51
|
Belli C, Trapani D, Viale G, D'Amico P, Duso BA, Della Vigna P, Orsi F, Curigliano G. Targeting the microenvironment in solid tumors. Cancer Treat Rev 2018; 65:22-32. [PMID: 29502037 DOI: 10.1016/j.ctrv.2018.02.004] [Citation(s) in RCA: 350] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 02/06/2018] [Accepted: 02/09/2018] [Indexed: 01/06/2023]
Abstract
Tumorigenesis is a complex and dynamic process involving different cellular and non-cellular elements composed of tumor microenvironment (TME). The interaction of TME with cancer cells is responsible for tumor development, progression and drug resistance. TME consists of non malignant cells of the tumor such as cancer associated fibroblasts (CAFs), endothelial cells and pericytes composing tumor vasculature, immune and inflammatory cells, bone marrow derived cells, and the extracellular matrix (ECM) establishing a complex cross-talk with tumor. These interactions contribute towards proliferation and invasion of the tumor by producing growth factors, chemokines and matrix-degrading enzymes. ECM is a complex system containing macromolecules with distinctive physical, biochemical and biomechanical properties. During tumorigenesis this system is deregulated favoring the generation of tumorigenic microenvironment enhancing tumor-associated angiogenesis and inflammation. An important step of anticancer treatment is the identification of the biological alterations present in TME in order to target these key molecular players. Multitargeted approaches, providing a simultaneous inhibition of TME components, may offer a more efficient way to treat cancer. In this manuscript we overview the function of each components of TME and the treatments targeting the key players.
Collapse
Affiliation(s)
- Carmen Belli
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy.
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Paolo D'Amico
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Bruno Achutti Duso
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Paolo Della Vigna
- Interventional Radiology Division, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Franco Orsi
- Interventional Radiology Division, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
52
|
Wang J, Luo L, Wang D, Guo B, Li J, Yang Z, Tang D. Combination adjuvant chemotherapy with targeted drugs for treatment of colorectal cancer: A network meta-analysis. J Cell Biochem 2018; 119:1521-1537. [PMID: 28771807 DOI: 10.1002/jcb.26312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/02/2017] [Indexed: 01/10/2023]
Abstract
Colorectal cancer (CRC) is one of the most fatal diseases in the world. The efficacy of present chemotherapy treatments are limited and the addition of targeted drugs have been put into practice. However, the preferred treatments among adjuvant chemotherapies still remain controversial and uncertain. To evaluate the efficacy of different adjuvant chemotherapies combined with or without targeted drugs to determine the optimal treatment for patients with CRC in clinical practice. PubMed and Embase were searched for eligible articles and only randomized controlled trials (RCTs) were included. R (Version 3.2.5) software was utilized to conduct the Bayesian network meta-analysis (NMA). Outcomes including overall survival (OS) and progression-free survival (PFS) were displayed using hazard ratios. And the rank probabilities of each treatment were evaluated using the surface under cumulative ranking curve. A total of 75 RCTs published after 1997 were included in the data analysis. Overall, FOLFIRI+ cetuximab was found to be the most effective treatment in terms of long-term survival and FOLFOX was the most effective pure chemotherapy treatment. The addition of targeted drugs will greatly improve the efficacy of chemotherapy. Targeted drug cetuximab combined with the chemotherapy regiment FOLFIRI is the preferable treatment for patients with CRC in clinical practice.
Collapse
Affiliation(s)
- Jinghui Wang
- Department of Oncological Hematology, First Affiliated Hospital of Guiyang College of TCM, Guiyang, Guizhou, China
| | - Li Luo
- Department of Oncological Hematology, First Affiliated Hospital of Guiyang College of TCM, Guiyang, Guizhou, China
| | - Dingxue Wang
- Department of Oncological Hematology, First Affiliated Hospital of Guiyang College of TCM, Guiyang, Guizhou, China
| | - Bin Guo
- Graduate College of Guiyang College of TCM, Guiyang, Guizhou, China
| | - Jun Li
- College of Basic Medicine of Guiyang College of TCM, Guiyang, Guizhou, China
| | - Zhu Yang
- Deanery of Guiyang College of TCM, Guiyang, Guizhou, China
| | - Dongxin Tang
- Department of Science and Education, First Affiliated Hospital of Guiyang College of TCM, Guiyang, Guizhou, China
| |
Collapse
|
53
|
Xu RH, Shen L, Wang KM, Wu G, Shi CM, Ding KF, Lin LZ, Wang JW, Xiong JP, Wu CP, Li J, Liu YP, Wang D, Ba Y, Feng JP, Bai YX, Bi JW, Ma LW, Lei J, Yang Q, Yu H. Famitinib versus placebo in the treatment of refractory metastatic colorectal cancer: a multicenter, randomized, double-blinded, placebo-controlled, phase II clinical trial. CHINESE JOURNAL OF CANCER 2017; 36:97. [PMID: 29273089 PMCID: PMC5741870 DOI: 10.1186/s40880-017-0263-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/05/2017] [Indexed: 12/13/2022]
Abstract
Background Metastatic colorectal cancer (mCRC) patients with progressive disease after all available standard therapies need new medication for further treatment. Famitinib is a small-molecule multikinase inhibitor, with promising anticancer activities. This multicenter, randomized, double-blinded, placebo-controlled, phase II clinical trial was designed to evaluate the safety and efficacy of famitinib in mCRC. Methods Famitinib or placebo was administered orally once daily. The primary endpoint was progression-free survival (PFS). Secondary endpoints included objective response rate (ORR), disease control rate (DCR), overall survival (OS), quality-of-life (QoL), and safety. Results Between July 18, 2012 and Jan 22, 2014, a total of 167 patients were screened, and 154 patients were randomized in a 2:1 ratio to receive either famitinib (n = 99) or placebo (n = 55). The median PFS was 2.8 and 1.5 months in the famitinib and placebo groups (hazard ratio = 0.60, 95% confidence interval = 0.41–0.86, P = 0.004). The DCR was 59.8% and 31.4% (P = 0.002) and the ORR was 2.2% and 0.0% (P = 0.540) in the famitinib and placebo groups, respectively. The most frequent grade 3–4 adverse events were hypertension (11.1%), hand-foot syndrome (10.1%), thrombocytopenia (10.1%), and neutropenia (9.1%). Serious adverse events occurred in 11 (11.1%) patients in the famitinib group and 5 (9.1%) in the placebo group (P = 0.788). The median OS of the famitinib and placebo groups was 7.4 and 7.2 months (P = 0.657). Conclusion Famitinib prolonged PFS in refractory mCRC patients with acceptable tolerability. Trial registration This study was registered on ClinicalTrials.gov (NCT01762293) and was orally presented in the 2015 ASCO-Gastrointestinal Symposium
Collapse
Affiliation(s)
- Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, Guangdong, P. R. China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No 52, Fucheng Road, Haidian District, Beijing, 100142, P. R. China.
| | - Ke-Ming Wang
- Department of Medical Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Gang Wu
- Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, P. R. China
| | - Chun-Mei Shi
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, P. R. China
| | - Ke-Feng Ding
- Department of Surgical Oncology, Second Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, P. R. China
| | - Li-Zhu Lin
- Department of Oncology, First Affiliated Hospital of Guangzhou Medical University of Chinese Medicine, Guangzhou, 510405, Guangdong, P. R. China
| | - Jin-Wan Wang
- Department of Medical Oncology, Chinese Academy of Medical Sciences Cancer Hospital, Beijing, 100021, P. R. China
| | - Jian-Ping Xiong
- Department of Medical Oncology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
| | - Chang-Ping Wu
- Department of Medical Oncology, First People's Hospital of Changzhou, Changzhou, 213003, Jiangsu, P. R. China
| | - Jin Li
- Department of Medical Oncology, Fudan University Cancer Hospital, Shanghai, 200032, P. R. China
| | - Yun-Peng Liu
- Department of Medical Oncology, First Hospital of China Medical University, Shenyang, 110001, Liaoning, P. R. China
| | - Dong Wang
- Cancer Center, Daping Hospital and Institute of Surgery Research, Third Military Medical University, Chongqing, 400042, P. R. China
| | - Yi Ba
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, P. R. China
| | - Jue-Ping Feng
- Department of Oncology, PuAi Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, P. R. China
| | - Yu-Xian Bai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, P. R. China
| | - Jing-Wang Bi
- Department of Oncology, Jinan Military General Hospital, Jinan, 250000, Shandong, P. R. China
| | - Li-Wen Ma
- Department of Tumor Chemotherapy and Radiology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Jian Lei
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, P. R. China
| | - Qing Yang
- Department of Clinical Medicine, Jiangsu Hengrui Medicine Co., Ltd, Lianyungang, 222047, Jiangsu, P. R. China
| | - Hao Yu
- Department of Epidemic and Health Statistics, Nanjing Medical University, Nanjing, 211166, Jiangsu, P. R. China
| |
Collapse
|
54
|
Bonnetain F, Borg C, Adams RR, Ajani JA, Benson A, Bleiberg H, Chibaudel B, Diaz-Rubio E, Douillard JY, Fuchs CS, Giantonio BJ, Goldberg R, Heinemann V, Koopman M, Labianca R, Larsen AK, Maughan T, Mitchell E, Peeters M, Punt CJA, Schmoll HJ, Tournigand C, de Gramont A. How health-related quality of life assessment should be used in advanced colorectal cancer clinical trials. Ann Oncol 2017; 28:2077-2085. [PMID: 28430862 DOI: 10.1093/annonc/mdx191] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Traditionally, the efficacy of cancer treatment in patients with advance or metastatic disease in clinical studies has been studied using overall survival and more recently tumor-based end points such as progression-free survival, measurements of response to treatment. However, these seem not to be the relevant clinical end points in current situation if such end points were no validated as surrogate of overall survival to demonstrate the clinical efficacy. Appropriate, meaningful, primary patient-oriented and patient-reported end points that adequately measure the effects of new therapeutic interventions are then crucial for the advancement of clinical research in metastatic colorectal cancer to complement the results of tumor-based end points. Health-related quality of life (HRQoL) is effectively an evaluation of quality of life and its relationship with health over time. HRQoL includes the patient report at least of the way a disease or its treatment affects its physical, emotional and social well-being. Over the past few years, several phase III trials in a variety of solid cancers have assessed the incremental value of HRQoL in addition to the traditional end points of tumor response and survival results. HRQoL could provide not only complementary clinical data to the primary outcomes, but also more precise predictive and prognostic value. This end point is useful for both clinicians and patients in order to achieve the dogma of precision medicine. The present article examines the use of HRQoL in phase III metastatic colorectal cancer clinical trials, outlines the importance of HRQoL assessment methods, analysis, and results presentation. Moreover, it discusses the relevance of including HRQoL as a primary/co-primary end point to support the progression-free survival results and to assess efficacy of treatment in the advanced disease setting.
Collapse
Affiliation(s)
- F Bonnetain
- Methodology and Quality of Life Unit, Oncology Department (INSERM UMR 1098), Quality of Life and Cancer Clinical Research Platform
| | - C Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon
- Centre d'Investigation Clinique en Biothérapie, CIC-1431, Nantes
- 11UMR1098 INSERM/Université de Franche Comté/Etablissement Français du Sang, Besançon
- Department of Oncology, University Hospital of Besançon, Besançon, France
| | - R R Adams
- Cardiff University and Velindre Cancer Centre, Cardiff, UK
| | - J A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - A Benson
- Division of Hematology/Oncology, Northwestern Medical Group, Chicago, USA
| | - H Bleiberg
- Montagne de Saint Job, Brussels, Belgium
| | - B Chibaudel
- Institut Hospitalier Franco-Britannique, Levallois-Perret, France
| | - E Diaz-Rubio
- Medical Oncology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - J Y Douillard
- Medical Oncology, Institut de Cancérologie de l'Ouest (ICO), Nantes St-Herblain, France
| | - C S Fuchs
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston
| | - B J Giantonio
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia
| | - R Goldberg
- Department of Medicine, The Ohio State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Columbus, USA
| | - V Heinemann
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - M Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R Labianca
- Cancer Center, Ospedale Giovanni XXIII, Bergamo, Italy
| | - A K Larsen
- Cancer Biology and Therapeutics, INSERM and Université Pierre et Marie Curie, Hôpital Saint-Antoine, Paris, France
| | - T Maughan
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Oxford, UK
| | - E Mitchell
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, USA
| | - M Peeters
- Department of Oncology, Center for Oncological Research Antwerp, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium
| | - C J A Punt
- Department of Medical Oncology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - H J Schmoll
- Department of Internal Medicine IV, University Clinic Halle, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - C Tournigand
- Department of Oncology, University of Paris Est Creteil; APHP, Henri-Mondor Hospital, Créteil, France
| | - A de Gramont
- Institut Hospitalier Franco-Britannique, Levallois-Perret, France
| |
Collapse
|
55
|
Rahbari NN, D'Angelica MI. Surgical salvage of recurrence after resection of colorectal liver metastases: incidence and outcomes. Hepat Oncol 2017; 4:25-33. [PMID: 28768424 PMCID: PMC6095401 DOI: 10.2217/hep-2017-0002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/27/2017] [Indexed: 12/12/2022] Open
Abstract
Surgical resection remains the primary curative treatment option for patients with colorectal liver metastases. While the majority of patients will develop tumor relapse within or outside of the liver after hepatic metastasectomy, a subset of these patients may be amenable to salvage surgical resection. However, outcomes for this approach are not well defined. In this article, we summarize the current evidence for the incidence, feasibility and outcomes of salvage resection for recurrence after initial resection of colorectal liver metastases.
Collapse
Affiliation(s)
- Nuh N Rahbari
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Michael I D'Angelica
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| |
Collapse
|
56
|
Prager G, Argilés G. Towards shedding some light on regorafenib treatment in refractory metastatic colorectal cancer. ESMO Open 2017; 2:e000205. [PMID: 29303156 PMCID: PMC5703386 DOI: 10.1136/esmoopen-2017-000205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 04/26/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
- Gerald Prager
- Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University Vienna, Vienna, Austria
| | - Guillem Argilés
- Vall d'Hebrón Institut of Oncology (VHIO), Vall d'Hebrón University Hospital, Barcelona, Spain
| |
Collapse
|
57
|
Antiangiogenic tyrosine kinase inhibitors in colorectal cancer: is there a path to making them more effective? Cancer Chemother Pharmacol 2017; 80:661-671. [PMID: 28721456 DOI: 10.1007/s00280-017-3389-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/10/2017] [Indexed: 01/07/2023]
Abstract
Antiangiogenic therapy has a proven survival benefit in metastatic colorectal cancer. Inhibition of the VEGF pathway using a variety of extracellular antibody approaches has clear benefit in combination with chemotherapy, while intracellular blockade using tyrosine kinase inhibitors (TKIs) such as sorafenib and regorafenib has had more limited success. Pharmacodynamic modeling using modalities such as DCE-MRI indicates potent antiangiogenic effects of these TKIs, yet numerous combination therapies, primarily with chemotherapy, have failed to demonstrate an additive benefit. The sole comparative study of a single agent TKI against placebo showed a survival benefit of regorafenib in patients with advanced, refractory disease. Preclinical data demonstrate synergy between antiantiogenic TKIs and targeted interventions including autophagy inhibition, and together with a renewed effort to define markers of susceptibility, such combinations may be a way to improve the limited efficacy of this once-promising drug class.
Collapse
|
58
|
Ramjiawan RR, Griffioen AW, Duda DG. Anti-angiogenesis for cancer revisited: Is there a role for combinations with immunotherapy? Angiogenesis 2017; 20:185-204. [PMID: 28361267 PMCID: PMC5439974 DOI: 10.1007/s10456-017-9552-y] [Citation(s) in RCA: 500] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/13/2017] [Indexed: 12/18/2022]
Abstract
Angiogenesis is defined as the formation of new blood vessels from preexisting vessels and has been characterized as an essential process for tumor cell proliferation and viability. This has led to the development of pharmacological agents for anti-angiogenesis to disrupt the vascular supply and starve tumor of nutrients and oxygen, primarily through blockade of VEGF/VEGFR signaling. This effort has resulted in 11 anti-VEGF drugs approved for certain advanced cancers, alone or in combination with chemotherapy or other targeted therapies. But this success had only limited impact on overall survival of cancer patients and rarely resulted in durable responses. Given the recent success of immunotherapies, combinations of anti-angiogenics with immune checkpoint blockers have become an attractive strategy. However, implementing such combinations will require a better mechanistic understanding of their interaction. Due to overexpression of pro-angiogenic factors in tumors, their vasculature is often tortuous and disorganized, with excessively branched leaky vessels. This enhances vascular permeability, which in turn is associated with high interstitial fluid pressure, and a reduction in blood perfusion and oxygenation. Judicious dosing of anti-angiogenic treatment can transiently normalize the tumor vasculature by decreasing vascular permeability and improving tumor perfusion and blood flow, and synergize with immunotherapy in this time window. However, anti-angiogenics may also excessively prune tumor vessels in a dose and time-dependent manner, which induces hypoxia and immunosuppression, including increased expression of the immune checkpoint programmed death receptor ligand (PD-L1). This review focuses on revisiting the concept of anti-angiogenesis in combination with immunotherapy as a strategy for cancer treatment.
Collapse
Affiliation(s)
- Rakesh R Ramjiawan
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom St, Cox-734, Boston, MA, 02114, USA
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Dan G Duda
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom St, Cox-734, Boston, MA, 02114, USA.
| |
Collapse
|
59
|
Stark DP, Cook A, Brown JM, Brundage MD, Embleton AC, Kaplan RS, Raja FA, Swart AMW, Velikova G, Qian W, Ledermann JA. Quality of life with cediranib in relapsed ovarian cancer: The ICON6 phase 3 randomized clinical trial. Cancer 2017; 123:2752-2761. [PMID: 28339098 PMCID: PMC5516140 DOI: 10.1002/cncr.30657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND The ICON6 trial showed that cediranib, an oral inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, improved clinical outcomes for patients with platinum-sensitive relapsed ovarian cancer when it was used with chemotherapy and was continued as maintenance therapy. This study describes health-related quality of life (QOL) during the first year of treatment. METHODS Four hundred fifty-six women were randomly allocated to receive standard chemotherapy only, chemotherapy with concurrent cediranib, or chemotherapy with cediranib administered concurrently and continued as maintenance. Patients completed QOL questionnaires until disease progression every 3 weeks during chemotherapy and then every 6 weeks to 1 year. Patients alive with disease progression completed a QOL form 1 year after randomization. The primary QOL endpoint was the global score from the Quality of Life Questionnaire Core 30 (of the European Organization for Research and Treatment of Cancer) at 1 year, with the standard chemotherapy group compared with the concurrent-maintenance cediranib group. RESULTS The rate of questionnaire compliance was 90% at the baseline and 76% at 1 year and was similar across the 3 groups. The mean global QOL score at 1 year was 62.6 points for the standard chemotherapy group and 68.7 points for the concurrent-maintenance group (+4.5; 95% confidence interval, -2.0 to 11.0; P = .18). Sensitivity analyses suggested that this finding was robust to the effect of missing data, and the improvement became statistically significant after adjustments for self-reported diarrhea. CONCLUSIONS The 6th study by the International Collaboration in Ovarian Neoplasm (ICON6) showed a significant improvement in progression-free survival with cediranib as concurrent and maintenance therapy. No QOL detriment with cediranib was found 1 year after treatment was commenced. The maintenance of QOL along with prolonged cancer control suggests that cediranib has a valuable role in the treatment of relapsed ovarian cancer. Cancer 2017;123:2752-61. © 2017 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
Collapse
Affiliation(s)
- Dan P Stark
- St. James's Institute of Oncology, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Adrian Cook
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Julia M Brown
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | | | - Andrew C Embleton
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Richard S Kaplan
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Fharat A Raja
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Ann Marie W Swart
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Galina Velikova
- St. James's Institute of Oncology, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Wendi Qian
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Jonathan A Ledermann
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| |
Collapse
|
60
|
Schuurhuizen C, Braamse A, Konings I, Sprangers M, Ket J, Dekker J, Verheul H. Does severe toxicity affect global quality of life in patients with metastatic colorectal cancer during palliative systemic treatment? A systematic review. Ann Oncol 2017; 28:478-486. [DOI: 10.1093/annonc/mdw617] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
61
|
Neuzillet C, Rousseau B, Kocher H, Bourget P, Tournigand C. Unravelling the pharmacologic opportunities and future directions for targeted therapies in gastro-intestinal cancers Part 1: GI carcinomas. Pharmacol Ther 2017; 174:145-172. [PMID: 28223233 DOI: 10.1016/j.pharmthera.2017.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Until the 1990s, cytotoxic chemotherapy has been the cornerstone of medical therapy for gastrointestinal (GI) cancers. Better understanding of the molecular biology of cancer cell has led to the therapeutic revolution of targeted therapies, i.e. monoclonal antibodies or small molecule inhibitors directed against proteins that are specifically overexpressed or mutated in cancer cells. These agents being more specific to cancer cells were expected to be less toxic than cytotoxic agents. Targeted agents have provided clinical benefit in many GI cancer types. For example, antiangiogenics and anti-EGFR therapies have significantly improved survival of patients affected by metastatic colorectal cancer and have deeply changed the therapeutic strategy in this disease. However, their effects have sometimes been disappointing, due to intrinsic or acquired resistance mechanisms (e.g., RAS mutation for anti-EGFR therapies), or to an activity restricted to some tumour settings (e.g., lack of activity in other cancer types, or on the microscopic residual disease in adjuvant setting). Many studies are negative in overall population but positive in some specific patient subgroups (e.g., trastuzumab in HER2-positive gastric cancer), illustrating the importance of patient selection and early identification of predictive biomarkers of response to these therapies. We propose a comprehensive two-part review providing a panoramic approach of the successes and failures of targeted agents in GI cancers to unravel the pharmacologic opportunities and future directions for these agents in GI oncology. In this first part, we will focus on adenocarcinomas and squamous cell carcinomas, for which targeted therapies are mostly used in combination with chemotherapy.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Medical Oncology, Henri Mondor University Hospital, AP-HP, Paris Est Créteil University (UPEC), 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London, E1 1BB, United Kingdom.
| | - Benoît Rousseau
- Department of Medical Oncology, Henri Mondor University Hospital, AP-HP, Paris Est Créteil University (UPEC), 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Hemant Kocher
- Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London, E1 1BB, United Kingdom
| | - Philippe Bourget
- Department of Clinical Pharmacy, Necker-Enfants Malades University Hospital, 149 Rue de Sèvres, 75015 Paris, France
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor University Hospital, AP-HP, Paris Est Créteil University (UPEC), 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| |
Collapse
|
62
|
Computational investigation of sphingosine kinase 1 (SphK1) and calcium dependent ERK1/2 activation downstream of VEGFR2 in endothelial cells. PLoS Comput Biol 2017; 13:e1005332. [PMID: 28178265 PMCID: PMC5298229 DOI: 10.1371/journal.pcbi.1005332] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/23/2016] [Indexed: 01/14/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a powerful regulator of neovascularization. VEGF binding to its cognate receptor, VEGFR2, activates a number of signaling pathways including ERK1/2. Activation of ERK1/2 is experimentally shown to involve sphingosine kinase 1 (SphK1) activation and its calcium-dependent translocation downstream of ERK1/2. Here we construct a rule-based computational model of signaling downstream of VEGFR2, by including SphK1 and calcium positive feedback mechanisms, and investigate their consequences on ERK1/2 activation. The model predicts the existence of VEGF threshold in ERK1/2 activation that can be continuously tuned by cellular concentrations of SphK1 and sphingosine 1 phosphate (S1P). The computer model also predicts powerful effects of perturbations in plasma and ER calcium pump rates and the current through the CRAC channels on ERK1/2 activation dynamics, highlighting the critical role of intracellular calcium in shaping the pERK1/2 signal. The model is then utilized to simulate anti-angiogenic therapeutic interventions targeting VEGFR2-ERK1/2 axis. Simulations indicate that monotherapies that exclusively target VEGFR2 phosphorylation, VEGF, or VEGFR2 are ineffective in shutting down signaling to ERK1/2. By simulating therapeutic strategies that target multiple nodes of the pathway such as Raf and SphK1, we conclude that combination therapy should be much more effective in blocking VEGF signaling to EKR1/2. The model has important implications for interventions that target signaling pathways in angiogenesis relevant to cancer, vascular diseases, and wound healing. Vascular endothelial growth factor (VEGF) signaling is a potent regulator of angiogenesis, the growth and development of new vessels out of a preexisting vascular network. Angiogenesis requires enhanced survival, proliferation, and motility of the vascular endothelial cells. Crucial signaling endpoints in VEGF-mediated angiogenic response include elevation in intracellular calcium and the activation of the proteins ERK1 and 2 (ERK1/2). In this study, we have developed a novel computer model for the activation of ERK1/2 and calcium downstream of VEGF receptor type 2 (VEGFR2). Our model is the first of its kind to incorporate and investigate the consequences of calcium elevation and the role of a cellular lipid modifier known as sphingosine kinase 1 (SphK1). We also utilize the model to simulate therapeutic strategies targeting VEGF signaling to ERK1/2 indicating inefficiency of single therapies known as tyrosine kinase inhibitors (TKI) that target receptor phosphorylation. Computer simulations indicate that combination therapy is essential for effective blockade of this important pathway. Our results have important implications for human diseases such as cancer where plethora of anti-VEGF therapies are currently employed. Overall, our computer model sheds new light on a complex feedback involving SphK1 and calcium that radically alters the response of cells to VEGF.
Collapse
|
63
|
Riechelmann R, Grothey A. Antiangiogenic therapy for refractory colorectal cancer: current options and future strategies. Ther Adv Med Oncol 2017; 9:106-126. [PMID: 28203302 PMCID: PMC5298403 DOI: 10.1177/1758834016676703] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Even though significant improvements in the treatment of colorectal cancer (CRC) have been made in recent years, survival rates for metastatic colorectal cancer (mCRC) are poor. Effective treatment options for metastatic colorectal cancer remain limited, and new therapeutic strategies are desperately needed. Several tyrosine kinase inhibitors (TKIs) and monoclonal antibodies (mAbs) that target angiogenesis, a critical process for facilitating tumor cell growth, invasion, and metastasis, are either approved or in clinical development for the treatment of mCRC. Many of these agents have shown efficacy in mCRC, both as single agents and in combination with standard chemotherapy regimens. However, there is a need for predictive markers of response to identify those patients most likely to benefit from antiangiogenic therapy, and, to date, no markers of this type have been validated in patients. Additionally, because antiangiogenic agents typically cause cytostatic as opposed to cytotoxic antitumor effects, it is important to determine the best strategies for evaluating therapeutic response in mCRC to ensure maximum clinical benefit. In this review, we summarize the efficacy and tolerability of approved and investigational antiangiogenic agents for the treatment of mCRC. We also discuss potential markers of response to antiangiogenic agents and how these markers, along with appropriate endpoint selection, can improve clinical trial design.
Collapse
Affiliation(s)
- Rachel Riechelmann
- Instituto do Cancer do Estado de São Paulo, Av. Doutor Arnaldo 251, 5° Andar, CEP 01246-000, São Paulo, SP, Brazil
| | | |
Collapse
|
64
|
Quidde J, Denne L, Kutscheidt A, Kindler M, Kirsch A, Kripp M, Petersen V, Schulze M, Seraphin J, Tummes D, Arnold D, Stein A. Baseline and On-Treatment Markers Determining Prognosis of First-Line Chemotherapy in Combination with Bevacizumab in Patients with Metastatic Colorectal Cancer. Oncol Res Treat 2017; 40:21-26. [DOI: 10.1159/000454774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 11/28/2016] [Indexed: 11/19/2022]
|
65
|
Ma WW, Saif MW, El‐Rayes BF, Fakih MG, Cartwright TH, Posey JA, King TR, von Borstel RW, Bamat MK. Emergency use of uridine triacetate for the prevention and treatment of life-threatening 5-fluorouracil and capecitabine toxicity. Cancer 2017; 123:345-356. [PMID: 27622829 PMCID: PMC5248610 DOI: 10.1002/cncr.30321] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/03/2016] [Accepted: 08/08/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Increased susceptibility to 5-fluorouracil (5-FU)/capecitabine can lead to rapidly occurring toxicity caused by impaired clearance, dihydropyrimidine dehydrogenase deficiency, and other genetic variations in the enzymes that metabolize 5-FU. Life-threatening 5-FU overdoses occur because of infusion pump errors, dosage miscalculations, and accidental or suicidal ingestion of capecitabine. Uridine triacetate (Vistogard) was approved in 2015 for adult and pediatric patients who exhibit early-onset severe or life-threatening 5-FU/capecitabine toxicities or present with an overdose. Uridine triacetate delivers high concentrations of uridine, which competes with toxic 5-FU metabolites. METHODS In 2 open-label clinical studies, patients who presented with a 5-FU/capecitabine overdose or an early onset of severe toxicities were treated. Patients received uridine triacetate as soon as possible (most within the first 96 hours after 5-FU/capecitabine). Outcomes included survival, resumption of chemotherapy, and safety. Their survival was compared with the survival of a historical cohort of overdose patients who received only supportive care. RESULTS A total of 137 of 142 overdose patients (96%) treated with uridine triacetate survived and had a rapid reversal of severe acute cardiotoxicity and neurotoxicity; in addition, mucositis and leukopenia were prevented, or the patients recovered from them. In the historical cohort, 21 of 25 patients (84%) died. Among the 141 uridine triacetate-treated overdose patients with a diagnosis of cancer (the noncancer patients included 6 intentional or accidental pediatric overdoses), 53 resumed chemotherapy in < 30 days (median time after 5-FU, 19.6 days), and this indicated a rapid recovery from toxicity. Adverse reactions in patients receiving uridine triacetate included vomiting (8.1%), nausea (4.6%), and diarrhea (3.5%). CONCLUSIONS In these studies, uridine triacetate was a safe and effective lifesaving antidote for capecitabine and 5-FU overexposure, and it facilitated the rapid resumption of chemotherapy. Cancer 2017;123:345-356. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Wen Wee Ma
- Department of OncologyMayo ClinicRochesterMinnesota
| | | | - Bassel F. El‐Rayes
- Department of Hematology and Oncology, Winship Cancer InstituteEmory UniversityAtlantaGeorgia
| | - Marwan G. Fakih
- Department of Medical OncologyCity of Hope Medical CenterDuarteCalifornia
| | | | - James A. Posey
- Department of Medical OncologyThomas Jefferson UniversityPhiladelphiaPennsylvania
| | | | | | | |
Collapse
|
66
|
Melsens E, Verberckmoes B, Rosseel N, Vanhove C, Descamps B, Pattyn P, Ceelen W. The VEGFR Inhibitor Cediranib Improves the Efficacy of Fractionated Radiotherapy in a Colorectal Cancer Xenograft Model. Eur Surg Res 2016; 58:95-108. [PMID: 28002822 DOI: 10.1159/000452741] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 10/19/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND/PURPOSE Radiotherapy (RT) increases local tumor control in locally advanced rectal cancer, but complete histological response is seen in only a minority of cases. Antiangiogenic therapy has been proposed to improve RT efficacy by "normalizing" the tumor microvasculature. Here, we examined whether cediranib, a pan-vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor, improves microvascular function and tumor control in combination with RT in a mouse colorectal cancer (CRC) model. METHODS CRC xenografts (HT29) were grown subcutaneously in mice. Animals were treated for 5 consecutive days with vehicle, RT (1.8 Gy daily), cediranib (6 mg/kg po), or combined therapy (cediranib 2 h prior to radiation). Tumor volume was measured with calipers. Vascular changes were analyzed by dynamic contrast-enhanced MRI, oxygenation and interstitial fluid pressure probes and histology. To investigate vascular changes more in detail, a second set of mice were fitted with titanium dorsal skinfold window chambers, wherein a HT29 tumor cell suspension was injected. In vivo fluorescence microscopy was performed before and after treatment (same treatment protocol). RESULTS In vivo microscopy analyses showed that VEGFR inhibition with cediranib led to a "normalization" of the vessel wall, with decreased microvessel permeability (p < 0.0001) and tortuosity (p < 0.01), and a trend to decreased vessel diameters. This seemed to lead to lower tumor hypoxia rates in the cediranib and combination groups compared to the control and RT groups. This led to an increased tumor control in the combination group compared to controls or monotherapy (p < 0.0001). CONCLUSIONS The combination of RT with cediranib enhances tumor control in a CRC xenograft mouse model. Microvascular analyses suggest that cediranib leads to vascular normalization and improved oxygenation.
Collapse
Affiliation(s)
- Elodie Melsens
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | | |
Collapse
|
67
|
Significant increase of synchronous disease in first-line metastatic colorectal cancer trials: Results of a systematic review. Eur J Cancer 2016; 69:166-177. [DOI: 10.1016/j.ejca.2016.09.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/19/2016] [Accepted: 09/25/2016] [Indexed: 01/08/2023]
|
68
|
Giuliani J, Bonetti A. The Pharmacological Costs of First-Line Therapies in Unselected Patients With Advanced Colorectal Cancer: A Review of Published Phase III Trials. Clin Colorectal Cancer 2016; 15:277-284. [DOI: 10.1016/j.clcc.2016.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/05/2016] [Indexed: 10/21/2022]
|
69
|
Lipitz-Snyderman A, Weingart SN, Anderson C, Epstein AS, Killen A, Classen D, Sima CS, Fortier E, Atoria CL, Pfister D, Lipitz-Snyderman A, Weingart SN, Anderson C, Epstein AS, Killen A, Classen D, Sima CS, Fortier E, Atoria CL, Pfister D. ReCAP: Detection of Potentially Avoidable Harm in Oncology From Patient Medical Records. J Oncol Pract 2016; 12:178-9; e224-30. [PMID: 26869656 DOI: 10.1200/jop.2015.006874] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Widespread consensus exists about the importance of addressing patient safety issues in oncology, yet our understanding of the frequency, spectrum, and preventability of adverse events (AEs) across cancer care is limited. METHODS We developed a screening tool to detect AEs across cancer care settings through medical record review. Members of the study team reviewed the scientific literature and obtained structured input from an external multidisciplinary panel of clinicians by using a modified Delphi process. RESULTS The screening tool comprises 76 triggers-readily identifiable findings to screen for possible AEs that occur during cancer care. Categories of triggers are general care, vital signs, medication related, laboratory tests, other orders, and consultations. CONCLUSION Although additional testing is required to assess its performance characteristics, this tool may offer an efficient mechanism for identifying possibly preventable AEs in oncology and serve as an instrument for quality improvement.
Collapse
Affiliation(s)
- Allison Lipitz-Snyderman
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Saul N Weingart
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Christopher Anderson
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Andrew S Epstein
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Aileen Killen
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - David Classen
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Camelia S Sima
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Elizabeth Fortier
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Coral L Atoria
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - David Pfister
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Allison Lipitz-Snyderman
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA.
| | - Saul N Weingart
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Christopher Anderson
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Andrew S Epstein
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Aileen Killen
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - David Classen
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Camelia S Sima
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Elizabeth Fortier
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - Coral L Atoria
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| | - David Pfister
- Memorial Sloan Kettering Cancer Center; Columbia University Medical Center; AIG, New York, NY; Tufts Medical Center, Boston, MA; Pascal Metrics, Washington, DC; University of Utah School of Medicine, Salt Lake City, UT; and Genentech, San Francisco, CA
| |
Collapse
|
70
|
Adamowicz K, Saad ED, Jassem J. Health-related quality of life assessment in contemporary phase III trials in advanced colorectal cancer. Cancer Treat Rev 2016; 50:194-199. [DOI: 10.1016/j.ctrv.2016.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 09/18/2016] [Accepted: 09/20/2016] [Indexed: 10/20/2022]
|
71
|
Ranji P, Salmani Kesejini T, Saeedikhoo S, Alizadeh AM. Targeting cancer stem cell-specific markers and/or associated signaling pathways for overcoming cancer drug resistance. Tumour Biol 2016; 37:13059-13075. [DOI: 10.1007/s13277-016-5294-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
|
72
|
Matos I, Elez E, Capdevila J, Tabernero J. Emerging tyrosine kinase inhibitors for the treatment of metastatic colorectal cancer. Expert Opin Emerg Drugs 2016; 21:267-82. [PMID: 27578253 DOI: 10.1080/14728214.2016.1220535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is a leading cause of cancer death worldwide. Over the last decade, the addition of antibodies that block the epidermal growth factor receptor (EGFR) or angiogenesis to the classic chemotherapy backbone has improved overall survival in metastatic colorectal cancer (mCRC). However, the role of the other major targeted therapy, the tyrosine kinase inhibitors (TKIs), is not yet fully clarified. AREAS COVERED This review discusses key published and ongoing studies with TKIs in mCRC, the mechanisms of resistance to standard treatments that are potentially targetable with these small molecules, along with the role of biomarkers in therapeutic decision-making process. EXPERT OPINION The current effectiveness of TKIs is limited by two principal reasons, firstly the use of combination chemotherapy necessitates lower dose-density to manage the toxicity profile and secondly, development of these drugs has mainly been performed in molecularly unselected populations. mCRC is a heterogeneous and dynamic disease, and clinical trials with TKIs must be designed on the basis of specific molecular alterations targeted by these drugs. Success with this approach relies on identifying mutations at the time of progression, raising the importance of minimally-invasive monitoring tools. Liquid biopsies are a promising option, although this technique remains to be validated. Overall, this approach contributes to the move towards personalized and precision therapeutic strategies.
Collapse
Affiliation(s)
- Ignacio Matos
- a Spain - Medical Oncology Department , Vall d'Hebron University Hospital , Barcelona , Spain
| | - Elena Elez
- a Spain - Medical Oncology Department , Vall d'Hebron University Hospital , Barcelona , Spain
| | - Jaume Capdevila
- a Spain - Medical Oncology Department , Vall d'Hebron University Hospital , Barcelona , Spain
| | - Josep Tabernero
- a Spain - Medical Oncology Department , Vall d'Hebron University Hospital , Barcelona , Spain
| |
Collapse
|
73
|
Zheng J, Bai X, Hong C, Gao H, Li X. Meta-analysis of the incidence and risk of arterial and venous thromboembolic events associated with anti-EGFR agents in non-small-cell lung cancer patients. Expert Rev Clin Pharmacol 2016; 9:1389-1395. [PMID: 27494194 DOI: 10.1080/17512433.2016.1220830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND To determine the risk of arterial and venous thromboembolic events (ATEs and VETs) associated with anti-epidermal growth factor receptor (EGFR) agents in non-small-cell lung cancer (NSCLC) patients. METHODS Prospective randomized trials evaluating therapy with or without anti-EGFR agents in NSCLC patients. Data on VTEs and ATEs were extracted. RESULTS A total of 8,410 patients from 12 trials were included for analysis. Anti-EGFR agents significantly increased the risk of all-grade and high-grade VTEs (Peto OR 1.50, 95%CI 1.16-1.95, P = 0.002; Peto OR 1.73, 95%CI: 1.32-2.26, p < 0.001, respectively), but not for all-grade and high-grade ATEs. CONCLUSION The use of anti-EGFR agents significantly increased the risk of all-grade and high-grade VTEs but not for ATEs in NSCLC patients.
Collapse
Affiliation(s)
- Jian Zheng
- a Liaoning Cancer Hospital & Insititute, Third Department of Internal Medicine , Cancer Hospital of China Medical University , Shenyang , China
| | - Xiaoru Bai
- b Liaoning Cancer Hospital & Insititute, The department of medical imaging , Cancer Hospital of China Medical University , Shenyang , China
| | - Chengyu Hong
- a Liaoning Cancer Hospital & Insititute, Third Department of Internal Medicine , Cancer Hospital of China Medical University , Shenyang , China
| | - Hong Gao
- a Liaoning Cancer Hospital & Insititute, Third Department of Internal Medicine , Cancer Hospital of China Medical University , Shenyang , China
| | - Xiaoling Li
- a Liaoning Cancer Hospital & Insititute, Third Department of Internal Medicine , Cancer Hospital of China Medical University , Shenyang , China
| |
Collapse
|
74
|
Ethier JL, Lheureux S, Oza A. The role of cediranib in ovarian cancer: current status and further investigation. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1196130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
75
|
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vessels, has been validated as a target in several tumour types through randomised trials, incorporating vascular endothelial growth factor (VEGF) pathway inhibitors into the therapeutic armoury. Although some tumours such as renal cell carcinoma, ovarian and cervical cancers, and pancreatic neuroendocrine tumours are sensitive to these drugs, others such as prostate cancer, pancreatic adenocarcinoma, and melanoma are resistant. Even when drugs have yielded significant results, improvements in progression-free survival, and, in some cases, overall survival, are modest. Thus, a crucial issue in development of these drugs is the search for predictive biomarkers-tests that predict which patients will, and will not, benefit before initiation of therapy. Development of biomarkers is important because of the need to balance efficacy, toxicity, and cost. Novel combinations of these drugs with other antiangiogenics or other classes of drugs are being developed, and the appreciation that these drugs have immunomodulatory and other modes of action will lead to combination regimens that capitalise on these newly understood mechanisms.
Collapse
Affiliation(s)
- Gordon C Jayson
- Institute of Cancer Sciences and Christie Hospital, University of Manchester, Manchester, UK.
| | - Robert Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Lee M Ellis
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adrian L Harris
- Department of Medical Oncology, Churchill Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
76
|
Benson AB, Kiss I, Bridgewater J, Eskens FALM, Sasse C, Vossen S, Chen J, Van Sant C, Ball HA, Keating A, Krivoshik A. BATON-CRC: A Phase II Randomized Trial Comparing Tivozanib Plus mFOLFOX6 with Bevacizumab Plus mFOLFOX6 in Stage IV Metastatic Colorectal Cancer. Clin Cancer Res 2016; 22:5058-5067. [PMID: 27401244 DOI: 10.1158/1078-0432.ccr-15-3117] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/20/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Tivozanib, a selective inhibitor of VEGFR-1, -2, and -3, plus mFOLFOX6 in an advanced gastrointestinal cancer phase Ib study had encouraging antineoplastic activity and a tolerable safety profile. This randomized, open-label, phase II trial of tivozanib/mFOLFOX6 versus bevacizumab/mFOLFOX6 in patients with previously untreated metastatic colorectal cancer (mCRC) evaluated tivozanib activity versus bevacizumab. EXPERIMENTAL DESIGN Treatment-naïve patients received mFOLFOX6 every 2 weeks of each 28-day cycle plus either tivozanib orally 1.5 mg once daily for 21 days or bevacizumab intravenously 5 mg/kg every 2 weeks. Investigator-assessed progression-free survival (PFS) was the primary endpoint; some secondary endpoints included safety, overall survival, overall response rate (ORR), duration of response, time to treatment failure, and biomarker subgroup analyses. RESULTS A prespecified interim futility analysis demonstrated that the futility boundary for superiority of tivozanib/mFOLFOX6 over bevacizumab/mFOLFOX6 for PFS in the intent-to-treat population was crossed; median PFS was 9.4 versus 10.7 months [HR = 1.091; confidence interval (CI), 0.693-1.718; P = 0.706]. Tivozanib/mFOLFOX6 resulted in PFS and ORR comparable with bevacizumab/mFOLFOX6; interim analyses biomarker results revealed no significant PFS association. Post hoc final analyses demonstrated a potential difference in tivozanib-specific PFS in patients with low neuropilin-1 (NRP-1), but not in patients with high NRP-1. Tivozanib/mFOLFOX6 was tolerable and adverse events were comparable with both bevacizumab/mFOLFOX6 and previous tivozanib studies. CONCLUSIONS The efficacy of tivozanib/mFOLFOX6 was comparable with but not superior to bevacizumab/mFOLFOX6 in patients with previously untreated mCRC. Since data from the prespecified interim analysis did not demonstrate superiority, this resulted in discontinuation of the study. The safety and tolerability profile of tivozanib/mFOLFOX6 was consistent with other tivozanib trials. NRP-1 is a potential predictive biomarker for tivozanib activity, but these results require further validation. Clin Cancer Res; 22(20); 5058-67. ©2016 AACR.
Collapse
Affiliation(s)
- Al B Benson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.
| | - Igor Kiss
- Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - John Bridgewater
- University College London Cancer Institute, London, United Kingdom
| | | | - Carolyn Sasse
- Astellas Pharma Global Development, Northbrook, Illinois
| | - Sandra Vossen
- Astellas Pharma Global Development, Northbrook, Illinois
| | - Jihong Chen
- Astellas Pharma Global Development, Northbrook, Illinois
| | - Chip Van Sant
- Astellas Pharma Global Development, Northbrook, Illinois
| | - Howard A Ball
- Astellas Pharma Global Development, Northbrook, Illinois
| | - Anne Keating
- Astellas Pharma Global Development, Northbrook, Illinois
| | | |
Collapse
|
77
|
Dong G, Vandemeulebroecke M. A modified varying-stage adaptive phase II/III clinical trial design. Pharm Stat 2016; 15:368-78. [DOI: 10.1002/pst.1753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Indexed: 11/08/2022]
|
78
|
Lewis CM, Wolf WA, Xun P, Sandler RS, He K. Racial differences in dietary changes and quality of life after a colorectal cancer diagnosis: a follow-up of the Study of Outcomes in Colorectal Cancer Survivors cohort. Am J Clin Nutr 2016; 103:1523-30. [PMID: 27099251 PMCID: PMC4880997 DOI: 10.3945/ajcn.115.126276] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/25/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Substantial racial disparities exist in colorectal cancer (CRC) survival. OBJECTIVE This was an exploratory study to assess the racial differences in dietary changes in relation to quality of life (QoL), recurrence, and survival after a CRC diagnosis. DESIGN Four hundred fifty-three stage II CRC patients were enrolled in the cohort study through the North Carolina Central Cancer Registry. Self-reported diet, physical activity, treatment, comorbidities, demographic characteristics, and QoL were collected at diagnosis and 12 and 24 mo after diagnosis. QoL was assessed with the Functional Assessment of Cancer Therapy-Colorectal (FACT-C) and the Medical Outcomes 12-Item Short Form Health Survey (SF-12) inventories. An overall dietary index score was calculated. Generalized estimating equations and logistic regression models were used to explore potential associations. Statistical power for this study was ∼50%. RESULTS African Americans (n = 81) were more likely to increase intakes of reduced-fat milk, vegetables, and fruit and decrease intakes of regular cheese, red meat, fried food, fast food, and fat (P < 0.05) than were Caucasians (n = 184) 24 mo after diagnosis. The least-squares means ± SEs for changes in dietary index were 6.05 ± 0.40 and 4.07 ± 0.27 for African Americans and Caucasians, respectively (P < 0.001). African Americans exhibited higher scores on portions of the FACT-C (colorectal cancer subscale: β = 1.04; 95% CI: 0.26, 1.82) and the SF-12 (Physical Component Summary: β = 2.49; 95% CI: 0.51, 4.48). Those who improved their dietary quality over 24 mo had lower risk of recurrence and mortality combined (OR: 0.42; 95% CI: 0.25, 0.72). CONCLUSIONS African Americans made more healthful changes in diet and had a higher QoL than did Caucasians in this underpowered study that used self-reported dietary data. No racial differences in recurrence or survival were evident, although improvements in dietary quality did reveal survival benefits overall. More prospective research on racial disparities in health behavior changes after diagnosis is desperately needed.
Collapse
Affiliation(s)
- Cari M Lewis
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana; and
| | - W Asher Wolf
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Pengcheng Xun
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana; and
| | - Robert S Sandler
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Ka He
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana; and
| |
Collapse
|
79
|
|
80
|
Folprecht G, Pericay C, Saunders MP, Thomas A, Lopez Lopez R, Roh JK, Chistyakov V, Höhler T, Kim JS, Hofheinz RD, Ackland SP, Swinson D, Kopp M, Udovitsa D, Hall M, Iveson T, Vogel A, Zalcberg JR. Oxaliplatin and 5-FU/folinic acid (modified FOLFOX6) with or without aflibercept in first-line treatment of patients with metastatic colorectal cancer: the AFFIRM study. Ann Oncol 2016; 27:1273-9. [PMID: 27091810 DOI: 10.1093/annonc/mdw176] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 04/10/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The combination of aflibercept with FOLFIRI has been shown to significantly prolong overall survival in patients with metastatic colorectal cancer (mCRC) after progression on oxaliplatin-based therapy. This trial evaluated the addition of aflibercept to oxaliplatin-based first-line treatment of patients with mCRC. PATIENTS AND METHODS Patients with mCRC were randomized to receive first-line therapy with mFOLFOX6 plus aflibercept (4 mg/kg) or mFOLFOX6 alone. The primary end point of this phase II study was the progression-free survival (PFS) rate at 12 months in each arm. The analysis of efficacy between the arms was a pre-planned secondary analysis. RESULTS Of 236 randomized patients, 227 and 235 patients were evaluable for the primary efficacy analysis and safety, respectively. The probabilities of being progression-free at 12 months were 25.8% [95% confidence interval (CI) 17.2-34.4] for the aflibercept/mFOLFOX6 arm and 21.2% (95% CI 12.2-30.3) for the mFOLFOX6 arm. The median PFS was 8.48 months (95% CI 7.89-9.92) for the aflibercept/mFOLFOX6 arm and 8.77 months (95% CI 7.62-9.27) for the mFOLFOX6 arm; the hazard ratio of aflibercept/mFOLFOX6 versus mFOLFOX6 was 1.00 (95% CI 0.74-1.36). The response rates were 49.1% (95% CI 39.7-58.6) and 45.9% (95% CI 36.4-55.7) for patients treated with and without aflibercept, respectively. The most frequent treatment-emergent grade 3/4 adverse events (AEs) excluding laboratory abnormalities reported for aflibercept/mFOLFOX6 versus mFOLFOX6 were neuropathy (16.8% versus 17.2%) and diarrhea (13.4% versus 5.2%). Neutropenia grade 3/4 occurred in 36.1% versus 29.3%. The most common vascular endothelial growth factor inhibition class-effect grade 3/4 AEs for aflibercept/mFOLFOX6 versus mFOLFOX6 were hypertension (35.3% versus 1.7%), proteinuria (9.2% versus 0%), deep vein thrombosis (5.9% versus 0.9%) and pulmonary embolism (5.9% versus 5.2%). CONCLUSION No difference in PFS rate was observed between treatment groups. Adding aflibercept to first-line mFOLFOX6 did not increase efficacy but was associated with higher toxicity. CLINICAL TRIAL NUMBER NCT00851084, www.clinicaltrials.gov, EudraCT 2008-004178-41.
Collapse
Affiliation(s)
- G Folprecht
- Medical Department I, University Cancer Center, University Hospital Carl Gustav Carus, Dresden, Germany
| | - C Pericay
- Hospital de Sabadell, Corporació Sanitaria Parc Taulí-Institut Universitari, Sabadell, Spain
| | - M P Saunders
- Department of Radiotherapy and Oncology, The Christie NHS Foundation Trust, Manchester
| | - A Thomas
- Department of Cancer Studies, University of Leicester, Leicester, UK
| | - R Lopez Lopez
- Department of Medical Oncology, Hospital Clinico Universitario e Instituto de Investigación, Santiago de Compostela, Spain
| | - J K Roh
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - T Höhler
- Department I of Internal Medicine, Prosper Hospital, Recklinghausen, Germany
| | - J-S Kim
- Department of Oncology and Hematology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - R-D Hofheinz
- Department III of Internal Medicine, University Hospital, Mannheim, Germany
| | - S P Ackland
- Department of Medical Oncology, Calvary Mater Hospital, Newcastle Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - D Swinson
- Department of Oncology, St James' Hospital, Leeds, UK
| | - M Kopp
- Samara Regional Oncology Dispensary, Samara
| | - D Udovitsa
- Oncological Dispensary #2, Sochi, Russia
| | - M Hall
- Cancer Services Division, Mount Vernon Cancer Centre, Middlesex
| | - T Iveson
- Department of Medical Oncology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - A Vogel
- Clinic of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - J R Zalcberg
- School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
81
|
Tampellini M, Sonetto C, Scagliotti GV. Novel anti-angiogenic therapeutic strategies in colorectal cancer. Expert Opin Investig Drugs 2016; 25:507-20. [PMID: 26938715 DOI: 10.1517/13543784.2016.1161754] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Anti-angiogenetic agents are currently the standard of care in metastatic CRC patients. Bevacizumab, aflibercept, regorafenib and recently ramucirumab have significantly improved both progression-free and overall survival in different lines of treatment. Since bevacizumab's approval, a number of novel anti-VEGF agents have been tested in preclinical and clinical models. AREAS COVERED This review is focused on the most recent clinical results of novel agents targeting VEGF and its receptors with a major focus on those investigated recently in clinical trials. EXPERT OPINION In the last 15 years, a number of new anti-angiogenetic agents have been tested. Unfortunately, most of them have demonstrated unacceptable toxicities or failed to show activity. When tested as single agents, encouraging preliminary results were reported with fruquintinib, famitinib, and nintedanib. Interesting novel mechanisms of action are also being explored: VGX-100 is a monoclonal antibody (mAb) which binds to VEGF-C, inhibiting activation of VEGFR-2 and VEGFR-3 when combined with bevacizumab; tanibirumab is a mAb which binds to VEGFR-2 and vanucizumab is a bispecific mAb binding both to VEGF-A and Angiopoietin-2. Data about the combination of these agents with chemotherapy are very encouraging, even though preliminary. However, the definition of specific predictive biomarkers remains a priority.
Collapse
Affiliation(s)
- M Tampellini
- a University of Turin , Department of Oncology, AOU San Luigi di Orbassano , Torino , Italy
| | - C Sonetto
- a University of Turin , Department of Oncology, AOU San Luigi di Orbassano , Torino , Italy
| | - G V Scagliotti
- a University of Turin , Department of Oncology, AOU San Luigi di Orbassano , Torino , Italy
| |
Collapse
|
82
|
Affiliation(s)
- John Norrie
- Centre for Healthcare Randomised Trials (CHaRT), Health Services Research Unit, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
83
|
Creutzfeldt A, Suling A, Oechsle K, Mehnert A, Atanackovic D, Kripp M, Arnold D, Stein A, Quidde J. Integrating patient reported measures as predictive parameters into decisionmaking about palliative chemotherapy: a pilot study. BMC Palliat Care 2016; 15:25. [PMID: 26928745 PMCID: PMC4772352 DOI: 10.1186/s12904-016-0101-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 02/25/2016] [Indexed: 12/19/2022] Open
Abstract
Background Systemic treatment has proven to improve physical symptoms in patients with advanced cancer. Relationship between quality of life (QoL) or symptom burden (SYB) and treatment efficacy (tumour response and survival) is poorly described. Therefore, we evaluated the predictive value of pretreatment QoL and SYB on treatment outcomes. Methods Eligible patients had metastatic gastrointestinal cancers and were about to receive 1st/2nd line palliative chemotherapy. 47 patients were consecutively enrolled. QoL and SYB were assessed by EORTC QLQ-C30 and MSKCC MSAS questionnaires before treatment and after first response evaluation after 8–12 weeks. Logistic regression analysis of QoL and SYB for prediction of objective treatment efficacy was performed. Patients were categorized according to response rate (RR) based on RECIST1.1 and progression free survival (PFS). PFS was categorized by a ratio (individual PFS/expected PFS) in above median (ratio ≥ 1) or below median PFS (ratio < 1). QoL and SYB were analysed for RR groups (partial response, stable or progressive disease) and PFS ratio (PFSR). Results Objective response to chemotherapy and increase in PFS were associated with better pretreatment QoL and less SYB. Patients with future objective treatment efficacy (PFSR ≥ 1) evidenced clinically relevant better role/emotional/cognitive/social functioning and less fatigue and appetite loss at baseline in comparison to PFSR < 1 (>10 points difference). Lowest scores in all functioning scales at treatment start were seen in patients with future PFSR < 1. Global health status (EORTC), PSYCH subscale and global distress index (MSAS) predicted PFSR, even if adjusted for gender, age, cancer type, ECOG and line of treatment (p < 0.05). Interestingly, improved QoL and SYB (subjective benefit) were noted even in patients with worse pretreatment status and no objective tumour response. Conclusion Future non-responders seem to show distinct QoL patterns before chemotherapy. This may facilitate early detection of patients deriving less or even no benefit from treatment regarding prolongation of survival. Even in patients with primarily progressive disease QoL and SYB may improve during treatment. Integration of QoL and SYB assessment into decision-making about palliative chemotherapy seem to be an important approach to improve patient outcome and should be further evaluated.
Collapse
Affiliation(s)
- Anna Creutzfeldt
- Department of Oncology, Hematology, BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumour Center - University Cancer Center Hamburg, Martinistr. 52, 20246, Hamburg, Germany.
| | - Anna Suling
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Karin Oechsle
- Department of Oncology, Hematology, BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Anja Mehnert
- Department of Medical Psychology and Medical Sociology, University Medical Center Leipzig, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Djordje Atanackovic
- Department of Oncology, Hematology, BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melanie Kripp
- Department of Oncology/Hematology, University Hospital Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Dirk Arnold
- Tumour Biology Center Freiburg, Breisacher Straße 117, 79106, Freiburg, Germany
| | - Alexander Stein
- Department of Oncology, Hematology, BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumour Center - University Cancer Center Hamburg, Martinistr. 52, 20246, Hamburg, Germany
| | - Julia Quidde
- Department of Oncology, Hematology, BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumour Center - University Cancer Center Hamburg, Martinistr. 52, 20246, Hamburg, Germany
| |
Collapse
|
84
|
Ferrara N, Adamis AP, Albeck M, Sredni B. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov 2016; 15:385-403. [PMID: 26775688 DOI: 10.1038/nrd.2015.17] [Citation(s) in RCA: 723] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The targeting of vascular endothelial growth factor A (VEGFA), a crucial regulator of both normal and pathological angiogenesis, has revealed innovative therapeutic approaches in oncology and ophthalmology. The first VEGFA inhibitor, bevacizumab, was approved by the US Food and Drug Administration in 2004 for the first-line treatment of metastatic colorectal cancer, and the first VEGFA inhibitors in ophthalmology, pegaptanib and ranibizumab, were approved in 2004 and 2006, respectively. To mark this tenth anniversary of anti-VEGFA therapy, we discuss the discovery of VEGFA, the successes and challenges in the development of VEGFA inhibitors and the impact of these agents on the treatment of cancers and ophthalmic diseases.
Collapse
Affiliation(s)
- Napoleone Ferrara
- University of California, San Diego, La Jolla, California, 92093, USA
| | | | | | | |
Collapse
|
85
|
Büning H, Hacker UT. Inhibitors of Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 917:261-85. [DOI: 10.1007/978-3-319-32805-8_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
86
|
Wasserstrum Y, Kornowski R, Raanani P, Leader A, Pasvolsky O, Iakobishvili Z. Hypertension in cancer patients treated with anti-angiogenic based regimens. CARDIO-ONCOLOGY 2015; 1:6. [PMID: 33530150 PMCID: PMC7837153 DOI: 10.1186/s40959-015-0009-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/25/2015] [Indexed: 02/08/2023]
Abstract
New anti-cancer drugs that inhibit the vascular endothelial growth factor (VEGF) signaling pathway are highly effective in the treatment of solid tumors, however concerns remain regarding their cardiovascular safety. The most common side effect of VEGF signaling pathway (VSP) inhibition is the development of systemic hypertension. We review the incidence, possible mechanisms, significance and management of hypertension in patients treated with VSP inhibitors.
Collapse
Affiliation(s)
- Yishay Wasserstrum
- Department of Cardiology, Rabin Medical Center, Petah Tikva, 49100, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, Petah Tikva, 49100, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pia Raanani
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avi Leader
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pasvolsky
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zaza Iakobishvili
- Department of Cardiology, Rabin Medical Center, Petah Tikva, 49100, Israel. .,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
87
|
Majithia N, Grothey A. Regorafenib in the treatment of colorectal cancer. Expert Opin Pharmacother 2015; 17:137-45. [DOI: 10.1517/14656566.2016.1118054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
88
|
Symonds RP, Gourley C, Davidson S, Carty K, McCartney E, Rai D, Banerjee S, Jackson D, Lord R, McCormack M, Hudson E, Reed N, Flubacher M, Jankowska P, Powell M, Dive C, West CML, Paul J. Cediranib combined with carboplatin and paclitaxel in patients with metastatic or recurrent cervical cancer (CIRCCa): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Oncol 2015; 16:1515-1524. [PMID: 26474517 PMCID: PMC4705431 DOI: 10.1016/s1470-2045(15)00220-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Patients treated with standard chemotherapy for metastatic or relapsed cervical cancer respond poorly to conventional chemotherapy (response achieved in 20-30% of patients) with an overall survival of less than 1 year. High tumour angiogenesis and high concentrations of intratumoural VEGF are adverse prognostic features. Cediranib is a potent tyrosine kinase inhibitor of VEGFR1, 2, and 3. In this trial, we aimed to assess the effect of the addition of cediranib to carboplatin and paclitaxel chemotherapy in patients with metastatic or recurrent cervical cancer. METHODS In this randomised, double-blind, placebo-controlled phase 2 trial, which was done in 17 UK cancer treatment centres, patients aged 18 years or older initially diagnosed with metastatic carcinoma or who subsequently developed metastatic disease or local pelvic recurrence after radical treatment that was not amenable to exenterative surgery were recruited. Eligible patients received carboplatin AUC of 5 plus paclitaxel 175 mg/m(2) by infusion every 3 weeks for a maximum of six cycles and were randomised centrally (1:1) through a minimisation approach to receive cediranib 20 mg or placebo orally once daily until disease progression. The stratification factors were disease site, disease-free survival after primary therapy or primary stage IVb disease, number of lines of previous treatment, Eastern Cooperative Oncology Group performance status, and investigational site. All patients, investigators, and trial personnel were masked to study drug allocation. The primary endpoint was progression-free survival. Efficacy analysis was by intention to treat, and the safety analysis included all patients who received at least one dose of study drug. This trial is registered with the ISCRTN registry, number ISRCTN23516549, and has been completed. FINDINGS Between Aug 19, 2010, and July 27, 2012, 69 patients were enrolled and randomly assigned to cediranib (n=34) or placebo (n=35). After a median follow-up of 24·2 months (IQR 21·9-29·5), progression-free survival was longer in the cediranib group (median 8·1 months [80% CI 7·4-8·8]) than in the placebo group (6·7 months [6·2-7·2]), with a hazard ratio (HR) of 0·58 (80% CI 0·40-0·85; one-sided p=0·032). Grade 3 or worse adverse events that occurred in the concurrent chemotherapy and trial drug period in more than 10% of patients were diarrhoea (five [16%] of 32 patients in the cediranib group vs one [3%] of 35 patients in the placebo group), fatigue (four [13%] vs two [6%]), leucopenia (five [16%] vs three [9%]), neutropenia (10 [31%] vs four [11%]), and febrile neutropenia (five [16%] vs none). The incidence of grade 2-3 hypertension was higher in the cediranib group than in the control group (11 [34%] vs four [11%]). Serious adverse events occurred in 18 patients in the placebo group and 19 patients in the cediranib group. INTERPRETATION Cediranib has significant efficacy when added to carboplatin and paclitaxel in the treatment of metastatic or recurrent cervical cancer. This finding was accompanied by an increase in toxic effects (mainly diarrhoea, hypertension, and febrile neutropenia). FUNDING Cancer Research UK and AstraZeneca.
Collapse
Affiliation(s)
- R Paul Symonds
- Department of Cancer Studies, University of Leicester, Leicester, UK.
| | - Charlie Gourley
- University of Edinburgh Cancer Research UK Centre, MRC IGMM, Edinburgh, UK
| | | | - Karen Carty
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Elaine McCartney
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Debbie Rai
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Susana Banerjee
- Gynaecology Unit, Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - David Jackson
- Institute of Oncology, Leeds Cancer Centre, St James's University Hospital, Leeds UK
| | - Rosemary Lord
- Clatterbridge Centre for Oncology, University of Liverpool, Liverpool, UK
| | - Mary McCormack
- Oncology Department, University College Hospital, London, UK
| | | | - Nicholas Reed
- Beatson West of Scotland Cancer Centre, Gartnavel General Hospital, Glasgow, UK
| | | | - Petra Jankowska
- Musgrove Park Hospital, Taunton and Somerset NHS Foundation Trust, Taunton, UK
| | | | - Caroline Dive
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | | | - James Paul
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
89
|
Grapsa D, Syrigos K, Saif MW. Bevacizumab in combination with fluoropyrimidine-irinotecan- or fluoropyrimidine-oxaliplatin-based chemotherapy for first-line and maintenance treatment of metastatic colorectal cancer. Expert Rev Anticancer Ther 2015; 15:1267-81. [PMID: 26506906 DOI: 10.1586/14737140.2015.1102063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite a slight decrease in mortality rates, recent advances in screening methods, diagnosis and overall improved therapeutic options, colorectal cancer (CRC) remains among the leading causes of cancer-related death worldwide. The major cause is the mortality related to metastatic status of CRC. Increasing clinical evidence derived from randomized trials strongly suggests that the efficacy of standard cytotoxic agents, including various combinations of 5-fluoouracil (5-FU)/leucovorin (LV), capecitabine, irinotecan and oxaliplatin, may be significantly augmented with concomitant administration of molecular agents targeting the vascular endothelial growth factor (VEGF) signaling pathways, such as bevacizumab. Herein, we critically discuss the current data on the efficacy and safety profile of bevacizumab in combination with fluoropyrimidine-based chemotherapy for first-line and maintenance treatment of metastatic CRC and briefly comment on existing controversies and future perspectives.
Collapse
Affiliation(s)
- Dimitra Grapsa
- a Oncology Unit, 3rd Department of Medicine, "Sotiria" General Hospital , Athens University School of Medicine , Athens , Greece
| | - Konstantinos Syrigos
- a Oncology Unit, 3rd Department of Medicine, "Sotiria" General Hospital , Athens University School of Medicine , Athens , Greece
| | - Muhammad Wasif Saif
- a Oncology Unit, 3rd Department of Medicine, "Sotiria" General Hospital , Athens University School of Medicine , Athens , Greece
| |
Collapse
|
90
|
Rosa B, de Jesus JP, de Mello EL, Cesar D, Correia MM. Effectiveness and safety of monoclonal antibodies for metastatic colorectal cancer treatment: systematic review and meta-analysis. Ecancermedicalscience 2015; 9:582. [PMID: 26557880 PMCID: PMC4631576 DOI: 10.3332/ecancer.2015.582] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The effectiveness of chemotherapy (CT) for select cases of metastatic colorectal cancer (MCRC) has been well established in the literature, however, it provides limited benefits and in many cases constitutes a treatment with high toxicity. The use of specific molecular biological treatments with monoclonal antibodies (MA) has been shown to be relevant, particularly for its potential for increasing the response rate of the host to the tumour, as these have molecular targets present in the cancerous cells and their microenvironment thereby blocking their development. The combination of MA and CT can bring a significant increase in the rate of resectability of metastases, the progression-free survival (PFS), and the global survival (GS) in MCRC patients. OBJECTIVE To assess the effectiveness and safety of MA in the treatment of MCRC. METHODS A systematic review was carried out with a meta-analysis of randomised clinical trials comparing the use of cetuximab, bevacizumab, and panitumumab in the treatment of MCRC. RESULTS Sixteen randomised clinical trials were selected. The quality of the evidence on the question was considered moderate and data from eight randomised clinical trials were included in this meta-analysis. The GS and PFS were greater in the groups which received the MA associated with CT, however, the differences were not statistically significant between the groups (mean of 17.7 months versus 17.1 months; mean difference of 1.09 (CI: 0.10-2.07); p = 0.84; and 7.4 versus 6.9 months. mean difference of 0.76 (CI: 0.08-1.44); p = 0.14 respectively). The meta-analysis was not done for any of the secondary outcomes. CONCLUSION The addition of MA to CT for patients with metastatic colorectal cancer does not prolong GS and PFS.
Collapse
Affiliation(s)
- Bruno Rosa
- Instituto Nacional de Câncer, Rio de Janeiro 20230-130, Brazil
| | | | | | - Daniel Cesar
- Instituto Nacional de Câncer, Rio de Janeiro 20230-130, Brazil
| | - Mauro M Correia
- Instituto Nacional de Câncer, Rio de Janeiro 20230-130, Brazil
| |
Collapse
|
91
|
Stone A, Macpherson E, Smith A, Jennison C. Model free audit methodology for bias evaluation of tumour progression in oncology. Pharm Stat 2015; 14:455-63. [PMID: 26435269 DOI: 10.1002/pst.1707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 06/15/2015] [Accepted: 08/11/2015] [Indexed: 11/12/2022]
Abstract
Many oncology studies incorporate a blinded independent central review (BICR) to make an assessment of the integrity of the primary endpoint, progression free survival. Recently, it has been suggested that, in order to assess the potential for bias amongst investigators, a BICR amongst only a sample of patients could be performed; if evidence of bias is detected, according to a predefined threshold, the BICR is then assessed in all patients, otherwise, it is concluded that the sample was sufficient to rule out meaningful levels of bias. In this paper, we present an approach that adapts a method originally created for defining futility bounds in group sequential designs. The hazard ratio ratio, the ratio of the hazard ratio (HR) for the treatment effect estimated from the BICR to the corresponding HR for the investigator assessments, is used as the metric to define bias. The approach is simple to implement and ensures a high probability that a substantial true bias will be detected. In the absence of bias, there is a high probability of accepting the accuracy of local evaluations based on the sample, in which case an expensive BICR of all patients is avoided. The properties of the approach are demonstrated by retrospective application to a completed Phase III trial in colorectal cancer. The same approach could easily be adapted for other disease settings, and for test statistics other than the hazard ratio.
Collapse
Affiliation(s)
| | | | - Ann Smith
- AstraZeneca, Alderley Park, Macclesfield, UK
| | | |
Collapse
|
92
|
Van Cutsem E, Prenen H, D'Haens G, Bennouna J, Carrato A, Ducreux M, Bouché O, Sobrero A, Latini L, Staines H, Oum'Hamed Z, Dressler H, Studeny M, Capdevila J. A phase I/II, open-label, randomised study of nintedanib plus mFOLFOX6 versus bevacizumab plus mFOLFOX6 in first-line metastatic colorectal cancer patients. Ann Oncol 2015; 26:2085-91. [PMID: 26272806 DOI: 10.1093/annonc/mdv286] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/27/2015] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND This randomised, open-label, phase I/II study evaluated the efficacy and safety of nintedanib, an oral, triple angiokinase inhibitor, combined with chemotherapy, relative to bevacizumab plus chemotherapy as first-line therapy in patients with metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Patients with histologically confirmed mCRC (adenocarcinoma), an Eastern Cooperative Oncology Group performance status ≤ 2 and adequate organ function were included. Patients were randomised 2:1 to receive nintedanib 150 mg or 200 mg b.i.d. plus mFOLFOX6 (oxaliplatin 85 mg/m(2), l-leucovorin 200 mg/m(2) or d,l-leucovorin 400 mg/m(2), 5-fluoruracil bolus 400 mg/m(2) followed by 2400 mg/m(2), every 2 weeks) or bevacizumab (5 mg/kg every 2 weeks) plus mFOLFOX6. During phase I, patients underwent a 3 + 3 dose-escalation schema to determine the maximum tolerated dose (MTD) of nintedanib in combination with mFOLFOX6. The primary end point was progression-free survival (PFS) rate at 9 months. Objective response (OR) was a secondary end point. RESULTS The nintedanib recommended phase II dose was 200 mg b.i.d. plus mFOLFOX6 based on safety data from phase I (n = 12). Of 128 patients randomised in the phase II part, 126 received treatment (nintedanib plus mFOLFOX6, n = 85; bevacizumab plus mFOLFOX6, n = 41). PFS at 9 months was 62.1% with nintedanib and 70.2% with bevacizumab [difference: -8.1% (95% confidence interval -27.8 to 11.5)]. Confirmed ORs were recorded in 63.5% and 56.1% of patients in the nintedanib and bevacizumab groups, respectively. The incidence of adverse events (AEs) considered related to treatment was 98.8% with nintedanib and 97.6% with bevacizumab; the incidence of serious AEs was 37.6% with nintedanib and 53.7% with bevacizumab. The pharmacokinetics of nintedanib and the components of mFOLFOX6 were unaffected by their combination. CONCLUSIONS Nintedanib in combination with mFOLFOX6 showed efficacy as first-line therapy in patients with mCRC with a manageable safety profile and further studies in this population are warranted.
Collapse
Affiliation(s)
- E Van Cutsem
- Digestive Oncology, University Hospitals Leuven and KULeuven, Leuven, Belgium
| | - H Prenen
- Digestive Oncology, University Hospitals Leuven and KULeuven, Leuven, Belgium
| | - G D'Haens
- Department of Gastroenterology and Hepatology, Academic Medical Centre, Amsterdam, The Netherlands Imelda GI Clinical Research Centre, Bonheiden, Belgium
| | - J Bennouna
- Department of Medical Oncology, Centre René Gauducheau, Nantes, France
| | - A Carrato
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - M Ducreux
- Gustave-Roussy Cancer Campus, Institut Gustave Roussy, Paris University of Paris-Sud, Le Kremlin Bicêtre, Paris
| | - O Bouché
- Department of Digestive Oncology, CHU Reims, Reims, France
| | - A Sobrero
- Department of Medical Oncology, San Martino Hospital, Genoa
| | - L Latini
- Oncology Unit, Macerata Hospital, Macerata, Italy
| | - H Staines
- Medical and Drug Regulatory Affairs, Boehringer Ingelheim France S.A.S., Reims, France
| | - Z Oum'Hamed
- Medical and Drug Regulatory Affairs, Boehringer Ingelheim France S.A.S., Reims, France
| | - H Dressler
- Global Pharmacovigilance, Boehringer Ingelheim, Ingelheim, Germany
| | - M Studeny
- Division of Medicine/Department of Clinical Development, Boehringer Ingelheim, Vienna, Austria
| | - J Capdevila
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
93
|
Jary M, Borg C, Bouché O, Kim S, André T, Bennouna J. [Anti-angiogenic treatments in metastatic colorectal cancer: Does a continuous angiogenic blockade make sense?]. Bull Cancer 2015; 102:758-71. [PMID: 26232849 DOI: 10.1016/j.bulcan.2015.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 09/10/2014] [Indexed: 01/07/2023]
Abstract
Ten years after the approval of bevacizumab in colorectal cancer patients, results from ML18147 and CORRECT studies have recently demonstrated the possibility to target angiogenesis in patients previously exposed to anti-VEGF. An increasing number of anti-angiogenic treatments are now available, however, no biomarker has yet succeeded in rationalizing our therapeutic strategies. Nevertheless, several lessons have been learned from preclinical and pivotal clinical studies. The first clinical trials demonstrated a survival benefit, adding VEGFA targeting monoclonal antibodies to chemotherapy in metastatic colorectal cancer patients (AVF2107, ECOG 3200). Many phase III clinical trials confirmed the interest of this strategy, in combination with chemotherapies containing irinotecan, oxaliplatin, or with 5-fluorouracil in monotherapy. To date, such results have not been reproduced with tyrosine kinase inhibitors targeting the angiogenesis pathways, with an increasing rate of chemotherapy related toxicities. Clinical trials performed in the adjuvant setting (AVANT, NSABPC08) failed to demonstrate any efficacy of the anti-VEGFA treatments on the micrometastatic disease, encouraging its prescription in the unresectable cases. On the other hand, a continuous inhibition of angiogenesis during the course of the metastatic disease was shown to be feasible and to extend colon cancer patient's survival in two recent randomized trials. For these patients, the continuation of bevacizumab beyond progression in first line improves overall survival. Lastly, results achieved by the CORRECT and CONCUR studies demonstrated that anti-angiogenics might be effective in colorectal cancers resistant to chemotherapy. This review presents the main results of preclinical and clinical studies sustaining the prescription of anti-angiogenics in metastatic colorectal cancers. The future challenge is to promote the development of biomarkers to enable the stratification of the different therapeutic strategies.
Collapse
Affiliation(s)
- Marine Jary
- CHRU de Besançon, IRFC, service d'oncologie médicale, 25000 Besançon, France.
| | - Christophe Borg
- CHRU de Besançon, IRFC, service d'oncologie médicale, 25000 Besançon, France; Unité Inserm 1098, université de Franche-Comté, 25000 Besançon, France
| | - Olivier Bouché
- CHU de Reims, services de gastroentérologie et d'oncologie médicale, 51100 Reims, France
| | - Stéfano Kim
- CHRU de Besançon, IRFC, service d'oncologie médicale, 25000 Besançon, France
| | - Thierry André
- Hôpital Saint-Antoine, service d'oncologie médicale, 75012 Paris, France
| | - Jaafar Bennouna
- Institut de cancérologie de l'Ouest, Nantes-Angers, 44805 Nantes, France
| |
Collapse
|
94
|
Lee YC, Michael M, Zalcberg JR. An overview of experimental and investigational multikinase inhibitors for the treatment of metastatic colorectal cancer. Expert Opin Investig Drugs 2015. [PMID: 26212373 DOI: 10.1517/13543784.2015.1070483] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
95
|
Li T, Wang B, Wang Z, Ragaz J, Zhang J, Sun S, Cao J, Lv F, Wang L, Zhang S, Ni C, Wu Z, Xie J, Hu X. Bevacizumab in Combination with Modified FOLFOX6 in Heavily Pretreated Patients with HER2/Neu-Negative Metastatic Breast Cancer: A Phase II Clinical Trial. PLoS One 2015; 10:e0133133. [PMID: 26186012 PMCID: PMC4506015 DOI: 10.1371/journal.pone.0133133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/19/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Bevacizumab combined with modified FOLFOX6 is a standard regimen for colorectal cancer. The present study was to determine the efficacy and safety of bevacizumab-modified FOLFOX6 regimen in heavily pretreated patients with human epidermal growth factor receptor 2 (HER2/neu)-negative MBC. METHODS Bevacizumab, 5 mg/kg every two weeks or 7.5 mg/kg every three weeks, was administered with modified FOLFOX6 (oxaliplatin 85 mg/m2, leucovorin 400 mg/m2, 5-FU 400 mg/m2 on day 1, followed by 5-FU 2400 mg/m2 intravenous infusion over 46 hours every 2 weeks) to patients who failed at least 1 chemotherapy regimen in the metastatic setting. The primary objective was progression free survival (PFS). Secondary objectives included objective response rate (ORR), clinical benefit rate (CBR), overall survival (OS), safety, and the change of tumor size and Eastern Cooperative Oncology Group (ECOG) performance status. RESULTS 69 patients were enrolled. The median PFS was 6.8 months (95% CI, 5.0 to 8.5 months), ORR was 50.0% and median OS was 10.5 months (95% CI, 7.9 to 13.1 months). Patients showing objective responses had a 4.2-month median PFS gain and 5.7-month median OS gain compared with those who did not (P < 0.05). Grade 3 or 4 adverse events occurring in more than one patient were neutropenia (53/69, 76.8%), leukopenia (36/69, 52.2%), thrombocytopenia (13/69, 18.8%), anemia (3/69, 4.3%) and hypertension (3/69, 4.3%). CONCLUSIONS Adding bevacizumab to modified FOLFOX6 does have significant anti-tumor activity and good safety profile in heavily pretreated HER2/neu-negative MBC patients. Further trials are required to confirm whether the high ORR can translate into a long-term PFS and even OS benefit. TRIAL REGISTRATION www.clinicaltrials.gov NCT01658033.
Collapse
Affiliation(s)
- Ting Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhonghua Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Joseph Ragaz
- Faculty of Medicine, School of Population & Public Health, University of British Columbia, Vancouver, B.C., V6T 1Z4, Canada
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Si Sun
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fangfang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Leiping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Sheng Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chen Ni
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenhua Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jie Xie
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- * E-mail:
| |
Collapse
|
96
|
Hecht JR, Mitchell EP, Yoshino T, Welslau M, Lin X, Chow Maneval E, Paolini J, Lechuga MJ, Kretzschmar A. 5-Fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) plus sunitinib or bevacizumab as first-line treatment for metastatic colorectal cancer: a randomized Phase IIb study. Cancer Manag Res 2015; 7:165-73. [PMID: 26109878 PMCID: PMC4474399 DOI: 10.2147/cmar.s61408] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Sunitinib is an oral inhibitor of tyrosine kinase receptors implicated in tumor proliferation, angiogenesis, and metastasis. In this randomized, multicenter, open-label Phase IIb study, sunitinib plus mFOLFOX6 (oxaliplatin plus leucovorin plus 5-fluorouracil) was compared with bevacizumab plus mFOLFOX6 as first-line therapy in patients with metastatic colorectal cancer. Methods Patients were stratified by performance status, baseline lactate dehydrogenase level, and prior adjuvant treatment, and randomized 1:1 to receive sunitinib 37.5 mg/day for 4 weeks on and 2 weeks off plus mFOLFOX6 every 2 weeks or bevacizumab 5 mg/kg every 2 weeks plus mFOLFOX6 every 2 weeks. The primary endpoint was progression-free survival. Secondary endpoints included objective response rate, overall survival, safety, and quality of life. Results Enrollment was closed early following accrual of 191 patients, based on an interim analysis showing an inferior trend in the primary progression-free survival efficacy endpoint for sunitinib. Ninety-six patients were randomized to sunitinib plus mFOLFOX6 and 95 to bevacizumab plus mFOLFOX6. Median progression-free survival was 9.3 months and 15.4 months, respectively, but the objective response rate was similar between the study arms. Median overall survival was 23.7 months and 34.1 months, respectively. Dose reductions and interruptions were more common with sunitinib. Hematologic toxicity was more common in the sunitinib arm. Conclusion While the results of the sunitinib arm are comparable with those of previously reported FOLFOX combinations, the sunitinib-based combination was associated with more toxicity than that observed with bevacizumab and mFOLFOX6. The bevacizumab arm had an unexpectedly good outcome, and was much better than that seen in the Phase III trials. Combination therapy with sunitinib plus mFOLFOX6 is not recommended for patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
| | - Edith P Mitchell
- Kimmel Cancer Center of Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Manfred Welslau
- Onkologische Praxis Klausmann/Welslau, Aschaffenburg, Germany
| | - Xun Lin
- Pfizer Oncology, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
97
|
Zhao Y, Adjei AA. Targeting Angiogenesis in Cancer Therapy: Moving Beyond Vascular Endothelial Growth Factor. Oncologist 2015; 20:660-73. [PMID: 26001391 DOI: 10.1634/theoncologist.2014-0465] [Citation(s) in RCA: 425] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Angiogenesis, or the formation of new capillary blood vessels, occurs primarily during human development and reproduction; however, aberrant regulation of angiogenesis is also a fundamental process found in several pathologic conditions, including cancer. As a process required for invasion and metastasis, tumor angiogenesis constitutes an important point of control of cancer progression. Although not yet completely understood, the complex process of tumor angiogenesis involves highly regulated orchestration of multiple signaling pathways. The proangiogenic signaling molecule vascular endothelial growth factor (VEGF) and its cognate receptor (VEGF receptor 2 [VEGFR-2]) play a central role in angiogenesis and often are highly expressed in human cancers, and initial clinical efforts to develop antiangiogenic treatments focused largely on inhibiting VEGF/VEGFR signaling. Such approaches, however, often lead to transient responses and further disease progression because angiogenesis is regulated by multiple pathways that are able to compensate for each other when single pathways are inhibited. The platelet-derived growth factor (PDGF) and PDGF receptor (PDGFR) and fibroblast growth factor (FGF) and FGF receptor (FGFR) pathways, for example, provide potential escape mechanisms from anti-VEGF/VEGFR therapy that could facilitate resumption of tumor growth. Accordingly, more recent treatments have focused on inhibiting multiple signaling pathways simultaneously. This comprehensive review discusses the limitations of inhibiting VEGF signaling alone as an antiangiogenic strategy, the importance of other angiogenic pathways including PDGF/PDGFR and FGF/FGFR, and the novel current and emerging agents that target multiple angiogenic pathways for the treatment of advanced solid tumors. IMPLICATIONS FOR PRACTICE Significant advances in cancer treatment have been achieved with the development of antiangiogenic agents, the majority of which have focused on inhibition of the vascular endothelial growth factor (VEGF) pathway. VEGF targeting alone, however, has not proven to be as efficacious as originally hoped, and it is increasingly clear that there are many interconnected and compensatory pathways that can overcome VEGF-targeted inhibition of angiogenesis. Maximizing the potential of antiangiogenic therapy is likely to require a broader therapeutic approach using a new generation of multitargeted antiangiogenic agents.
Collapse
Affiliation(s)
- Yujie Zhao
- Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Alex A Adjei
- Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
98
|
Petrelli F, Coinu A, Ghilardi M, Cabiddu M, Zaniboni A, Barni S. Efficacy of oxaliplatin-based chemotherapy + bevacizumab as first-line treatment for advanced colorectal cancer: a systematic review and pooled analysis of published trials. Am J Clin Oncol 2015; 38:227-33. [PMID: 25806713 DOI: 10.1097/coc.0b013e3182a2d7b8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Oxaliplatin and either capecitabine or infusional/bolus 5-fluorouracil (5FU)-based chemotherapy + bevacizumab (XELOX + B and FOLFOX + B) represent 2 of the approved first-line treatments for advanced colorectal cancer (CRC). However, the addition of B did not offer a survival benefit compared with FOLFOX/XELOX alone in the phase III, NO16966 trial. The aim of this review was to aggregate all published data on the efficacy of XELOX and FOLFOX-B in prospective and retrospective studies as first-line therapy for stage IV CRC. METHODS We performed a systematic review, through PubMed and EMBASE, of all published studies exploring the efficacy of fluoropyrimidines + oxaliplatin + B-based chemotherapy as first-line chemotherapy in advanced CRC patients. Pooled estimates of the response rates, weighted medians of progression-free survival, and overall survival from all FOLFOX + B and XELOX + B arms were calculated. RESULTS A total of 25 studies were retrieved, with a total of 7878 patients. Overall, the pooled response rates (n=20 publications) was 49.1%. The median progression-free survival and overall survival (n=21 and 22 publications, respectively) were 10.3 and 23.7 months, respectively. The pooled median rate of surgical resection of metastases (n=13 publications) was 14%. CONCLUSIONS XELOX + B and FOLFOX + B are active combinations as first-line treatment of advanced CRC. The efficacy is confirmed for the first time from this pooled analysis of 25 trials. Both the XELOX + B and the FOLFOX + B arms represent 2 of the cornerstone combinations when B is used as first-line therapy.
Collapse
Affiliation(s)
- Fausto Petrelli
- *Oncology Department, Medical Oncology Unit, Azienda Ospedaliera Treviglio-Caravaggio, Treviglio (BG) †UO Oncologia, Dipartimento Oncologico, Istituto Ospedaliero Fondazione Poliambulanza, Brescia (BS), Italy
| | | | | | | | | | | |
Collapse
|
99
|
Lin Z, Yang Y, Huang Y, Liang J, Lu F, Lao X. Vascular endothelial growth factor receptor tyrosine kinase inhibitors versus bevacizumab in metastatic colorectal cancer: A systematic review and meta-analysis. Mol Clin Oncol 2015; 3:959-967. [PMID: 26171215 DOI: 10.3892/mco.2015.572] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/24/2015] [Indexed: 01/23/2023] Open
Abstract
Bevacizumab has demonstrated a survival benefit in patients with metastatic colorectal cancer (mCRC) when combined with chemotherapy. Several randomized clinical trials comparing the efficacy and toxicity of vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) against bevacizumab have been reported. The present meta-analysis was conducted to identify the potentially significant benefit of the combined treatment regimens in patients with mCRC. PubMed, Embase and Cochrane Library databases were searched for the randomized controlled trials published on or before September 2014, which compared the efficacy and toxicity of VEGFR TKIs with bevacizumab in combination with chemotherapy in patients with mCRC. The primary endpoints included progression-free survival (PFS), overall survival (OS) and overall response rate (ORR), and secondary endpoints were the toxicity profiles. Relative risks (RRs) with 95% confidence intervals (CIs) for response rate and adverse events (AEs) were calculated, as well as hazard ratios (HRs) for PFS and OS. The final analysis included 4 studies comprising a total of 1,929 intent-to-treat patients with mCRC, which compared VEGFR TKIs (cediranib and axitinib) plus chemotherapy with bevacizumab plus chemotherapy. Results demonstrated that VEGFR TKIs plus chemotherapy significantly resulted in a modest but significantly shorter PFS [hazard ratio (HR), 1.12; 95% CI, 1.00-1.25; P=0.05] compared with that of bevacizumab plus chemotherapy but not in OS (HR, 1.10; 95% CI, 0.88-1.17; P=0.87) and ORR (RR, 0.95; 95% CI, 0.85-1.05; P=0.30). VEGFR TKIs treatment showed a less favorable AE profile compared with bevacizumab, with higher rates of grade-III/IV diarrhea, fatigue, hypertension, neutropenia and thrombocytopenia, whereas a higher incidence of peripheral neuropathy associated with the bevacizumab group was observed. In conclusion, the addition of VEGFR TKIs to chemotherapy resulted in a modest but significantly shorter PFS but not in OS and ORR compared with bevacizumab. The VEGFR TKIs group showed a less favorable AE profile with higher rates of diarrhea, fatigue, hypertension, neutropenia and thrombocytopenia, whereas a higher incidence of peripheral neuropathy associated with the bevacizumab was observed.
Collapse
Affiliation(s)
- Zexin Lin
- Department of General Surgery, The First Clinical Medicine School of Jinan University, Affiliated Shantou Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Yilin Yang
- Department of General Surgery, The First Clinical Medicine School of Jinan University, Guangzhou, Guangdong, P.R. China
| | - Yongliang Huang
- Department of General Surgery, The First Clinical Medicine School of Jinan University, Guangzhou, Guangdong, P.R. China
| | - Junjie Liang
- Department of General Surgery, The First Clinical Medicine School of Jinan University, Guangzhou, Guangdong, P.R. China
| | - Fang Lu
- Department of Orthopedics, The First Clinical Medicine School of Jinan University, Guangzhou, Guangdong, P.R. China
| | - Xuejun Lao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
100
|
Nakayama N, Sato A, Tanaka S, Shimada K, Konishi K, Sasaki E, Hibi K, Ichikawa H, Kikuchi Y, Sakuyama T, Sekikawa T, Hayashi K, Nishina H. A phase II study of bevacizumab with modified OPTIMOX1 as first-line therapy for metastatic colorectal cancer: the TCOG-GI 0802 study. Invest New Drugs 2015; 33:954-62. [PMID: 25937430 DOI: 10.1007/s10637-015-0239-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/31/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Although bevacizumab plus FOLFOX is a standard treatment for metastatic colorectal cancer, oxaliplatin must be withdrawn in many patients because of cumulative neurotoxicity. We postulated that a reduced dose of oxaliplatin and modified treatment schedule would prolong the time to treatment failure and evaluated bevacizumab combined with a modified OPTIMOX1 regimen (mOPTIMOX1, oxaliplatin dose: 85 mg/m(2)). METHODS Eligible patients had a histologically confirmed diagnosis of metastatic colorectal cancer and a performance status of 0-1. Patients were excluded if they had grade 1 or higher peripheral sensory neuropathy or had previously received chemotherapy for metastatic colorectal cancer. Patients received bevacizumab plus mFOLFOX6 every 2 weeks for 6 cycles, followed by 12 cycles of a simplified biweekly regimen of leucovorin and fluorouracil (sLV5FU2) plus bevacizumab. Oxaliplatin was then reintroduced, and bevacizumab plus mFOLFOX6 was continued until progressive disease. RESULTS The median duration of disease control was 11.7 months (95 % confidence interval [CI], 9.7-13.5 months). The median overall survival was 23.1 months (95 % CI, 18.8-27.9 months). The overall response rate according to both the RECIST and WHO criteria was 51.3 %. The most common grade 3 or 4 toxicities were neutropaenia (32.5 %), hypertension (17.5 %), leukocytopaenia, sensory neuropathy, and diarrhoea (10.0 %). There were no treatment-related deaths. CONCLUSIONS Bevacizumab plus mFOLFOX6 was well tolerated, and patients could continue chemotherapy for longer than with conventional FOLFOX regimens. This regimen might be an effective treatment option for patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Norisuke Nakayama
- Department of Gastroenterology, Kanagawa Cancer Center Hospital, 2-3-2,Nakao, Asahi-ku, Yokohama City, 241-8515, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|