1
|
An Y, Chen W, Cao Y, Chen B, Li Q, Zhou X, Huang W. The mechanism of folic acid on N-methyl-N’-nitro-N-nitrosoguanidine-induced chronic atrophic gastritis through the PI3K/Akt pathway. INVESTIGACIÓN CLÍNICA 2024; 65:48-58. [DOI: 10.54817/ic.v65n1a05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Chronic atrophic gastritis (CAG) is a precancerous atrophic gastritis of the stomach, which generates an urge to develop novel therapeu-tic schedules. This study aimed to investigate the effect of experimental folic acid administration on N-methyl-N’-nitro-N-nitrosoguanidine (MNNG)-induced CAG through the PI3K/Akt pathway in rats. The rats were divided into a Model Group, a Folic Acid Group and a Blank Group. Rats in the Model Group were induced by MNNG and given 10 mL/kg/d distilled water by gavage, while rats in the Folic Acid Group were induced by MNNG and given 5 mg/kg/d folic acid suspension by gavage. As a control, rats in the Blank Group were given the same amount of distilled water as MNNG and 10 mL/kg/d distilled water by gavage. The levels of gastrin (GAS) and motilin (MTL) in serum were measured by enzyme-linked immunosorbent assay (ELISA), and the mRNA and protein ex-pressions were detected by quantitative polymerase chain reaction (q-PCR) and Western blot. According to hematoxylin and eosin (H&E) pathological analysis, there were inflammatory factors infiltration and derangement of mucosal epi-thelial cells in the model group, while the gastric tissue injury in the folic acid group was improved. Folic acid could decrease the content of GAS, increase the content of MTL in the serum of the rats, and regulate the expression of PI3K and AKT signal pathways. Folic acid can have a therapeutic effect on CAG by reducing the concentration of GAS in serum and increasing the concentration of MLT in serum. Our study would lay a theoretical foundation for using folic acid to investigate new therapies for CAG in humans.
Collapse
Affiliation(s)
- Yun An
- Department of Digestive Internal Medicine, Panyu District Traditional Chinese Medicine Hospital, Guangzhou,China
| | - Weigang Chen
- Department of Digestive Internal Medicine, Panyu District Traditional Chinese Medicine Hospital, Guangzhou,China
| | - Yong Cao
- Department of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Boshen Chen
- Department of Digestive Internal Medicine, Panyu District Traditional Chinese Medicine Hospital, Guangzhou,China
| | - Qiangbin Li
- Department of Digestive Internal Medicine, Panyu District Traditional Chinese Medicine Hospital, Guangzhou,China
| | - Xia Zhou
- Department of Digestive Internal Medicine, Panyu District Traditional Chinese Medicine Hospital, Guangzhou,China
| | - Weihan Huang
- Department of Digestive Internal Medicine, Panyu District Traditional Chinese Medicine Hospital, Guangzhou,China
| |
Collapse
|
2
|
Yang Z, Yuan H, He H, Qi S, Zhu X, Hu X, Jin M, Zhang XX, Yuan ZG. Unlocking the role of EIF5A: A potential diagnostic marker regulating the cell cycle and showing negative correlation with immune infiltration in lung adenocarcinoma. Int Immunopharmacol 2024; 126:111227. [PMID: 37977067 DOI: 10.1016/j.intimp.2023.111227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Despite EIF5A upregulation related to tumor progression in LUAD (lung adenocarcinoma), the underlying mechanisms remain elusive. In addition, there are few comprehensive analyses of EIF5A in LUAD. METHODS We investigated the EIF5A expression level in LUAD patients using data from the TCGA and GEO databases. We employed qRT-PCR and western blot to verify EIF5A expression in cell lines, while immunohistochemistry was utilized for clinical sample analysis. We analyzed EIF5A expression in tumor-infiltrating immune cells using the TISCH database and assessed its association with immune infiltration in LUAD using the "ESTIMATE" R package. Bioinformatics approaches were developed to discover the EIF5A-related genes and explore EIF5A potential mechanisms in LUAD. Proliferation ability was verified through CCK-8, clone formation, and EdU assays, while flow cytometry assessed apoptosis and cell cycle. Western blot was used to detect the expression of pathway-related proteins. RESULTS EIF5A was significantly upregulated in LUAD. Moreover, we constructed a MAZ-hsa-miR-424-3p-EIF5A transcriptional network. We explored the potential mechanism of EIF5A in LUAD and further investigated the cAMP signaling pathway and the cell cycle. Finally, we proved that EIF5A silencing induced G1/S Cell Cycle arrest, promoted apoptosis, and inhibited proliferation via the cAMP/PKA/CREB signaling pathway. CONCLUSION EIF5A serves as a prognostic biomarker with a negative correlation to immune infiltrates in LUAD. It regulated the cell cycle in LUAD by inhibiting the cAMP/PKA/CREB signaling pathway.
Collapse
Affiliation(s)
- Zipeng Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China
| | - Hao Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China
| | - Houjing He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China
| | - Shuting Qi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China
| | - Xiaojing Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China
| | - Xiaoyu Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China
| | - Mengyuan Jin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China
| | - Xiu-Xiang Zhang
- College of Agriculture, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China.
| | - Zi-Guo Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, Guangdong Province 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong Province 510642, PR China.
| |
Collapse
|
3
|
Sanaei MJ, Baghery Saghchy Khorasani A, Pourbagheri-Sigaroodi A, Shahrokh S, Zali MR, Bashash D. The PI3K/Akt/mTOR axis in colorectal cancer: Oncogenic alterations, non-coding RNAs, therapeutic opportunities, and the emerging role of nanoparticles. J Cell Physiol 2021; 237:1720-1752. [PMID: 34897682 DOI: 10.1002/jcp.30655] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/02/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is one of the deadliest human malignancies worldwide. Several molecular pathways have been demonstrated to be involved in the initiation and development of CRC which among them, the overactivation of the phosphatidyl-inositol 3-kinase (PI3K)/Akt/mTOR axis is of importance. The current review aims to unravel the mechanisms by which the PI3K/Akt/mTOR pathway affects CRC progression; and also, to summarize the original data obtained from international research laboratories on the oncogenic alterations and polymorphisms affecting this pathway in CRC. Besides, we provide a special focus on the regulatory role of noncoding RNAs targeting the PI3K/Akt/mTOR pathway in this malignancy. Questions on how this axis is involved in the inhibition of apoptosis, in the induction of drug resistance, and the angiogenesis, epithelial to mesenchymal transition, and metastasis are also responded. We also discussed the PI3K/Akt pathway-associated prognostic and predictive biomarkers in CRC. In addition, we provide a general overview of PI3K/Akt/mTOR pathway inhibition whether by chemical-based drugs or by natural-based medications in the context of CRC, either as monotherapy or in combination with other therapeutic agents; however, those treatments might have life-threatening side effects and toxicities. To the best of our knowledge, the current review is one of the first ones highlighting the emerging roles of nanotechnology to overcome challenges related to CRC therapy in the hope that providing a promising platform for the treatment of CRC.
Collapse
Affiliation(s)
- Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Ugai T, Zhao M, Shimizu T, Akimoto N, Shi S, Takashima Y, Zhong R, Lau MC, Haruki K, Arima K, Fujiyoshi K, Langworthy B, Masugi Y, da Silva A, Nosho K, Baba Y, Song M, Chan AT, Wang M, Meyerhardt JA, Giannakis M, Väyrynen JP, Nowak JA, Ogino S. Association of PIK3CA mutation and PTEN loss with expression of CD274 (PD-L1) in colorectal carcinoma. Oncoimmunology 2021; 10:1956173. [PMID: 34377593 PMCID: PMC8331006 DOI: 10.1080/2162402x.2021.1956173] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy targeting the CD274 (PD-L1)/PDCD1 (PD-1) immune checkpoint axis has emerged as a promising treatment strategy for various cancers. Experimental evidence suggests that phosphatidylinositol-4,5-bisphosphonate 3-kinase (PI3K) signaling may upregulate CD274 expression. Thus, we hypothesized that PIK3CA mutation, PTEN loss, or their combined status might be associated with CD274 overexpression in colorectal carcinoma. We assessed tumor CD274 and PTEN expression by immunohistochemistry and assessed PIK3CA mutation by pyrosequencing in 753 patients among 4,465 incident rectal and colon cancer cases that had occurred in two U.S.-wide prospective cohort studies. To adjust for potential confounders and selection bias due to tissue availability, inverse probability weighted multivariable ordinal logistic regression analyses used the 4,465 cases and tumoral data including microsatellite instability, CpG island methylator phenotype, KRAS and BRAF mutations. PIK3CA mutation and loss of PTEN expression were detected in 111 of 753 cases (15%) and 342 of 585 cases (58%), respectively. Tumor CD274 expression was negative in 306 (41%), low in 195 (26%), and high in 252 (33%) of 753 cases. PTEN loss was associated with CD274 overexpression [multivariable odds ratio (OR) 1.83; 95% confidence interval (CI), 1.22–2.75; P = .004]. PIK3CA mutation was statistically-insignificantly (P = .036 with the stringent alpha level of 0.005) associated with CD274 overexpression (multivariable OR, 1.54; 95% CI, 1.03–2.31). PIK3CA-mutated PTEN-lost tumors (n = 33) showed higher prevalence of CD274-positivity (82%) than PIK3CA-wild-type PTEN-lost tumors (n = 204; 70% CD274-positivity) and PTEN-expressed tumors (n = 147; 50% CD274-positivity) (P = .003). Our findings support the role of PI3K signaling in the CD274/PDCD1 pathway.
Collapse
Affiliation(s)
- Tomotaka Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Melissa Zhao
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Takashi Shimizu
- Department of Cardiovascular Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Naohiko Akimoto
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shanshan Shi
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yasutoshi Takashima
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rong Zhong
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mai Chan Lau
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Koichiro Haruki
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kota Arima
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kenji Fujiyoshi
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Benjamin Langworthy
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yohei Masugi
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Annacarolina da Silva
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Katsuhiko Nosho
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshifumi Baba
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Juha P Väyrynen
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.,Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, MA, USA
| |
Collapse
|
5
|
Zhao Y, Feng X, Chen Y, Selfridge JE, Gorityala S, Du Z, Wang JM, Hao Y, Cioffi G, Conlon RA, Barnholtz-Sloan JS, Saltzman J, Krishnamurthi SS, Vinayak S, Veigl M, Xu Y, Bajor DL, Markowitz SD, Meropol NJ, Eads JR, Wang Z. 5-Fluorouracil Enhances the Antitumor Activity of the Glutaminase Inhibitor CB-839 against PIK3CA-Mutant Colorectal Cancers. Cancer Res 2020; 80:4815-4827. [PMID: 32907836 DOI: 10.1158/0008-5472.can-20-0600] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/06/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022]
Abstract
PIK3CA encodes the p110α catalytic subunit of PI3K and is frequently mutated in human cancers, including ∼30% of colorectal cancer. Oncogenic mutations in PIK3CA render colorectal cancers more dependent on glutamine. Here we report that the glutaminase inhibitor CB-839 preferentially inhibits xenograft growth of PIK3CA-mutant, but not wild-type (WT), colorectal cancers. Moreover, the combination of CB-839 and 5-fluorouracil (5-FU) induces PIK3CA-mutant tumor regression in xenograft models. CB-839 treatment increased reactive oxygen species and caused nuclear translocation of Nrf2, which in turn upregulated mRNA expression of uridine phosphorylase 1 (UPP1). UPP1 facilitated the conversion of 5-FU to its active compound, thereby enhancing the inhibition of thymidylate synthase. Consistently, knockout of UPP1 abrogated the tumor inhibitory effect of combined CB-839 and 5-FU administration. A phase I clinical trial showed that the combination of CB-839 and capecitabine, a prodrug of 5-FU, was well tolerated at biologically-active doses. Although not designed to test efficacy, an exploratory analysis of the phase I data showed a trend that PIK3CA-mutant patients with colorectal cancer might derive greater benefit from this treatment strategy as compared with PIK3CA WT patients with colorectal cancer. These results effectively demonstrate that targeting glutamine metabolism may be an effective approach for treating patients with PIK3CA-mutant colorectal cancers and warrants further clinical evaluation. SIGNIFICANCE: Preclinical and clinical trial data suggest that the combination of CB-839 with capecitabine could serve as an effective treatment for PIK3CA-mutant colorectal cancers.
Collapse
Affiliation(s)
- Yiqing Zhao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Xiujing Feng
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Yicheng Chen
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - J Eva Selfridge
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Zhanwen Du
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Janet M Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Yujun Hao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Gino Cioffi
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Ronald A Conlon
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Joel Saltzman
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Smitha S Krishnamurthi
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Shaveta Vinayak
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Martina Veigl
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, Ohio
| | - David L Bajor
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Sanford D Markowitz
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Neal J Meropol
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Flatiron Health, New York, New York
| | - Jennifer R Eads
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio. .,Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
6
|
Morales-Oyarvide V, Yuan C, Babic A, Zhang S, Niedzwiecki D, Brand-Miller JC, Sampson-Kent L, Ye X, Li Y, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Wu K, Willett WC, Giovannucci EL, Wolpin BM, Meyerhardt JA, Fuchs CS, Ng K. Dietary Insulin Load and Cancer Recurrence and Survival in Patients With Stage III Colon Cancer: Findings From CALGB 89803 (Alliance). J Natl Cancer Inst 2020; 111:170-179. [PMID: 30726946 DOI: 10.1093/jnci/djy098] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/08/2018] [Accepted: 04/27/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Evidence suggests that diets inducing postprandial hyperinsulinemia may be associated with increased cancer-related mortality. The goal of this study was to assess the influence of postdiagnosis dietary insulin load and dietary insulin index on outcomes of stage III colon cancer patients. METHODS We conducted a prospective observational study of 1023 patients with resected stage III colon cancer enrolled in an adjuvant chemotherapy trial who reported dietary intake halfway through and six months after chemotherapy. We evaluated the association of dietary insulin load and dietary insulin index with cancer recurrence and survival using Cox proportional hazards regression adjusted for potential confounders; statistical tests were two-sided. RESULTS High dietary insulin load had a statistically significant association with worse disease-free survival (DFS), comparing the highest vs lowest quintile (adjusted hazard ratio [HR] = 2.77, 95% confidence interval [CI] = 1.90 to 4.02, Ptrend < .001). High dietary insulin index was also associated with worse DFS (highest vs lowest quintile, HR = 1.75, 95% CI = 1.22 to 2.51, Ptrend= .01). The association between higher dietary insulin load and worse DFS differed by body mass index and was strongest among patients with obesity (HR = 3.66, 95% CI = 1.88 to 7.12, Pinteraction = .04). The influence of dietary insulin load on cancer outcomes did not differ by mutation status of KRAS, BRAF, PIK3CA, TP53, or microsatellite instability. CONCLUSIONS Patients with resected stage III colon cancer who consumed a high-insulinogenic diet were at increased risk of recurrence and mortality. These findings support the importance of dietary management following resection of colon cancer, and future research into underlying mechanisms of action is warranted.
Collapse
Affiliation(s)
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Jennie C Brand-Miller
- Institute of Obesity, Nutrition, and Exercise, University of Sydney, Sydney, Australia
| | - Laura Sampson-Kent
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Xing Ye
- Alliance Statistics and Data Center, Duke University, Durham, NC
| | - Yanping Li
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | | | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Rex B Mowat
- Toledo Community Hospital Oncology Program, Toledo, OH
| | | | | | - Al Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | - Michael Messino
- Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC
| | - Hedy Kindler
- University of Chicago Comprehensive Cancer Center, Chicago, IL
| | - Alan Venook
- University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Department of Pathology, Harvard Medical School, Boston, MA
| | - Kana Wu
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Walter C Willett
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Charles S Fuchs
- Yale Cancer Center, Smilow Cancer Hospital and Yale School of Medicine, New Haven, CT
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
7
|
Jin J, Shi Y, Zhang S, Yang S. PIK3CA mutation and clinicopathological features of colorectal cancer: a systematic review and Meta-Analysis. Acta Oncol 2020; 59:66-74. [PMID: 31545109 DOI: 10.1080/0284186x.2019.1664764] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: There is conflicting evidence regarding the association between PIK3CA mutations and clinicopathological features of colorectal cancer (CRC). We performed a comprehensive meta-analysis investigating the association between PIK3CA mutations and clinicopathological features in CRC, including subgroup analysis of mutations in exons 9 and 20, to elucidate the role of PIK3CA mutations in CRC.Materials and Methods: A detailed literature search was performed within the PubMed, Web of Science, and Embase databases, examining the associations between PIK3CA mutations and demographic characteristics, clinicopathologic parameters, and molecular features in patients with CRC. The odds ratios with 95% confidence intervals were used to estimate the effect of PIK3CA mutations on outcome parameters.Results: Forty-four studies enrolling 17621 patients were eligible for inclusion. PIK3CA mutations were associated with proximal tumor location, mucinous differentiation, KRAS mutations, and microsatellite instability (MSI). Subgroup analysis demonstrated that PIK3CA exon 9 mutations were positively associated with proximal tumor location and KRAS mutations, and negatively associated with BRAF mutations and MSI; exon 20 mutations were associated with proximal tumor location, KRAS mutations, BRAF mutations and MSI.Conclusions: Our findings suggest that overall or exon-specific PIK3CA mutations showed null associations with key clinicopathological parameters, including disease stage and tumor differentiation, indicating that PIK3CA mutations do not predict aggressive clinicopathological characteristics in CRC. As PIK3CA mutations were found to be closely associated with KRAS mutations, their relationship warrants further investigation. Since PIK3CA exon 9 and 20 mutations showed different tendencies with regard to BRAF mutation and MSI status, they may have distinct molecular impacts on CRC.
Collapse
Affiliation(s)
- Juan Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yaqin Shi
- Department of Medical Oncology, the First Hospital Affiliated to Soochow University, Suzhou, China
| | - Shu Zhang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
8
|
PIK3CA Gene Mutations in Solid Malignancies: Association with Clinicopathological Parameters and Prognosis. Cancers (Basel) 2019; 12:cancers12010093. [PMID: 31905960 PMCID: PMC7017171 DOI: 10.3390/cancers12010093] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023] Open
Abstract
Phosphoinositide kinases (PIKs) are a group of lipid kinases that are important upstream activators of various significant signaling pathways. Hyperactivation of the PI3K/AKT/mTOR pathways—either via mutations or genomic amplification—confers key oncogenic activity, essential for the development and progression of several solid tumors. Alterations in the PIK3CA gene are associated with poor prognosis of solid malignancies. Although the literature reports contradictory prognostic values of PIK3CA in aggressive cancers, most of the available data highlight the important role of PIK3CA mutation in mediating tumorigenesis via increased signaling of the PI3K/AKT/mTOR survival pathway. Several inhibitors of PI3K/AKT/mTOR pathways are investigated as potential therapeutic options in solid malignancies. This article reviews the role of PIK3CA mutations and inhibitors of PI3K/AKT/mTOR pathways in major cancer types and examines its association with clinicopathological parameters and prognosis.
Collapse
|
9
|
Guercio BJ, Zhang S, Niedzwiecki D, Li Y, Babic A, Morales-Oyarvide V, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Zoltick ES, Stampfer M, Ng K, Wu K, Willett WC, Giovannucci EL, Meyerhardt JA, Fuchs CS. Associations of artificially sweetened beverage intake with disease recurrence and mortality in stage III colon cancer: Results from CALGB 89803 (Alliance). PLoS One 2018; 13:e0199244. [PMID: 30024889 PMCID: PMC6053135 DOI: 10.1371/journal.pone.0199244] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Purpose Observational studies have demonstrated increased colon cancer recurrence and mortality in states of excess energy balance, as denoted by factors including sedentary lifestyle, diabetes, increased dietary glycemic load, and increased intake of sugar-sweetened beverages. Nonetheless, the relation between artificially sweetened beverages, a popular alternative for sugar-sweetened beverages, and colon cancer recurrence and survival is unknown. Methods We analyzed data from 1,018 patients with stage III colon cancer who prospectively reported dietary intake during and after chemotherapy while enrolled in a National Cancer Institute-sponsored trial of adjuvant chemotherapy. Using Cox proportional hazards regressions, we assessed associations of artificially sweetened beverage intake with cancer recurrence and mortality. Results Patients consuming one or more 12-ounce servings of artificially sweetened beverages per day experienced an adjusted hazard ratio for cancer recurrence or mortality of 0.54 (95% confidence interval, 0.36 to 0.80) when compared to those who largely abstained (Ptrend = .004). Similarly, increasing artificially sweetened beverage intake was also associated with a significant improvement in both recurrence-free survival (Ptrend = .005) and overall survival (Ptrend = .02). Substitution models demonstrated that replacing a 12-ounce serving of a sugar-sweetened beverage with an isovolumetric serving of an artificially sweetened beverage per day was associated with a 23% lower risk of cancer recurrence and mortality (relative risk, 0.77; 95% confidence interval, 0.63 to 0.95; P = .02). Conclusion Higher artificially sweetened beverage consumption may be associated with significantly reduced cancer recurrence and death in patients with stage III colon cancer. This association may be mediated by substitution for sugar-sweetened alternatives. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Brendan J. Guercio
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Sui Zhang
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | - Donna Niedzwiecki
- Alliance Statistics and Data Center, Duke University, Durham, North Carolina, United States of America
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Ana Babic
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | | | - Leonard B. Saltz
- Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Robert J. Mayer
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | - Rex B. Mowat
- Toledo Community Hospital Oncology Program, Toledo, Ohio, United States of America
| | | | - Alexander Hantel
- Edward-Elmhurst Healthcare, Naperville, Illinois, United States of America
| | - Al Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, United States of America
| | - Daniel Atienza
- Virginia Oncology Associates, Norfolk, Virginia, United States of America
| | - Michael Messino
- Southeast Clinical Oncology Research (SCOR) Consortium, Mission Hospitals, Incorporated, Asheville, North Carolina, United States of America
| | - Hedy Kindler
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois, United States of America
| | - Alan Venook
- University of California at San Francisco Comprehensive Cancer Center, San Francisco, California, United States of America
| | - Shuji Ogino
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Division of Molecular Pathological Epidemiology (MPE), Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Emilie S. Zoltick
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Meir Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kimmie Ng
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Walter C. Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Edward L. Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Charles S. Fuchs
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
10
|
Van Blarigan EL, Fuchs CS, Niedzwiecki D, Ye X, Zhang S, Song M, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Giovannucci EL, Meyerhardt JA. Marine ω-3 Polyunsaturated Fatty Acid and Fish Intake after Colon Cancer Diagnosis and Survival: CALGB 89803 (Alliance). Cancer Epidemiol Biomarkers Prev 2018; 27:438-445. [PMID: 29358223 PMCID: PMC5939380 DOI: 10.1158/1055-9965.epi-17-0689] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/06/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
Background: Marine ω-3 polyunsaturated fatty acids (PUFAs), primarily found in dark fish, may prevent colorectal cancer progression, in part through inhibition of prostaglandin-endoperoxide synthase 2 (PTGS2). However, data in humans are limited.Methods: We examined marine ω-3 PUFAs and fish intake and survival among 1,011 colon cancer patients enrolled in Cancer and Leukemia Group B 89803 between 1999 and 2001 and followed through 2009. Diet was assessed during and 6 months after chemotherapy. We used Cox proportional hazards regression to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for disease-free (DFS), recurrence-free (RFS), and overall survival (OS).Results: We observed 343 recurrences and 305 deaths (median follow-up: 7 years). Patients in the highest vs. lowest quartile of marine ω-3 PUFA intake had an HR for DFS of 0.72 (95% CI, 0.54-0.97; Ptrend = 0.03). Individuals who consumed dark fish ≥1/week versus never had longer DFS (HR 0.65; 95% CI, 0.48-0.87; P-value = 0.007), RFS (HR 0.61; 95% CI, 0.46-0.86; Ptrend = 0.007), and OS (HR 0.68; 95% CI, 0.48-0.96; Ptrend = 0.04). In a subset of 510 patients, the association between marine ω-3 PUFA intake and DFS appeared stronger in patients with high PTGS2 expression (HR 0.32; 95% CI, 0.11-0.95; Ptrend = 0.01) compared with patients with absent/low PTGS2 expression (HR 0.78; 95% CI, 0.48-1.27; Ptrend = 0.35; Pinteraction = 0.19).Conclusions: Patients with high intake of marine ω-3 PUFAs and dark fish after colon cancer diagnosis may have longer DFS.Impact: Randomized controlled trials examining dark fish and/or marine ω-3 PUFA supplements and colon cancer recurrence/survival are needed. Cancer Epidemiol Biomarkers Prev; 27(4); 438-45. ©2018 AACR.
Collapse
Affiliation(s)
- Erin L. Van Blarigan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California,Department of Urology, University of California, San Francisco, California
| | - Charles S. Fuchs
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Donna Niedzwiecki
- Alliance Statistics and Data Center, Duke University Medical Center, Durham, North Carolina
| | - Xing Ye
- Alliance Statistics and Data Center, Duke University Medical Center, Durham, North Carolina
| | - Sui Zhang
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Robert J. Mayer
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Rex B. Mowat
- Toledo Community Hospital Oncology Program, Toledo, Ohio
| | - Renaud Whittom
- Hôpital du Sacré-Coeur de Montréal, Montréal, QC, Canada
| | - Alexander Hantel
- Loyola University Stritch School of Medicine, Naperville, Illinois
| | - Al Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | | | - Michael Messino
- Southeast Clinical Oncology Research (SCOR) Consortium, Mission Hospitals-Memorial Campus, Asheville, North Carolina
| | - Hedy Kindler
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| | - Alan Venook
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California,Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Shuji Ogino
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut,Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Edward L. Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
11
|
Fadelu T, Zhang S, Niedzwiecki D, Ye X, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson AB, Atienza DM, Messino M, Kindler HL, Venook A, Ogino S, Ng K, Wu K, Willett W, Giovannucci E, Meyerhardt J, Bao Y, Fuchs CS. Nut Consumption and Survival in Patients With Stage III Colon Cancer: Results From CALGB 89803 (Alliance). J Clin Oncol 2018; 36:1112-1120. [PMID: 29489429 PMCID: PMC5891130 DOI: 10.1200/jco.2017.75.5413] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Observational studies have reported increased colon cancer recurrence and mortality in patients with states of hyperinsulinemia, including type 2 diabetes, obesity, sedentary lifestyle, and high glycemic load diet. Nut intake has been associated with a lower risk of type 2 diabetes, metabolic syndrome, and insulin resistance. However, the effect of nut intake on colon cancer recurrence and survival is not known. Patients and Methods We conducted a prospective, observational study of 826 eligible patients with stage III colon cancer who reported dietary intake on food frequency questionnaires while enrolled onto a randomized adjuvant chemotherapy trial. Using Cox proportional hazards regression, we assessed associations of nut intake with cancer recurrence and mortality. Results After a median follow-up of 6.5 years, compared with patients who abstained from nuts, individuals who consumed two or more servings of nuts per week experienced an adjusted hazard ratio (HR) for disease-free survival of 0.58 (95% CI, 0.37 to 0.92; Ptrend = .03) and an HR for overall survival of 0.43 (95% CI, 0.25 to 0.74; Ptrend = .01). In subgroup analysis, the apparent benefit was confined to tree nut intake (HR for disease-free survival, 0.54; 95% CI, 0.34 to 0.85; Ptrend = .04; and HR for overall survival, 0.47; 95% CI, 0.27 to 0.82; Ptrend = .04). The association of total nut intake with improved outcomes was maintained across other known or suspected risk factors for cancer recurrence and mortality. Conclusion Diets with a higher consumption of nuts may be associated with a significantly reduced incidence of cancer recurrence and death in patients with stage III colon cancer.
Collapse
Affiliation(s)
- Temidayo Fadelu
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Sui Zhang
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Donna Niedzwiecki
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Xing Ye
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Leonard B Saltz
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Robert J Mayer
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Rex B Mowat
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Renaud Whittom
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Alexander Hantel
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Al B Benson
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Daniel M Atienza
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Michael Messino
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Hedy L Kindler
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Alan Venook
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Shuji Ogino
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Kimmie Ng
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Kana Wu
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Walter Willett
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Edward Giovannucci
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Jeffrey Meyerhardt
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Ying Bao
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Charles S Fuchs
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| |
Collapse
|
12
|
Li AJ, Li HG, Tang EJ, Wu W, Chen Y, Jiang HH, Lin MB, Yin L. PIK3CA and TP53 mutations predict overall survival of stage II/III colorectal cancer patients. World J Gastroenterol 2018; 24:631-640. [PMID: 29434452 PMCID: PMC5799864 DOI: 10.3748/wjg.v24.i5.631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/13/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the predictive value of PIK3CA and TP53 mutation status in colorectal cancer (CRC) patients treated with 5-fluorouracil-based chemotherapy.
METHODS In this study, a total of 315 patients with histologically proven CRC were enrolled from Yangpu Hospital affiliated to Shanghai Tongji University between 2007 and 2011. Of these patients, 241 with stage II/III CRC received 5-fluorouracil-based adjuvant chemotherapy. Formalin-fixed paraffin-embedded lesion samples of the patients with curatively resected CRC were collected. Next-generation sequencing was performed to identify somatic gene mutations. The correlation of PIK3CA and TP53 mutation status with overall survival (OS) was analyzed using a Cox proportional hazard model and the Kaplan-Meier method.
RESULTS Among the 241 patients with stage II/III in this cohort, the PIK3CA and/or TP53 mutation was detected in 177 patients, among which 54 patients had PIK3CA and TP53 double mutations. The PIK3CA or TP53 mutation was not significantly correlated with OS in univariate and multivariate analyses. Compared with patients without PIK3CA and TP53 mutations, those with double PIK3CA-TP53 mutations showed a significantly worse survival (univariate HR = 2.21; 95%CI: 1.15-4.24; multivariate HR = 2.02; 95%CI: 1.04-3.91). The PIK3CA mutation located in the kinase domain showed a trend toward a shorter OS compared with wild-type tumors (multivariate HR = 1.56; 95%CI: 1.00-2.44; P = 0.052). The Kaplan-Meier curve showed that patients harboring the PIK3CA mutation located in the kinase domain had a worse clinical outcome than those with wild-type status (Log-rank P = 0.041)
CONCLUSION Double mutation of PIK3CA and TP53 is correlated with a shorter OS in stage II/III CRC patients treated with 5-fluorouracil-based therapy.
Collapse
Affiliation(s)
- A-Jian Li
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Hua-Guang Li
- Center for Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Er-Jiang Tang
- Center for Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Wei Wu
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Ying Chen
- Center for Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Hui-Hong Jiang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Mou-Bin Lin
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Lu Yin
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
13
|
Ogino S, Jhun I, Mata DA, Soong TR, Hamada T, Liu L, Nishihara R, Giannakis M, Cao Y, Manson JE, Nowak JA, Chan AT. Integration of pharmacology, molecular pathology, and population data science to support precision gastrointestinal oncology. NPJ Precis Oncol 2017; 1. [PMID: 29552640 PMCID: PMC5856171 DOI: 10.1038/s41698-017-0042-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Precision medicine has a goal of customizing disease prevention and treatment strategies. Under the precision medicine paradigm, each patient has unique pathologic processes resulting from cellular genomic, epigenomic, proteomic, and metabolomic alterations, which are influenced by pharmacological, environmental, microbial, dietary, and lifestyle factors. Hence, to realize the promise of precision medicine, multi-level research methods that can comprehensively analyze many of these variables are needed. In order to address this gap, the integrative field of molecular pathology and population data science (i.e., molecular pathological epidemiology) has been developed to enable such multi-level analyses, especially in gastrointestinal cancer research. Further integration of pharmacology can improve our understanding of drug effects, and inform decision-making of drug use at both the individual and population levels. Such integrative research demonstrated potential benefits of aspirin in colorectal carcinoma with PIK3CA mutations, providing the basis for new clinical trials. Evidence also suggests that HPGD (15-PDGH) expression levels in normal colon and the germline rs6983267 polymorphism that relates to tumor CTNNB1 (β-catenin)/WNT signaling status may predict the efficacy of aspirin for cancer chemoprevention. As immune checkpoint blockade targeting the CD274 (PD-L1)/PDCD1 (PD-1) pathway for microsatellite instability-high (or mismatch repair-deficient) metastatic gastrointestinal or other tumors has become standard of care, potential modifying effects of dietary, lifestyle, microbial, and environmental factors on immunotherapy need to be studied to further optimize treatment strategies. With its broad applicability, our integrative approach can provide insights into the interactive role of medications, exposures, and molecular pathology, and guide the development of precision medicine.
Collapse
Affiliation(s)
- Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Iny Jhun
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Douglas A Mata
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thing Rinda Soong
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Li Liu
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Reiko Nishihara
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marios Giannakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yin Cao
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Grancher A, Michel P, Di Fiore F, Sefrioui D. [Aspirin and colorectal cancer]. Bull Cancer 2017; 105:171-180. [PMID: 29153543 DOI: 10.1016/j.bulcan.2017.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/18/2017] [Accepted: 09/08/2017] [Indexed: 12/17/2022]
Abstract
Colorectal cancer is a worldwide public health problem. Aspirin has been identified as a protective factor against the apparition of colorectal cancer. There are several mechanisms about the actions by aspirin on colorectal tumorogenesis. These are not perfectly known nowadays. On one hand, there are direct mechanisms on colorectal mucosa, on the other hand there are indirect mechanisms through platelet functions. Aspirin also plays a role by its anti-inflammatory action and the stimulation of antitumor immunity. Several studies show that long-term treatment with low-doses of aspirin decreases the incidence of adenomas and colorectal cancers. In the United States, aspirin is currently recommended for primary prevention of the risk of colorectal cancer in all patients aged 50 to 59, with a 10-year risk of cardiovascular event greater than 10 %. However, primary prevention with aspirin should not be a substitute for screening in colorectal cancer. Furthermore, aspirin seems to be beneficial when used in post-diagnosis of colorectal cancer. It could actually decrease the risk of metastasis in case of a localized colorectal cancer, and increase the survival in particular, concerning PIK3CA mutated tumors. The association of aspirin with neoadjuvant treatment of colorectal cancer by radiochimiotherapy seems to have beneficial effects. French prospective randomized study is currently being conducted to investigate postoperative aspirin in colorectal cancers with a PIK3CA mutation.
Collapse
Affiliation(s)
- Adrien Grancher
- Normandie université, UNIROUEN, hôpital universitaire de Rouen, service d'hépato-gastroenterologie, 76000 Rouen, France
| | - Pierre Michel
- Normandie université, UNIROUEN, Inserm 1245, IRON group, hôpital universitaire de Rouen, service d'hépato-gastroentérologie, 76000 Rouen, France.
| | - Frédéric Di Fiore
- Normandie université, UNIROUEN, Inserm 1245, IRON group, hôpital universitaire de Rouen, centre Henri-Becquerel, département d'oncolgie médicale, service d'hépato-gastroentérologie, 76000 Rouen, France
| | - David Sefrioui
- Normandie université, UNIROUEN, Inserm 1245, IRON group, hôpital universitaire de Rouen, service d'hépato-gastroentérologie, 76000 Rouen, France
| |
Collapse
|
15
|
Abstract
Colorectal cancers develop through at least 3 major pathways, including chromosomal instability, mismatch repair, and methylator phenotype. These pathways can coexist in a single individual and occur in both sporadic and inherited colorectal cancers. In spite of the unique molecular and genetic signatures of colorectal cancers, nonspecific chemotherapy based on the antineoplastic effects of 5-fluorouracil is the cornerstone of therapy for stage III and some stage II disease. Techniques to recognize colorectal cancer at the molecular level have facilitated development of new signature drugs designed to inhibit the unique pathways of colorectal cancer growth and immunity.
Collapse
|
16
|
Abstract
The ability to identify a specific cancer using minimally invasive biopsy holds great promise for improving the diagnosis, treatment selection, and prediction of prognosis in cancer. Using whole-genome methylation data from The Cancer Genome Atlas (TCGA) and machine learning methods, we evaluated the utility of DNA methylation for differentiating tumor tissue and normal tissue for four common cancers (breast, colon, liver, and lung). We identified cancer markers in a training cohort of 1,619 tumor samples and 173 matched adjacent normal tissue samples. We replicated our findings in a separate TCGA cohort of 791 tumor samples and 93 matched adjacent normal tissue samples, as well as an independent Chinese cohort of 394 tumor samples and 324 matched adjacent normal tissue samples. The DNA methylation analysis could predict cancer versus normal tissue with more than 95% accuracy in these three cohorts, demonstrating accuracy comparable to typical diagnostic methods. This analysis also correctly identified 29 of 30 colorectal cancer metastases to the liver and 32 of 34 colorectal cancer metastases to the lung. We also found that methylation patterns can predict prognosis and survival. We correlated differential methylation of CpG sites predictive of cancer with expression of associated genes known to be important in cancer biology, showing decreased expression with increased methylation, as expected. We verified gene expression profiles in a mouse model of hepatocellular carcinoma. Taken together, these findings demonstrate the utility of methylation biomarkers for the molecular characterization of cancer, with implications for diagnosis and prognosis.
Collapse
|
17
|
Sepulveda AR, Hamilton SR, Allegra CJ, Grody W, Cushman-Vokoun AM, Funkhouser WK, Kopetz SE, Lieu C, Lindor NM, Minsky BD, Monzon FA, Sargent DJ, Singh VM, Willis J, Clark J, Colasacco C, Bryan Rumble R, Temple-Smolkin R, B Ventura C, Nowak JA. Molecular Biomarkers for the Evaluation of Colorectal Cancer: Guideline From the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and American Society of Clinical Oncology. Arch Pathol Lab Med 2017; 141:625-657. [PMID: 28165284 DOI: 10.5858/arpa.2016-0554-cp] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES - To develop evidence-based guideline recommendations through a systematic review of the literature to establish standard molecular biomarker testing of colorectal cancer (CRC) tissues to guide epidermal growth factor receptor (EGFR) therapies and conventional chemotherapy regimens. METHODS - The American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and American Society of Clinical Oncology convened an expert panel to develop an evidence-based guideline to establish standard molecular biomarker testing and guide therapies for patients with CRC. A comprehensive literature search that included more than 4,000 articles was conducted. RESULTS - Twenty-one guideline statements were established. CONCLUSIONS - Evidence supports mutational testing for EGFR signaling pathway genes, since they provide clinically actionable information as negative predictors of benefit to anti-EGFR monoclonal antibody therapies for targeted therapy of CRC. Mutations in several of the biomarkers have clear prognostic value. Laboratory approaches to operationalize CRC molecular testing are presented.
Collapse
Affiliation(s)
- Antonia R Sepulveda
- From the 1 Department of Pathology and Cell Biology, Columbia University, New York, NY
| | | | - Carmen J Allegra
- 5 Division of Hematology and Oncology, University of Florida Medical Center, Gainesville
| | - Wayne Grody
- 6 Departments of Pathology and Laboratory Medicine, Pediatrics, and Human Genetics, UCLA Medical Center, Los Angeles, CA
| | | | - William K Funkhouser
- 8 Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill
| | | | - Christopher Lieu
- 9 Division of Medical Oncology, University of Colorado Denver School of Medicine, Denver
| | | | - Bruce D Minsky
- 4 Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston
| | | | - Daniel J Sargent
- 12 Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | - Joseph Willis
- 14 Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Jennifer Clark
- 15 ASCP Institute for Science, Technology, and Policy, American Society for Clinical Pathology, Washington, DC
| | - Carol Colasacco
- 16 Laboratory and Pathology Quality Center, College of American Pathologists, Northfield, IL
| | - R Bryan Rumble
- 17 American Society of Clinical Oncology, Alexandria, VA
| | | | - Christina B Ventura
- 16 Laboratory and Pathology Quality Center, College of American Pathologists, Northfield, IL
| | - Jan A Nowak
- From the 1 Department of Pathology and Cell Biology, Columbia University, New York, NY
- 2 Department of Pathology
- 3 Department of Gastrointestinal (GI) Medical Oncology
- 4 Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston
- 5 Division of Hematology and Oncology, University of Florida Medical Center, Gainesville
- 6 Departments of Pathology and Laboratory Medicine, Pediatrics, and Human Genetics, UCLA Medical Center, Los Angeles, CA
- 7 Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha
- 8 Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill
- 9 Division of Medical Oncology, University of Colorado Denver School of Medicine, Denver
- 10 Department of Medical Genetics, Mayo Clinic, Scottsdale, AZ
- 11 Castle Biosciences, Friendswood, TX
- 12 Department of Health Sciences Research, Mayo Clinic, Rochester, MN
- 13 Biocept, San Diego, CA
- 14 Department of Pathology, Case Western Reserve University, Cleveland, OH
- 15 ASCP Institute for Science, Technology, and Policy, American Society for Clinical Pathology, Washington, DC
- 16 Laboratory and Pathology Quality Center, College of American Pathologists, Northfield, IL
- 17 American Society of Clinical Oncology, Alexandria, VA
- 18 Association for Molecular Pathology, Bethesda, MD
- 19 Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, NY
| |
Collapse
|
18
|
Taieb J, Le Malicot K, Shi Q, Penault-Llorca F, Bouché O, Tabernero J, Mini E, Goldberg RM, Folprecht G, Luc Van Laethem J, Sargent DJ, Alberts SR, Emile JF, Laurent Puig P, Sinicrope FA. Prognostic Value of BRAF and KRAS Mutations in MSI and MSS Stage III Colon Cancer. J Natl Cancer Inst 2017; 109:2760215. [PMID: 28040692 PMCID: PMC6075212 DOI: 10.1093/jnci/djw272] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/04/2016] [Accepted: 10/12/2016] [Indexed: 02/06/2023] Open
Abstract
Background The prognostic value of BRAF and KRAS mutations within microsatellite-unstable (MSI) and microsatellite-stable (MSS) subgroups of resected colon carcinoma patients remains controversial. We examined this question in prospectively collected biospecimens from stage III colon cancer with separate analysis of MSI and MSS tumors from patients receiving adjuvant FOLFOX +/- cetuximab in two adjuvant therapy trials. Methods Three groups were defined: BRAF Mutant, KRAS Mutant, and double wild-type. The analytic strategy involved estimation of study-specific effects, assessment of homogeneity of results, and then analysis of pooled data as no differences in patient outcome were found between treatment arms in both trials. Associations of mutations with patient outcome were analyzed, and multivariable models were adjusted for treatment and relevant factors. Results Four thousand four hundred eleven tumors were evaluable for BRAF and KRAS mutations and mismatch repair status; 3934 were MSS and 477 were MSI. In MSS patients, all BRAF V600E mutations (hazard ratio [HR] = 1.54, 95% confidence interval [CI] = 1.23 to 1.92, P < .001), KRAS codon 12 alterations, and p.G13D mutations (HR = 1.60, 95% CI = 1.40 to 1.83, P < .001) were associated with shorter time to recurrence (TTR) and shorter survival after relapse (SAR; HR = 3.02 , 95% CI = 2.32 to 3.93, P < .001, and HR = 1.20, 95% CI = 1.01 to 1.44, P = .04, respectively). Overall survival (OS) in MSS patients was poorer for BRAF-mutant patients (HR = 2.01, 95% CI = 1.56 to 2.57, P < .001) and KRAS-mutant patients (HR = 1.62, 95% CI = 1.38 to 1.91, P < .001) vs wild-type. No prognostic role of KRAS or BRAF mutations was seen in MSI patients. Furthermore, no interaction was found between treatment arm (with or without cetuximab) and KRAS and BRAF mutations for TTR or OS in MSS patients. Conclusions In a pooled analysis of resected stage III colon cancer patients receiving adjuvant FOLFOX, BRAF or KRAS mutations are independently associated with shorter TTR, SAR, and OS in patients with MSS, but not MSI, tumors. Future clinical trials in the adjuvant setting should consider these mutations as important stratification factors.
Collapse
Affiliation(s)
- Julien Taieb
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Karine Le Malicot
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Qian Shi
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Frédérique Penault-Llorca
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Olivier Bouché
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Josep Tabernero
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Enrico Mini
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Richard M Goldberg
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Gunnar Folprecht
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Jean Luc Van Laethem
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Daniel J Sargent
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Steven R Alberts
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Jean Francois Emile
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Pierre Laurent Puig
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| | - Frank A Sinicrope
- Affiliations of authors: Department of Gastroenterology and GI Oncology, Université Paris Descartes, Hopital Européen Georges Pompidou, Paris, France (JTai); Fédération Francophone de Cancérologie Digestive (FFCD), Dijon, France (KLM); Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN (QS, DJS); Department of Pathology, Centre Jean Perrin, EA ERTICa, Université d'Auvergne, Clermont-Ferrand, France (FPL); Centre Hospitalier Universitaire Robert Debré, Reims, France (OB); Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain (JTab); Spanish Gastrointestinal Tumors TTD Group, Universitat Autònoma de Barcelona, Barcelona, Spain (JTab); Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy (EM); Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); First Medical Department, University Hospital Carl Gustav Carus, Dresden, Germany (GF); Department of Gastroenterology, Hôpital Universitaire Erasme, Brussels, Belgium (JLVL); Department of Oncology, Mayo Clinic and Mayo Cancer Center, Rochester, MN (SRA, FAS); Pathology Department, Ambroise Paré Hospital, Boulogne, France (JFE); Université Paris Descartes, Sorbonne Paris Cité, France (PLP); Assistance Publique Hôpitaux de Paris, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France (PLP); INSERM UMR-S1147, Paris, France (PLP)
| |
Collapse
|
19
|
Abstract
Colorectal cancer (CRC) is a leading cause of cancer deaths worldwide. CRC develops as a consequence of genomic instability, characterized by various genetic and epigenetic alterations. Its molecular heterogeneity explains the large variability in patient prognosis and treatment response, emphasizing the need for development of accurate prognostic and predictive biomarkers. This article delineates the different pathways of colorectal carcinogenesis and its molecular subtype classification. With this review, we aim to provide a comprehensive overview of the current and future biomarkers guiding clinical decision-making and CRC treatment.
Collapse
Affiliation(s)
- Pieter-Jan Cuyle
- Department of Gastroenterology/Digestive Oncology, Imelda General Hospital, Bonheiden, Belgium
- Department of Gastroenterology/Digestive Oncology, University Hospitals Gasthuisberg Leuven, Leuven, Belgium
| | - Hans Prenen
- Department of Gastroenterology/Digestive Oncology, University Hospitals Gasthuisberg Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Sepulveda AR, Hamilton SR, Allegra CJ, Grody W, Cushman-Vokoun AM, Funkhouser WK, Kopetz SE, Lieu C, Lindor NM, Minsky BD, Monzon FA, Sargent DJ, Singh VM, Willis J, Clark J, Colasacco C, Rumble RB, Temple-Smolkin R, Ventura CB, Nowak JA. Molecular Biomarkers for the Evaluation of Colorectal Cancer. Am J Clin Pathol 2017; 147:221-260. [PMID: 28165529 PMCID: PMC7263311 DOI: 10.1093/ajcp/aqw209] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objectives: To develop evidence-based guideline recommendations through a systematic review of the literature to establish standard molecular biomarker testing of colorectal cancer (CRC) tissues to guide epidermal growth factor receptor (EGFR) therapies and conventional chemotherapy regimens. Methods: The American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and American Society of Clinical Oncology convened an expert panel to develop an evidence-based guideline to establish standard molecular biomarker testing and guide therapies for patients with CRC. A comprehensive literature search that included more than 4,000 articles was conducted. Results: Twenty-one guideline statements were established. Conclusions: Evidence supports mutational testing for EGFR signaling pathway genes, since they provide clinically actionable information as negative predictors of benefit to anti-EGFR monoclonal antibody therapies for targeted therapy of CRC. Mutations in several of the biomarkers have clear prognostic value. Laboratory approaches to operationalize CRC molecular testing are presented.
Collapse
Affiliation(s)
- Antonia R. Sepulveda
- From theDepartment of Pathology and Cell Biology, Columbia University, New York, NY; Departments of
| | | | - Carmen J. Allegra
- Division of Hematology and Oncology, University of Florida Medical Center, Gainesville
| | - Wayne Grody
- Departments of Pathology and Laboratory Medicine, Pediatrics, and Human Genetics UCLA Medical Center, Los Angeles, CA
| | | | - William K. Funkhouser
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill
| | | | - Christopher Lieu
- Division of Medical Oncology, University of Colorado Denver School of Medicine, Denver
| | | | - Bruce D. Minsky
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston
| | | | | | | | - Joseph Willis
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Jennifer Clark
- ASCP Institute for Science, Technology, and Policy, American Society for Clinical Pathology, Washington, DC
| | - Carol Colasacco
- Laboratory and Pathology Quality Center, College of American Pathologists, Northfield, IL
| | | | | | - Christina B. Ventura
- Laboratory and Pathology Quality Center, College of American Pathologists, Northfield, IL
| | - Jan A. Nowak
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, NY
| |
Collapse
|
21
|
Sepulveda AR, Hamilton SR, Allegra CJ, Grody W, Cushman-Vokoun AM, Funkhouser WK, Kopetz SE, Lieu C, Lindor NM, Minsky BD, Monzon FA, Sargent DJ, Singh VM, Willis J, Clark J, Colasacco C, Rumble RB, Temple-Smolkin R, Ventura CB, Nowak JA. Molecular Biomarkers for the Evaluation of Colorectal Cancer: Guideline From the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and American Society of Clinical Oncology. J Mol Diagn 2017; 19:187-225. [PMID: 28185757 PMCID: PMC5971222 DOI: 10.1016/j.jmoldx.2016.11.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To develop evidence-based guideline recommendations through a systematic review of the literature to establish standard molecular biomarker testing of colorectal cancer (CRC) tissues to guide epidermal growth factor receptor (EGFR) therapies and conventional chemotherapy regimens. METHODS The American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and American Society of Clinical Oncology convened an expert panel to develop an evidence-based guideline to establish standard molecular biomarker testing and guide therapies for patients with CRC. A comprehensive literature search that included more than 4,000 articles was conducted. RESULTS Twenty-one guideline statements were established. CONCLUSIONS Evidence supports mutational testing for EGFR signaling pathway genes, since they provide clinically actionable information as negative predictors of benefit to anti-EGFR monoclonal antibody therapies for targeted therapy of CRC. Mutations in several of the biomarkers have clear prognostic value. Laboratory approaches to operationalize CRC molecular testing are presented. Key Words: Molecular diagnostics; Gastrointestinal; Histology; Genetics; Oncology.
Collapse
Affiliation(s)
- Antonia R Sepulveda
- Department of Pathology and Cell Biology, Columbia University, New York, NY.
| | - Stanley R Hamilton
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Carmen J Allegra
- Division of Hematology and Oncology, University of Florida Medical Center, Gainesville
| | - Wayne Grody
- Departments of Pathology and Laboratory Medicine, Pediatrics, and Human Genetics, UCLA Medical Center, Los Angeles, CA
| | | | - William K Funkhouser
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill
| | - Scott E Kopetz
- Department of Gastrointestinal (GI) Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Christopher Lieu
- Division of Medical Oncology, University of Colorado Denver School of Medicine, Denver
| | | | - Bruce D Minsky
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston
| | | | - Daniel J Sargent
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | - Joseph Willis
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Jennifer Clark
- ASCP Institute for Science, Technology, and Policy, American Society for Clinical Pathology, Washington, DC
| | - Carol Colasacco
- Laboratory and Pathology Quality Center, College of American Pathologists, Northfield, IL
| | | | | | - Christina B Ventura
- Laboratory and Pathology Quality Center, College of American Pathologists, Northfield, IL
| | - Jan A Nowak
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, NY
| |
Collapse
|
22
|
Sepulveda AR, Hamilton SR, Allegra CJ, Grody W, Cushman-Vokoun AM, Funkhouser WK, Kopetz SE, Lieu C, Lindor NM, Minsky BD, Monzon FA, Sargent DJ, Singh VM, Willis J, Clark J, Colasacco C, Rumble RB, Temple-Smolkin R, Ventura CB, Nowak JA. Molecular Biomarkers for the Evaluation of Colorectal Cancer: Guideline From the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and the American Society of Clinical Oncology. J Clin Oncol 2017; 35:1453-1486. [PMID: 28165299 DOI: 10.1200/jco.2016.71.9807] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Molecular testing of colorectal cancers (CRCs) to improve patient care and outcomes of targeted and conventional therapies has been the center of many recent studies, including clinical trials. Evidence-based recommendations for the molecular testing of CRC tissues to guide epidermal growth factor receptor (EGFR) -targeted therapies and conventional chemotherapy regimens are warranted in clinical practice. The purpose of this guideline is to develop evidence-based recommendations to help establish standard molecular biomarker testing for CRC through a systematic review of the literature. Methods The American Society for Clinical Pathology (ASCP), College of American Pathologists (CAP), Association for Molecular Pathology (AMP), and the American Society of Clinical Oncology (ASCO) convened an Expert Panel to develop an evidence-based guideline to help establish standard molecular biomarker testing, guide targeted therapies, and advance personalized care for patients with CRC. A comprehensive literature search that included over 4,000 articles was conducted to gather data to inform this guideline. Results Twenty-one guideline statements (eight recommendations, 10 expert consensus opinions and three no recommendations) were established. Recommendations Evidence supports mutational testing for genes in the EGFR signaling pathway, since they provide clinically actionable information as negative predictors of benefit to anti-EGFR monoclonal antibody therapies for targeted therapy of CRC. Mutations in several of the biomarkers have clear prognostic value. Laboratory approaches to operationalize molecular testing for predictive and prognostic molecular biomarkers involve selection of assays, type of specimens to be tested, timing of ordering of tests and turnaround time for testing results. Additional information is available at: www.asco.org/CRC-markers-guideline and www.asco.org/guidelineswiki.
Collapse
Affiliation(s)
- Antonia R Sepulveda
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Stanley R Hamilton
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Carmen J Allegra
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Wayne Grody
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Allison M Cushman-Vokoun
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - William K Funkhouser
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Scott E Kopetz
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Christopher Lieu
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Noralane M Lindor
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Bruce D Minsky
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Federico A Monzon
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Daniel J Sargent
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Veena M Singh
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Joseph Willis
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Jennifer Clark
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Carol Colasacco
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - R Bryan Rumble
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Robyn Temple-Smolkin
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Christina B Ventura
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| | - Jan A Nowak
- Antonia R. Sepulveda, Columbia University, New York, NY; Stanley R. Hamilton, Scott E. Kopetz, and Bruce D. Minsky, University of Texas MD Anderson Cancer Center, Houston, TX; Carmen J. Allegra, University of Florida Medical Center, Gainesville, FL; Wayne Grody, UCLA Medical Center, Los Angeles, CA; Allison M. Cushman-Vokoun, University of Nebraska Medical Center, Omaha, NE; William K. Funkhouser, University of North Carolina School of Medicine, Chapel Hill, NC; Christopher Lieu, University of Colorado Denver School of Medicine, Denver, CO; Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ; Federico A. Monzon, Castle Biosciences, Friendswood, TX; Daniel J. Sargent, Mayo Clinic, Rochester, MN; Veena M. Singh, Biocept, San Diego, CA; Joseph Willis, Case Western Reserve University, Cleveland, OH; Jennifer Clark, American Society for Clinical Pathology, Washington, DC; Carol Colasacco and Christina B. Ventura, College of American Pathologists, Northfield, IL; R. Bryan Rumble, American Society of Clinical Oncology, Alexandria, VA; Robyn Temple-Smolkin, Association for Molecular Pathology, Bethesda, MD; and Jan A. Nowak, Roswell Park Cancer Institute, Buffalo, NY
| |
Collapse
|
23
|
Mei ZB, Duan CY, Li CB, Cui L, Ogino S. Prognostic role of tumor PIK3CA mutation in colorectal cancer: a systematic review and meta-analysis. Ann Oncol 2016; 27:1836-48. [PMID: 27436848 DOI: 10.1093/annonc/mdw264] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/28/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Somatic mutations in the phosphatidylinositol-4,5-bisphosphate 3-kinase/AKT pathway play a vital role in carcinogenesis. Approximately 15%-20% of colorectal cancers (CRCs) harbor activating mutations in PIK3CA, making it one of the most frequently mutated genes in CRC. We thus carried out a systematic review and meta-analysis investigating the prognostic significance of PIK3CA mutations in CRC. MATERIALS AND METHODS Electronic databases were searched from inception through May 2015. We extracted the study characteristics and prognostic data of each eligible study. The hazard ratio (HR) and 95% confidence interval (CI) were derived and pooled using the random-effects Mantel-Haenszel model. RESULTS Twenty-eight studies enrolling 12 747 patients were eligible for inclusion. Data on overall survival (OS) and progression-free survival (PFS) were available from 19 and 10 studies, respectively. Comparing PIK3CA-mutated CRC patients with PIK3CA-wild-type CRC patients, the summary HRs for OS and PFS were 0.96 (95% CI 0.83-1.12) and 1.20 (95% CI 0.98-1.46), respectively. The trim-and-fill, Copas model and subgroup analyses stratified by the study characteristics confirmed the robustness of the results. Five studies reported the CRC prognosis for PIK3CA mutations in exons 9 and 20 separately; neither exon 9 mutation nor exon 20 mutation in PIK3CA was significantly associated with patient survival. CONCLUSIONS Our findings suggest that PIK3CA mutation has the neutral prognostic effects on CRC OS and PFS. Evidence was accumulating for the establishment of CRC survival between PIK3CA mutations and patient-specific clinical or molecular profiles.
Collapse
Affiliation(s)
- Z B Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai
| | - C Y Duan
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing
| | - C B Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - L Cui
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - S Ogino
- Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston Department of Medical Oncology, Dana-Farber Cancer Institute, Boston Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| |
Collapse
|
24
|
Zhu L, Dong C, Cao Y, Fang X, Zhong C, Li D, Yuan Y. Prognostic Role of BRAF Mutation in Stage II/III Colorectal Cancer Receiving Curative Resection and Adjuvant Chemotherapy: A Meta-Analysis Based on Randomized Clinical Trials. PLoS One 2016; 11:e0154795. [PMID: 27138801 PMCID: PMC4854379 DOI: 10.1371/journal.pone.0154795] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/19/2016] [Indexed: 02/07/2023] Open
Abstract
Background and Objective Studies examining the prognostic value of the BRAF mutation on relapse-free survival (RFS), disease-free survival (DFS) and overall survival (OS) in stage II/III colorectal cancer (CRC) patients receiving curative resection and adjuvant chemotherapy so far showed discrepant results. Therefore, a meta-analysis of relevant studies was performed for clarification. Methods Randomized trials of stage II/III colorectal cancer treated with curative resection followed by adjuvant chemotherapy were selected to conduct a meta-analysis. The necessary descriptive and statistical information such as hazard ratios (HRs) and 95% confidence intervals (CIs) were derived from published survival data. Results Seven phase III randomized clinical trials (RCTs) including 1,035 BRAF mutation stage II/III CRC patients receiving curative resection and adjuvant chemotherapy were analyzed. Overall, BRAF mutation resulted in poorer OS (HR = 1.42, 95% CI: 1.25–1.60; P < 0.00001), and poorer DFS (HR = 1.26, 95% CI: 1.07–1.48, P = 0.006) compared with BRAF wild-type CRC. The prognostic role on RFS could not be elucidated in the meta-analysis because of limited data. Conclusions BRAF mutation was significantly related with shorter DFS and OS among stage II/III CRC patients receiving adjuvant chemotherapy after curative resection. Its prognostic role for RFS needs to be further analyzed when more data is available.
Collapse
Affiliation(s)
- Lizhen Zhu
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Caixia Dong
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Cao
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xuefeng Fang
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Chenhan Zhong
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Dan Li
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- * E-mail:
| |
Collapse
|
25
|
Goldberg RM. Genomic Profiling in Gastrointestinal Cancer: Are We Ready To Use These Data to Make Treatment Decisions? Oncologist 2015; 20:1448-56. [PMID: 26512044 PMCID: PMC4679091 DOI: 10.1634/theoncologist.2015-0263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/03/2015] [Indexed: 01/27/2023] Open
Abstract
Remarkable strides have been made in the past 10-15 years in identifying the molecular events that drive cancer. With an enormous amount of new data, including those from The Cancer Genome Atlas Project, therapies are increasingly being developed and tested in clinical trials specifically designed to target some of these molecular events. Often, molecular signatures have become more important than the histologic features in making treatment choices. The success rate of these therapies depends on many factors but, perhaps most importantly, on patient selection according to the genetic analysis results of their individual tumors.
Collapse
Affiliation(s)
- Richard M Goldberg
- The Ohio State James Comprehensive Cancer Center and Solove Research Institute, Columbus, Ohio, USA
| |
Collapse
|
26
|
Markers in Colorectal Cancer and Clinical Trials Based Upon Them. CURRENT COLORECTAL CANCER REPORTS 2015. [DOI: 10.1007/s11888-015-0298-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
27
|
Chang SC, Lin PC, Lin JK, Lin CH, Yang SH, Liang WY, Chen WS, Jiang JK. Mutation Spectra of Common Cancer-Associated Genes in Different Phenotypes of Colorectal Carcinoma Without Distant Metastasis. Ann Surg Oncol 2015; 23:849-55. [PMID: 26471487 DOI: 10.1245/s10434-015-4899-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is a heterogeneous disease caused by genetic and epigenetic alterations. This study aimed to describe the mutation frequency of 12 genes in different CRC phenotypes. METHODS Patients who underwent surgery at the Taipei Veterans General Hospital during 2000-2010 for CRC (n = 1249) were enrolled. The endpoint was overall survival. The prognostic value was determined with the log-rank test and Cox regression analysis. RESULTS We found 1836 mutations of 12 genes in 997 (79.8%) tumors. Mutations were most frequently in KRAS (485, 38.8%), TP53 (373, 29.9%), APC (363, 29.0%), and PIK3CA (179, 14.3%); 137 (11.0%) cancers had high microsatellite instability (MSI). Women had significantly higher high MSI (14.3%) and BRAF mutation (6.3%) frequencies. The abnormal MSI (21.7%) and KRAS (44.6%), BRAF (8.6%), PIK3CA (19.4%), AKT1 (2.2%), and TGF - βR (9.6%) mutation frequencies were significantly higher in proximal colon cancer. The high MSI (35.6%) and BRAF (20.3%), TGF - βR (18.6%), PTEN (5.1%), and AKT1 (3.4%) mutation frequencies were significantly higher in 59 (4.7%) poorly differentiated tumors. The high MSI (21.3%) and KRAS (51.9%), BRAF (8.3%), PIK3CA (25.0%), AKT1 (4.6%), and SMAD4 (8.3%) mutation frequencies were significantly higher in 108 mucinous tumors. TNM stage, lymphovascular invasion, and mucinous histology were significantly associated with patient outcomes in univariate and multivariate analyses. Only NRAS mutation (hazard ratio 1.59, 95% confidence interval 1.06-2.38) affected patient survival. CONCLUSIONS Mutational spectra differ significantly between CRC subtypes, implying diverse carcinogenetic pathways. The NRAS mutation is important, despite its low frequency.
Collapse
Affiliation(s)
- Shih-Ching Chang
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Endoscopy Center for Diagnosis and Treatment, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pei-Ching Lin
- Department of Clinical Pathology, Yang-Ming Campus, Taipei City Hospital, Taipei, Taiwan
| | - Jen-Kou Lin
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Hsing Lin
- Division of Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Shung-Haur Yang
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Yi Liang
- Department of Pathology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Shone Chen
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jeng-Kai Jiang
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,Department of Surgery, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
28
|
Abstract
Sporadic colorectal cancer (CRC) is a somatic genetic disease in which pathogenesis is influenced by the local colonic environment and the patient's genetic background. Consolidating the knowledge of genetic and epigenetic events that occur with initiation, progression, and metastasis of sporadic CRC has identified some biomarkers that might be utilized to predict behavior and prognosis beyond staging, and inform treatment approaches. Modern next-generation sequencing of sporadic CRCs has confirmed prior identified genetic alterations and has classified new alterations. Each patient's CRC is genetically unique, propelled by 2-8 driver gene alterations that have accumulated within the CRC since initiation. Commonly observed alterations across sporadic CRCs have allowed classification into a (1) hypermutated group that includes defective DNA mismatch repair with microsatellite instability and POLE mutations in ∼15%, containing multiple frameshifted genes and BRAF(V600E); (2) nonhypermutated group with multiple somatic copy number alterations and aneuploidy in ∼85%, containing oncogenic activation of KRAS and PIK3CA and mutation and loss of heterozygosity of tumor suppressor genes, such as APC and TP53; (3) CpG island methylator phenotype CRCs in ∼20% that overlap greatly with microsatellite instability CRCs and some nonhypermutated CRCs; and (4) elevated microsatellite alterations at selected tetranucleotide repeats in ∼60% that associates with metastatic behavior in both hypermutated and nonhypermutated groups. Components from these classifications are now used as diagnostic, prognostic, and treatment biomarkers. Additional common biomarkers may come from genome-wide association studies and microRNAs among other sources, as well as from the unique alteration profile of an individual CRC to apply a precision medicine approach to care.
Collapse
Affiliation(s)
- John M Carethers
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Barbara H Jung
- Division of Gastroenterology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| |
Collapse
|
29
|
Erstad DJ, Tumusiime G, Cusack JC. Prognostic and Predictive Biomarkers in Colorectal Cancer: Implications for the Clinical Surgeon. Ann Surg Oncol 2015. [DOI: 10.1245/s10434-015-4706-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
30
|
Stec R, Semeniuk-Wojtaś A, Charkiewicz R, Bodnar L, Korniluk J, Smoter M, Chyczewski L, Nikliński J, Szczylik C. Mutation of the PIK3CA gene as a prognostic factor in patients with colorectal cancer. Oncol Lett 2015; 10:1423-1429. [PMID: 26622684 DOI: 10.3892/ol.2015.3398] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 05/28/2015] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, with ~700,000 mortalities occurring due to CRC in 2012. The treatment options are effective in a small percentage of patients, and it is important to identify specific biomarkers in order to determine patients for whom the available therapies will be beneficial. It has been hypothesised that the PIK3CA gene mutation may affect the response to therapy of patients with metastatic CRC. In the present study, primary tumour specimens were collected from 156 patients with CRC who were treated in the Military Institute of Medicine in Warsaw (Warsaw, Poland). Codons 12 and 13 of exon 1 of KRAS, exons 11 and 15 of BRAF and exons 9 and 20 of PIK3CA were analysed for mutation using direct sequencing. The prognostic value of each mutation and the clinical and pathological variables of these tumours were estimated. The results revealed that PIK3CA mutations were present in 15 patients (9.6%), of whom seven (46.7%) possessed mutations in codon 9 and eight (53.3%) possessed mutations in codon 20. Mutation in the PIK3CA gene was detected in six patients with KRAS gene mutations, which accounted for 40% of PIK3CA-mutated tumours, and in one patient with BRAF mutations, which accounted for 6.6% of PIK3CA-mutated tumours. No significant differences were identified between the overall survival (OS) rates of patients with PIK3CA mutations (median OS, 56.7 months) and those with wild-type PIK3CA genes (median OS, 47.6 months) (P=0.1270). Univariate analysis identified that the following prognostic factors affected the OS rate in the current patient cohort: Gender, female patients survived for 57.5 months compared with 39.3 months for male patients (P=0.0111); and lymph node involvement grade, as survival of patients without lymph node metastases was 61.4 months compared with 45.4 months in patients presenting with metastases (P=0.0122). The findings of the present analysis indicate that PIK3CA mutation status is not a prognostic factor in CRC patients. In addition, no statistically significant association exists between tumours with PIK3CA mutations and clinical or pathological factors.
Collapse
Affiliation(s)
- Rafał Stec
- Department of Oncology, Military Institute of Medicine, Warsaw 04-141, Poland
| | | | - Radosław Charkiewicz
- Department of Clinical Molecular Biology, Medical University of Bialystok, Bialystok 15-089, Poland
| | - Lubomir Bodnar
- Department of Oncology, Military Institute of Medicine, Warsaw 04-141, Poland
| | - Jan Korniluk
- Department of Oncology, Military Institute of Medicine, Warsaw 04-141, Poland
| | - Marta Smoter
- Department of Oncology, Military Institute of Medicine, Warsaw 04-141, Poland
| | - Lech Chyczewski
- Department of Clinical Pathology, Medical University of Bialystok, Bialystok 15-089, Poland
| | - Jacek Nikliński
- Department of Clinical Molecular Biology, Medical University of Bialystok, Bialystok 15-089, Poland
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, Warsaw 04-141, Poland
| |
Collapse
|
31
|
Gleeson FC, Kipp BR, Voss JS, Campion MB, Minot DM, Tu ZJ, Klee EW, Sciallis AP, Graham RP, Lazaridis KN, Henry MR, Levy MJ. Endoscopic ultrasound fine-needle aspiration cytology mutation profiling using targeted next-generation sequencing: personalized care for rectal cancer. Am J Clin Pathol 2015; 143:879-88. [PMID: 25972331 DOI: 10.1309/ajcpu3j7fgayqbrl] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES In an era of precision medicine, our aim was to determine the frequency and theranostic potential of mutations identified in malignant lymph nodes (LNs) sampled by endoscopic ultrasound fine-needle aspiration (EUS FNA) of patients with rectal cancer by targeted next-generation sequencing (NGS). METHODS The NGS Ion AmpliSeq Cancer Hotspot Panel v2 (Life Technologies, Carlsbad, CA) and MiSeq (Illumina, San Diego, CA) sequencers were used to sequence and assess for 2,800 or more possible mutations in 50 established cancer-associated genes. RESULTS Among 102 patients, 89% had 194 pathogenic alterations identified in 19 genes. The identification of KRAS, NRAS, or BRAF mutations suggests that 42% are likely nonresponders to anti-epidermal growth factor receptor therapy. Among KRAS, NRAS, or BRAF wild-type patients, alterations in eight genes linked to alternative therapies were identified in 44%. CONCLUSIONS Our data demonstrate the successful ability to apply a single multiplex test to allow multigene mutation detection from malignant LN cytology specimen DNA collected by EUS FNA.
Collapse
Affiliation(s)
- Ferga C. Gleeson
- Divison of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | - Jesse S. Voss
- Department of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | | | | | - Zheng J. Tu
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Eric W. Klee
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | | | - Konstantinos N. Lazaridis
- Divison of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | - Michael J. Levy
- Divison of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| |
Collapse
|
32
|
Phipps AI, Ahnen DJ, Cheng I, Newcomb PA, Win AK, Burnett T. PIK3CA Somatic Mutation Status in Relation to Patient and Tumor Factors in Racial/Ethnic Minorities with Colorectal Cancer. Cancer Epidemiol Biomarkers Prev 2015; 24:1046-51. [PMID: 25994739 DOI: 10.1158/1055-9965.epi-15-0204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/20/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Approximately 10% to 20% of colorectal cancers exhibit somatic mutations in the phosphoinositide-3-kinase, catalytic, alpha polypeptide gene (PIK3CA). We evaluated the relationship of PIK3CA mutation status in colorectal cancer with race/ethnicity, colorectal cancer survival, and other patient and tumor factors. METHODS This study comprised 377 racial/ethnic minorities with incident invasive colorectal cancer, enrolled in the Colon Cancer Family Registry via population-based cancer registries. Tumor specimens were tested for PIK3CA mutations in exon 9 and 20 hotspots, BRAF p.V600E mutations, and DNA mismatch repair (MMR). In logistic regression models, we evaluated the association between PIK3CA mutation status and race/ethnicity, overall, and by mutation site. Using Cox regression, we evaluated the association between PIK3CA mutation status and survival after colorectal cancer diagnosis. RESULTS PIK3CA mutations were detected in 42 cases (11%), with a similar prevalence across racial/ethnic groups. Individuals with PIK3CA-mutated colorectal cancer were significantly more likely than those with PIK3CA-wildtype disease to have proximal colon cancer, MMR-deficient tumors, and a germline MMR mutation (P ≤ 0.01). There was no evidence for an association between PIK3CA and overall survival (HR, 0.77; 95% confidence interval, 0.43-1.39). CONCLUSIONS The prevalence of PIK3CA mutation status in colorectal cancer does not differ according to race/ethnicity, but may vary according to other relevant clinicopathologic and etiologic factors, including germline MMR mutation status, tumor MMR status, and tumor site. IMPACT These findings underscore the importance of PIK3CA mutation status in colorectal cancer epidemiology and provide evidence that the prevalence of such mutations is similar across several racial/ethnic groups.
Collapse
Affiliation(s)
- Amanda I Phipps
- Epidemiology Department, University of Washington, Seattle, Washington. Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Dennis J Ahnen
- Division of Gastroenterology, University of Colorado School of Medicine, Denver, Colorado. Department of Medicine, Veterans Affairs Medical Center, Denver, Colorado
| | - Iona Cheng
- Cancer Prevention Institute of California, Fremont, California
| | - Polly A Newcomb
- Epidemiology Department, University of Washington, Seattle, Washington. Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Aung Ko Win
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Terrilea Burnett
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| |
Collapse
|
33
|
Manceau G, Marisa L, Boige V, Duval A, Gaub MP, Milano G, Selves J, Olschwang S, Jooste V, le Legrain M, Lecorre D, Guenot D, Etienne-Grimaldi MC, Kirzin S, Martin L, Lepage C, Bouvier AM, Laurent-Puig P. PIK3CA mutations predict recurrence in localized microsatellite stable colon cancer. Cancer Med 2015; 4:371-82. [PMID: 25641861 PMCID: PMC4380963 DOI: 10.1002/cam4.370] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 09/28/2014] [Accepted: 09/29/2014] [Indexed: 12/25/2022] Open
Abstract
PIK3CA, which encodes the p110α catalytic subunit of PI3Kα, is one of the most frequently altered oncogenes in colon cancer (CC), but its prognostic value is still a matter of debate. Few reports have addressed the association between PIK3CA mutations and survival and their results are controversial. In the present study, we aimed to clarify the prognostic impact of PIK3CA mutations in stage I-III CC according to mismatch repair status. Fresh frozen tissue samples from two independent cohorts with a total of 826 patients who underwent curative surgical resection of CC were analyzed for microsatellite instability and screened for activating point mutations in exon 9 and 20 of PIK3CA by direct sequencing. Overall, 693 tumors (84%) exhibited microsatellite stability (MSS) and 113 samples (14%) harbored PIK3CA mutation. In the retrospective training cohort (n = 433), patients with PIK3CA-mutated MSS tumors (n = 47) experienced a significant increased 5-year relapse-free interval compared with PIK3CA wild-type MSS tumors (n = 319) in univariate analysis (94% vs. 68%, Log-rank P = 0. 0003) and in multivariate analysis (HR = 0.12; 95% confidence interval, 0.029-0.48; P = 0.0027). In the prospective validation cohort (n = 393), the favorable prognostic impact of PIK3CA mutations in MSS tumors (n = 327) was confirmed (83% vs. 67%, Log-rank P = 0.04). Our study showed that PIK3CA mutations are associated with a good prognosis in patients with MSS stage I-III CC.
Collapse
Affiliation(s)
- Gilles Manceau
- Unité Mixte de Recherche S1147, Paris Sorbonne Cité, Université Paris Descartes, INSERM, Paris, France; Assistance Publique-Hôpitaux de Paris, Service de Chirurgie Digestive et Hépato-Bilio-Pancréatique, Hôpital Pitié-Salpêtrière, Paris, France; Institut Universitaire de Cancérologie, Université Pierre et Marie Curie-Paris 6, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yaeger R, Cowell E, Chou JF, Gewirtz AN, Borsu L, Vakiani E, Solit DB, Rosen N, Capanu M, Ladanyi M, Kemeny N. RAS mutations affect pattern of metastatic spread and increase propensity for brain metastasis in colorectal cancer. Cancer 2014; 121:1195-203. [PMID: 25491172 DOI: 10.1002/cncr.29196] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/03/2014] [Accepted: 11/17/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND RAS and PIK3CA mutations in metastatic colorectal cancer (mCRC) have been associated with worse survival. We sought to evaluate the impact of RAS and PIK3CA mutations on cumulative incidence of metastasis to potentially curable sites of liver and lung and other sites such as bone and brain. METHODS We performed a computerized search of the electronic medical record of our institution for mCRC cases genotyped for RAS or PIK3CA mutations from 2008 to 2012. Cases were reviewed for patient characteristics, survival, and site-specific metastasis. RESULTS Among the 918 patients identified, 477 cases were RAS wild type, and 441 cases had a RAS mutation (394 at KRAS exon 2, 29 at KRAS exon 3 or 4, and 18 in NRAS). RAS mutation was significantly associated with shorter median overall survival (OS) and on multivariate analysis independently predicted worse OS (HR, 1.6; P < .01). RAS mutant mCRC exhibited a significantly higher cumulative incidence of lung, bone, and brain metastasis and on multivariate analysis was an independent predictor of involvement of these sites (HR, 1.5, 1.6, and 3.7, respectively). PIK3CA mutations occurred in 10% of the 786 cases genotyped, did not predict for worse survival, and did not exhibit a site-specific pattern of metastatic spread. CONCLUSIONS The metastatic potential of CRC varies with the presence of RAS mutation. RAS mutation is associated with worse OS and increased incidence of lung, bone, and brain metastasis. An understanding of this site-specific pattern of spread may help to inform physicians' assessment of symptoms in patients with mCRC.
Collapse
Affiliation(s)
- Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zou Y, Huang MZ, Liu FY, Yang BC, Wang LQ, Wang F, Yu XH, Wan L, Wan XDI, Xu XY, Li W, Huang OP, He M. Absence of DICER1, CTCF, RPL22, DNMT3A, TRRAP, IDH1 and IDH2 hotspot mutations in patients with various subtypes of ovarian carcinomas. Biomed Rep 2014; 3:33-37. [PMID: 25469243 DOI: 10.3892/br.2014.378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/24/2014] [Indexed: 12/12/2022] Open
Abstract
Cancer is caused by multiple genetic alterations within cells. Recently, large-scale sequencing has identified frequent ribonuclease type III (DICER1), CCCTC-binding factor (CTCF), ribosomal protein L22 (RPL22), DNA (cytosine-5-)-methyltransferase 3α (DNMT3A), transformation/transcription domain-associated protein (TRRAP), isocitrate dehydrogenase (IDH)1 and IDH2 hotspot mutations in diverse types of cancer. However, it remains largely unknown whether these mutations also exist in ovarian carcinomas. In the present study, a collection of 251 patients with distinct subtypes of ovarian carcinomas were recruited and sequenced for the presence of these hotspot mutations. However, no mutations in the seven genes were detected in the samples. These negative results, together with certain recent reports, indicate that the hotspot mutations in the CTCF, RPL22, DNMT3A, TRRAP, IDH1 and IDH2 genes may not be actively involved in the carcinogenesis of ovarian carcinoma. Of note, the DICER1 mutation frequency in Sertoli-Leydig cell tumor in the present study was significantly lower compared to prior observation, and therefore, it is speculated that this discrepancy may be mainly due to the small sample size analyzed in the study. In addition, among these samples, frequent polymerase (DNA directed) ε, catalytic subunit (POLE1) and ring finger protein 43 (RNF43) mutations were identified in endometrioid and mucinous ovarian carcinomas, respectively; thus DICER1, CTCF, RPL22, DNMT3A, TRRAP, IDH1 and IDH2 hotspot mutations may not play synergistic roles with POLE1 or RNF43 mutations in the carcinogenesis of endometrioid or mucinous ovarian carcinomas.
Collapse
Affiliation(s)
- Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China ; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Mei-Zhen Huang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China ; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China ; Graduate School of Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Fa-Ying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China ; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Bi-Cheng Yang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China
| | - Li-Qun Wang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China
| | - Feng Wang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China ; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Hong Yu
- Department of Pathology, Jiangxi Provincial Maternal and Child Health Hospital, P.R. China
| | - Lei Wan
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China
| | - Xi-DI Wan
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China
| | - Xiao-Yun Xu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China ; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China ; Graduate School of Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Wei Li
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China ; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China ; Graduate School of Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Ou-Ping Huang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, P.R. China ; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Ming He
- Department of Pharmacology and Molecular Therapeutics, Nanchang University School of Pharmaceutical Science, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
36
|
Martini M, De Santis MC, Braccini L, Gulluni F, Hirsch E. PI3K/AKT signaling pathway and cancer: an updated review. Ann Med 2014; 46:372-83. [PMID: 24897931 DOI: 10.3109/07853890.2014.912836] [Citation(s) in RCA: 823] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite development of novel agents targeting oncogenic pathways, matching targeted therapies to the genetic status of individual tumors is proving to be a daunting task for clinicians. To improve the clinical efficacy and to reduce the toxic side effects of treatments, a deep characterization of genetic alterations in different tumors is required. The mutational profile often evidences a gain of function or hyperactivity of phosphoinositide 3-kinases (PI3Ks) in tumors. These enzymes are activated downstream tyrosine kinase receptors (RTKs) and/or G proteins coupled receptors (GPCRs) and, via AKT, are able to induce mammalian target of rapamycin (mTOR) stimulation. Here, we elucidate the impact of class I (p110α, β, γ, and δ) catalytic subunit mutations on AKT-mediated cellular processes that control crucial mechanisms in tumor development. Moreover, the interrelation of PI3K signaling with mTOR, ERK, and RAS pathways will be discussed, exploiting the potential benefits of PI3K signaling inhibitors in clinical use.
Collapse
Affiliation(s)
- Miriam Martini
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin , Italy
| | | | | | | | | |
Collapse
|
37
|
Konda K, Konishi K, Yamochi T, Ito YM, Nozawa H, Tojo M, Shinmura K, Kogo M, Katagiri A, Kubota Y, Muramoto T, Yano Y, Kobayashi Y, Kihara T, Tagawa T, Makino R, Takimoto M, Imawari M, Yoshida H. Distinct molecular features of different macroscopic subtypes of colorectal neoplasms. PLoS One 2014; 9:e103822. [PMID: 25093594 PMCID: PMC4122357 DOI: 10.1371/journal.pone.0103822] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 07/01/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Colorectal adenoma develops into cancer with the accumulation of genetic and epigenetic changes. We studied the underlying molecular and clinicopathological features to better understand the heterogeneity of colorectal neoplasms (CRNs). METHODS We evaluated both genetic (mutations of KRAS, BRAF, TP53, and PIK3CA, and microsatellite instability [MSI]) and epigenetic (methylation status of nine genes or sequences, including the CpG island methylator phenotype [CIMP] markers) alterations in 158 CRNs including 56 polypoid neoplasms (PNs), 25 granular type laterally spreading tumors (LST-Gs), 48 non-granular type LSTs (LST-NGs), 19 depressed neoplasms (DNs) and 10 small flat-elevated neoplasms (S-FNs) on the basis of macroscopic appearance. RESULTS S-FNs showed few molecular changes except SFRP1 methylation. Significant differences in the frequency of KRAS mutations were observed among subtypes (68% for LST-Gs, 36% for PNs, 16% for DNs and 6% for LST-NGs) (P<0.001). By contrast, the frequency of TP53 mutation was higher in DNs than PNs or LST-Gs (32% vs. 5% or 0%, respectively) (P<0.007). We also observed significant differences in the frequency of CIMP between LST-Gs and LST-NGs or PNs (32% vs. 6% or 5%, respectively) (P<0.005). Moreover, the methylation level of LINE-1 was significantly lower in DNs or LST-Gs than in PNs (58.3% or 60.5% vs. 63.2%, P<0.05). PIK3CA mutations were detected only in LSTs. Finally, multivariate analyses showed that macroscopic morphologies were significantly associated with an increased risk of molecular changes (PN or LST-G for KRAS mutation, odds ratio [OR] 9.11; LST-NG or DN for TP53 mutation, OR 5.30; LST-G for PIK3CA mutation, OR 26.53; LST-G or DN for LINE-1 hypomethylation, OR 3.41). CONCLUSION We demonstrated that CRNs could be classified into five macroscopic subtypes according to clinicopathological and molecular differences, suggesting that different mechanisms are involved in the pathogenesis of colorectal tumorigenesis.
Collapse
Affiliation(s)
- Kenichi Konda
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kazuo Konishi
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- * E-mail:
| | - Toshiko Yamochi
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Yoichi M. Ito
- Department of Biostatistics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hisako Nozawa
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Tojo
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kensuke Shinmura
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Mari Kogo
- Department of Hospital Pharmaceutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Atsushi Katagiri
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yutaro Kubota
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takashi Muramoto
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yuichiro Yano
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yoshiya Kobayashi
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Toshihiro Kihara
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Teppei Tagawa
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Reiko Makino
- Clinical Collaborating laboratory, Showa University School of Medicine, Tokyo, Japan
| | - Masafumi Takimoto
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Michio Imawari
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hitoshi Yoshida
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
38
|
Stintzing S, Lenz HJ. A small cog in a big wheel: PIK3CA mutations in colorectal cancer. J Natl Cancer Inst 2013; 105:1775-6. [PMID: 24231451 DOI: 10.1093/jnci/djt330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Sebastian Stintzing
- Affiliation of authors: USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA (SS, H-JL)
| | | |
Collapse
|