51
|
Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet 2019; 394:1467-1480. [PMID: 31631858 DOI: 10.1016/s0140-6736(19)32319-0] [Citation(s) in RCA: 2907] [Impact Index Per Article: 484.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 07/19/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
Several decades ago, colorectal cancer was infrequently diagnosed. Nowadays, it is the world's fourth most deadly cancer with almost 900 000 deaths annually. Besides an ageing population and dietary habits of high-income countries, unfavourable risk factors such as obesity, lack of physical exercise, and smoking increase the risk of colorectal cancer. Advancements in pathophysiological understanding have increased the array of treatment options for local and advanced disease leading to individual treatment plans. Treatments include endoscopic and surgical local excision, downstaging preoperative radiotherapy and systemic therapy, extensive surgery for locoregional and metastatic disease, local ablative therapies for metastases, and palliative chemotherapy, targeted therapy, and immunotherapy. Although these new treatment options have doubled overall survival for advanced disease to 3 years, survival is still best for those with non-metastasised disease. As the disease only becomes symptomatic at an advanced stage, worldwide organised screening programmes are being implemented, which aim to increase early detection and reduce morbidity and mortality from colorectal cancer.
Collapse
Affiliation(s)
- Evelien Dekker
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands.
| | - Pieter J Tanis
- Department of Surgery, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
| | - Jasper L A Vleugels
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands; Department of Internal Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
| | - Pashtoon M Kasi
- Department of Medical Oncology, University of Iowa, Iowa City, IA, USA
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
52
|
Symonds LK, Cohen SA. Use of perioperative chemotherapy in colorectal cancer metastatic to the liver. Gastroenterol Rep (Oxf) 2019; 7:301-311. [PMID: 31687149 PMCID: PMC6821343 DOI: 10.1093/gastro/goz035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/06/2019] [Accepted: 07/18/2019] [Indexed: 12/22/2022] Open
Abstract
A curative-intent approach may improve survival in carefully selected patients with oligometastatic colorectal cancer. Aggressive treatments are most frequently administered to patients with isolated liver metastasis, though they may be judiciously considered for other sites of metastasis. To be considered for curative intent with surgery, patients must have disease that can be definitively treated while leaving a sufficient functional liver remnant. Neoadjuvant chemotherapy may be used for upfront resectable disease as a test of tumor biology and/or for upfront unresectable disease to increase the likelihood of resectability (so-called 'conversion' chemotherapy). While conversion chemotherapy in this setting aims to improve survival, the choice of a regimen remains a complex and highly individualized decision. In this review, we discuss the role of RAS status, primary site, sidedness, and other clinical features that affect chemotherapy treatment selection as well as key factors of patients that guide individualized patient-treatment recommendations for colorectal-cancer patients being considered for definitive treatment with metastasectomy.
Collapse
Affiliation(s)
- Lynn K Symonds
- Division of Oncology, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Stacey A Cohen
- Division of Oncology, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
53
|
Van Blarigan EL, Fuchs CS, Niedzwiecki D, Zhang S, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Giovannucci EL, Ng K, Meyerhardt JA. Association of Survival With Adherence to the American Cancer Society Nutrition and Physical Activity Guidelines for Cancer Survivors After Colon Cancer Diagnosis: The CALGB 89803/Alliance Trial. JAMA Oncol 2019; 4:783-790. [PMID: 29710284 DOI: 10.1001/jamaoncol.2018.0126] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Importance The American Cancer Society Nutrition and Physical Activity Guidelines for Cancer Survivors (ACS guidelines) include maintaining (1) a healthy body weight; (2) physical activity; and (3) a diet that includes vegetables, fruits, and whole grains. It is not known whether patients with colon cancer who follow these guidelines have improved survival. Objective To examine whether a lifestyle consistent with the ACS guidelines is associated with improved survival rates after colon cancer. Design, Setting, and Participants This prospective cohort study included 992 patients with stage III colon cancer who were enrolled in the CALGB 89803 randomized adjuvant chemotherapy trial from 1999 through 2001. Data for the present study were analyzed between November 2016 and December 2017. Exposures We assigned an ACS guidelines score for each included patient based on body mass index; physical activity; and intake of vegetables, fruits, whole grains, and red/processed meats (score range, 0-6, with higher score indicating healthier behaviors). Secondarily, we examined a score that also included alcohol intake in addition to the other factors (range, 0-8). Lifestyle was assessed during and 6 months after chemotherapy. Main Outcomes and Measures Hazard ratios (HRs) and 95% confidence intervals (CIs) for disease-free, recurrence-free, and overall survival. Results Of the 992 patients enrolled in the study, 430 (43%) were women, and the mean (SD) age was 59.6 (11.2) years (range, 21-85 years). Over a 7-year median follow-up, we observed 335 recurrences and 299 deaths (43 deaths without recurrence). Compared with patients with a 0 to 1 ACS guidelines score (n = 262; 26%), patients with a 5 to 6 score (n = 91; 9%) had a 42% lower risk of death during the study period (HR, 0.58; 95% CI, 0.34-0.99; P = .01 for trend) and improved disease-free survival (HR, 0.69; 95% CI, 0.45-1.06; P = .03 for trend). When alcohol consumption was included in the score, the adjusted HRs comparing patients with scores of 6 to 8 (n = 162; 16%) vs those with scores of 0 to 2 (187; 91%) were 0.49 for overall survival (95% CI, 0.32-0.76; P = .002 for trend), 0.58 for disease-free survival (95% CI, 0.40, 0.84; P = .01 for trend), and 0.64 for recurrence-free survival (95% CI, 0.44-0.94; P = .05 for trend). Conclusions and Relevance Having a healthy body weight, being physically active, and eating a diet rich in vegetables, fruits, and whole grains after diagnosis of stage III colon cancer was associated with a longer survival. Trial Registration clinicaltrials.gov Identifier: NCT00003835.
Collapse
Affiliation(s)
- Erin L Van Blarigan
- Department of Epidemiology and Biostatistics, University of California, San Francisco.,Department of Urology, University of California, San Francisco
| | - Charles S Fuchs
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Donna Niedzwiecki
- Alliance Statistics and Data Center, Duke University, Durham, North Carolina
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Rex B Mowat
- Toledo Community Hospital Oncology Program, Toledo, Ohio
| | - Renaud Whittom
- Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, 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 Consortium, Mission Hospitals Inc, Asheville, North Carolina
| | - Hedy Kindler
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| | - Alan Venook
- Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Program in Molecular Pathology Epidemiology (MPE), 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 Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
54
|
Merloni F, Ranallo N, Scortichini L, Giampieri R, Berardi R. Tailored therapy in patients treated with fluoropyrimidines: focus on the role of dihydropyrimidine dehydrogenase. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:787-802. [PMID: 35582578 PMCID: PMC8992529 DOI: 10.20517/cdr.2018.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/01/2019] [Accepted: 06/04/2019] [Indexed: 01/19/2023]
Abstract
Fluoropyrimidines are widely used in the treatment of solid tumors, mainly gastrointestinal, head and neck and breast cancer. Dihydropyrimidine dehydrogenase (DPD) is the rate-limiting enzyme for catabolism of 5-FU and it is encoded by DPYD gene. To date, many known polymorphisms cause DPD deficiency and subsequent increase of 5-FU toxicity. In addition, reduced inactivation of 5-FU could lead to increased 5-FU intracellular concentration and augmented efficacy of this drugs. Therefore DPD expression, particularly intratumoral, has been investigated as predictive and prognostic marker in 5-FU treated patients. There also seems to be a tendency to support the correlation between DPD expression and response/survival in patients treated with fluoropyrimidine even if definitive conclusions cannot be drawn considering that some studies are conflicting. Therefore, the debate on intratumoral DPD expression as a potential predictor and prognostic marker in patients treated with fluoropyrimidines is still open. Four DPD-polymorphisms are the most relevant for their frequency in population and clinical relevance. Many studies demonstrate that treating a carrier of one of these polymorphisms with a full dose of fluoropyrimidine can expose patient to a severe, even life-threatening, toxicity. Severe toxicity is reduced if this kind of patients received a dose-adjustment after being genotyped. CPIC (Clinical Pharmacogenetics Implementation Consortium) is an International Consortium creating guidelines for facilitating use of pharmacogenetic tests for patient care and helps clinicians ensuring a safer drug delivery to the patient. Using predictive DPD deficiency tests in patients receiving 5FU-based chemotherapy, in particular for colorectal cancer, has proven to be a cost-effective strategy.
Collapse
Affiliation(s)
- Filippo Merloni
- Scuola di Specializzazione in Oncologia, Università Politecnica delle Marche, Ancona 60121, Italy
| | - Nicoletta Ranallo
- Scuola di Specializzazione in Oncologia, Università Politecnica delle Marche, Ancona 60121, Italy
| | - Laura Scortichini
- Scuola di Specializzazione in Oncologia, Università Politecnica delle Marche, Ancona 60121, Italy
| | - Riccardo Giampieri
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti, Ancona 60126, Italy
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti, Ancona 60126, Italy
| |
Collapse
|
55
|
Gelibter AJ, Caponnetto S, Urbano F, Emiliani A, Scagnoli S, Sirgiovanni G, Napoli VM, Cortesi E. Adjuvant chemotherapy in resected colon cancer: When, how and how long? Surg Oncol 2019; 30:100-107. [PMID: 31500770 DOI: 10.1016/j.suronc.2019.06.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 02/11/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022]
Abstract
The benefit of adjuvant chemotherapy has been clearly established in the adjuvant setting for node-positive colon cancer. A number of trials in the adjuvant setting have analyzed the efficacy of multiple-agent combinations, including irinotecan, oxaliplatin, bevacizumab and cetuximab. Only oxaliplatin added to fluorouracil/capecitabine has been shown to be superior beyond a fluropyrimidine alone in the adjuvant setting. As such, standard treatment options include fluorouracil (FU) or capecitabine with or without oxaliplatin. However, oxaliplatin is associated with cumulative dose-dependent neurotoxicity, characterized by distal or perioral paresthesias or dysesthesias; for this reason, in this review we discuss the results of the International Duration Evaluation of Adjuvant Chemotherapy (IDEA) trial. The IDEA trail is the largest prospective clinical trial ever conducted in colorectal cancer, wherein patients were treated with either 3 months or 6 months of adjuvant chemotherapy. In the era of cancer gene expression-based subtyping, the Colorectal Cancer Subtyping Consortium has proposed a four-subgroup molecular classification system for colorectal cancer, consisting of CMS1 (immune), CMS2 (canonical), CMS3 (metabolic) and CMS4 (mesenchymal). In this review, we present and analyze the available data on efficacy and toxicity of the combination regimen approved for treatment of resected colon cancer, and discuss the questions of when, how and how long we need to treat such patients.
Collapse
Affiliation(s)
- Alain J Gelibter
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy.
| | - Salvatore Caponnetto
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy
| | - Federica Urbano
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy
| | - Alessandra Emiliani
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy
| | - Simone Scagnoli
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy
| | - Grazia Sirgiovanni
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy
| | - Valerio M Napoli
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy
| | - Enrico Cortesi
- Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Italy
| |
Collapse
|
56
|
Guan Y, Shen Y, Xu Y, Li C, Wang J, Gu W, Lian P, Huang D, Cai S, Zhang Z, Zhu J. An expansion study of genotype-driven weekly irinotecan and capecitabine in combination with neoadjuvant radiotherapy for locally advanced rectal cancer with UGT1A1 *1*1 genotype. Therap Adv Gastroenterol 2019; 12:1756284819852293. [PMID: 31217818 PMCID: PMC6557009 DOI: 10.1177/1756284819852293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/29/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND In our previous dose-escalation study, we uncovered the maximum tolerated dose (MTD) of weekly irinotecan was escalated to 80 mg/m2 and 65 mg/m2 for UDP glucuronosyltransferase family 1 member A1 (UGT1A1) *1*1 and *1*28 rectal cancer patients in neoadjuvant chemoradiotherapy (nCRT). This is an expansion study for *1*1 patients. METHODS Patients with clinical stage T3-4, N0-2 rectal cancer eligible for preoperative chemoradiotherapy were screened for the UGT1A1*28 genotype. A total of 52 patients with the *1*1 genotype were enrolled. Whole-pelvic intensity-modulated radiation therapy was given in 50 Gy/25 fractions. Concurrently, irinotecan of 80 mg/m2 and capecitabine of 625 mg/m2 twice daily from Monday to Friday were administered weekly. Primary endpoint was toxicities; secondary endpoints included pathological complete response (pCR), tumour-regression grading, treatment compliance, overall survival, local recurrence and disease-free survival. RESULTS All patients completed capecitabine-based radiotherapy as scheduled, and 42 (81%) patients completed more than three cycles of weekly irinotecan. Overall, grade 3/4 toxicities were observed in 20 cases, including 11 leucopenia, 10 neutropenia and 12 diarrhoea. Forty-three patients (83%) underwent a radical surgery, and 12 were evaluated as pCR. Another four patients accepted a watch-and-wait strategy because of clinical complete response (CCR). CONCLUSIONS Our data demonstrated manageable toxicities and an encouraging CCR rate for UGT1A1 *1*1 genotype in an enhanced neoadjuvant therapy. A phase III trial is ongoing to evaluate the value of irinotecan in neoadjuvant therapy (CinClare) [ClinicalTrials.gov identifier: NCT02605265].
Collapse
Affiliation(s)
- Yun Guan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
- Department of Neurosurgery, Fudan University, Shanghai, China
- Department of Cyberknife, Fudan University, Shanghai, China
| | - Yunzhu Shen
- Department of Oncology, Nanjing Medical University, Nanjing, China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
| | - Chao Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
| | - Jingwen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
| | - Weilie Gu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
| | - Peng Lian
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Fudan University, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Centre, No. 270, Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ji Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Centre, No. 270, Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
57
|
Taieb J, André T, Auclin E. Refining adjuvant therapy for non-metastatic colon cancer, new standards and perspectives. Cancer Treat Rev 2019; 75:1-11. [DOI: 10.1016/j.ctrv.2019.02.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
|
58
|
Rovers KP, Bakkers C, Simkens GAAM, Burger JWA, Nienhuijs SW, Creemers GJM, Thijs AMJ, Brandt-Kerkhof ARM, Madsen EVE, Ayez N, de Boer NL, van Meerten E, Tuynman JB, Kusters M, Sluiter NR, Verheul HMW, van der Vliet HJ, Wiezer MJ, Boerma D, Wassenaar ECE, Los M, Hunting CB, Aalbers AGJ, Kok NFM, Kuhlmann KFD, Boot H, Chalabi M, Kruijff S, Been LB, van Ginkel RJ, de Groot DJA, Fehrmann RSN, de Wilt JHW, Bremers AJA, de Reuver PR, Radema SA, Herbschleb KH, van Grevenstein WMU, Witkamp AJ, Koopman M, Haj Mohammad N, van Duyn EB, Mastboom WJB, Mekenkamp LJM, Nederend J, Lahaye MJ, Snaebjornsson P, Verhoef C, van Laarhoven HWM, Zwinderman AH, Bouma JM, Kranenburg O, van 't Erve I, Fijneman RJA, Dijkgraaf MGW, Hemmer PHJ, Punt CJA, Tanis PJ, de Hingh IHJT. Perioperative systemic therapy and cytoreductive surgery with HIPEC versus upfront cytoreductive surgery with HIPEC alone for isolated resectable colorectal peritoneal metastases: protocol of a multicentre, open-label, parallel-group, phase II-III, randomised, superiority study (CAIRO6). BMC Cancer 2019; 19:390. [PMID: 31023318 PMCID: PMC6485075 DOI: 10.1186/s12885-019-5545-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/28/2019] [Indexed: 02/08/2023] Open
Abstract
Background Upfront cytoreductive surgery with HIPEC (CRS-HIPEC) is the standard treatment for isolated resectable colorectal peritoneal metastases (PM) in the Netherlands. This study investigates whether addition of perioperative systemic therapy to CRS-HIPEC improves oncological outcomes. Methods This open-label, parallel-group, phase II-III, randomised, superiority study is performed in nine Dutch tertiary referral centres. Eligible patients are adults who have a good performance status, histologically or cytologically proven resectable PM of a colorectal adenocarcinoma, no systemic colorectal metastases, no systemic therapy for colorectal cancer within six months prior to enrolment, and no previous CRS-HIPEC. Eligible patients are randomised (1:1) to perioperative systemic therapy and CRS-HIPEC (experimental arm) or upfront CRS-HIPEC alone (control arm) by using central randomisation software with minimisation stratified by a peritoneal cancer index of 0–10 or 11–20, metachronous or synchronous PM, previous systemic therapy for colorectal cancer, and HIPEC with oxaliplatin or mitomycin C. At the treating physician’s discretion, perioperative systemic therapy consists of either four 3-weekly neoadjuvant and adjuvant cycles of capecitabine with oxaliplatin (CAPOX), six 2-weekly neoadjuvant and adjuvant cycles of 5-fluorouracil/leucovorin with oxaliplatin (FOLFOX), or six 2-weekly neoadjuvant cycles of 5-fluorouracil/leucovorin with irinotecan (FOLFIRI) followed by four 3-weekly (capecitabine) or six 2-weekly (5-fluorouracil/leucovorin) adjuvant cycles of fluoropyrimidine monotherapy. Bevacizumab is added to the first three (CAPOX) or four (FOLFOX/FOLFIRI) neoadjuvant cycles. The first 80 patients are enrolled in a phase II study to explore the feasibility of accrual and the feasibility, safety, and tolerance of perioperative systemic therapy. If predefined criteria of feasibility and safety are met, the study continues as a phase III study with 3-year overall survival as primary endpoint. A total of 358 patients is needed to detect the hypothesised 15% increase in 3-year overall survival (control arm 50%; experimental arm 65%). Secondary endpoints are surgical characteristics, major postoperative morbidity, progression-free survival, disease-free survival, health-related quality of life, costs, major systemic therapy related toxicity, and objective radiological and histopathological response rates. Discussion This is the first randomised study that prospectively compares oncological outcomes of perioperative systemic therapy and CRS-HIPEC with upfront CRS-HIPEC alone for isolated resectable colorectal PM. Trial registration Clinicaltrials.gov/NCT02758951, NTR/NTR6301, ISRCTN/ISRCTN15977568, EudraCT/2016–001865-99.
Collapse
Affiliation(s)
- Koen P Rovers
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Checca Bakkers
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Geert A A M Simkens
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Jacobus W A Burger
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Simon W Nienhuijs
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Geert-Jan M Creemers
- Department of Medical Oncology, Catharina Hospital, PO Box 1350, 5602, Eindhoven, ZA, Netherlands
| | - Anna M J Thijs
- Department of Medical Oncology, Catharina Hospital, PO Box 1350, 5602, Eindhoven, ZA, Netherlands
| | | | - Eva V E Madsen
- Department of Surgical Oncology, Erasmus Medical Centre, PO Box 2040, 3000, Rotterdam, CA, Netherlands
| | - Ninos Ayez
- Department of Surgical Oncology, Erasmus Medical Centre, PO Box 2040, 3000, Rotterdam, CA, Netherlands
| | - Nadine L de Boer
- Department of Surgical Oncology, Erasmus Medical Centre, PO Box 2040, 3000, Rotterdam, CA, Netherlands
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000, Rotterdam, CA, Netherlands
| | - Jurriaan B Tuynman
- Department of Surgery, Amsterdam University Medical Centres, Location VUMC, PO Box 7057, 1007, Amsterdam, MB, Netherlands
| | - Miranda Kusters
- Department of Surgery, Amsterdam University Medical Centres, Location VUMC, PO Box 7057, 1007, Amsterdam, MB, Netherlands
| | - Nina R Sluiter
- Department of Surgery, Amsterdam University Medical Centres, Location VUMC, PO Box 7057, 1007, Amsterdam, MB, Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Amsterdam University Medical Centres, Location VUMC, PO Box 7057, 1007, Amsterdam, MB, Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, Amsterdam University Medical Centres, Location VUMC, PO Box 7057, 1007, Amsterdam, MB, Netherlands
| | - Marinus J Wiezer
- Department of Surgery, St. Antonius Hospital, PO Box 2500, 3430, Nieuwegein, EM, Netherlands
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, PO Box 2500, 3430, Nieuwegein, EM, Netherlands
| | - Emma C E Wassenaar
- Department of Surgery, St. Antonius Hospital, PO Box 2500, 3430, Nieuwegein, EM, Netherlands
| | - Maartje Los
- Department of Medical Oncology, St. Antonius Hospital, PO Box 2500, 3430, Nieuwegein, EM, Netherlands
| | - Cornelis B Hunting
- Department of Medical Oncology, St. Antonius Hospital, PO Box 2500, 3430, Nieuwegein, EM, Netherlands
| | - Arend G J Aalbers
- Department of Surgical Oncology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Niels F M Kok
- Department of Surgical Oncology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Koert F D Kuhlmann
- Department of Surgical Oncology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Henk Boot
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Myriam Chalabi
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Schelto Kruijff
- Department of Surgery, University Medical Centre Groningen, PO Box 30001, 9700 RB, Groningen, RB, Netherlands
| | - Lukas B Been
- Department of Surgery, University Medical Centre Groningen, PO Box 30001, 9700 RB, Groningen, RB, Netherlands
| | - Robert J van Ginkel
- Department of Surgery, University Medical Centre Groningen, PO Box 30001, 9700 RB, Groningen, RB, Netherlands
| | - Derk Jan A de Groot
- Department of Medical Oncology, University Medical Centre Groningen, PO Box 30001, 9700, Groningen, RB, Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Centre Groningen, PO Box 30001, 9700, Groningen, RB, Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Centre, PO Box 9101, 6500, Nijmegen, HB, Netherlands
| | - Andreas J A Bremers
- Department of Surgery, Radboud University Medical Centre, PO Box 9101, 6500, Nijmegen, HB, Netherlands
| | - Philip R de Reuver
- Department of Surgery, Radboud University Medical Centre, PO Box 9101, 6500, Nijmegen, HB, Netherlands
| | - Sandra A Radema
- Department of Medical Oncology, Radboud University Medical Centre, PO Box 9101, 6500, Nijmegen, HB, Netherlands
| | - Karin H Herbschleb
- Department of Medical Oncology, Radboud University Medical Centre, PO Box 9101, 6500, Nijmegen, HB, Netherlands
| | | | - Arjen J Witkamp
- Department of Surgery, University Medical Centre Utrecht, PO Box 85500, 3508, Utrecht, GA, Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Centre Utrecht, PO Box 85500, 3508, Utrecht, GA, Netherlands
| | - Nadia Haj Mohammad
- Department of Medical Oncology, University Medical Centre Utrecht, PO Box 85500, 3508, Utrecht, GA, Netherlands
| | - Eino B van Duyn
- Department of Surgery, Medisch Spectrum Twente, PO Box 50000, 7500, Enschede, KA, Netherlands
| | - Walter J B Mastboom
- Department of Surgery, Medisch Spectrum Twente, PO Box 50000, 7500, Enschede, KA, Netherlands
| | - Leonie J M Mekenkamp
- Department of Medical Oncology, Medisch Spectrum Twente, PO Box 50000, 7500, Enschede, KA, Netherlands
| | - Joost Nederend
- Department of Radiology, Catharina Hospital, PO Box 1350, 5602, Eindhoven, ZA, Netherlands
| | - Max J Lahaye
- Department of Radiology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus Medical Centre, PO Box 2040, 3000, Rotterdam, CA, Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Amsterdam University Medical Centres, Location AMC, PO Box 22660, 1100, Amsterdam, DD, Netherlands
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Biostatistics & Bioinformatics, Amsterdam University Medical Centres, Location AMC, PO Box 22660, 1100, Amsterdam, DD, Netherlands
| | - Jeanette M Bouma
- Clinical Trial Department, Netherlands Comprehensive Cancer Organisation (IKNL), PO Box 19079, 3501, Utrecht, DB, Netherlands
| | - Onno Kranenburg
- UMC Utrecht Cancer Centre, University Medical Centre Utrecht, PO Box 85500, 3508, Utrecht, GA, Netherlands
| | - Iris van 't Erve
- Department of Pathology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Remond J A Fijneman
- Department of Pathology, Netherlands Cancer Institute, PO Box 90203, 1006, Amsterdam, BE, Netherlands
| | - Marcel G W Dijkgraaf
- Department of Clinical Epidemiology, Biostatistics & Bioinformatics, Amsterdam University Medical Centres, Location AMC, PO Box 22660, 1100, Amsterdam, DD, Netherlands
| | - Patrick H J Hemmer
- Department of Surgery, University Medical Centre Groningen, PO Box 30001, 9700 RB, Groningen, RB, Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Amsterdam University Medical Centres, Location AMC, PO Box 22660, 1100, Amsterdam, DD, Netherlands
| | - Pieter J Tanis
- Department of Surgery, Amsterdam University Medical Centres, Location AMC, PO Box 22660, 1100, Amsterdam, DD, Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, Netherlands.
| | | | | |
Collapse
|
59
|
Paschke S, Hebart H, Goeb R, Staib L, Fleck U, Henne-Bruns D, Sander S, Link KH, Kornmann M. Adjuvant Chemotherapy of Locally Advanced Colon Cancer: Final Results of a Randomized Trial Comparing 5-Fluorouracil and Folinic Acid with Folfiri. Visc Med 2019; 35:124-132. [PMID: 31192246 PMCID: PMC6514490 DOI: 10.1159/000491648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND There is still the need to optimize adjuvant treatment of colon cancer (CC). Standard adjuvant chemotherapy using 5-fluorouracil (FU) and folinic acid (FA) was compared with a combination including irinotecan (Folfiri). The aim of the present report was to analyze overall survival (OS) after long-term follow-up, to summarize final recurrence rates and toxicity data, and to identify possible clinical and pathological factors associated with prognosis. METHODS Patients (CC stage IIb and III) were randomized to a 6-month treatment with FUFA or Folfiri. The trial was closed after 275 of 588 planned patients, 269 of which were included in the intention-to-treat analysis. RESULTS 133 and 136 patients received FUFA and Folfiri, respectively. Adjuvant therapy was not completed for 16 FUFA (12.0%) and 44 Folfiri (32.4%) patients. Toxicities grade III and IV were observed in 17 (12.8%) patients treated with FUFA and in 50 (36.8%) patients treated with Folfiri. Recurrences occurred in 46 of 133 (34.6%) and in 47 of 136 (34.6%) patients who received FUFA and Folfiri, respectively. 5-year OS rates were 69.9% (95% confidence interval (CI): 61.2-77.1) for FUFA and 72.7% (95% CI: 63.9-79.8) for Folfiri. OS was associated with tumor grading (1 & 2 vs. 3), tumor sub-stage (II vs. IIIa vs. IIIb vs. IIIc), and tumor location (left vs. right colon). CONCLUSION Folfiri cannot be generally recommended for adjuvant chemotherapy of CC. Besides tumor grading and sub-staging, prognosis of CC may depend on tumor location. Left-sided tumors had a significantly better prognosis irrespective of treatment.
Collapse
Affiliation(s)
- Stephan Paschke
- Clinic of General and Visceral Surgery, University Hospital of Ulm, Ulm, Germany
| | - Holger Hebart
- Center for Internal Medicine, Stauferklinikum, Schwäbisch Gmünd, Germany
| | - Roland Goeb
- Department of General and Visceral Surgery, Katholisches Krankenhaus St. Johann Nepomuk, Erfurt, Deutschland
| | - Ludger Staib
- Department of General and Visceral Surgery, Städtisches Klinikum Esslingen, Esslingen, Germany
| | - Ullrich Fleck
- Department of General and Visceral Surgery, Krankenhaus Luckenwalde, Luckenwalde, Germany
| | - Doris Henne-Bruns
- Clinic of General and Visceral Surgery, University Hospital of Ulm, Ulm, Germany
| | - Silvia Sander
- Institute of Epidemiology and Medical Biometry, University Hospital of Ulm, Ulm, Germany
| | | | - Marko Kornmann
- Clinic of General and Visceral Surgery, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
60
|
Abstract
A 55-year-old man with no medical history presents to his primary care physician with fatigue and dark stools and is found to have anemia. He is referred for diagnostic colonoscopy and found to have an ulcerated mass in the ascending colon. Metastatic workup is negative. He is referred to a colorectal surgeon and undergoes an uneventful laparoscopic right hemicolectomy. Final pathology reveals a 4-cm ulcerated mass involving the muscularis propria with 3 positive lymph nodes out of 12 (T2N1bM0, stage IIIA). The patient arrives at his postoperative appointment with questions about chemotherapy.
Collapse
|
61
|
Buccafusca G, Proserpio I, Tralongo AC, Rametta Giuliano S, Tralongo P. Early colorectal cancer: diagnosis, treatment and survivorship care. Crit Rev Oncol Hematol 2019; 136:20-30. [PMID: 30878125 DOI: 10.1016/j.critrevonc.2019.01.023] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/29/2018] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
CRC is the third most commonly diagnosed malignancy and the fourth leading cause of cancer-related death in the world. With advances in treatment, colorectal cancer is being transformed from a deadly disease to an illness that is increasingly curable. With this transformation has come increased interest in the unique problems, risks, needs, and concerns of survivors who have completed treatment and are cancer-free. They often suffer late/long-term side effects of therapies that may compromise their QoL such as fatigue, sleep difficulty, fear of recurrence, anxiety, depression, negative body image, sensory neuropathy, gastrointestinal problems, urinary incontinence, and sexual dysfunction. In this review, we discuss what is known about early colorectal diagnosis, staging, treatments and their long-term effects on quality of life and survivorship care.
Collapse
Affiliation(s)
- Gabriella Buccafusca
- UOC Oncologia Medica, Ospedale Umberto I, Via Giuseppe Testaferrata 1, 96100, Siracusa, Italy
| | - Ilaria Proserpio
- UOC Oncologia Medica, ASST Settelaghi, Ospedale di Circolo e Fondazione Macchi, Via Francesco Guicciardini 9, 21100, Varese, Italy
| | - Antonino Carmelo Tralongo
- UOC Oncologia Medica, ASST Settelaghi, Ospedale di Circolo e Fondazione Macchi, Via Francesco Guicciardini 9, 21100, Varese, Italy
| | | | - Paolo Tralongo
- UOC Oncologia Medica, Ospedale Umberto I, Via Giuseppe Testaferrata 1, 96100, Siracusa, Italy.
| |
Collapse
|
62
|
Burotto M, Wilkerson J, Stein WD, Bates SE, Fojo T. Adjuvant and neoadjuvant cancer therapies: A historical review and a rational approach to understand outcomes. Semin Oncol 2019; 46:83-99. [DOI: 10.1053/j.seminoncol.2019.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
|
63
|
Azrad M, Demark-Wahnefried W. Too Early to Worry About Blueberries. J Natl Cancer Inst 2019; 111:103-104. [DOI: 10.1093/jnci/djy101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 04/30/2018] [Indexed: 11/14/2022] Open
Affiliation(s)
- Maria Azrad
- Department of Human Nutrition, University of Alabama (UA), Tuscaloosa, AL
| | | |
Collapse
|
64
|
Paulík A, Nekvindová J, Filip S. Irinotecan toxicity during treatment of metastatic colorectal cancer: focus on pharmacogenomics and personalized medicine. TUMORI JOURNAL 2018; 106:87-94. [PMID: 30514181 DOI: 10.1177/0300891618811283] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Colorectal cancer, one of the most frequent types of cancer worldwide, has a high mortality rate. Irinotecan (CPT-11) has been approved for the treatment of advanced or metastatic disease either as a single agent or, more commonly, as part of combined chemotherapeutic regimens. Treatment with irinotecan is often accompanied by severe toxicity (e.g. neutropenia and diarrhea) that can result in treatment interruption or cessation, thus jeopardizing the patient's prognosis and quality of life. Irinotecan is bioactivated into its metabolite SN-38, which is subsequently detoxified by uridine diphosphate-glucuronosyl transferases (mainly UGT1A1). Further, ABC transporters (i.e. ABCB1, ABCC1-ABCC6, and ABCG2) are responsible for drug efflux into bile and urine whereas OATP transporters (SLCO1B1) enable its influx from blood into hepatocytes. Genetic polymorphisms in these enzymes/pumps may result in increased systemic SN-38 level, directly correlating with toxicity. Contemporary research is focused on the clinical implementation of genetic screenings for validated gene variations prior to treatment onset, allowing tailored individual doses or treatment regimens.
Collapse
Affiliation(s)
- Adam Paulík
- Charles University, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové, Department of Oncology and Radiotherapy, Czech Republic
| | - Jana Nekvindová
- University Hospital Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Czech Republic
| | - Stanislav Filip
- Charles University, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové, Department of Oncology and Radiotherapy, Czech Republic
| |
Collapse
|
65
|
Dahdaleh FS, Sherman SK, Poli EC, Vigneswaran J, Polite BN, Sharma MR, Catenacci DV, Maron SB, Turaga KK. Obstruction predicts worse long-term outcomes in stage III colon cancer: A secondary analysis of the N0147 trial. Surgery 2018; 164:1223-1229. [DOI: 10.1016/j.surg.2018.06.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022]
|
66
|
Lunenburg CATC, Henricks LM, van Kuilenburg ABP, Mathijssen RHJ, Schellens JHM, Gelderblom H, Guchelaar HJ, Swen JJ. Diagnostic and Therapeutic Strategies for Fluoropyrimidine Treatment of Patients Carrying Multiple DPYD Variants. Genes (Basel) 2018; 9:E585. [PMID: 30487465 PMCID: PMC6316498 DOI: 10.3390/genes9120585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/16/2023] Open
Abstract
DPYD genotyping prior to fluoropyrimidine treatment is increasingly implemented in clinical care. Without phasing information (i.e., allelic location of variants), current genotype-based dosing guidelines cannot be applied to patients carrying multiple DPYD variants. The primary aim of this study is to examine diagnostic and therapeutic strategies for fluoropyrimidine treatment of patients carrying multiple DPYD variants. A case series of patients carrying multiple DPYD variants is presented. Different genotyping techniques were used to determine phasing information. Phenotyping was performed by dihydropyrimidine dehydrogenase (DPD) enzyme activity measurements. Publicly available databases were queried to explore the frequency and phasing of variants of patients carrying multiple DPYD variants. Four out of seven patients carrying multiple DPYD variants received a full dose of fluoropyrimidines and experienced severe toxicity. Phasing information could be retrieved for four patients. In three patients, variants were located on two different alleles, i.e., in trans. Recommended dose reductions based on the phased genotype differed from the phenotype-derived dose reductions in three out of four cases. Data from publicly available databases show that the frequency of patients carrying multiple DPYD variants is low (< 0.2%), but higher than the frequency of the commonly tested DPYD*13 variant (0.1%). Patients carrying multiple DPYD variants are at high risk of developing severe toxicity. Additional analyses are required to determine the correct dose of fluoropyrimidine treatment. In patients carrying multiple DPYD variants, we recommend that a DPD phenotyping assay be carried out to determine a safe starting dose.
Collapse
Affiliation(s)
- Carin A T C Lunenburg
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Linda M Henricks
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
| | - André B P van Kuilenburg
- Department of Clinical Chemistry, Amsterdam University Medical Centre, 1105 AZ Amsterdam, The Netherlands.
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands.
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
67
|
Dasari A, Grothey A, Kopetz S. Circulating Tumor DNA-Defined Minimal Residual Disease in Solid Tumors: Opportunities to Accelerate the Development of Adjuvant Therapies. J Clin Oncol 2018; 36:JCO2018789032. [PMID: 30376428 PMCID: PMC6286158 DOI: 10.1200/jco.2018.78.9032] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Arvind Dasari
- Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Axel Grothey, The University of Tennessee West Cancer Center, Memphis, TN; and Scott Kopetz, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Axel Grothey
- Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Axel Grothey, The University of Tennessee West Cancer Center, Memphis, TN; and Scott Kopetz, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Scott Kopetz
- Arvind Dasari, The University of Texas MD Anderson Cancer Center, Houston, TX; Axel Grothey, The University of Tennessee West Cancer Center, Memphis, TN; and Scott Kopetz, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
68
|
Van Blarigan EL, Ou FS, Niedzwiecki D, Zhang S, Fuchs CS, Saltz L, Mayer RJ, Venook A, Ogino S, Song M, Benson A, Hantel A, Atkins JN, Giovannucci EL, Meyerhardt JA. Dietary Fat Intake after Colon Cancer Diagnosis in Relation to Cancer Recurrence and Survival: CALGB 89803 (Alliance). Cancer Epidemiol Biomarkers Prev 2018; 27:1227-1230. [PMID: 30038051 PMCID: PMC6170705 DOI: 10.1158/1055-9965.epi-18-0487] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/11/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022] Open
Abstract
Background: Higher intake of long-chain ω-3 polyunsaturated fatty acids and nuts, rich plant sources of unsaturated fats, after colon cancer diagnosis are associated with improved survival. It is not known whether the amount or the distribution of other types of fat is associated with survival after colon cancer.Methods: We prospectively examined postdiagnostic total, animal, and vegetable fats, as well as the saturated, monounsaturated, polyunsaturated, and trans fat in relation to disease-free survival among 1,011 patients with stage III colon cancer. Patients were enrolled between 1999 and 2001 at the onset of adjuvant chemotherapy and followed for recurrence or death through 2009.Results: During median follow-up of 7 years, we observed 305 deaths and 81 recurrences (total events: 386). Neither total nor any specific type of dietary fat examined was statistically significantly associated with risk of cancer recurrence or death from any cause (disease-free survival) after stage III colon cancer.Conclusions: The amount and type (animal, vegetable, saturated, monounsaturated, polyunsaturated, and trans) of dietary fat consumed after colon cancer does not appear to be substantially associated with risk of recurrence or survival.Impact: Neither total nor major types (animal, vegetable, saturated, monounsaturated, polyunsaturated, and trans) of dietary fat consumed after colon cancer was associated with cancer recurrence or survival. Cancer Epidemiol Biomarkers Prev; 27(10); 1227-30. ©2018 AACR.
Collapse
Affiliation(s)
- Erin L. Van Blarigan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA,Department of Urology, University of California, San Francisco, CA
| | - Fang-Shu Ou
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC
| | - Sui Zhang
- Dana-Farber/Partners CancerCare, Boston, MA
| | | | - Leonard Saltz
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Alan Venook
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA,Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Shuji Ogino
- Dana-Farber/Partners CancerCare, Boston, MA,Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Al Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | - James N Atkins
- Southeast Clinical Oncology Research (SCOR) Consortium NCORP, Winston-Salem, NC
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | | |
Collapse
|
69
|
Hishinuma E, Narita Y, Saito S, Maekawa M, Akai F, Nakanishi Y, Yasuda J, Nagasaki M, Yamamoto M, Yamaguchi H, Mano N, Hirasawa N, Hiratsuka M. Functional Characterization of 21 Allelic Variants of Dihydropyrimidine Dehydrogenase Identified in 1070 Japanese Individuals. Drug Metab Dispos 2018; 46:1083-1090. [PMID: 29769267 DOI: 10.1124/dmd.118.081737] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/11/2018] [Indexed: 11/22/2022] Open
Abstract
Dihydropyrimidine dehydrogenase (DPD, EC 1.3.1.2), encoded by the DPYD gene, is the rate-limiting enzyme in the degradation pathway of endogenous pyrimidine and fluoropyrimidine drugs such as 5-fluorouracil (5-FU). DPD catalyzes the reduction of uracil, thymine, and 5-FU. In Caucasians, DPYD mutations, including DPYD*2A, DPYD*13, c.2846A>T, and c.1129-5923C>G/hapB3, are known to contribute to interindividual variations in the toxicity of 5-FU; however, none of these DPYD polymorphisms has been identified in the Asian population. Recently, 21 DPYD allelic variants, including some novel single-nucleotide variants (SNVs), were identified in 1070 healthy Japanese individuals by analyzing their whole-genome sequences (WGSs), but the functional alterations caused by these variants remain unknown. In this study, in vitro analysis was performed on 22 DPD allelic variants by transiently expressing wild-type DPD and 21 DPD variants in 293FT cells and characterizing their enzymatic activities using 5-FU as a substrate. DPD expression levels and dimeric forms were determined using immunoblotting and blue-native PAGE, respectively. Additionally, the values of three kinetic parameters-the Michaelis constant (Km ), maximum velocity (Vmax ), and intrinsic clearance (CLint = Vmax/Km )-were determined for the reduction of 5-FU. Eleven variants exhibited significantly decreased intrinsic clearance compared with wild-type DPD. Moreover, the band patterns observed in the immunoblots of blue-native gels indicated that DPD dimerization is required for enzymatic activity in DPD. Thus, the detection of rare DPYD variants might facilitate severe adverse effect prediction of 5-FU-based chemotherapy in the Japanese population.
Collapse
Affiliation(s)
- Eiji Hishinuma
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Yoko Narita
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Sakae Saito
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masamitsu Maekawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Fumika Akai
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Yuya Nakanishi
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Jun Yasuda
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masao Nagasaki
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masayuki Yamamoto
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Hiroaki Yamaguchi
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Nariyasu Mano
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| | - Masahiro Hiratsuka
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences (E.H., Yo.N., F.A., Yu.N., N.H., M.H.), and Tohoku Medical Megabank Organization (S.S., J.Y., M.N., M.Y., M.H.), and Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan (M.M., H.Y., N.M., M.H.)
| |
Collapse
|
70
|
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
|
71
|
Cecchin E, De Mattia E, Ecca F, Toffoli G. Host genetic profiling to increase drug safety in colorectal cancer from discovery to implementation. Drug Resist Updat 2018; 39:18-40. [PMID: 30075835 DOI: 10.1016/j.drup.2018.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Adverse events affect the pharmacological treatment of approximately 90% of colorectal cancer (CRC) patients at any stage of the disease. Chemotherapy including fluoropyrimidines, irinotecan, and oxaliplatin is the cornerstone of the pharmacological treatment of CRC. The introduction of novel targeted agents, as anti-EGFR (i.e. cetuximab, panitumumab) and antiangiogenic (i.e. bevacizumab, ziv-aflibercept, regorafenib, and ramucirumab) molecules, into the oncologist's toolbox has led to significant improvements in the life expectancy of advanced CRC patients, but with a substantial increase in toxicity burden. In this respect, pharmacogenomics has largely been applied to the personalization of CRC chemotherapy, focusing mainly on the study of inhered polymorphisms in genes encoding phase I and II enzymes, ATP-binding cassette (ABC)/solute carrier (SLC) membrane transporters, proteins involved in DNA repair, folate pathway and immune response. These research efforts have led to the identification of some validated genetic markers of chemotherapy toxicity, for fluoropyrimidines and irinotecan. No validated genetic determinants of oxaliplatin-specific toxicity, as peripheral neuropathy, has thus far been established. The contribution of host genetic markers in predicting the toxicity associated with novel targeted agents' administration is still controversial due to the heterogeneity of published data. Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. However, these recommendations are still poorly applied at the patient's bedside. Several ongoing projects in the U.S. and Europe are currently evaluating how pharmacogenomics can be implemented successfully in daily clinical practice. The majority of drug-related adverse events are still unexplained, and a great deal of ongoing research is aimed at improving knowledge of the role of pharmacogenomics in increasing treatment safety. In this review, the issue of pre-treatment identification of CRC patients at risk of toxicity via the analysis of patients' genetic profiles is addressed. Available pharmacogenomics guidelines with ongoing efforts to implement them in clinical practice and new exploratory markers for clinical validation are described.
Collapse
Affiliation(s)
- Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Fabrizio Ecca
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy.
| |
Collapse
|
72
|
Fisch MJ, McNeill LH, Basen-Engquist KM. Helping Colorectal Cancer Survivors Benefit From Changing Lifestyle Behaviors: Implementation Science and Private Industry Collaboration to the Rescue. JAMA Oncol 2018; 4:777-778. [PMID: 29801068 DOI: 10.1001/jamaoncol.2018.0124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Michael J Fisch
- Medical Oncology and Genetics, AIM Specialty Health, Chicago, Illinois.,Department of General Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Lorna H McNeill
- Center for Community-Engaged Translational Research, Department of Health Disparities Research, University of Texas MD Anderson Cancer Center, Houston
| | - Karen M Basen-Engquist
- Department of Behavioral Science, Center for Energy Balance in Cancer Prevention and Survivorship, University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
73
|
Brown JC, Zhang S, Niedzwiecki D, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Li Y, Zhang X, Ng K, Willett WC, Giovannucci EL, Fuchs CS, Meyerhardt JA. Grain Intake and Clinical Outcome in Stage III Colon Cancer: Results From CALGB 89803 (Alliance). JNCI Cancer Spectr 2018; 2:pky017. [PMID: 29877501 PMCID: PMC5977856 DOI: 10.1093/jncics/pky017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 12/15/2022] Open
Abstract
Background Energy balance–related risk factors for colon cancer recurrence and mortality—type II diabetes, hyperinsulinemia, inflammation, and visceral obesity—are positively correlated with consumption of refined grains and negatively correlated with consumption of whole grains. We examined the relationship between the consumption of refined and whole grains with cancer recurrence and mortality in a cohort of patients with colon cancer. Methods We conducted a prospective observational study of 1024 patients with stage III colon cancer who participated in a randomized trial of postoperative chemotherapy. Patients reported consumption of refined and whole grains using a food frequency questionnaire during and six months after chemotherapy. The primary outcome was disease-free survival (DFS). Multivariable-adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using Cox regression models. All P values are two-sided. Results During a median follow-up of 7.3 years, 394 patients experienced a DFS event. The hazard ratio for DFS was 1.56 (95% CI = 1.09 to 2.24) for patients consuming three or more servings per day of refined grains compared with patients consuming less than one serving per day (Ptrend = .005). The hazard ratio for DFS was 0.89 (95% CI = 0.66 to 1.20) for patients consuming three or more servings per day of whole grains compared with patients consuming less than one serving per day (Ptrend = .54). The hazard ratio for DFS of substituting one serving per day of refined grain with one serving per day of whole grain was 0.87 (95% CI = 0.79 to 0.96, P = .007). Conclusions The choice of grain consumed may be associated with cancer recurrence and mortality. Future studies are necessary to confirm our findings and to inform the design of randomized trials.
Collapse
Affiliation(s)
| | - Sui Zhang
- Dana-Farber/Partners CancerCare, 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 (SCOR) Consortium, Mission Hospitals, Inc., Asheville, NC
| | - Hedy Kindler
- University of Chicago Comprehensive Cancer, Chicago, IL
| | - Alan Venook
- University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA
| | - Shuji Ogino
- Dana-Farber/Partners CancerCare, Boston, MA.,Harvard T.H. Chan School of Public Health, Boston, MA.,Brigham and Women's Hospital, Boston, MA
| | - Yanping Li
- Harvard T.H. Chan School of Public Health, Boston, MA
| | | | - Kimmie Ng
- Dana-Farber/Partners CancerCare, Boston, MA
| | | | - Edward L Giovannucci
- Harvard T.H. Chan School of Public Health, Boston, MA.,Brigham and Women's Hospital, Boston, MA
| | | | | |
Collapse
|
74
|
Traylor M, Walker JL, Corrigan AA, Hernandez MA, Newhouse SJ, Folarin AA, Patel H, Ross PJ, Sanderson JD, Spicer J, Prescott NJ, Mathew CG, Marinaki AM, Lewis CM. Exome array analysis of adverse reactions to fluoropyrimidine-based therapy for gastrointestinal cancer. PLoS One 2018; 13:e0188911. [PMID: 29715290 PMCID: PMC5929530 DOI: 10.1371/journal.pone.0188911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/15/2017] [Indexed: 12/31/2022] Open
Abstract
Fluoropyrimidines, including 5-fluororacil (5FU) and its pro-drug Capecitabine, are the common treatment for colorectal, breast, neck and head cancers—either as monotherapy or in combination therapy. Adverse reactions (ADRs) to the treatment are common and often result in treatment discontinuation or dose reduction. Factors contributing to ADRs, including genetic variation, are poorly characterized. We performed exome array analysis to identify genetic variants that contribute to adverse reactions. Our final dataset consisted of 504 European ancestry individuals undergoing fluoropyrimidine-based therapy for gastrointestinal cancer. A subset of 254 of these were treated with Capecitabine. All individuals were genotyped on the Illumina HumanExome Array. Firstly, we performed SNP and gene-level analyses of protein-altering variants on the array to identify novel associations the following ADRs, which were grouped into four phenotypes based on symptoms of diarrhea, mucositis, and neutropenia and hand-and-foot syndrome. Secondly, we performed detailed analyses of the HLA region on the same phenotypes after imputing the HLA alleles and amino acids. No protein-altering variants, or sets of protein-altering variants collapsed into genes, were associated with the main outcomes after Bonferroni correction. We found evidence that the HLA region was enriched for associations with Hand-and-Foot syndrome (p = 0.023), but no specific SNPs or HLA alleles were significant after Bonferroni correction. Larger studies will be required to characterize the genetic contribution to ADRs to 5FU. Future studies that focus on the HLA region are likely to be fruitful.
Collapse
Affiliation(s)
- Matthew Traylor
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- * E-mail:
| | - Jemma L. Walker
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Adele A. Corrigan
- Purine Research Laboratory, GSTS Pathology, Guy’s and St. Thomas’ Hospital NHS Foundation Trust, St. Thomas Hospital, London, United Kingdom
| | - Monica A. Hernandez
- Purine Research Laboratory, GSTS Pathology, Guy’s and St. Thomas’ Hospital NHS Foundation Trust, St. Thomas Hospital, London, United Kingdom
| | - Stephen J. Newhouse
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health at South London and Maudsley NHS Foundation Trust and (Institute of Psychiatry), King’s College London, London, United Kingdom
| | - Amos A. Folarin
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health at South London and Maudsley NHS Foundation Trust and (Institute of Psychiatry), King’s College London, London, United Kingdom
| | - Hamel Patel
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health at South London and Maudsley NHS Foundation Trust and (Institute of Psychiatry), King’s College London, London, United Kingdom
| | - Paul J. Ross
- Department of Gastroenterology, Guy’s and St. Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Jeremy D. Sanderson
- Department of Gastroenterology, Guy’s and St. Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
| | - James Spicer
- Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Natalie J. Prescott
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
| | - Christopher G. Mathew
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
| | - Anthony M. Marinaki
- Purine Research Laboratory, GSTS Pathology, Guy’s and St. Thomas’ Hospital NHS Foundation Trust, St. Thomas Hospital, London, United Kingdom
| | - Cathryn M. Lewis
- Department of Medical and Molecular Genetics, King’s College London, London, United Kingdom
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
75
|
Venook AP. Advances in Adjuvant Therapy for Colon Cancer: P value or Practical Value. J Clin Oncol 2018; 36:1461-1462. [PMID: 29620996 DOI: 10.1200/jco.2018.77.8423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Alan P Venook
- Alan P. Venook, University of California San Francisco, San Francisco, CA
| |
Collapse
|
76
|
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
|
77
|
Tang M, Price TJ, Shapiro J, Gibbs P, Haller DG, Arnold D, Peeters M, Segelov E, Roy A, Tebbutt N, Pavlakis N, Karapetis C, Burge M. Adjuvant therapy for resected colon cancer 2017, including the IDEA analysis. Expert Rev Anticancer Ther 2018; 18:339-349. [DOI: 10.1080/14737140.2018.1444481] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Monica Tang
- NHMRC Clinical Trials Centre, The University of Sydney, Camperdown, Australia
| | | | - Jeremy Shapiro
- Medical Oncology, Cabrini Hospital, Malvern, Australia
- Medical Oncology, Monash University, Melbourne, Australia
| | - Peter Gibbs
- Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Medical Oncology, Flinders University, Bedford Park, Australia
| | - Daniel G. Haller
- Abramson Cancer Center at the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dirk Arnold
- Medical Oncology, Asklepios Tumorzentrum Hamburg, Hamburg, Germany
| | - Marc Peeters
- Medical Oncology, University Hospital Antwerp, Oncology, Edegem, Belgium
| | - Eva Segelov
- Medical Oncology, Monash Medical Centre, Clayton, Australia
| | - Amitesh Roy
- Medical Oncology, Flinders Centre for Innovation in Cancer, Bedford Park, Australia
| | - Niall Tebbutt
- Medical Oncology, Austin Health, Heidelberg, Australia
| | - Nick Pavlakis
- Medical Oncology, Royal North Shore Hospital, St Leonards, Australia
| | - Chris Karapetis
- Medical Oncology, Flinders Medical Centre, Bedford Park, Australia
| | - Matthew Burge
- Medical Oncology, University of Queensland, Brisbane, Australia
| |
Collapse
|
78
|
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
|
79
|
Holmer C, Kreis ME. [Update on colon cancer 2017]. MMW Fortschr Med 2018; 159:54-62. [PMID: 29468509 DOI: 10.1007/s15006-017-9592-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Christoph Holmer
- Klinik für Allgemein-, Viszeral- und Gefäßchirurgie, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200, Berlin, Deutschland.
| | - Martin E Kreis
- Klinik für Allgemein-, Viszeral- und Gefäßchirurgie, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200, Berlin, Deutschland
| |
Collapse
|
80
|
Fuchs MA, Yuan C, Sato K, Niedzwiecki D, Ye X, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Innocenti F, Warren RS, Bertagnolli MM, Ogino S, Giovannucci EL, Horvath E, Meyerhardt JA, Ng K. Predicted vitamin D status and colon cancer recurrence and mortality in CALGB 89803 (Alliance). Ann Oncol 2018; 28:1359-1367. [PMID: 28327908 DOI: 10.1093/annonc/mdx109] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background Observational studies suggest that higher levels of 25-hydroxyvitamin D3 (25(OH)D) are associated with a reduced risk of colorectal cancer and improved survival of colorectal cancer patients. However, the influence of vitamin D status on cancer recurrence and survival of patients with stage III colon cancer is unknown. Patients and methods We prospectively examined the influence of post-diagnosis predicted plasma 25(OH)D on outcome among 1016 patients with stage III colon cancer who were enrolled in a National Cancer Institute-sponsored adjuvant therapy trial (CALGB 89803). Predicted 25(OH)D scores were computed using validated regression models. We examined the influence of predicted 25(OH)D scores on cancer recurrence and mortality (disease-free survival; DFS) using Cox proportional hazards. Results Patients in the highest quintile of predicted 25(OH)D score had an adjusted hazard ratio (HR) for colon cancer recurrence or mortality (DFS) of 0.62 (95% confidence interval [CI], 0.44-0.86), compared with those in the lowest quintile (Ptrend = 0.005). Higher predicted 25(OH)D score was also associated with a significant improvement in recurrence-free survival and overall survival (Ptrend = 0.01 and 0.0004, respectively). The benefit associated with higher predicted 25(OH)D score appeared consistent across predictors of cancer outcome and strata of molecular tumor characteristics, including microsatellite instability and KRAS, BRAF, PIK3CA, and TP53 mutation status. Conclusion Higher predicted 25(OH)D levels after a diagnosis of stage III colon cancer may be associated with decreased recurrence and improved survival. Clinical trials assessing the benefit of vitamin D supplementation in the adjuvant setting are warranted. ClinicalTrials.gov Identifier NCT00003835.
Collapse
Affiliation(s)
- M A Fuchs
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston
| | - C Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston
| | - K Sato
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston
| | - D Niedzwiecki
- Alliance Statistics and Data Center, Duke University Medical Center, Durham
| | - X Ye
- Alliance Statistics and Data Center, Duke University Medical Center, Durham
| | - L B Saltz
- Memorial Sloan-Kettering Cancer Center, New York
| | - R J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston
| | - R B Mowat
- Toledo Community Hospital Oncology Program, Toledo, USA
| | - R Whittom
- Hôpital du Sacré-Coeur de Montréal, Montreal, Canada
| | - A Hantel
- Edward Cancer Center, Naperville
| | - A Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago
| | - D Atienza
- Virginia Oncology Associates, Norfolk
| | - M Messino
- Southeast Cancer Control Consortium, Mission Hospitals-Memorial Campus, Asheville
| | | | - A Venook
- University of California at San Francisco Comprehensive Cancer Center, San Francisco
| | - F Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill
| | - R S Warren
- University of California at San Francisco Comprehensive Cancer Center, San Francisco
| | - M M Bertagnolli
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston.,Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston
| | - S Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston.,Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston
| | - E L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston
| | - E Horvath
- Alliance Protocol Operations Office, Chicago, USA
| | - J A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston
| | - K Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston
| |
Collapse
|
81
|
Radiation Therapy in Colon Carcinoma. Radiat Oncol 2018. [DOI: 10.1007/978-3-319-52619-5_46-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
82
|
Peng J, Zhang R, Zhao Y, Wu X, Chen G, Wan D, Lu Z, Pan Z. Prognostic value of preoperative prognostic nutritional index and its associations with systemic inflammatory response markers in patients with stage III colon cancer. CHINESE JOURNAL OF CANCER 2017; 36:96. [PMID: 29268783 PMCID: PMC5740941 DOI: 10.1186/s40880-017-0260-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/21/2017] [Indexed: 02/08/2023]
Abstract
Background The prognostic nutritional index (PNI) has been widely applied for predicting survival outcomes of patients with various malignant tumors. Although a low PNI predicts poor prognosis in patients with colorectal cancer after tumor resection, the prognostic value remains unknown in patients with stage III colon cancer undergoing curative tumor resection followed by adjuvant chemotherapy. This study aimed to investigate the prognostic value of PNI in patients with stage III colon cancer. Methods Medical records of 274 consecutive patients with stage III colon cancer undergoing curative tumor resection followed by adjuvant chemotherapy with oxaliplatin and capecitabine between December 2007 and December 2013 were reviewed. The optimal PNI cutoff value was determined using receiver operating characteristic (ROC) curve analysis. The associations of PNI with systemic inflammatory response markers, including lymphocyte-to-monocyte ratio (LMR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and C-reactive protein (CRP) level, and clinicopathologic characteristics were assessed using the Chi square or Fisher’s exact test. Correlation analysis was performed using Spearman’s correlation coefficient. Disease-free survival (DFS) and overall survival (OS) stratified by PNI were analyzed using Kaplan–Meier method and log-rank test, and prognostic factors were identified by Cox regression analyses. Results The preoperative PNI was positively correlated with LMR (r = 0.483, P < 0.001) and negatively correlated with NLR (r = − 0.441, P < 0.001), PLR (r = − 0.607, P < 0.001), and CRP level (r = − 0.333, P < 0.001). A low PNI (≤ 49.22) was significantly associated with short OS and DFS in patients with stage IIIC colon cancer but not in patients with stage IIIA/IIIB colon cancer. In addition, patients with a low PNI achieved a longer OS and DFS after being treated with 6–8 cycles of adjuvant chemotherapy than did those with < 6 cycles. Multivariate analyses revealed that PNI was independently associated with DFS (hazard ratios 2.001; 95% confidence interval 1.157–3.462; P = 0.013). Conclusion The present study identified preoperative PNI as a valuable predictor for survival outcomes in patients with stage III colon cancer receiving curative tumor resection followed by adjuvant chemotherapy.
Collapse
Affiliation(s)
- Jianhong Peng
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - Rongxin Zhang
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - Yixin Zhao
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - Xiaojun Wu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - Gong Chen
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - Desen Wan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - Zhenhai Lu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Zhizhong Pan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| |
Collapse
|
83
|
Zhu J, Li X, Shen Y, Guan Y, Gu W, Lian P, Sheng W, Cai S, Zhang Z. Genotype-driven phase I study of weekly irinotecan in combination with capecitabine-based neoadjuvant chemoradiation for locally advanced rectal cancer. Radiother Oncol 2017; 129:143-148. [PMID: 29273261 DOI: 10.1016/j.radonc.2017.11.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 10/26/2017] [Accepted: 11/13/2017] [Indexed: 11/26/2022]
Abstract
PURPOSE We aimed to identify the maximum tolerated dose (MTD) of weekly irinotecan in combination with capecitabine-based neoadjuvant chemoradiation according to the UGT1A1∗28 genotype in patients with locally advanced rectal cancer. PATIENTS AND METHODS Patients with clinical stage T3-4, N0-2 who were eligible for preoperative chemoradiotherapy were screened for the UGT1A1∗28 genotype. Twenty-six patients with either the ∗1∗1 or ∗1∗28 genotype were eligible for dose escalation of irinotecan, and patients with a ∗28∗28 genotype were excluded. The starting dose of weekly irinotecan was 50 mg/m2 for the two genotype groups, whereas the dose of capecitabine was fixed at 625 mg/m2. Intensity-modulated radiation therapy (IMRT) was applied to the whole pelvis (total dose of 50 Gy in 25 fractions). RESULTS The dose of weekly irinotecan was escalated to 95 mg/m2 in patients with the ∗1∗1 genotype and to 80 mg/m2 in those with the ∗1∗28 genotype. Dose-limiting toxicities (DLTs) were observed in 2/2 ∗1∗1 patients at 95 mg/m2 and 2/3 ∗1∗28 patients at 80 mg/m2. No DLT cases were observed among the three ∗1∗1 patients at 80 mg/m2, and one DLT case was observed among the six patients with ∗1∗28 at 65 mg/m2. Hence, 80 mg/m2 and 65 mg/m2 were the MTDs for the two groups. The most common grade 3 to 4 toxicities were neutropenia and diarrhea. CONCLUSION A higher dose of weekly irinotecan in combination with capecitabine-based CRT is feasible under the guidance of the UGT1A1∗28 genotype. Further clinical trials at these dose levels are warranted.
Collapse
Affiliation(s)
- Ji Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Yunzhu Shen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, China
| | - Yun Guan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Weilie Gu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Peng Lian
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, China; Department of Oncology, Shanghai Medical College, Fudan University, China.
| |
Collapse
|
84
|
Henricks LM, Opdam FL, Beijnen JH, Cats A, Schellens JHM. DPYD genotype-guided dose individualization to improve patient safety of fluoropyrimidine therapy: call for a drug label update. Ann Oncol 2017; 28:2915-2922. [PMID: 29045513 DOI: 10.1093/annonc/mdx411] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The fluoropyrimidine anticancer drugs, especially 5-fluorouracil (5-FU) and capecitabine, are frequently prescribed for several types of cancer, including breast, colorectal, head and neck and gastric cancer. In the current drug labels of 5-FU and capecitabine in the European Union and the United States, no adaptive dosing strategies are incorporated for polymorphic metabolism of 5-FU. Although treatment with fluoropyrimidines is generally well tolerated, a major clinical limitation is that a proportion of the treated population experiences severe, sometimes life-threatening, fluoropyrimidine-related toxicity. This toxicity is strongly affected by interindividual variability in activity of dihydropyrimidine dehydrogenase (DPD), the main metabolic enzyme for inactivation of fluoropyrimidines, with an estimated 3%-8% of the population being partially DPD deficient. A reduced functional or abrogated DPD enzyme is often caused by genetic polymorphisms in DPYD, the gene encoding for DPD, and heterozygous carriers of such DPYD polymorphisms have a partial DPD deficiency. When these partially DPD deficient patients are treated with a full dose of fluoropyrimidines, they are generally exposed to toxic levels of 5-FU and its metabolites, and the risk of developing severe treatment-related toxicity is therefore significantly increased.Currently, functional and clinical validity is well established for four DPYD variants (DPYD*2A, c.2846A>T, c.1679T>G and c.1236G>A), as those variants have retrospectively and in a large population study prospectively been shown to be associated with increased risk of fluoropyrimidine-associated toxicity. Patient safety of fluoropyrimidine treatment can be significantly improved by pre-emptive screening for DPYD genotype variants and dose reductions in heterozygous DPYD variant allele carriers, thereby normalizing 5-FU exposure. Based on the critical appraisal of currently available data, adjusting the labels of capecitabine and 5-FU by including recommendations on pre-emptive screening for DPYD variants and DPYD genotype-guided dose adjustments should be the new standard of care.
Collapse
Affiliation(s)
- L M Henricks
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology
| | - F L Opdam
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| | - A Cats
- Department of Gastroenterology and Hepatology, Division of Medical Oncology
| | - J H M Schellens
- Division of Pharmacology.,Department of Clinical Pharmacology, Division of Medical Oncology.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht
| |
Collapse
|
85
|
The American Society of Colon and Rectal Surgeons Clinical Practice Guidelines for the Treatment of Colon Cancer. Dis Colon Rectum 2017; 60:999-1017. [PMID: 28891842 DOI: 10.1097/dcr.0000000000000926] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The American Society of Colon and Rectal Surgeons is dedicated to ensuring high-quality patient care by advancing the science, prevention, and management of disorders and diseases of the colon, rectum, and anus. The Clinical Practice Guidelines Committee is composed of society members who are chosen because they have demonstrated expertise in the specialty of colon and rectal surgery. This committee was created to lead international efforts in defining quality care for conditions related to the colon, rectum, and anus. This is accompanied by developing Clinical Practice Guidelines based on the best available evidence. These guidelines are inclusive and not prescriptive. Their purpose is to provide information on which decisions can be made, rather than to dictate a specific form of treatment. These guidelines are intended for the use of all practitioners, health care workers, and patients who desire information about the management of the conditions addressed by the topics covered in these guidelines. It should be recognized that these guidelines should not be deemed inclusive of all proper methods of care or exclusive of methods of care reasonably directed to obtaining the same results. The ultimate judgment regarding the propriety of any specific procedure must be made by the physician in light of all the circumstances presented by the individual patient.
Collapse
|
86
|
Targeting tumor multicellular aggregation through IGPR-1 inhibits colon cancer growth and improves chemotherapy. Oncogenesis 2017; 6:e378. [PMID: 28920928 PMCID: PMC5623903 DOI: 10.1038/oncsis.2017.77] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/11/2022] Open
Abstract
Adhesion to extracellular matrix (ECM) is crucially important for survival of normal epithelial cells as detachment from ECM triggers specific apoptosis known as anoikis. As tumor cells lose the requirement for anchorage to ECM, they rely on cell-cell adhesion 'multicellular aggregation' for survival. Multicellular aggregation of tumor cells also significantly determines the sensitivity of tumor cells to the cytotoxic effects of chemotherapeutics. In this report, we demonstrate that expression of immunoglobulin containing and proline-rich receptor-1 (IGPR-1) is upregulated in human primary colon cancer. Our study demonstrates that IGPR-1 promotes tumor multicellular aggregation, and interfering with its adhesive function inhibits multicellular aggregation and, increases cell death. IGPR-1 supports colon carcinoma tumor xenograft growth in mouse, and inhibiting its activity by shRNA or blocking antibody inhibits tumor growth. More importantly, IGPR-1 regulates sensitivity of tumor cells to the chemotherapeutic agent, doxorubicin/adriamycin by a mechanism that involves doxorubicin-induced AKT activation and phosphorylation of IGPR-1 at Ser220. Our findings offer novel insight into IGPR-1's role in colorectal tumor growth, tumor chemosensitivity, and as a possible novel anti-cancer target.
Collapse
|
87
|
Nitsche U, Stöss C, Friess H. Effect of Adjuvant Chemotherapy on Elderly Colorectal Cancer Patients: Lack of Evidence. Gastrointest Tumors 2017; 4:11-19. [PMID: 29071260 DOI: 10.1159/000479318] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Adjuvant chemotherapy has become the standard form of treatment for all patients with stage III colorectal cancer and is also recommended for patients with stage II disease and defined risk factors. However, clinical studies that evaluate the effect of adjuvant treatment regimens have a selection bias in favor of younger patients, so that even retrospective subgroup analyses cannot define the best therapeutic procedure in elderly patients with comorbidities. SUMMARY As long as the role of adjuvant chemotherapy in elderly colorectal cancer patients is not investigated in comprehensive trials, no clear recommendations are possible. KEY MESSAGE An exploratory review of the relevant literature revealed that a formal meta-analysis concerning adjuvant chemotherapy in elderly patients with colorectal cancer is not feasible due to varying definitions of elderly patients, inclusion and exclusion criteria, and a plethora of chemotherapeutic regimens. PRACTICAL IMPLICATIONS Given the high incidence of colorectal cancer and the median age of 68 years for patients at the time of diagnosis, health economic considerations should promote randomized controlled trials regarding the role of adjuvant chemotherapy in the elderly.
Collapse
Affiliation(s)
- Ulrich Nitsche
- Klinik und Poliklinik für Chirurgie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Stöss
- Klinik und Poliklinik für Chirurgie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Helmut Friess
- Klinik und Poliklinik für Chirurgie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
88
|
Saleh K, Khalife-Saleh N, Kourie HR, Chahine G. How and when adjuvant treatment should be intensified in stage III colorectal cancers? Future Oncol 2017; 13:1999-2006. [PMID: 28829195 DOI: 10.2217/fon-2017-0197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The adjuvant chemotherapy (FOLFOX) represents the standard of care in stage III colon cancer with some exceptions in old patients. Adjuvant treatment must also be discussed in high-risk stage II colon cancer. However, 40-50% of patients develop disease recurrence after curative R0 surgical resection. The liver was the most common site of recurrence followed by peritoneum. Although adjuvant chemotherapy improved disease-free survival and overall survival, 5-year overall survival remains less than 55% in stage III colon cancer. Different strategies could be adopted to escalate the standard adjuvant chemotherapy in these patients going from aggressive intravenous chemotherapy, hepatic arterial infusion chemotherapy, hyperthermic intraperitoneal chemotherapy to adding targeted therapies or immunotherapies. We reported in this review the published and ongoing trials evaluating these treatment modalities in colon cancer.
Collapse
Affiliation(s)
- Khalil Saleh
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Nadine Khalife-Saleh
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | | | - Georges Chahine
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
89
|
Hishinuma E, Akai F, Narita Y, Maekawa M, Yamaguchi H, Mano N, Oda A, Hirasawa N, Hiratsuka M. Functional characterization of 21 allelic variants of dihydropyrimidinase. Biochem Pharmacol 2017. [PMID: 28642038 DOI: 10.1016/j.bcp.2017.06.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dihydropyrimidinase (DHP, EC 3.5.2.2), encoded by the gene DPYS, is the second enzyme in the catabolic pathway of pyrimidine and of fluoropyrimidine drugs such as 5-fluorouracil, which are commonly used in anticancer treatment; DHP catalyzes the hydrolytic ring opening of dihydrouracil and dihydro-5-fluorouracil. DPYS mutations are known to contribute to interindividual variations in the toxicity of fluoropyrimidine drugs, but the functional characterization of DHP allelic variants remains inadequate. In this study, in vitro analysis was performed on 22 allelic variants of DHP by transiently expressing wild-type DHP and 21 DHP variants in 293FT cells and characterizing their enzymatic activities by using dihydrouracil and dihydro-5-fluorouracil as substrates. DHP expression levels and oligomeric forms were determined using immunoblotting and blue native PAGE, respectively, and the stability of the DHP variants was assessed by examining the proteins in variant-transfected cells treated with cycloheximide or bortezomib. Moreover, three kinetic parameters, Km, Vmax, and intrinsic clearance (Vmax/Km), for the hydrolysis of dihydrouracil and dihydro-5-fluorouracil were determined. We found that 5/21 variants showed significantly decreased intrinsic clearance as compared to wild-type DHP, and that 9/21 variants were expressed at low levels and were inactive due to proteasome-mediated degradation. The band patterns observed in the immunoblotting of blue native gels corresponded to DHP activity, and, notably, 18/21 DHP variants exhibited decreased or null enzymatic activity and these variants also showed a drastically reduced ability to form large oligomers. Thus, detection of DPYS genetic polymorphisms might facilitate the prediction severe adverse effects of fluoropyrimidine-based treatments.
Collapse
Affiliation(s)
- Eiji Hishinuma
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Fumika Akai
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Yoko Narita
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Akifumi Oda
- Department of Biophysical Chemistry, Faculty of Pharmacy, Meijo University, Nagoya 468-8503, Japan
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masahiro Hiratsuka
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8575, Japan.
| |
Collapse
|
90
|
The current value of determining the mismatch repair status of colorectal cancer: A rationale for routine testing. Crit Rev Oncol Hematol 2017; 116:38-57. [PMID: 28693799 DOI: 10.1016/j.critrevonc.2017.05.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/02/2017] [Accepted: 05/15/2017] [Indexed: 02/08/2023] Open
Abstract
Colorectal Cancer (CRC) is the third most prevalent cancer in men and women. Up to 15% of CRCs display microsatellite instability (MSI). MSI is reflective of a deficient mismatch repair (MMR) system and is most commonly caused by hypermethylation of the MLH1 promoter. However, it may also be due to autosomal dominant constitutional mutations in DNA MMR, termed Lynch Syndrome. MSI may be diagnosed via polymerase chain reaction (PCR) or alternatively, immunohistochemistry (IHC) can identify MMR deficiency (dMMR). Many institutions now advocate universal tumor screening of CRC via either PCR for MSI or IHC for dMMR to guide Lynch Syndrome testing. The association of sporadic MSI with methylation of the MLH1 promoter and an activating BRAF mutation may offer further exclusion criteria for genetic testing. Aside from screening for Lynch syndrome, MMR testing is important because of its prognostic and therapeutic implications. Several studies have shown MSI CRCs exhibit different clinicopathological features and prognosis compared to microsatellite-stable (MSS) CRCs. For example, response to conventional chemotherapy has been reported to be less in MSI tumours. More recently, MSI tumours have been shown to be responsive to immune-checkpoint inhibition providing a novel therapeutic strategy. This provides a rationale for routine testing for MSI or dMMR in CRC.
Collapse
|
91
|
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
|
92
|
Zhang Q, Freidlin B, Korn EL, Halabi S, Mandrekar S, Dignam JJ. Comparison of futility monitoring guidelines using completed phase III oncology trials. Clin Trials 2017; 14:48-58. [PMID: 27590208 PMCID: PMC5300958 DOI: 10.1177/1740774516666502] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Futility (inefficacy) interim monitoring is an important component in the conduct of phase III clinical trials, especially in life-threatening diseases. Desirable futility monitoring guidelines allow timely stopping if the new therapy is harmful or if it is unlikely to demonstrate to be sufficiently effective if the trial were to continue to its final analysis. There are a number of analytical approaches that are used to construct futility monitoring boundaries. The most common approaches are based on conditional power, sequential testing of the alternative hypothesis, or sequential confidence intervals. The resulting futility boundaries vary considerably with respect to the level of evidence required for recommending stopping the study. PURPOSE We evaluate the performance of commonly used methods using event histories from completed phase III clinical trials of the Radiation Therapy Oncology Group, Cancer and Leukemia Group B, and North Central Cancer Treatment Group. METHODS We considered published superiority phase III trials with survival endpoints initiated after 1990. There are 52 studies available for this analysis from different disease sites. Total sample size and maximum number of events (statistical information) for each study were calculated using protocol-specified effect size, type I and type II error rates. In addition to the common futility approaches, we considered a recently proposed linear inefficacy boundary approach with an early harm look followed by several lack-of-efficacy analyses. For each futility approach, interim test statistics were generated for three schedules with different analysis frequency, and early stopping was recommended if the interim result crossed a futility stopping boundary. For trials not demonstrating superiority, the impact of each rule is summarized as savings on sample size, study duration, and information time scales. RESULTS For negative studies, our results show that the futility approaches based on testing the alternative hypothesis and repeated confidence interval rules yielded less savings (compared to the other two rules). These boundaries are too conservative, especially during the first half of the study (<50% of information). The conditional power rules are too aggressive during the second half of the study (>50% of information) and may stop a trial even when there is a clinically meaningful treatment effect. The linear inefficacy boundary with three or more interim analyses provided the best results. For positive studies, we demonstrated that none of the futility rules would have stopped the trials. CONCLUSION The linear inefficacy boundary futility approach is attractive from statistical, clinical, and logistical standpoints in clinical trials evaluating new anti-cancer agents.
Collapse
Affiliation(s)
- Qiang Zhang
- Statistics and Data Management Center, NRG Oncology, Philadelphia, PA, USA
- Department of Biostatistics and Epidemiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Boris Freidlin
- Biometric Research Program, National Cancer Institute, Bethesda, MD, USA
| | - Edward L Korn
- Biometric Research Program, National Cancer Institute, Bethesda, MD, USA
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27705, USA
| | - Sumithra Mandrekar
- Division of Biomedical Statistics and Informatics, Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - James J Dignam
- Statistics and Data Management Center, NRG Oncology, Philadelphia, PA, USA
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
93
|
Abstract
In 2015, ramucirumab and TAS-102 became the 10th and 11th drugs approved by the Food and Drug administration for the treatment of patients with colorectal cancer, not counting leucovorin, and yet only 3 agents, 5-fluorouracil, capecitabine, and oxaliplatin, have proven benefit in adjuvant treatment. In fact, there have been no additions (and 1 subtraction levamisole) to our arsenal of therapies for patients with stages II and III colon cancer for more than a decade. How did we get here? Are we stuck? And how do we move forward?
Collapse
|
94
|
Kumar S, Noel MS, Khorana AA. Advances in adjuvant therapy of colon cancer. SEMINARS IN COLON AND RECTAL SURGERY 2016. [DOI: 10.1053/j.scrs.2016.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
95
|
Kotani D, Kuboki Y, Yoshino T. Adjuvant Chemotherapy for Colon Cancer: Guidelines and Clinical Trials in Japan. CURRENT COLORECTAL CANCER REPORTS 2016. [DOI: 10.1007/s11888-016-0336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
96
|
Niedzwiecki D, Hasson RM, Lenz HJ, Ye C, Redston M, Ogino S, Fuchs CS, Compton CC, Mayer RJ, Goldberg RM, Colacchio TA, Saltz LB, Warren RS, Bertagnolli MM. A Study of Thymidylate Synthase Expression as a Biomarker for Resectable Colon Cancer: Alliance (Cancer and Leukemia Group B) 9581 and 89803. Oncologist 2016; 22:107-114. [PMID: 27821793 PMCID: PMC5313270 DOI: 10.1634/theoncologist.2016-0215] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/26/2016] [Indexed: 11/28/2022] Open
Abstract
Tumor thymidylate synthase (TS) levels were prospectively evaluated in two adjuvant therapy trials for patients with resected stage II or III colon cancer. Results indicated that high tumor TS levels were associated with improved disease‐free survival and overall survival following adjuvant therapy for colon cancer, although tumor TS expression did not predict benefit of 5‐fluorouracil‐based chemotherapy. Purpose. Tumor levels of thymidylate synthase (TS), a target of 5‐fluorouracil (5‐FU)‐based chemotherapy for colorectal cancer, have been studied as a predictive or prognostic biomarker with mixed results. Patients and Methods. Tumor TS levels were prospectively evaluated in two adjuvant therapy trials for patients with resected stage II or III colon cancer. TS expression was determined by standard immunohistochemistry and by automated quantitative analysis. Tumor mismatch repair deficiency (MMR‐D) and BRAF c.1799T > A (p.V600E) mutation status were also examined. Relationships between tumor TS, MMR‐D, and BRAF mutation status, overall survival (OS), and disease‐free survival (DFS) were investigated in the subset of stage III patients. Results. Patients whose tumors demonstrated high TS expression experienced better treatment outcomes, with DFS hazard ratio (HR) = 0.67, 95% confidence interval (CI) = 0.53, 0.84; and OS HR = 0.68, 95% CI = 0.53, 0.88, for high versus low TS expression, respectively. No significant interaction between TS expression and stage was observed (DFS: interaction HR = 0.94; OS: interaction HR = 0.94). Tumors with high TS expression were more likely to demonstrate MMR‐D (22.2% vs. 12.8%; p = .0003). Patients whose tumors demonstrated both high TS and MMR‐D had a 7‐year DFS of 77%, compared with 58% for those whose tumors had low TS and were non‐MMR‐D (log‐rank p = .0006). Tumor TS expression did not predict benefit of a particular therapeutic regimen. Conclusion. This large prospective analysis showed that high tumor TS levels were associated with improved DFS and OS following adjuvant therapy for colon cancer, although tumor TS expression did not predict benefit of 5‐FU‐based chemotherapy. Implications for Practice. This study finds that measurement of tumor levels of thymidylate synthase is not helpful in assigning specific adjuvant treatment for colorectal cancer. It also highlights the importance of using prospective analyses within treatment clinical trials as the optimal method of determining biomarker utility.
Collapse
Affiliation(s)
- Donna Niedzwiecki
- Alliance Statistics and Data Center and Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Rian M Hasson
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Heinz-Josef Lenz
- University of Southern California Norris Cancer Center, Los Angeles, California, USA
| | - Cynthia Ye
- Alliance Statistics and Data Center and Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Mark Redston
- Pathology, Department of Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Shuji Ogino
- Pathology, Department of Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Charles S Fuchs
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Thomas A Colacchio
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Leonard B Saltz
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert S Warren
- Department of Surgery, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
97
|
Kerr RS, Love S, Segelov E, Johnstone E, Falcon B, Hewett P, Weaver A, Church D, Scudder C, Pearson S, Julier P, Pezzella F, Tomlinson I, Domingo E, Kerr DJ. Adjuvant capecitabine plus bevacizumab versus capecitabine alone in patients with colorectal cancer (QUASAR 2): an open-label, randomised phase 3 trial. Lancet Oncol 2016; 17:1543-1557. [PMID: 27660192 DOI: 10.1016/s1470-2045(16)30172-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Antiangiogenic agents have established efficacy in the treatment of metastatic colorectal cancer. We investigated whether bevacizumab could improve disease-free survival in the adjuvant setting after resection of the primary tumour. METHODS For the open-label, randomised, controlled QUASAR 2 trial, which was done at 170 hospitals in seven countries, we recruited patients aged 18 years or older with WHO performance status scores of 0 or 1 who had undergone potentially curative surgery for histologically proven stage III or high-risk stage II colorectal cancer. Patients were randomly assigned (1:1) to receive eight 3-week cycles of oral capecitabine alone (1250 mg/m2 twice daily for 14 days followed by a break for 7 days) or the same regimen of oral capecitabine plus 16 cycles of 7·5 mg/kg bevacizumab by intravenous infusion over 90 min on day 1 of each cycle. Randomisation was done by a computer-generated schedule with use of minimisation with a random element stratified by age, disease stage, tumour site, and country. The study was open label and no-one was masked to treatment assignment. The primary endpoint was 3-year disease-free survival, assessed in the intention-to-treat population. Toxic effects were assessed in patients who received at least one dose of randomised treatment. This trial is registered with the ISRCTN registry, number ISRCTN45133151. FINDINGS Between April 25, 2005, and Oct 12, 2010, 1952 eligible patients were enrolled, of whom 1941 had assessable data (968 in the capecitabine alone group and 973 in the capecitabine and bevacizumab group). Median follow-up was 4·92 years (IQR 4·00-5·16). Disease-free survival at 3 years did not differ between the groups (75·4%, 95% CI 72·5-78·0 in the capecitabine and bevacizumab group vs 78·4%, 75·7-80·9 in the capecitabine alone group; hazard ratio 1·06, 95% CI 0·89-1·25, p=0·54). The most common grade 3-4 adverse events were hand-foot syndrome (201 [21%] of 963 in the capecitabine alone group vs 257 [27%] of 959 in the capecitabine and bevacizumab group) and diarrhoea (102 [11%] vs 104 [11%]), and, with the addition of bevacizumab, expected increases were recorded in all-grade hypertension (320 [33%] vs 75 [8%]), proteinuria (197 [21%] vs 49 [5%]), and wound healing problems (30 [3%] vs 17 [2%]). 571 serious adverse events were reported (221 with capecitabine alone and 350 with capecitabine and bevacizumab). Most of these were gastrointestinal (n=245) or cardiovascular (n=169). 23 deaths within 6 months of randomisation were classified as being related to treatment, eight in the capecitabine alone group and 15 in the capecitabine and bevacizumab group. INTERPRETATION The addition of bevacizumab to capecitabine in the adjuvant setting for colorectal cancer yielded no benefit in the treatment of an unselected population and should not be used. FUNDING Roche.
Collapse
Affiliation(s)
- Rachel S Kerr
- Department of Oncology, University of Oxford, Oxford, UK.
| | - Sharon Love
- Department of Oncology, University of Oxford, Oxford, UK
| | - Eva Segelov
- Department of Medicine, University of New South Wales, Sydney, NSW, Australia
| | | | | | - Peter Hewett
- Queen Elizabeth Hospital, Adelaide, SA, Australia
| | | | - David Church
- Department of Oncology, University of Oxford, Oxford, UK
| | - Claire Scudder
- Department of Oncology, University of Oxford, Oxford, UK
| | - Sarah Pearson
- Department of Oncology, University of Oxford, Oxford, UK
| | - Patrick Julier
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Ian Tomlinson
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Enric Domingo
- Department of Oncology, University of Oxford, Oxford, UK; Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - David J Kerr
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
98
|
Fontana E, Homicsko K, Eason K, Sadanandam A. Molecular Classification of Colon Cancer: Perspectives for Personalized Adjuvant Therapy. CURRENT COLORECTAL CANCER REPORTS 2016. [DOI: 10.1007/s11888-016-0341-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
99
|
Saltz LB. Bevacizumab in colorectal cancer: it should have worked. Lancet Oncol 2016; 17:1469-1470. [PMID: 27660191 DOI: 10.1016/s1470-2045(16)30213-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 01/30/2023]
Affiliation(s)
- Leonard B Saltz
- GastroIntestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
100
|
Loree JM, Cheung WY. Optimizing adjuvant therapy and survivorship care of stage III colon cancer. Future Oncol 2016; 12:2021-35. [DOI: 10.2217/fon-2016-0109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The MOSAIC trial demonstrated nearly a decade ago that the addition of oxaliplatin to 5-fluorouracil improves outcomes in the adjuvant treatment of colon cancer, but no new agents have been shown to be superior to standard FOLFOX therapy. Oncologists have refined the use of oxaliplatin containing regimens to optimize outcomes, improved patient selection for multi-agent chemotherapy and expanded survivorship care to meet the needs of the growing number of survivors. In this article, we review the historical contexts of current therapy, appropriate staging investigations, the importance of timely initiation of therapy and key survivorship issues. We also discuss exciting opportunities for change, including reduced duration of adjuvant chemotherapy and the use of circulating tumor cells and DNA in surveillance.
Collapse
Affiliation(s)
- Jonathan M Loree
- Division of Medical Oncology, University of British Columbia, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, British Columbia, V5Z 4E6, Canada
| | - Winson Y Cheung
- Division of Medical Oncology, University of British Columbia, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, British Columbia, V5Z 4E6, Canada
| |
Collapse
|