa Lutz
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany
| | - Justin Mastroianni
- Faculty of Biology, ALU, Freiburg, Germany. Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany
| | - Martin Klose
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany
| | - Nicola Bittermann
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany
| | - Silke Kowar
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Silke Lassmann
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hauke Busch
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Boerries
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tilman Brummer
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg.
| |
Collapse
|
141
|
Venderbosch S, Nagtegaal ID, Maughan TS, Smith CG, Cheadle JP, Fisher D, Kaplan R, Quirke P, Seymour MT, Richman SD, Meijer GA, Ylstra B, Heideman DAM, de Haan AFJ, Punt CJA, Koopman M. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res 2014. [PMID: 25139339 DOI: 10.1158/1078?0432.ccr?14?0332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE To determine the prevalence and prognostic value of mismatch repair (MMR) status and its relation to BRAF mutation (BRAF(MT)) status in metastatic colorectal cancer (mCRC). EXPERIMENTAL DESIGN A pooled analysis of four phase III studies in first-line treatment of mCRC (CAIRO, CAIRO2, COIN, and FOCUS) was performed. Primary outcome parameter was the hazard ratio (HR) for median progression-free survival (PFS) and overall survival (OS) in relation to MMR and BRAF. For the pooled analysis, Cox regression analysis was performed on individual patient data. RESULTS The primary tumors of 3,063 patients were analyzed, of which 153 (5.0%) exhibited deficient MMR (dMMR) and 250 (8.2%) a BRAF(MT). BRAF(MT) was observed in 53 (34.6%) of patients with dMMR tumors compared with 197 (6.8%) of patients with proficient MMR (pMMR) tumors (P < 0.001). In the pooled dataset, median PFS and OS were significantly worse for patients with dMMR compared with pMMR tumors [HR, 1.33; 95% confidence interval (CI), 1.12-1.57 and HR, 1.35; 95% CI, 1.13-1.61, respectively), and for patients with BRAF(MT) compared with BRAF wild-type (BRAF(WT)) tumors (HR, 1.34; 95% CI, 1.17-1.54 and HR, 1.91; 95% CI, 1.66-2.19, respectively). PFS and OS were significantly decreased for patients with BRAF(MT) within the group of patients with pMMR, but not for BRAF status within dMMR, or MMR status within BRAF(WT) or BRAF(MT). CONCLUSIONS Prevalence of dMMR and BRAF(MT) in patients with mCRC is low and both biomarkers confer an inferior prognosis. Our data suggest that the poor prognosis of dMMR is driven by the BRAF(MT) status.
Collapse
Affiliation(s)
- Sabine Venderbosch
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands. Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tim S Maughan
- Department of Oncology, Gray Institute for Radiation Oncology, Biology University of Oxford, Oxford, United Kingdom
| | - Christopher G Smith
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jeremy P Cheadle
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - David Fisher
- MRC Clinical Trials Unit, Aviation House, London, United Kingdom
| | - Richard Kaplan
- MRC Clinical Trials Unit, Aviation House, London, United Kingdom
| | - Philip Quirke
- Department of Pathology, Anatomy and Tumor Biology, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
| | - Matthew T Seymour
- Department of Medical Oncology, Leeds Institute of Cancer and Pathology, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Susan D Richman
- Department of Pathology, Anatomy and Tumor Biology, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
| | - Gerrit A Meijer
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | | | - Anton F J de Haan
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
142
|
Venderbosch S, Nagtegaal ID, Maughan TS, Smith CG, Cheadle JP, Fisher D, Kaplan R, Quirke P, Seymour MT, Richman SD, Meijer GA, Ylstra B, Heideman DAM, de Haan AFJ, Punt CJA, Koopman M. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res 2014; 20:5322-30. [PMID: 25139339 DOI: 10.1158/1078-0432.ccr-14-0332] [Citation(s) in RCA: 542] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE To determine the prevalence and prognostic value of mismatch repair (MMR) status and its relation to BRAF mutation (BRAF(MT)) status in metastatic colorectal cancer (mCRC). EXPERIMENTAL DESIGN A pooled analysis of four phase III studies in first-line treatment of mCRC (CAIRO, CAIRO2, COIN, and FOCUS) was performed. Primary outcome parameter was the hazard ratio (HR) for median progression-free survival (PFS) and overall survival (OS) in relation to MMR and BRAF. For the pooled analysis, Cox regression analysis was performed on individual patient data. RESULTS The primary tumors of 3,063 patients were analyzed, of which 153 (5.0%) exhibited deficient MMR (dMMR) and 250 (8.2%) a BRAF(MT). BRAF(MT) was observed in 53 (34.6%) of patients with dMMR tumors compared with 197 (6.8%) of patients with proficient MMR (pMMR) tumors (P < 0.001). In the pooled dataset, median PFS and OS were significantly worse for patients with dMMR compared with pMMR tumors [HR, 1.33; 95% confidence interval (CI), 1.12-1.57 and HR, 1.35; 95% CI, 1.13-1.61, respectively), and for patients with BRAF(MT) compared with BRAF wild-type (BRAF(WT)) tumors (HR, 1.34; 95% CI, 1.17-1.54 and HR, 1.91; 95% CI, 1.66-2.19, respectively). PFS and OS were significantly decreased for patients with BRAF(MT) within the group of patients with pMMR, but not for BRAF status within dMMR, or MMR status within BRAF(WT) or BRAF(MT). CONCLUSIONS Prevalence of dMMR and BRAF(MT) in patients with mCRC is low and both biomarkers confer an inferior prognosis. Our data suggest that the poor prognosis of dMMR is driven by the BRAF(MT) status.
Collapse
Affiliation(s)
- Sabine Venderbosch
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands. Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tim S Maughan
- Department of Oncology, Gray Institute for Radiation Oncology, Biology University of Oxford, Oxford, United Kingdom
| | - Christopher G Smith
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jeremy P Cheadle
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - David Fisher
- MRC Clinical Trials Unit, Aviation House, London, United Kingdom
| | - Richard Kaplan
- MRC Clinical Trials Unit, Aviation House, London, United Kingdom
| | - Philip Quirke
- Department of Pathology, Anatomy and Tumor Biology, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
| | - Matthew T Seymour
- Department of Medical Oncology, Leeds Institute of Cancer and Pathology, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Susan D Richman
- Department of Pathology, Anatomy and Tumor Biology, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
| | - Gerrit A Meijer
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | | | - Anton F J de Haan
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
143
|
Tie J, Desai J. Targeting BRAF mutant metastatic colorectal cancer: clinical implications and emerging therapeutic strategies. Target Oncol 2014; 10:179-88. [PMID: 25119972 DOI: 10.1007/s11523-014-0330-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 07/24/2014] [Indexed: 12/16/2022]
Abstract
Increasing knowledge of the underlying signaling pathways and molecular defects involved in colorectal cancer growth or progression enabled the discovery of several prognostic and predictive biomarkers, leading to the development of novel molecularly targeted therapies. The mitogen-activated protein kinase (MAPK) signaling pathway plays a critical role in colorectal cancer progression. Mutations in BRAF, a principal effector of Ras in this signaling cascade, are found in 10 % of colorectal cancer and play a clear pathogenic role, particularly in patients with metastatic disease. Intense efforts have therefore focused on targeting BRAF as an oncogenic driver, with mixed early results. This article summarizes the molecular and clinical features of BRAF mutant colorectal cancer, the prognostic and predictive role of BRAFV600E mutation in colorectal cancer, initial clinical trial results in targeting BRAFV600E, and the more recent preclinical insights into potential mechanisms of resistance to BRAF inhibition that have now led to a number of rationale-driven combination therapeutic strategies.
Collapse
Affiliation(s)
- Jeanne Tie
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC, Australia
| | | |
Collapse
|
144
|
BRAF mutations in patients with non-small cell lung cancer: a systematic review and meta-analysis. PLoS One 2014; 9:e101354. [PMID: 24979348 PMCID: PMC4076330 DOI: 10.1371/journal.pone.0101354] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 06/06/2014] [Indexed: 12/23/2022] Open
Abstract
Background BRAF mutations have been well described in non-small cell lung cancer (NSCLC) for several years, but the clinical features of patients harboring BRAF mutations are still not well described. We performed a meta-analysis to identify common clinical features in NSCLC patients carrying BRAF mutations. Methods We identified clinical studies that examined the association between BRAF mutations and features of NSCLC within PubMed, Embase and ISI Science Citation Index database up to October 2013. The effect size of clinical features was estimated by odds ratios (ORs) with 95% confidence interval (CI) for each study, using a fixed-effects or random-effects model. Results Ten studies with a total of 5599 NSCLC patients were included. There was a 3% (170/5599) BRAF mutation rate. BRAF mutations in NSCLC were significantly associated with adenocarcinomas (ADCs) (compared with non-ADCs, OR = 4.96, 95%CI = 2.29–10.75). There were no significant differences in gender, smoking and stage in patients with and without BRAF mutations. The BRAFV600E mutation was more frequent in women than non-BRAFV600E mutations (OR = 0.27, 95%CI = 0.12–0.59), and was closely related to never smokers (OR = 0.14, 95%CI = 0.05–0.42). Conclusions These findings have important implications for the prediction of the NSCLC sub-types more accurately combined with other genetic changes.
Collapse
|
145
|
Morris V, Overman MJ, Jiang ZQ, Garrett C, Agarwal S, Eng C, Kee B, Fogelman D, Dasari A, Wolff R, Maru D, Kopetz S. Progression-free survival remains poor over sequential lines of systemic therapy in patients with BRAF-mutated colorectal cancer. Clin Colorectal Cancer 2014; 13:164-71. [PMID: 25069797 DOI: 10.1016/j.clcc.2014.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND BRAF mutations occur in 5% to 10% of metastatic colorectal cancers and are biomarkers associated with a poor prognosis. However, the outcomes with standard chemotherapy over sequential lines of therapy in a large cohort of patients with BRAF-mutant tumors have not been described. PATIENTS AND METHODS We searched the M.D. Anderson Cancer Center databases for patients with colorectal cancer and identified BRAF mutations between December 2003 and May 2012. Patients were analyzed for clinical characteristics, PFS, overall survival, and chemotherapeutic agents used. Survival was estimated according to the Kaplan-Meier method. RESULTS Among the 1567 patients tested for BRAF mutations at our institution, 127 (8.1%) had tumors with BRAF mutations. The 71 patients who presented with metastatic disease received a median of 2 lines of chemotherapy. For the first 3 lines of chemotherapy, median PFS was 6.3 months (n = 69 patients; 95% confidence interval [CI], 4.9-7.7 months), 2.5 months (n = 58 patients; 95% CI, 1.8-3.0 months), and 2.6 months (n = 31 patients; 95% CI, 1.0-4.2 months), respectively. Median PFS was not affected by the backbone chemotherapeutic agent in the first-line setting, whether oxaliplatin-based or irinotecan-based (6.4 months vs. 5.4 months, respectively; P = .99). CONCLUSION PFS is expectedly poor for patients with BRAF-mutated metastatic colorectal cancer. Despite the ascertainment bias present (with testing preferentially performed in patients suitable for clinical trials in refractory disease), these data provide historic controls suitable for future study design and support the idea that novel therapeutic options are essential in this population.
Collapse
Affiliation(s)
- Van Morris
- Department of Cancer Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Zhi-Qin Jiang
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Christopher Garrett
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Shweta Agarwal
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Cathy Eng
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - David Fogelman
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Robert Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Dipen Maru
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX.
| |
Collapse
|
146
|
Schirripa M, Cremolini C, Loupakis F, Morvillo M, Bergamo F, Zoratto F, Salvatore L, Antoniotti C, Marmorino F, Sensi E, Lupi C, Fontanini G, De Gregorio V, Giannini R, Basolo F, Masi G, Falcone A. Role of NRAS mutations as prognostic and predictive markers in metastatic colorectal cancer. Int J Cancer 2014; 136:83-90. [PMID: 24806288 DOI: 10.1002/ijc.28955] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/25/2014] [Indexed: 02/06/2023]
Abstract
NRAS mutations occur in 3-5% of colorectal cancer. Differently from KRAS and BRAF mutations, the role of NRAS mutations as prognostic and predictive markers in metastatic colorectal cancer (mCRC) has been investigated to a lesser extent. A retrospective series suggested the role of NRAS mutations as predictors of resistance to anti-EGFR monoclonal antibodies (MoAbs) in chemo-refractory patients with mCRC. In our study, KRAS codons 12, 13, 61 and BRAF codon 600 mutational status were evaluated in mCRCs referred to our Institution from 2009 to 2012. NRAS codons 12, 13 and 61 mutational status was analyzed in KRAS/BRAF wt patients. We collected pathological and clinical features in the overall population and outcome data in a subset of NRAS mutated chemo-refractory patients treated with anti-EGFR MoAbs in advanced lines. NRAS was mutated in 47/786 (6%) mCRCs. NRAS and KRAS mutated tumors did not show significant differences in terms of clinical and pathological characteristics, except for a lower prevalence of mucinous histology (p = 0.012) and lung metastases (p = 0.012) among NRAS mutated tumors. In the uni- and multivariate model, NRAS mutations were associated with shorter overall survival (OS) compared to all wt patients (median OS 25.6 vs 42.7 months; univ: HR = 1.91, 95% CI 1.39-3.86, p = 0.0013; multiv: HR = 1.75, 95% CI 1.1.3-2.72, p = 0.013). None of the chemo-refractory NRAS mutated patients evaluable for response to anti-EGFRs achieved response. In conclusion, NRAS mutations have a relevant incidence in patients with mCRC and showed an association with specific clinical and pathological features. NRAS mutations affect mCRC patients' prognosis and predict lack of response to anti-EGFRs.
Collapse
Affiliation(s)
- Marta Schirripa
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Day F, Muranyi A, Singh S, Shanmugam K, Williams D, Byrne D, Pham K, Palmieri M, Tie J, Grogan T, Gibbs P, Sieber O, Waring P, Desai J. A mutant BRAF V600E-specific immunohistochemical assay: correlation with molecular mutation status and clinical outcome in colorectal cancer. Target Oncol 2014; 10:99-109. [PMID: 24859797 PMCID: PMC4363480 DOI: 10.1007/s11523-014-0319-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 05/09/2014] [Indexed: 12/12/2022]
Abstract
The B-type Raf kinase (BRAF) V600E mutation is a well-established biomarker for poor prognosis in metastatic colorectal cancer (mCRC) and is a highly attractive drug target. A barrier to the development of new therapies targeting BRAF V600E in mCRC is the low prevalence of mutations (approximately 10 %) and the current need for access to sequencing-based technologies which are not routinely available outside of large cancer centres. Availability of a standardised immunohistochemistry (IHC) test, more suited to routine pathology practice, would provide much broader access to patient identification. We sought to evaluate the accuracy and clinical utility of a recently developed BRAF V600E IHC method as a prognostic biomarker in a large cohort of community-based CRC patients. Archival tumour samples from 505 patients with stage I–IV CRC were immunohistochemically tested with two antibodies, pBR1 for total BRAF and VE1 for BRAF V600E. Cases were assessed by two blinded pathologists, and results were compared to BRAF V600E mutation status determined using DNA sequencing. Discordant cases were retested with a BRAF V600E SNaPshot assay. BRAF mutation status was correlated with overall survival (OS) in stage IV CRC. By DNA sequencing and IHC, 505 and 477 patients were respectively evaluable. Out of 477 patients, 56 (11. 7 %) had BRAF V600E mutations detected by sequencing and 63 (13.2 %) by IHC. Using DNA sequencing results as the reference, sensitivity and specificity for IHC were 98.2 % (55/56) and 98.1 % (413/421), respectively. IHC had a positive predictive value (PPV) of 87.3 % (55/63) and a negative predictive value (NPV) of 99.8 % (413/414). Compared to DNA sequencing plus retesting of available discordant cases by SNaPshot assay, IHC using the VE1 antibody had a 100 % sensitivity (59/59), specificity (416/416), NPV (416/416) and PPV (59/59). Stage IV CRC patients with BRAF V600E protein detected by IHC exhibited a significantly shorter overall survival (hazard ratio = 2.20, 95 % CI 1.26–3.83, p = 0.005), consistent with other published series. Immunohistochemistry using the BRAF V600E VE1 antibody is an accurate diagnostic assay in CRC. The test provides a simple, clinically applicable method of testing for the BRAF V600E mutation in routine practice.
Collapse
Affiliation(s)
- Fiona Day
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
| | | | | | | | - David Williams
- Department of Pathology, Austin Health, Heidelberg, VIC Australia
| | - David Byrne
- Pathology Department, Peter MacCallum Cancer Centre, East Melbourne, VIC Australia
| | - Kym Pham
- Faculty of Medicine, Dentistry and Health Sciences, Department of Pathology, University of Melbourne, Parkville, VIC Australia
| | - Michelle Palmieri
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
| | - Jeanne Tie
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
- Department of Medical Oncology, Royal Melbourne Hospital, Parkville, VIC Australia
- Department of Medical Oncology, Western Hospital, Footscray, VIC Australia
| | | | - Peter Gibbs
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
- Department of Medical Oncology, Royal Melbourne Hospital, Parkville, VIC Australia
- Department of Medical Oncology, Western Hospital, Footscray, VIC Australia
| | - Oliver Sieber
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
| | - Paul Waring
- Faculty of Medicine, Dentistry and Health Sciences, Department of Pathology, University of Melbourne, Parkville, VIC Australia
| | - Jayesh Desai
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
- Department of Medical Oncology, Royal Melbourne Hospital, Parkville, VIC Australia
- Department of Medical Oncology, Western Hospital, Footscray, VIC Australia
| |
Collapse
|
148
|
Kim JH, Kang GH. Molecular and prognostic heterogeneity of microsatellite-unstable colorectal cancer. World J Gastroenterol 2014; 20:4230-4243. [PMID: 24764661 PMCID: PMC3989959 DOI: 10.3748/wjg.v20.i15.4230] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/30/2014] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancers (CRCs) with a high level of microsatellite instability (MSI-H) are clinicopathologically distinct tumors characterized by predominance in females, proximal colonic localization, poor differentiation, mucinous histology, tumor-infiltrating lymphocytes, a Crohn’s-like lymphoid reaction and a favorable prognosis. In terms of their molecular features, MSI-H CRCs are heterogeneous tumors associated with various genetic and epigenetic alterations, including DNA mismatch repair deficiency, target microsatellite mutations, BRAF mutations, a CpG island methylator phenotype-high (CIMP-H) status, and a low level of genomic hypomethylation. The molecular heterogeneity of MSI-H CRCs also depends on ethnic differences; for example, in Eastern Asian countries, relatively low frequencies of CIMP-H and BRAF mutations have been observed in MSI-H CRCs compared to Western countries. Although the prognostic features of MSI-H CRCs include a favorable survival of patients and low benefit of adjuvant chemotherapy, there may be prognostic differences based on the molecular heterogeneity of MSI-H CRCs. Here, we have reviewed and discussed the molecular and prognostic features of MSI-H CRCs, as well as several putative prognostic or predictive molecular markers, including HSP110 expression, beta2-microglobulin mutations, myosin 1a expression, CDX2/CK20 expression, SMAD4 expression, CIMP status and LINE-1 methylation levels.
Collapse
|
149
|
Chen D, Huang JF, Liu K, Zhang LQ, Yang Z, Chuai ZR, Wang YX, Shi DC, Huang Q, Fu WL. BRAFV600E mutation and its association with clinicopathological features of colorectal cancer: a systematic review and meta-analysis. PLoS One 2014; 9:e90607. [PMID: 24594804 PMCID: PMC3940924 DOI: 10.1371/journal.pone.0090607] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/03/2014] [Indexed: 01/02/2023] Open
Abstract
Background Colorectal cancer (CRC) is a heterogeneous disease with multiple underlying causative genetic mutations. The B-type Raf proto-oncogene (BRAF) plays an important role in the mitogen-activated protein kinase (MAPK) signaling cascade during CRC. The presence of BRAFV600E mutation can determine the response of a tumor to chemotherapy. However, the association between the BRAFV600E mutation and the clinicopathological features of CRC remains controversial. We performed a systematic review and meta-analysis to estimate the effect of BRAFV600E mutation on the clinicopathological characteristics of CRC. Methods We identified studies that examined the effect of BRAFV600E mutation on CRC within the PubMed, ISI Science Citation Index, and Embase databases. The effect of BRAFV600E on outcome parameters was estimated by odds ratios (ORs) with 95% confidence intervals (CIs) for each study using a fixed effects or random effects model. Results 25 studies with a total of 11,955 CRC patients met inclusion criteria. The rate of BRAFV600 was 10.8% (1288/11955). The BRAFV600E mutation in CRC was associated with advanced TNM stage, poor differentiation, mucinous histology, microsatellite instability (MSI), CpG island methylator phenotype (CIMP). This mutation was also associated with female gender, older age, proximal colon, and mutL homolog 1 (MLH1) methylation. Conclusions This meta-analysis demonstrated that BRAFV600E mutation was significantly correlated with adverse pathological features of CRC and distinct clinical characteristics. These data suggest that BRAFV600E mutation could be used to supplement standard clinical and pathological staging for the better management of individual CRC patients, and could be considered as a poor prognostic marker for CRC.
Collapse
Affiliation(s)
- Dong Chen
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Jun-Fu Huang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Kai Liu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Li-Qun Zhang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Zhao Yang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Zheng-Ran Chuai
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Yun-Xia Wang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Da-Chuan Shi
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Qing Huang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
- * E-mail: (FW); (HQ)
| | - Wei-Ling Fu
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, PR China
- * E-mail: (FW); (HQ)
| |
Collapse
|
150
|
Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, Lugli A, Zlobec I, Hartmann A, Bifulco C, Nagtegaal ID, Palmqvist R, Masucci GV, Botti G, Tatangelo F, Delrio P, Maio M, Laghi L, Grizzi F, Asslaber M, D'Arrigo C, Vidal-Vanaclocha F, Zavadova E, Chouchane L, Ohashi PS, Hafezi-Bakhtiari S, Wouters BG, Roehrl M, Nguyen L, Kawakami Y, Hazama S, Okuno K, Ogino S, Gibbs P, Waring P, Sato N, Torigoe T, Itoh K, Patel PS, Shukla SN, Wang Y, Kopetz S, Sinicrope FA, Scripcariu V, Ascierto PA, Marincola FM, Fox BA, Pagès F. Towards the introduction of the 'Immunoscore' in the classification of malignant tumours. J Pathol 2014; 232:199-209. [PMID: 24122236 PMCID: PMC4255306 DOI: 10.1002/path.4287] [Citation(s) in RCA: 1023] [Impact Index Per Article: 102.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 02/06/2023]
Abstract
The American Joint Committee on Cancer/Union Internationale Contre le Cancer (AJCC/UICC) TNM staging system provides the most reliable guidelines for the routine prognostication and treatment of colorectal carcinoma. This traditional tumour staging summarizes data on tumour burden (T), the presence of cancer cells in draining and regional lymph nodes (N) and evidence for distant metastases (M). However, it is now recognized that the clinical outcome can vary significantly among patients within the same stage. The current classification provides limited prognostic information and does not predict response to therapy. Multiple ways to classify cancer and to distinguish different subtypes of colorectal cancer have been proposed, including morphology, cell origin, molecular pathways, mutation status and gene expression-based stratification. These parameters rely on tumour-cell characteristics. Extensive literature has investigated the host immune response against cancer and demonstrated the prognostic impact of the in situ immune cell infiltrate in tumours. A methodology named ‘Immunoscore’ has been defined to quantify the in situ immune infiltrate. In colorectal cancer, the Immunoscore may add to the significance of the current AJCC/UICC TNM classification, since it has been demonstrated to be a prognostic factor superior to the AJCC/UICC TNM classification. An international consortium has been initiated to validate and promote the Immunoscore in routine clinical settings. The results of this international consortium may result in the implementation of the Immunoscore as a new component for the classification of cancer, designated TNM-I (TNM-Immune). © 2013 The Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jérôme Galon
- INSERM, U872, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie Paris 6, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|