1
|
Tanzawa S, Yoshioka H, Misumi T, Miyauchi E, Ninomiya K, Murata Y, Takeshita M, Kinoshita F, Fujishita T, Sugawara S, Kawashima Y, Hashimoto K, Mori M, Miyanaga A, Hayashi A, Tanaka H, Honda R, Nojiri M, Sato Y, Hata A, Ishikawa N, Kozuki T, Kawamura T, Saito G, Yamaguchi T, Asada K, Tetsumoto S, Tanaka H, Watanabe S, Umeda Y, Yamaguchi K, Nishii K, Tsuruno K, Misumi Y, Kuraishi H, Yoshihara K, Nakao A, Kubo A, Yokoyama T, Watanabe K, Seki N. Clinical impact of hypomagnesemia induced by necitumumab plus cisplatin and gemcitabine treatment in patients with advanced lung squamous cell carcinoma: a subanalysis of the NINJA study. Ther Adv Med Oncol 2025; 17:17588359251318850. [PMID: 39957806 PMCID: PMC11829289 DOI: 10.1177/17588359251318850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Background The clinical impact of hypomagnesemia induced by necitumumab plus gemcitabine and cisplatin (GCN) as a second-line or later therapy is unclear. Objective We aimed to evaluate the clinical characteristics and survival impact of hypomagnesemia induced by this therapy. Design This was a sub-analysis of the retrospective multicenter NINJA study. Methods Among the 93 patients enrolled in the NINJA study, this subanalysis included 75 patients with baseline serum magnesium concentrations. Results The incidence of grade ⩾2 hypomagnesemia was 18.0% in the patients with normal baseline serum magnesium concentrations and 42.8% in those with low concentrations (p = 0.073). The discontinuation rates of GCN treatment owing to hypomagnesemia in each group were 0% and 7.1%, respectively (p = 0.187). The number of necitumumab doses and severity of hypomagnesemia were positively correlated (r = 0.389, p < 0.001). Patients who developed hypomagnesemia in fewer than 21 days after the first dose of GCN (n = 12) had significantly poorer progression-free survival (PFS) than those without the condition (n = 63; median: 4.1 vs 4.4 months, p = 0.048). A similar trend was observed for OS (median: 9.7 vs 15.7 months, p = 0.062). These results were maintained after multivariate analyses (PFS: hazard ratio (HR) 2.46, p = 0.014; OS: HR 2.78, p = 0.021). Conclusion GCN as a second-line or later therapy may be tolerable regardless of the patient's baseline serum magnesium concentration. On the other hand, early serum magnesium reduction with this therapy is associated with a poor prognosis. However, caution should be needed because our results lacked sufficient information for confounding variables other than those analyzed here that may influence the correlation between hypomagnesemia and survival.
Collapse
Affiliation(s)
- Shigeru Tanzawa
- Division of Medical Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Hiroshige Yoshioka
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| | - Toshihiro Misumi
- Department of Data Science, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Kiichiro Ninomiya
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Okayama, Japan
| | - Yasunori Murata
- Department of Respiratory Medicine, Ichinomiya Nishi Hospital, Ichinomiya, Aichi, Japan
| | - Masafumi Takeshita
- Department of Respiratory Medicine, Ichinomiya Nishi Hospital, Ichinomiya, Aichi, Japan
| | - Fumihiko Kinoshita
- Department of Thoracic Oncology, NHO Kyushu Cancer Center, Fukuoka, Fukuoka, Japan
| | - Takatoshi Fujishita
- Department of Thoracic Oncology, NHO Kyushu Cancer Center, Fukuoka, Fukuoka, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Miyagi, Japan
| | - Yosuke Kawashima
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Miyagi, Japan
| | - Kazuki Hashimoto
- Department of Thoracic Oncology, NHO Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Masahide Mori
- Department of Thoracic Oncology, NHO Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Akihiko Miyanaga
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Anna Hayashi
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Hisashi Tanaka
- Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Ryoichi Honda
- Department of Respiratory Medicine, Asahi General Hospital, Asahi, Chiba, Japan
| | - Masafumi Nojiri
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Yuki Sato
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Akito Hata
- Division of Thoracic Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Hyogo, Japan
| | - Nobuhisa Ishikawa
- Department of Respiratory Medicine, Hiroshima Prefectural Hospital, Minami-ku, Hiroshima, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology and Medicine, NHO Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Takahisa Kawamura
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Osaka, Japan
| | - Go Saito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Teppei Yamaguchi
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Kazuhiro Asada
- Department of Respiratory Medicine, Shizuoka General Hospital, Aoi-ku, Shizuoka, Japan
| | - Satoshi Tetsumoto
- Department of Respiratory Medicine and Clinical Immunology, Suita Municipal Hospital, Suita, Osaka, Japan
| | - Hiroshi Tanaka
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Niigata, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Yukihiro Umeda
- Department of Respiratory Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida, Fukui, Japan
| | - Kakuhiro Yamaguchi
- Department of Respiratory Medicine, Hiroshima University Hospital, Hiroshima, Hiroshima, Japan
| | - Kazuya Nishii
- Department of Respiratory Medicine, NHO Iwakuni Clinical Center, Iwakuni, Yamaguchi, Japan
| | - Kosuke Tsuruno
- Department of Respiratory Medicine, Iizuka Hospital, Iizuka, Fukuoka, Japan
| | - Yuki Misumi
- Department of Respiratory Medicine, Yokohama Municipal Citizen’s Hospital, Yokohama, Kanagawa, Japan
| | - Hiroshi Kuraishi
- Department of Pulmonary Medicine, Nagano Red Cross Hospital, Nagano, Nagano, Japan
| | - Ken Yoshihara
- Department of Internal Medicine, Division of Medical Oncology and Respiratory Medicine, Shimane University Faculty of Medicine, Izumo, Shimane, Japan
| | - Akira Nakao
- Department of Respiratory Medicine, Fukuoka University Hospital, Fukuoka, Fukuoka, Japan
| | - Akihito Kubo
- Department of Respiratory Medicine and Allergology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Toshihiko Yokoyama
- Department of Respiratory Medicine, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Aichi, Japan
| | - Kana Watanabe
- Department of Respiratory Medicine, Miyagi Cancer Center, Natori, Miyagi, Japan
| | - Nobuhiko Seki
- Division of Medical Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Itabashi-ku, Tokyo, Japan
| |
Collapse
|
2
|
Tydings CW, Singh B, Smith AW, Ledwitch KV, Brown BP, Lovly CM, Walker AS, Meiler J. Analysis of EGFR binding hotspots for design of new EGFR inhibitory biologics. Protein Sci 2024; 33:e5141. [PMID: 39275996 PMCID: PMC11400634 DOI: 10.1002/pro.5141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/16/2024]
Abstract
The epidermal growth factor (EGF) receptor (EGFR) is activated by the binding of one of seven EGF-like ligands to its ectodomain. Ligand binding results in EGFR dimerization and stabilization of the active receptor conformation subsequently leading to activation of downstream signaling. Aberrant activation of EGFR contributes to cancer progression through EGFR overexpression/amplification, modulation of its positive and negative regulators, and/or activating mutations within EGFR. EGFR targeted therapeutic antibodies prevent dimerization and interaction with endogenous ligands by binding the ectodomain of EGFR. However, these antibodies have had limited success in the clinic, partially due to EGFR ectodomain resistance mutations, and are only applicable to a subset of patients with EGFR-driven cancers. These limitations suggest that alternative EGFR targeted biologics need to be explored for EGFR-driven cancer therapy. To this end, we analyze the EGFR interfaces of known inhibitory biologics with determined structures in the context of endogenous ligands, using the Rosetta macromolecular modeling software to highlight the most important interactions on a per-residue basis. We use this analysis to identify the structural determinants of EGFR targeted biologics. We suggest that commonly observed binding motifs serve as the basis for rational design of new EGFR targeted biologics, such as peptides, antibodies, and nanobodies.
Collapse
Affiliation(s)
- Claiborne W. Tydings
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Bhuminder Singh
- Department of Medicine – Division of Gastroenterology, Hepatology, and NutritionVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Adam W. Smith
- Department of Chemistry and BiochemistryTexas Tech UniversityLubbockTexasUSA
| | - Kaitlyn V. Ledwitch
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Benjamin P. Brown
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Christine M. Lovly
- Department of Medicine – Division of Hematology and OncologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Allison S. Walker
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Department of Biological SciencesVanderbilt UniversityNashvilleTennesseeUSA
| | - Jens Meiler
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
- Institute for Drug DiscoveryLeipzig University Medical SchoolLeipzigSACGermany
| |
Collapse
|
3
|
Jiang Y, Zhao M, Tang W, Zheng X. Comparison of systemic treatments for previously treated patients with unresectable colorectal liver metastases: a systematic review and network meta-analysis. Front Oncol 2024; 14:1293598. [PMID: 39050571 PMCID: PMC11266080 DOI: 10.3389/fonc.2024.1293598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Background There is limited evidence of comparative results among different treatments for patients with unresectable colorectal liver metastases (CRLM) who have failed at least one line of previous systemic therapy. We aimed to compare the efficacy of systemic treatments among these patients through this investigation. Methods We collected randomized controlled trials (RCTs) reported in English up until July 2023, from databases including PubMed, Embase, Cochrane Library, ClinicalTrials.gov, and prominent conference databases, for this Bayesian network meta-analysis. Phase II or III trials that evaluated at least two therapeutic regimens were included. Primary outcome was overall survival (OS), secondary outcome was progression-free survival (PFS). Hazards ratios (HRs) with 95% confidence intervals (CIs) were used as effect size. Subgroup analysis was performed based on metastatic sites. The current systematic review protocol was registered on PROSPERO (CRD42023420498). Results 30 RCTs were included, with a total of 13,511 patients. Compared to chemotherapy, multi-targeted therapy (HR 0.57, 95% CI 0.37-0.87) and targeted therapy plus chemotherapy (HR 0.78, 95% CI 0.67-0.91) show significant advantages. Targeted therapy (HR 0.92, 95% CI 0.54-1.57) and local treatment plus chemotherapy (HR 1.03, 95% CI 0.85-1.23) had comparable performance. For patients with liver metastases, TAS-102 plus bevacizumab, aflibercept plus fluorouracil-based combination chemotherapy (CTFU), and bevacizumab plus capecitabine-based combination chemotherapy (CTCA) showed the best outcomes in terms of OS. Bevacizumab plus intensified CTFU, bevacizumab plus CTCA, and HAI followed by single-agent chemotherapy (SingleCT) performed the best regarding PFS. For patients with liver-limited metastases, aflibercept plus CTFU is the optimal choice in OS. For PFS, the best options were HAI followed by SingleCT, aflibercept plus CTFU, and panitumumab plus CTFU. For patients with multiple-site metastases, the best treatments were TAS-102 plus bevacizumab, bevacizumab plus CTCA, bevacizumab plus CTFU, and aflibercept plus CTFU. Conclusion Multi-targeted therapy and targeted therapy plus chemotherapy are the best treatment mechanisms. TAS-102 plus bevacizumab is superior in OS, the combination of anti-VEGF drugs like bevacizumab and aflibercept with standard chemotherapy is the preferred option for CRLM patients.
Collapse
Affiliation(s)
- Yunlin Jiang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingye Zhao
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenxi Tang
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xueping Zheng
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
4
|
Chai Y, Liu JL, Zhang S, Li N, Xu DQ, Liu WJ, Fu RJ, Tang YP. The effective combination therapies with irinotecan for colorectal cancer. Front Pharmacol 2024; 15:1356708. [PMID: 38375031 PMCID: PMC10875015 DOI: 10.3389/fphar.2024.1356708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 01/19/2024] [Indexed: 02/21/2024] Open
Abstract
Colorectal cancer is the third most common type of cancer worldwide and has become one of the major human disease burdens. In clinical practice, the treatment of colorectal cancer has been closely related to the use of irinotecan. Irinotecan combines with many other anticancer drugs and has a broader range of drug combinations. Combination therapy is one of the most important means of improving anti-tumor efficacy and overcoming drug resistance. Reasonable combination therapy can lead to better patient treatment options, and inappropriate combination therapy will increase patient risk. For the colorectal therapeutic field, the significance of combination therapy is to improve the efficacy, reduce the adverse effects, and improve the ease of treatment. Therefore, we explored the clinical advantages of its combination therapy based on mechanism or metabolism and reviewed the rationale basis and its limitations in conducting exploratory clinical trials on irinotecan combination therapy, including the results of clinical trials on the combination potentiation of cytotoxic drugs, targeted agents, and herbal medicine. We hope that these can evoke more efforts to conduct irinotecan in the laboratory for further studies and evaluations, as well as the possibility of more in-depth development in future clinical trials.
Collapse
Affiliation(s)
- Yun Chai
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Jing-Li Liu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Shuo Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Na Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Ding-Qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Wen-Juan Liu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Rui-Jia Fu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
5
|
Yang L, Zhang W, Fan N, Cao P, Cheng Y, Zhu L, Luo S, Zong H, Bai Y, Zhou J, Deng Y, Ba Y, Liu T, Aili M, Yin X, Gu K, Dai G, Ying J, Shi J, Gao Y, Li W, Yu G, Xie L, Gai W, Wang Y, Meng P, Shi Y. Efficacy, safety and genomic analysis of SCT200, an anti-EGFR monoclonal antibody, in patients with fluorouracil, irinotecan and oxaliplatin refractory RAS and BRAF wild-type metastatic colorectal cancer: a phase Ⅱ study. EBioMedicine 2024; 100:104966. [PMID: 38217945 PMCID: PMC10826138 DOI: 10.1016/j.ebiom.2024.104966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/03/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Limited therapeutic options are available for metastatic colorectal cancer (mCRC) patients after failure of first- and second-line therapies, representing an unmet medical need for novel therapies. METHODS This is an open-label, single arm, multicenter, phase Ⅱ study aiming to perform the efficacy, safety and genomic analysis of SCT200, a noval fully humanized IgG1 anti-epidermal growth factor receptor (EGFR) monoclonal antibody, in patients with fluorouracil, irinotecan and oxaliplatin refractory RAS and BRAF wild-type mCRC. SCT200 (6 mg/kg) was given weekly for the first six weeks, followed by a higher dose of 8 mg/kg every two weeks until disease progression or unacceptable toxicity. Primary endpoint was independent review committee (IRC)-assessed objective response rate (ORR) and secondary endpoints included ORR in patients with left-sided tumor, disease control rate (DCR), duration of response (DoR), time to response (TTR), progression-free survival (PFS), overall survival (OS) and safety. FINDINGS From February 12, 2018 to December 1, 2019, a total of 110 patients aged between 26 and 77 years (median: 55; interquartile range [IQR]: 47-63) with fluorouracil, oxaliplatin, and irinotecan refractory RAS and BRAF wild-type mCRC were enrolled from 22 hospitals in China. As the data cut-off date on May 15, 2020, the IRC-assessed ORR and DCR was 31% (34/110, 95% confidence interval [CI] 22-40%) and 75% (82/110, 95% CI 65-82%), respectively. Thirty one percent (34/110) patients achieved confirmed partial response (PR). The median PFS and median OS were 5.1 months (95% CI 3.4-5.2) and 16.2 months (95% CI 11.1-not available [NA]), respectively. The most common ≥ grade 3 treatment-related adverse events (TRAEs) were hypomagnesemia (17%, 19/110) and acneiform dermatitis (11%, 12/110). No deaths occurred. Genomic analysis suggested positive association between MYC amplification and patients' response (P = 0.0058). RAS/RAF mutation and MET amplification were the most frequently detected resistance mechanisms. Patients with high circulating tumor DNA (ctDNA) at baseline or without ctDNA clearance at the 7th week after the first dose of SCT200 administration before receiving SCT200 had worse PFS and OS. INTERPRETATION SCT200 exhibited promising clinical efficacy and manageable safety profiles in RAS and BRAF wild-type mCRC patients progressed on fluorouracil, irinotecan and oxaliplatin treatment. The baseline ctDNA and ctDNA clearance status at the 7th week after the first dose of SCT200 administration before receiving SCT200 could be a potential prognostic biomarker for RAS and BRAF wild-type mCRC patients with SCT200 therapy. FUNDING This study was sponsored by Sinocelltech Ltd., Beijing, China and partly supported by the National Science and Technology Major Project for Key New Drug Development (2019ZX09732001-006, 2017ZX09304015).
Collapse
Affiliation(s)
- Lin Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Nanfeng Fan
- Department of Abdominal Oncology, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Peiguo Cao
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ying Cheng
- Department of Oncology, Cancer Hospital of Jilin Province, Changchun, China
| | - Lingjun Zhu
- Department of Oncology, Jiangsu Province Hospital, Nanjing, China
| | - Suxia Luo
- Department of Medical Oncology, Henan Provincial Cancer Hospital, Zhengzhou, China
| | - Hong Zong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuxian Bai
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianfeng Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yi Ba
- Department of Gastroenterology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Mayinuer Aili
- The Third Department of Oncology, Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xianli Yin
- Department of Gastroenterology, Hunan Cancer Hospital, Changsha, China
| | - Kangsheng Gu
- Department of Medical Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guanghai Dai
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, China
| | - Jieer Ying
- Department of Abdominal Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jianhua Shi
- Department of Medical Oncology, Linyi Cancer Hospital, Linyi, China
| | - Yajie Gao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Guohua Yu
- Department of Oncology, Weifang People's Hospital, Weifang, China
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Wenlin Gai
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Yan Wang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Peng Meng
- Burning Rock Biotech, Shanghai, China
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China.
| |
Collapse
|
6
|
Liu T, Jiang S, Teng X, Zhong L, Liu M, Jin Y, Dong M. A comparison of panitumumab and cetuximab in the treatment of KRAS wild-type metastatic colorectal cancer: a systematic review and meta-analysis. Immunopharmacol Immunotoxicol 2023; 45:1-9. [PMID: 35950851 DOI: 10.1080/08923973.2022.2112222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AIM Cetuximab and panitumumab are common antibodies against epidermal growth factor receptor (EGFR) that can be used in combination with chemotherapy for the treatment of metastatic colorectal cancer (mCRC). Although these two drugs are considered to be very similar, differences in the efficacy and safety of cetuximab and panitumumab are still unclear. We conducted this meta-analysis to explore the effects and adverse reactions of cetuximab and panitumumab in the treatment of mCRC. METHODS We searched PubMed, the Cochrane Library, Embase, Web of Science, China national knowledge infrastructure (CNKI) and WanFang databases to identify records related to the efficacy and safety of cetuximab and panitumumab in the treatment of mCRC. The search terms were "cetuximab," "panitumumab," and "colorectal cancer." The deadline of searching was April 2022. Review manager 5.4 software was used to perform the statistical analysis for this meta-analysis. Pooled hazard ratio (HR) with 95% confidence intervals (CI) were calculated to evaluate the overall survival (OS) and progression free survival (PFS) of cetuximab and panitumumab in the treatment of mCRC. RESULTS There was no significant difference in OS, PFS, and response rate (RR) between cetuximab arm and panitumumab arm (OS: HR = 0.91, 95% CI = 0.81-1.03, p = .14; PFS: HR = 0.92, 95% CI = 0.83-1.02, p = .11; RR: OR = 1.22, 95% CI = 0.96-1.61, p = .14). We also did not observe any statistical difference between both arms in incidence of acneiform rash, severe acneiform rash, diarrhea, and severe diarrhea (acneiform rash: OR = 1.09, 95% CI = 0.84-1.42, p = .51; severe acneiform rash: OR = 1.50, 95% CI = 0.80-2.81, p = .21; diarrhea: OR = 1.08, 95% CI = 0.82-1.42, p = .58; severe diarrhea: OR = 0.90, 95% CI = 0.44-1.84, p = .77). The incidence of paronychia was decreased in the panitumumab arm, but that of hypomagnesemia and severe hypomagnesemia were decreased in the cetuximab arm. (paronychia: OR = 0.74, 95% CI = 0.55-1.00, p = .05; hypomagnesemia: OR = 1.85, 95% CI =1.41-2.41, p < .00001; severe hypomagnesemia: OR = 2.66, 95% CI = 1.52-4.67, p = .0006). CONCLUSION There was no significant difference in OS, PFS and RR between the cetuximab arm and panitumumab arm in the treatment of mCRC. For adverse reactions, the incidence of paronychia was decreased in the panitumumab arm, and the incidence of hypomagnesemia was deceased in the cetuximab arm.
Collapse
Affiliation(s)
- Tong Liu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuai Jiang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Teng
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lu Zhong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengmeng Liu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yao Jin
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
7
|
Li D, Pan H, Wang W, Xue Y, Fang Y, Lou H, Pan Q, Jin W, Zheng Y, Han W, Zhu K, Zhao X, Xu R, Han J, Pan H. A phase Ia dose-escalation trial of Ametumumab (a fully human monoclonal antibody against epidermal growth factor receptor) in patients with advanced solid malignancies. Ther Adv Med Oncol 2023; 15:17588359231165968. [PMID: 37025261 PMCID: PMC10071157 DOI: 10.1177/17588359231165968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/06/2023] [Indexed: 04/03/2023] Open
Abstract
Background: Epidermal growth factor receptor (EGFR) is a well-known target for cancer treatment. However, the authorized anti-EGFR monoclonal antibodies generally cause several toxic effects, especially severe cutaneous toxicities as well as infusion reactions, and the clinical indications are limited. Here we developed Ametumumab, a fully human recombinant anti-EGFR monoclonal antibody. Objectives: To assess the safety, tolerability, pharmacokinetics (PK), and immunogenicity of Ametumumab. Design: A first-in-human phase Ia dose escalation study of Ametumumab in patients with advanced solid malignancies. Methods: An open-label, first-in-human dose escalation study was done in 22 patients with advanced malignancies who received six ascending dosages ranging from 75 to 750 mg/m2. Following a single dosage and a 28-day dose-limiting toxicity (DLT) monitoring period, patients were given repeated doses weekly. Blood samples were taken to determine the PK parameters of Ametumumab and anti-drug antibody concentrations. Every 8 weeks, radiographic tumor evaluations were conducted. Results: In this trial, no DLT was observed, and the maximum tolerated dose was not reached at doses up to 750 mg/m2. There were no severe adverse events but mild and moderate adverse effects, such as headache, proteinuria, and rash. Single-dose PK results demonstrated a straightforward linear relationship with dosage escalation. The medication concentrations accumulated and attained steady-state after four rounds of injections. It was calculated that 10 patients with disease control would be observed in the 22 evaluable patients. The disease control rate was 45.5%. Conclusion: The Ametumumab was well tolerated and safe in patients with advanced solid malignancies, exhibiting minimal immunogenicity, a long half-life, high levels of drug exposure in the blood, and preliminary effectiveness. Registration: The trial was registered with CTR20170343 on 10 April 2017, The China Center for Drug Evaluation.
Collapse
Affiliation(s)
- Da Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Hong Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Wei Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Yanan Xue
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Haizhou Lou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Qin Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Wei Jin
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Yu Zheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Kongli Zhu
- Shanghai Celfuture Biotech Co., Ltd., Shanghai, China
| | - Xianfeng Zhao
- Shanghai Celfuture Biotech Co., Ltd., Shanghai, China
| | - Rong Xu
- Shanghai Celfuture Biotech Co., Ltd., Shanghai, China
| | - Jin Han
- Shanghai Celfuture Biotech Co., Ltd., No. 280 Juli Rd. Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, No. 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
8
|
Poad H, Khan S, Wheaton L, Thomas A, Sweeting M, Bujkiewicz S. The Validity of Surrogate Endpoints in Sub Groups of Metastatic Colorectal Cancer Patients Defined by Treatment Class and KRAS Status. Cancers (Basel) 2022; 14:5391. [PMID: 36358810 PMCID: PMC9654686 DOI: 10.3390/cancers14215391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Background and Aim: Findings from the literature suggest that the validity of surrogate endpoints in metastatic colorectal cancer (mCRC) may depend on a treatments' mechanism of action. We explore this and the impact of Kirsten rat sarcoma (KRAS) status on surrogacy patterns in mCRC. Methods: A systematic review was undertaken to identify randomized controlled trials (RCTs) for pharmacological therapies in mCRC. Bayesian meta-analytic methods for surrogate endpoint evaluation were used to evaluate surrogate relationships across all RCTs, by KRAS status and treatment class. Surrogate endpoints explored were progression free survival (PFS) as a surrogate endpoint for overall survival (OS), and tumour response (TR) as a surrogate for PFS and OS. Results: 66 RCTs were identified from the systematic review. PFS showed a strong surrogate relationship with OS across all data and in subgroups by KRAS status. The relationship appeared stronger within individual treatment classes compared to the overall analysis. The TR-PFS and TR-OS relationships were found to be weak overall but stronger within the Epidermal Growth Factor Receptor + Chemotherapy (EGFR + Chemo) treatment class; both overall and in the wild type (WT) patients for TR-PFS, but not in patients with the mutant (MT) KRAS status where data were limited. Conclusions: PFS appeared to be a good surrogate endpoint for OS. TR showed a moderate surrogate relationship with PFS and OS for the EGFR + Chemo treatment class. There was some evidence of impact of the mechanism of action on the strength of the surrogacy patterns in mCRC, but little evidence of the impact of KRAS status on the validity of surrogate endpoints.
Collapse
Affiliation(s)
- Heather Poad
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Sam Khan
- Leicester Cancer Research Centre, University of Leicester, Leicester LE1 7RH, UK
| | - Lorna Wheaton
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Anne Thomas
- Leicester Cancer Research Centre, University of Leicester, Leicester LE1 7RH, UK
| | - Michael Sweeting
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Sylwia Bujkiewicz
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
9
|
Tanaka R, Ishikawa H, Sato J, Aoyama T, Shikamura Y, Shino M. Prevention of Acne-Like Eruption Caused by Panitumumab Treatment through Oral Administration of Non-steroidal Anti-inflammatory Drugs. Biol Pharm Bull 2022; 45:1531-1536. [PMID: 36184512 DOI: 10.1248/bpb.b22-00404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acne-like eruption caused by anti-epidermal growth factor receptor (EGFR) antibodies such as panitumumab reduces treatment adherence and patient QOL; an alternative therapy is desired. Meanwhile, the usefulness of oral Non-steroidal Anti-inflammatory Drugs (NSAIDs) for acne-like eruptions caused by low-molecular-weight EGFR inhibitors such as erlotinib has been reported in the treatment of lung cancer. This study aimed to investigate whether the combined use of oral NSAIDs and panitumumab for colorectal cancer patients helps prevent acne-like eruption. We retrospectively investigated 167 colorectal cancer patients who had been treated with panitumumab for three cycles or more. The observation period was set from the start of panitumumab treatment to the end of three cycles. Within this period, the incidence and severity of acne-like eruptions were compared. A total of 59 and 108 patients were in the NSAIDs use and non-use groups, respectively, showing differences in the incidence of acne-like eruption rates (78.0 vs. 90.7%, respectively; p = 0.033). In the use group, eruption severity grades 0, 1, 2, and 3 were observed in 13, 33, 13, and 0 patients, respectively; the corresponding values in the non-use group were 10, 60, 36, and 2, respectively (p = 0.007). Oral NSAIDs may help prevent acne-like eruptions caused by panitumumab.
Collapse
Affiliation(s)
- Rei Tanaka
- Department of Pharmacy, Shizuoka Cancer Center.,Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | | | - Junya Sato
- Department of Pharmacy, Shizuoka Cancer Center.,Department of Pharmacy, International University of Health and Welfare Hospital.,School of Pharmacy, International University of Health and Welfare
| | - Takao Aoyama
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | | | | |
Collapse
|
10
|
Strickler JH, Yoshino T, Graham RP, Siena S, Bekaii-Saab T. Diagnosis and Treatment of ERBB2-Positive Metastatic Colorectal Cancer: A Review. JAMA Oncol 2022; 8:760-769. [PMID: 35238866 DOI: 10.1001/jamaoncol.2021.8196] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Amplification of ERBB2 (formerly referred to as HER2) is present in nearly 3% of patients with metastatic colorectal cancer overall and 5% of patients with KRAS and NRAS wild-type tumors. Despite the availability of several ERBB2-targeted therapeutic options for patients with ERBB2-positive breast and gastric/gastroesophageal tumors, to date, there are currently no approved therapies for patients with ERBB2-positive metastatic colorectal cancer, although ERBB2-targeted therapies are recommended in National Comprehensive Cancer Network guidelines. Recent evidence indicates that anti-ERBB2 therapeutic strategies are active in patients with ERBB2-positive metastatic colorectal cancer and could potentially represent a new standard-of-care. Observations The protein ERBB2 is a member of a family of epidermal growth factor receptors that also includes epidermal growth factor receptor (ERBB1), ERBB3, and ERBB4. Amplification of ERBB2 leads to overexpression of the ERBB2 tyrosine kinase receptor, resulting in aberrant signaling and cell migration, growth, adhesion, and differentiation. Colorectal tumors that harbor ERBB2 amplification are more likely to originate on the left side of the colon, are associated with primary and acquired resistance to anti-epidermal growth factor receptor therapies, and have increased incidence of central nervous system metastases. Using immunohistochemistry, fluorescence in situ hybridization, next-generation sequencing, and liquid biopsy techniques, several randomized clinical trials have evaluated the efficacy of ERBB2-targeted therapies in patients with ERBB2-positive metastatic colorectal cancer. These therapies include monoclonal antibodies, antibody-drug conjugates, and tyrosine kinase inhibitors, many of which were associated with favorable efficacy and safety profiles when treating patients with ERBB2-positive metastatic colorectal cancer. Conclusions and Relevance The results of this review suggest the ERBB2 receptor is a promising target for patients with metastatic colorectal cancer; however, to date, no therapies are approved for use in this patient population. Therefore, it is imperative to continue to work to address this unmet need so that patients with ERBB2-positive metastatic colorectal cancer have therapeutic options should they become refractory to treatment with standard therapies.
Collapse
Affiliation(s)
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Rondell P Graham
- Division of Laboratory Genetics and Genomics, Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda and Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
11
|
Martins D, Rodrigues J, Redondo P, Julião I, Faustino C. Evaluating the Optimal Sequence of Treatment With EGFR Inhibitors and Bevacizumab in RAS Wild-Type Metastatic Colorectal Cancer. Cureus 2022; 14:e23543. [PMID: 35494924 PMCID: PMC9042308 DOI: 10.7759/cureus.23543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2022] [Indexed: 11/05/2022] Open
|
12
|
Siddiqui Z, Ahmed S, Vickers M. Epidermal Growth Factor Receptor Inhibitor-Induced Hypomagnesaemia: Is There a Best Replacement Strategy? EUROPEAN MEDICAL JOURNAL 2021. [DOI: 10.33590/emj/21-00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Monoclonal antibodies targeting the epidermal growth factor receptor (EGFRI), such as cetuximab and panitumumab, are commonly used systemic therapies for advanced colorectal and head and neck cancers. Hypomagnesaemia is a common side effect of these therapies and occurs in up to 30% of patients. Interruption of EGFR signalling in the distal convoluted tubule leads to inactivation of the transcellular transporter transient receptor potential channel melastatin member 6 and increased renal magnesium excretion. This paper describes the incidence, risk factors, and the emerging management options for EGFRI-induced hypomagnesaemia.
Collapse
Affiliation(s)
- Zeba Siddiqui
- Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Sumaiya Ahmed
- Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Michael Vickers
- Department of Medicine, Division of Medical Oncology, The Ottawa Hospital, Ottawa, Canada
| |
Collapse
|
13
|
Baysal H, De Pauw I, Zaryouh H, Peeters M, Vermorken JB, Lardon F, De Waele J, Wouters A. The Right Partner in Crime: Unlocking the Potential of the Anti-EGFR Antibody Cetuximab via Combination With Natural Killer Cell Chartering Immunotherapeutic Strategies. Front Immunol 2021; 12:737311. [PMID: 34557197 PMCID: PMC8453198 DOI: 10.3389/fimmu.2021.737311] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Cetuximab has an established role in the treatment of patients with recurrent/metastatic colorectal cancer and head and neck squamous cell cancer (HNSCC). However, the long-term effectiveness of cetuximab has been limited by the development of acquired resistance, leading to tumor relapse. By contrast, immunotherapies can elicit long-term tumor regression, but the overall response rates are much more limited. In addition to epidermal growth factor (EGFR) inhibition, cetuximab can activate natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC). In view of the above, there is an unmet need for the majority of patients that are treated with both monotherapy cetuximab and immunotherapy. Accumulated evidence from (pre-)clinical studies suggests that targeted therapies can have synergistic antitumor effects through combination with immunotherapy. However, further optimizations, aimed towards illuminating the multifaceted interplay, are required to avoid toxicity and to achieve better therapeutic effectiveness. The current review summarizes existing (pre-)clinical evidence to provide a rationale supporting the use of combined cetuximab and immunotherapy approaches in patients with different types of cancer.
Collapse
Affiliation(s)
- Hasan Baysal
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Ines De Pauw
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Hannah Zaryouh
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.,Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Jan Baptist Vermorken
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.,Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Jorrit De Waele
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - An Wouters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
14
|
Kato T, Kagawa Y, Kuboki Y, Gamoh M, Komatsu Y, Yasui H, Satake H, Oki E, Tanioka H, Kotaka M, Makiyama A, Denda T, Goto M, Yoshino T, Yamazaki K, Soeda J, Shibuya K, Iwata M, Oba K, Yamaguchi K. Safety and efficacy of panitumumab in combination with trifluridine/tipiracil for pre-treated patients with unresectable, metastatic colorectal cancer with wild-type RAS: The phase 1/2 APOLLON study. Int J Clin Oncol 2021; 26:1238-1247. [PMID: 33928486 PMCID: PMC8213662 DOI: 10.1007/s10147-021-01902-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/17/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND We aimed to assess the safety and efficacy of combination treatment with panitumumab plus trifluridine/tipiracil (FTD/TPI) in patients with wild-type RAS metastatic colorectal cancer (mCRC) who were refractory/intolerant to standard therapies other than anti-epidermal growth factor receptor therapy. METHODS APOLLON was an open-label, multicentre, phase 1/2 trial. In the phase 1 part, 3 + 3 de-escalation design was used to investigate the recommended phase 2 dose (RP2D); all patients in the phase 2 part received the RP2D. The primary endpoint was investigator-assessed progression-free survival (PFS) rate at 6 months. Secondary endpoints included PFS, overall survival (OS), overall response rate (ORR), disease control rate (DCR), time to treatment failure (TTF), and safety. RESULTS Fifty-six patients were enrolled (phase 1, n = 7; phase 2, n = 49) at 25 Japanese centres. No dose-limiting toxicities were observed in patients receiving panitumumab (6 mg/kg every 2 weeks) plus FTD/TPI (35 mg/m2 twice daily; days 1-5 and 8-12 in a 28-day cycle), which became RP2D. PFS rate at 6 months was 33.3% (90% confidence interval [CI] 22.8-45.3). Median PFS, OS, ORR, DCR, and TTF were 5.8 months (95% CI 4.5-6.5), 14.1 months (95% CI 12.2-19.3), 37.0% (95% CI 24.3-51.3), 81.5% (95% CI 68.6-90.8), and 5.8 months (95% CI 4.29-6.21), respectively. Neutrophil count decreased (47.3%) was the most common Grade 3/4 treatment-emergent adverse event. No treatment-related deaths occurred. CONCLUSION Panitumumab plus FTD/TPI exhibited favourable anti-tumour activity with a manageable safety profile and may be a therapeutic option for pre-treated mCRC patients.
Collapse
Affiliation(s)
- Takeshi Kato
- National Hospital Organization Osaka National Hospital, 2 Chome-1-14 Hoenzaka, Chuo Ward, Osaka, 540-0006, Japan
| | - Yoshinori Kagawa
- Kansai Rosai Hospital, 3 Chome-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Yasutoshi Kuboki
- National Cancer Center Hospital East, 6 Chome-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Makio Gamoh
- Osaki Citizen Hospital, Furukawa Honami, 3 Chome, Osaki, 989-6183, Japan
| | - Yoshito Komatsu
- Hokkaido University Hospital, 5 Chome Kita 14 Jonishi, Kita Ward, Sapporo, Hokkaido, 060-8648, Japan
| | - Hirofumi Yasui
- Shizuoka Cancer Center, 1007 Shimonagakubo, Nagaizumi, Sunto District, Shizuoka, 411-0934, Japan
| | - Hironaga Satake
- Kobe City Medical Center General Hospital, 2 Chome-1-1 Minatojima Minamimachi, Chuo Ward, Kobe, Hyogo, 650-0047, Japan
- Cancer Treatment Center, Kansai Medical University Hospital, 2 Chome-3-1 Shinmachi, Hirakata, Osaka, 573-1191, Japan
| | - Eiji Oki
- Kyushu University, Maidashi 3 Chome-1-3, Higashi Ward, Fukuoka, 812-0053, Japan
| | - Hiroaki Tanioka
- Okayama Rosai Hospital, 1 Chome-10-25 Chikkomidorimachi, Minami Ward, Okayama, 702-8055, Japan
- Medical Oncology Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Masahito Kotaka
- Sano Hospital, 2 Chome-5-1 Shimizugaoka, Tarumi Ward, Kobe, Hyogo, 655-0031, Japan
| | - Akitaka Makiyama
- Japan Community Healthcare Organization Kyushu Hospital, 1 Chome-8-1 Kishinoura, Yahatanishi Ward, Kitakyushu, Fukuoka, 806-8501, Japan
- Gifu University Hospital, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Tadamichi Denda
- Chiba Cancer Center, 666-2 Nitona-cho, Chuo Ward, Chiba, 260-8717, Japan
| | - Masahiro Goto
- Osaka Medical College Hospital, 2-7 Daigakumachi, Takatsuki, Osaka, 569-0096, Japan
| | - Takayuki Yoshino
- National Cancer Center Hospital East, 6 Chome-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Kentaro Yamazaki
- Shizuoka Cancer Center, 1007 Shimonagakubo, Nagaizumi, Sunto District, Shizuoka, 411-0934, Japan
| | - Junpei Soeda
- Takeda Pharmaceutical Company, Ltd, Nihonbashi-Honcho 2 Chome-1-1, Chuo Ward, Tokyo, 103-8668, Japan
| | - Kazunori Shibuya
- Takeda Pharmaceutical Company, Ltd, Nihonbashi-Honcho 2 Chome-1-1, Chuo Ward, Tokyo, 103-8668, Japan
| | - Masaru Iwata
- Takeda Pharmaceutical Company, Ltd, Nihonbashi-Honcho 2 Chome-1-1, Chuo Ward, Tokyo, 103-8668, Japan
| | - Koji Oba
- University of Tokyo, 7 Chome-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Kensei Yamaguchi
- Gastroenterological Chemotherapy Department, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3 Chome-8-31, Ariake, Koto, Tokyo, 135-8550, Japan.
| |
Collapse
|
15
|
Predictive and prognostic value of magnesium serum level in FOLFIRI plus cetuximab or bevacizumab treated patients with stage IV colorectal cancer: results from the FIRE-3 (AIO KRK-0306) study. Anticancer Drugs 2021; 31:856-865. [PMID: 32639280 DOI: 10.1097/cad.0000000000000965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Magnesium wasting is a frequent side effect of epidermal growth factor receptor (EGFR)-antibody treatment as magnesium-absorption mechanisms are dependent on EGFR signaling. EGFR-inhibition results in decreased renal reabsorption. There is evidence that hypomagnesemia during cetuximab treatment correlates with response. The prognostic role of hypomagnesemia during bevacizumab treatment has not been studied yet. Here, we evaluate the prognostic value of hypomagnesemia in patients with metastatic colorectal cancer treated with FOLFIRI plus cetuximab or bevacizumab as first-line therapy. A total of 391 of 752 patients of the firstline irinotecan study population had magnesium levels measured at baseline and for the first three cycles (6 weeks) of treatment. Of those, 240 had Rat Sarkoma wildtype tumors. Overall hypomagnesemia was more common in the cetuximab compared to the bevacizumab arm (80 vs. 43%, P < 0.005). During therapy, magnesium showed a time-dependent decrease to 80% of baseline in the cetuximab and to 89% in the bevacizumab arm. Whereas magnesium continued to decrease over time in the cetuximab-treated patients, it remained stable in the bevacizumab-treated. Overall response rate (ORR) was associated with higher magnesium at week 6 (20.9 vs. 79.1%, P = 0.041). Bevacizumab-treated patients with magnesium levels below the median value at week 6 had a significantly longer progression-free survival (PFS; 11.7 vs. 9.9 months, P = 0.034; hazard ratio 0.73) and a trend towards longer overall survival (OS) (29.6 vs. 23.2 months, P = 0.089; hazard ratio 0.77). Hypomagnesemia at predefined time points and magnesium nadir had no significant effect on ORR, OS and PFS in the cetuximab arm. Our data show different magnesium kinetics in patients with metastatic colorectal cancer treated with cetuximab or bevacizumab. For patients treated with cetuximab, hypomagnesemia did not have an impact on response and survival. Hypomagnesemia might have a prognostic value in bevacizumab treatment.
Collapse
|
16
|
Coronel-Hernández J, Salgado-García R, Cantú-De León D, Jacobo-Herrera N, Millan-Catalan O, Delgado-Waldo I, Campos-Parra AD, Rodríguez-Morales M, Delgado-Buenrostro NL, Pérez-Plasencia C. Combination of Metformin, Sodium Oxamate and Doxorubicin Induces Apoptosis and Autophagy in Colorectal Cancer Cells via Downregulation HIF-1α. Front Oncol 2021; 11:594200. [PMID: 34123772 PMCID: PMC8187873 DOI: 10.3389/fonc.2021.594200] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/30/2021] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death worldwide in both sexes. Current therapies include surgery, chemotherapy, and targeted therapy; however, prolonged exposure to chemical agents induces toxicity in patients and drug resistance. So, we implemented a therapeutic strategy based on the combination of doxorubicin, metformin, and sodium oxamate called triple therapy (Tt). We found that Tt significantly reduced proliferation by inhibiting the mTOR/AKT pathway and promoted apoptosis and autophagy in CRC derived cells compared with doxorubicin. Several autophagy genes were assessed by western blot; ULK1, ATG4, and LC3 II were overexpressed by Tt. Interestingly, ULK1 was the only one autophagy-related protein gradually overexpressed during Tt administration. Thus, we assumed that there was a post-transcriptional mechanism mediating by microRNAs that regulate UKL1 expression during autophagy activation. Through bioinformatics approaches, we ascertained that ULK1 could be targeted by mir-26a, which is overexpressed in advanced stages of CRC. In vitro experiments revealed that overexpression of mir-26a decreased significantly ULK1, mRNA, and protein expression. Contrariwise, the Tt recovered ULK1 expression by mir-26a decrease. Due to triple therapy repressed mir-26a expression, we hypothesized this drug combination could be involved in mir-26a transcription regulation. Consequently, we analyzed the mir-26a promoter sequence and found two HIF-1α transcription factor recognition sites. We developed two different HIF-1α stabilization models. Both showed mir-26a overexpression and ULK1 reduction in hypoxic conditions. Immunoprecipitation experiments were performed and HIF-1α enrichment was observed in mir-26a promoter. Surprisingly, Tt diminished HIF-1α detection and restored ULK1 mRNA expression. These results reveal an important regulation mechanism controlled by the signaling that activates HIF-1α and that in turn regulates mir-26a transcription.
Collapse
Affiliation(s)
- Jossimar Coronel-Hernández
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-Iztacala, UNAM, Tlalnepantla, Mexico,Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico
| | | | - David Cantú-De León
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico
| | | | | | | | | | | | | | - Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-Iztacala, UNAM, Tlalnepantla, Mexico,Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico,*Correspondence: Carlos Pérez-Plasencia,
| |
Collapse
|
17
|
Navigating metastatic colorectal treatment options in the USA: a survey of patient acceptance of skin toxicities associated with Vectibix. Support Care Cancer 2021; 29:6731-6740. [PMID: 33973081 PMCID: PMC8464558 DOI: 10.1007/s00520-021-06134-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 03/04/2021] [Indexed: 11/16/2022]
Abstract
Abstract Purpose To understand the extent to which metastatic colorectal cancer (mCRC) patients receive education on the prevention and management associated with skin rash following Vectibix treatment. Furthermore, to investigate how this adverse event affects a patient’s quality of life (QoL) and influences their treatment decisions. Methods A cross-sectional survey was administered to 200 mCRC patients (100 Vectibix users and 100 Vectibix non-users). After excluding respondents who had used cetuximab, 61 Vectibix users and 56 Vectibix non-users remained. Results Most Vectibix users (79%) experienced a skin rash in response to treatment of which 65% considered the rash moderate, 27% mild, and 8% severe. Vectibix users generally felt they were adequately informed about the rash (83%), with the most common messages received related to sun protection. However, sunscreen was used by only 42% of patients prior to rash and 60% of patients following the appearance of rash. The use of oral antibiotics was low prior to rash (21%) and following rash (46%). Among patients experiencing a rash within the past week (n=16), 75% reported the rash had a large negative impact on their QoL based on the Dermatology Life Quality Index. Conclusion There was a disconnect between patients feeling they were adequately informed and use of prevention and management strategies such as sun protection. This suggests a gap in patient education and adoption currently exists on management strategies both prior to and following the appearance of rash. Given the negative impact that skin toxicity has on the patient’s quality of life, it is essential that patients receive and subsequently utilize all information that can minimize rash severity.
Collapse
|
18
|
Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Farkas L, Garrido-Laguna I, Grem JL, Gunn A, Hecht JR, Hoffe S, Hubbard J, Hunt S, Johung KL, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Gregory KM, Gurski LA. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19:329-359. [PMID: 33724754 DOI: 10.6004/jnccn.2021.0012] [Citation(s) in RCA: 914] [Impact Index Per Article: 228.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This selection from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Colon Cancer focuses on systemic therapy options for the treatment of metastatic colorectal cancer (mCRC), because important updates have recently been made to this section. These updates include recommendations for first-line use of checkpoint inhibitors for mCRC, that is deficient mismatch repair/microsatellite instability-high, recommendations related to the use of biosimilars, and expanded recommendations for biomarker testing. The systemic therapy recommendations now include targeted therapy options for patients with mCRC that is HER2-amplified, or BRAF V600E mutation-positive. Treatment and management of nonmetastatic or resectable/ablatable metastatic disease are discussed in the complete version of the NCCN Guidelines for Colon Cancer available at NCCN.org. Additional topics covered in the complete version include risk assessment, staging, pathology, posttreatment surveillance, and survivorship.
Collapse
Affiliation(s)
- Al B Benson
- 1Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Alan P Venook
- 2UCSF Helen Diller Family Comprehensive Cancer Center
| | | | | | | | | | - Stacey Cohen
- 6Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | - Linda Farkas
- 9UT Southwestern Simmons Comprehensive Cancer Center
| | | | | | | | | | | | | | - Steven Hunt
- 16Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Smitha Krishnamurthi
- 19Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | - Eric D Miller
- 22The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Mary F Mulcahy
- 1Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | - Katrina Pedersen
- 16Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
IMPORTANCE Colorectal cancer (CRC) is the third most common cause of cancer mortality worldwide with more than 1.85 million cases and 850 000 deaths annually. Of new colorectal cancer diagnoses, 20% of patients have metastatic disease at presentation and another 25% who present with localized disease will later develop metastases. OBSERVATIONS Colorectal cancer is the third most common cause of cancer mortality for men and women in the United States, with 53 200 deaths projected in 2020. Among people diagnosed with metastatic colorectal cancer, approximately 70% to 75% of patients survive beyond 1 year, 30% to 35% beyond 3 years, and fewer than 20% beyond 5 years from diagnosis. The primary treatment for unresectable metastatic CRC is systemic therapy (cytotoxic chemotherapy, biologic therapy such as antibodies to cellular growth factors, immunotherapy, and their combinations.) Clinical trials completed in the past 5 years have demonstrated that tailoring treatment to the molecular and pathologic features of the tumor improves overall survival. Genomic profiling to detect somatic variants is important because it identifies the treatments that may be effective. For the 50% of patients with metastatic CRC with KRAS/NRAS/BRAF wild-type tumors, cetuximab and panitumumab (monoclonal antibodies to the epithelial growth factor receptor [EGFR]), in combination with chemotherapy, can extend median survival by 2 to 4 months compared with chemotherapy alone. However, for the 35% to 40% of patients with KRAS or NRAS sequence variations (formerly termed mutations), effective targeted therapies are not yet available. For the 5% to 10% with BRAF V600E sequence variations, targeted combination therapy with BRAF and EGFR inhibitors extended overall survival to 9.3 months, compared to 5.9 months for those receiving standard chemotherapy. For the 5% with microsatellite instability (the presence of numerous insertions or deletions at repetitive DNA units) or mismatch repair deficiency, immunotherapy may be used in the first or subsequent line and has improved treatment outcomes with a median overall survival of 31.4 months in previously treated patients. CONCLUSIONS AND RELEVANCE Advances in molecular profiling of metastatic CRC facilitate the ability to direct treatments to the biologic features of the tumor for specific patient subsets. Although cures remain uncommon, more patients can anticipate extended survival. Genomic profiling allows treatment selection so that more patients derive benefit and fewer are exposed to toxicity from ineffective therapies.
Collapse
Affiliation(s)
- Leah H Biller
- Division of Gastrointestinal Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Deborah Schrag
- Division of Gastrointestinal Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
- Division of Population Sciences, Dana Farber Cancer Institute, Boston, Massachusetts
- Associate Editor, JAMA
| |
Collapse
|
20
|
Franken GAC, Adella A, Bindels RJM, de Baaij JHF. Mechanisms coupling sodium and magnesium reabsorption in the distal convoluted tubule of the kidney. Acta Physiol (Oxf) 2021; 231:e13528. [PMID: 32603001 PMCID: PMC7816272 DOI: 10.1111/apha.13528] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/29/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Hypomagnesaemia is a common feature of renal Na+ wasting disorders such as Gitelman and EAST/SeSAME syndrome. These genetic defects specifically affect Na+ reabsorption in the distal convoluted tubule, where Mg2+ reabsorption is tightly regulated. Apical uptake via TRPM6 Mg2+ channels and basolateral Mg2+ extrusion via a putative Na+ -Mg2+ exchanger determines Mg2+ reabsorption in the distal convoluted tubule. However, the mechanisms that explain the high incidence of hypomagnesaemia in patients with Na+ wasting disorders of the distal convoluted tubule are largely unknown. In this review, we describe three potential mechanisms by which Mg2+ reabsorption in the distal convoluted tubule is linked to Na+ reabsorption. First, decreased activity of the thiazide-sensitive Na+ /Cl- cotransporter (NCC) results in shortening of the segment, reducing the Mg2+ reabsorption capacity. Second, the activity of TRPM6 and NCC are determined by common regulatory pathways. Secondary effects of NCC dysregulation such as hormonal imbalance, therefore, might disturb TRPM6 expression. Third, the basolateral membrane potential, maintained by the K+ permeability and Na+ -K+ -ATPase activity, provides the driving force for Na+ and Mg2+ extrusion. Depolarisation of the basolateral membrane potential in Na+ wasting disorders of the distal convoluted tubule may therefore lead to reduced activity of the putative Na+ -Mg2+ exchanger SLC41A1. Elucidating the interconnections between Mg2+ and Na+ transport in the distal convoluted tubule is hampered by the currently available models. Our analysis indicates that the coupling of Na+ and Mg2+ reabsorption may be multifactorial and that advanced experimental models are required to study the molecular mechanisms.
Collapse
Affiliation(s)
- Gijs A. C. Franken
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| | - Anastasia Adella
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| | - René J. M. Bindels
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| | - Jeroen H. F. de Baaij
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| |
Collapse
|
21
|
Gürsoy P, Çakar B, Almuradova E, Karateke M, Doğanavşargil B, Sezak M, Harman M, Karabulut B. The effectiveness of cetuximab and panitumumab when combined with FOLFIRI in second-line treatment of KRAS wild type metastatic colorectal cancers. Single centre experience. J Chemother 2020; 33:180-186. [PMID: 33349195 DOI: 10.1080/1120009x.2020.1861531] [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: 10/22/2022]
Abstract
Panitumumab and cetuximab are monoclonal antibodies known to be effective in metastatic colorectal cancer (mCRC). Although the survival benefits when combined with chemotherapy have been determined, there are no studies comparing the two agents with chemotherapy in the second-line treatment. In this study, we aimed to compare the efficacy of cetuximab vs panitumumab in patients who previously received chemotherapy. Who progressed after first-line treatment for K-ras wild type mCRC were analyzed. The efficacy of cetuximab vs panitumumab on overall survival (OS) and progression-free survival (PFS) when combined with FOLFIRI regimen was compared retrospectively. Median PFS was 6.9 months in the cetuximab group and 4.7 months in the panitumumab group. Median OS cetuximab and panitumumab groups were 18.4 and 12.2 months, respectively. In the second-line treatment of K-ras wild type mCRC, both PFS and OS were found to be longer in patients receiving cetuximab than in patients receiving panitumumab, but no statistically significant difference was found.
Collapse
Affiliation(s)
- Pınar Gürsoy
- Tulay Aktas Oncology Hospital, Ege University School of Medicine, IZMIR, Turkey
| | - Burcu Çakar
- Tulay Aktas Oncology Hospital, Ege University School of Medicine, IZMIR, Turkey
| | - Elvina Almuradova
- Tulay Aktas Oncology Hospital, Ege University School of Medicine, IZMIR, Turkey
| | - Murat Karateke
- Tulay Aktas Oncology Hospital, Ege University School of Medicine, IZMIR, Turkey
| | | | - Murat Sezak
- Department Pathology, Ege University School of Medicine, IZMIR, Turkey
| | - Mustafa Harman
- Department Radiology, Ege University School of Medicine, IZMIR, Turkey
| | - Bülent Karabulut
- Tulay Aktas Oncology Hospital, Ege University School of Medicine, IZMIR, Turkey
| |
Collapse
|
22
|
Fernández-Montes A, Grávalos C, Pericay C, Safont MJ, Benavides M, Élez E, García-Alfonso P, García-Paredes B, Carrato A, Aranda E. Current Options for Third-line and Beyond Treatment of Metastatic Colorectal Cancer. Spanish TTD Group Expert Opinion. Clin Colorectal Cancer 2020; 19:165-177. [PMID: 32507561 DOI: 10.1016/j.clcc.2020.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is a public health problem: it is the third most common cancer in men (746,000 new cases/year) and the second in women (614,000 new cases/year), representing the second leading cause of death by cancer worldwide. The survival of patients with metastatic CRC (mCRC) has increased prominently in recent years, reaching a median of 25 to 30 months. A growing number of patients with mCRC are candidates to receive a treatment in third line or beyond, although the optimal drug regimen and sequence are still unknown. In this situation of refractoriness, there are several alternatives: (1) To administer sequentially the 2 oral drugs approved in this indication: trifluridine/tipiracil and regorafenib, which have shown a statistically significant benefit in progression-free survival and overall survival with a different toxicity profile. (2) To administer cetuximab or panitumumab in treatment-naive patients with RAS wild type, which is increasingly rare because these drugs are usually indicated in first- or second-line. (3) To reuse drugs already administered that were discontinued owing to toxicity or progression (oxaliplatin, irinotecan, fluoropyrimidine, antiangiogenics, anti-epidermal growth factor receptor [if RAS wild-type]). High-quality evidence is limited, but this strategy is often used in routine clinical practice in the absence of alternative therapies especially in patients with good performance status. (4) To use specific treatments for very selected populations, such as trastuzumab/lapatinib in mCRC human epidermal growth factor receptor 2-positive, immunotherapy in microsatellite instability, intrahepatic therapies in limited disease or primarily located in the liver, although the main recommendation is to include patients in clinical trials.
Collapse
Affiliation(s)
- Ana Fernández-Montes
- Medical Oncology, Complejo Hospitalario Universitario de Ourense, Orense, Spain.
| | - Cristina Grávalos
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Carles Pericay
- Medical Oncology, Hospital de Sabadell, Corporación Sanitaria Parc Tauli, Sabadell, Barcelona, Spain
| | - Ma José Safont
- Medical Oncology, Hospital General Universitario de Valencia, València, Spain
| | - Manuel Benavides
- Medical Oncology, Hospital Universitario Regional y Virgen de la Victoria, Málaga, Spain
| | - Elena Élez
- Medical Oncology, Hospital Universitari Vall d'Hebron, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | | | - Beatriz García-Paredes
- Medical Oncology, Hospital Clínico San Carlos, Instituto de Investigación Hospital Clínico San Carlos (IdISSC). CIBERONC, Madrid, Spain
| | - Alfredo Carrato
- Medical Oncology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá de Henares, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), CIBERONC, Madrid, Spain
| | - Enrique Aranda
- Medical Oncology, Hospital Reina Sofía, University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), CIBERONC, Córdoba, Spain
| |
Collapse
|
23
|
Druzhkova I, Shirmanova M, Kuznetsova D, Lukina М, Zagaynova Е. Modern Approaches to Testing Drug Sensitivity of Patients' Tumors (Review). Sovrem Tekhnologii Med 2020; 12:91-102. [PMID: 34795997 PMCID: PMC8596271 DOI: 10.17691/stm2020.12.4.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Indexed: 11/19/2022] Open
Abstract
Drug therapy is still one of the basic techniques used to treat cancers of different etiology. However, tumor resistance to drugs is a pressing problem limiting drug treatment efficacy. It is obvious for both modern fundamental and clinical oncology that there is the need for an individual approach to treating cancer taking into account the biological properties of a tumor when prescribing chemo- and targeted therapy. One of the promising strategies is to increase the antitumor therapy efficacy by developing predictive tests, which enable to evaluate the sensitivity of a particular tumor to a specific drug or a drug combination before the treatment initiation and, thus, make individual therapy selection possible. The present review considers the main approaches to drug sensitivity assessment of patients' tumors: molecular genetic profiling of tumor cells, and direct efficiency testing of the drugs on tumor cells isolated from surgical or biopsy material. There were analyzed the key directions in research and clinical studies such as: the search for predictive molecular markers, the development of methods to maintain tumor cells or tissue sections viable, i.e. in a condition maximum close to their physiological state, the development of high throughput systems to assess therapy efficiency. Special attention was given to a patient-centered approach to drug therapy in colorectal cancer.
Collapse
Affiliation(s)
- I.N. Druzhkova
- Junior Researcher, Fluorescent Bio-imaging Laboratory, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - M.V. Shirmanova
- Deputy Director for Science, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Head of Fluorescent Bio-imaging Laboratory, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - D.S. Kuznetsova
- Researcher, Regenerative Medicine Laboratory, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - М.М. Lukina
- Junior Researcher, Fluorescent Bio-imaging Laboratory, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - Е.V. Zagaynova
- Corresponding Member of Russian Academy of Sciences, Rector; National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia Chief Researcher, Laboratory of Optical Coherence Tomography, Research Institute of Experimental Oncology and Biomedical Technologies Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
24
|
Efficacy of Panitumumab and Cetuximab in Patients with Colorectal Cancer Previously Treated with Bevacizumab; a Combined Analysis of Individual Patient Data from ASPECCT and WJOG6510G. Cancers (Basel) 2020; 12:cancers12071715. [PMID: 32605298 PMCID: PMC7407286 DOI: 10.3390/cancers12071715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/20/2020] [Accepted: 06/25/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Phase-III ASPECCT and randomised phase-II WJOG6510G trials demonstrated the noninferiority of panitumumab, when compared with cetuximab, for overall survival in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer. Methods: The subgroup that received bevacizumab either prior to panitumumab or cetuximab monotherapy (ASPECCT) or in combination with irinotecan (WJOG6510G) was included. Multivariate Cox models were created, including the treatment arms as covariates together with patient, disease and treatment characteristics. Results: We included 185 and 189 patients in the panitumumab and cetuximab arms, respectively. The median overall survival was 12.8 and 10.1 months [p = 0.0031; log-rank test, stratified by trial; hazard ratio (HR), 0.72; 95% confidence interval (CI), 0.58–0.90], and the median progression-free survival was 4.7 and 4.1 months, in the panitumumab and cetuximab arms, respectively (p = 0.0207; HR, 0.79; 95% CI, 0.64–0.97). The treatment regimen was an independent prognostic factor of overall survival (adjusted HR, 0.69; 95% CI, 0.54–0.87; p = 0.0013). Conclusions: Panitumumab significantly prolonged the overall survival and progression-free survival, when compared with cetuximab in the cohort that previously received bevacizumab in the included studies. Clinical Trial Registration: ASPECCT trial registered with ClinicalTrials.gov (NCT01001377) and WJOG6510G trial registered with UMIN-CTR (UMIN000006643).
Collapse
|
25
|
Randomised phase II study of panitumumab plus irinotecan versus cetuximab plus irinotecan in patients with KRAS wild-type metastatic colorectal cancer refractory to fluoropyrimidine, irinotecan and oxaliplatin (WJOG 6510G). Eur J Cancer 2020; 135:11-21. [PMID: 32526634 DOI: 10.1016/j.ejca.2020.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/26/2020] [Accepted: 04/04/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Cetuximab has been shown to be clinically active when given in combination with irinotecan in patients with irinotecan-refractory metastatic colorectal cancer (mCRC). However, it has remained unclear whether panitumumab is effective when combined with irinotecan. We compared efficacies of both regimens in this randomised phase II study. PATIENTS AND METHODS Patients with wild-type KRAS exon 2 mCRC previously treated with fluorouracil-, oxaliplatin- and irinotecan-based chemotherapies were randomised (1:1) to either panitumumab plus irinotecan (panitumumab arm) or cetuximab plus irinotecan (cetuximab arm). The primary end-point was progression-free survival (PFS). The planned sample size was 120, expecting a hazard ratio (HR) of 1.0 with non-inferiority margin of 1.3 (one-sided alpha error 0.2 and power 0.7). Major secondary end-points were overall survival (OS), response rate and safety. RESULTS From December 2011 to September 2014, 121 patients were enrolled, and 61 and 59 patients were randomised to the panitumumab and cetuximab arms, respectively (1 patient excluded). Most patients (97%) had received prior chemotherapies containing bevacizumab. The median PFS was 5.42 months in the panitumumab arm and 4.27 months in the cetuximab arm (HR = 0.64, 95% confidence interval [CI] = 0.44-0.94, P < 0.001 for non-inferiority, P = 0.058 for superiority), and median OS was 14.85 and 11.53 months (HR = 0.66, 95% CI = 0.44-1.00, P = 0.050 for superiority), respectively. The incidence of grade 3 or 4 hypomagnesaemia was higher in the panitumumab arm than that in the cetuximab arm (17% vs. 7%). CONCLUSION Panitumumab may be non-inferior to cetuximab in combination with irinotecan in survival of patients with irinotecan-refractory mCRC.
Collapse
|
26
|
Current and New Predictors for Treatment Response in Metastatic Colorectal Cancer. The Role of Circulating miRNAs as Biomarkers. Int J Mol Sci 2020; 21:ijms21062089. [PMID: 32197436 PMCID: PMC7139554 DOI: 10.3390/ijms21062089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequently diagnosed cancer in the world. More than half of all CRC patients will eventually develop metastases and require treatment accordingly, but few validated predictive factors for response to systemic treatments exist. In order to ascertain which patients benefit from specific treatments, there is a strong need for new and reliable biomarkers. We conducted a comprehensive search using the PUBMED database, up to December 2019, in order to identify relevant studies on predictive biomarkers for treatment response in metastatic CRC. We will herein present the currently used and potential biomarkers for treatment response and bring up-to-date knowledge on the role of circulating microRNAs, associated with chemotherapy and targeted therapy regimens used in metastatic CRC treatment. Molecular, tumor-related, disease-related, clinical, and laboratory predictive markers for treatment response were identified, mostly proposed, with few validated. Several circulating microRNAs have already proven their role of prediction for treatment response in CRC, but future clinical studies are needed to confirm their role as biomarkers across large cohorts of patients.
Collapse
|
27
|
Kafatos G, Dube S, Burdon P, Demonty G, Flinois A, Leclerc M, Lowe K, Feudjo-Tepie M, Segaert S. Management of EGFR Inhibitor-induced Skin Toxicity and Factors Impacting Patients' Adherence to Skin Toxicity Treatment: Health Care Provider and Patient Surveys in European Oncology Centers. Clin Colorectal Cancer 2020; 19:100-108.e9. [PMID: 32113902 DOI: 10.1016/j.clcc.2020.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION This study aimed to provide a description of existing measures for the prevention and management of epidermal growth factor receptor inhibitor monoclonal antibody-induced skin toxicities and factors impacting patients' adherence to those measures in France, Germany, and Spain. MATERIALS AND METHODS The study consisted of 2 separate surveys. Health care professionals (HCPs; oncologists and nurses) in France, Germany, and Spain were interviewed, and patients with metastatic colorectal cancer and head-and-neck cancer in France and Germany self-completed questionnaires. The study was conducted between February and July 2018. RESULTS A total of 53 oncologists, 44 nurses, and 143 patients participated in the study. HCPs stated that skin toxicities moderately (52%) or severely (28%) impacted patient care. Ninety percent of HCPs reported routine provision of prophylactic measures. The great majority of patients self-reported adherence with the prophylactic (80% to 88% depending on the type of measures) and reactive (93% to drug prescription) skin toxicity recommendations. HCPs estimated patient adherence to be 45% for full adherence and 40% for partial adherence. Most HCPs reported a positive or very positive impact of preventive measures and recommendations on skin toxicity incidence and severity, patients' quality of life, and various aspects of quality of anti-cancer treatment. CONCLUSIONS Skin toxicities are an important adversity negatively impacting on patient care. However, despite the positive perception of the effectiveness of skin toxicity prophylaxis, almost one-third of oncology centers did not provide formal guidelines, and 10% of HCPs did not provide routine prophylactic measures. Patient adherence appears to be high for epidermal growth factor receptor inhibitor monoclonal antibody-induced skin toxicity prevention measures.
Collapse
Affiliation(s)
- George Kafatos
- Center for Observational Research, Amgen Ltd, Uxbridge, United Kingdom.
| | - Sabada Dube
- Center for Observational Research, Amgen Ltd, Uxbridge, United Kingdom
| | - Peter Burdon
- EU Medical Affairs, Amgen (Europe) GmbH, Rotkreuz, Switzerland
| | | | | | | | - Kimberly Lowe
- Center for Observational Research, Amgen Inc, Thousand Oaks, CA
| | | | | |
Collapse
|
28
|
Abstract
Background The incorporation of novel biomarkers into therapy selection for patients with metastatic colorectal cancer (mcrc) has significantly improved outcomes. Optimal treatment planning now takes into account diverse characteristics of patients and their tumours to create personalized therapeutic plans. Discussion This review is split into two sections. In the first section, we review the prognostic and predictive significance of expanded RAS mutation testing, BRAF mutations, ERBB2 (her2) amplification, microsatellite instability (msi) and deficient mismatch repair (dmmr) protein, NTRK fusions, PIK3CA mutations, and met amplifications. The therapeutic implication of each of those biomarkers for personalizing therapies for each patient with mcrc is discussed. In the second section, we touch on testing methods and considerations of relevance to clinicians when they interpret companion diagnostics meant to guide therapy selection. The advantages and pitfalls of various methods are evaluated, and we also look at the potential of liquid biopsies and circulating tumour dna (ctdna) to change the landscape of therapeutic choice and biologic understanding of the disease. Summary Routine testing for extended RAS, BRAF, dmmr or high msi, and NTRK fusions is necessary to determine the best sequencing of chemotherapy and biologic agents for patients with mcrc. Although next-generation sequencing and ctdna are increasingly being adopted, other techniques such as immunohistochemistry retain their relevance in detection of her2 amplification, NTRK fusions, and dmmr.
Collapse
|
29
|
García-Foncillas J, Sunakawa Y, Aderka D, Wainberg Z, Ronga P, Witzler P, Stintzing S. Distinguishing Features of Cetuximab and Panitumumab in Colorectal Cancer and Other Solid Tumors. Front Oncol 2019; 9:849. [PMID: 31616627 PMCID: PMC6763619 DOI: 10.3389/fonc.2019.00849] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/19/2019] [Indexed: 12/27/2022] Open
Abstract
Cetuximab and panitumumab are two distinct monoclonal antibodies (mAbs) targeting the epidermal growth factor receptor (EGFR), and both are widely used in combination with chemotherapy or as monotherapy to treat patients with RAS wild-type metastatic colorectal cancer. Although often considered interchangeable, the two antibodies have different molecular structures and can behave differently in clinically relevant ways. More specifically, as an immunoglobulin (Ig) G1 isotype mAb, cetuximab can elicit immune functions such as antibody-dependent cell-mediated cytotoxicity involving natural killer cells, T-cell recruitment to the tumor, and T-cell priming via dendritic cell maturation. Panitumumab, an IgG2 isotype mAb, does not possess these immune functions. Furthermore, the two antibodies have different binding sites on the EGFR, as evidenced by mutations on the extracellular domain that can confer resistance to one of the two therapeutics or to both. We consider a comparison of the properties of these two antibodies to represent a gap in the literature. We therefore compiled a detailed, evidence-based educational review of the known molecular, clinical, and functional differences between the two antibodies and concluded that they are distinct therapeutic agents that should be considered individually during treatment planning. Available data for one agent can only partly be extrapolated to the other. Looking to the future, the known immune activity of cetuximab may provide a rationale for this antibody as a combination partner with investigational chemotherapy plus immunotherapy regimens for colorectal cancer.
Collapse
Affiliation(s)
- Jesús García-Foncillas
- Cancer Institute, University Hospital Fundacion Jimenez Diaz, Autonomous University of Madrid, Madrid, Spain
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Dan Aderka
- Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Zev Wainberg
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | | | | | - Sebastian Stintzing
- Department of Hematology, Oncology, and Tumor Immunology (CCM) Charité Universitaetsmedizin, Berlin, Germany
| |
Collapse
|
30
|
Gyawali B, Tessema FA, Jung EH, Kesselheim AS. Assessing the Justification, Funding, Success, and Survival Outcomes of Randomized Noninferiority Trials of Cancer Drugs: A Systematic Review and Pooled Analysis. JAMA Netw Open 2019; 2:e199570. [PMID: 31469391 PMCID: PMC6724156 DOI: 10.1001/jamanetworkopen.2019.9570] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/30/2019] [Indexed: 12/12/2022] Open
Abstract
Importance Noninferiority trials test whether a new intervention is not worse than the comparator by a given margin. Objectives To study the characteristics of published randomized noninferiority trials in oncology with overall survival as an end point, to assess the association of justification and success in achieving noninferiority with the funding of these trials, and to evaluate the association of such trials with patient survival. Data Sources A systematic search of PubMed and Google Scholar databases was conducted in March 2018, with no date restrictions. Study Selection Randomized noninferiority trials of cancer drug therapies with overall survival as an end point were included. Trials of decision support, supportive care, and nondrug treatment in both arms were excluded. Data Extraction and Synthesis This study followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines for meta-epidemiological studies. Studies were screened for eligibility criteria, and data on criteria for noninferiority, funding, success (achieving noninferiority), and hazard ratios with confidence intervals for overall survival were extracted. Hazard ratios for overall survival were pooled across trials using a random-effects model. Main Outcomes and Measures Associations of the justification for using a noninferiority design and success in achieving noninferiority with the source of funding were assessed. Overall pooled hazard ratios and confidence intervals for overall survival were calculated. Results Among 74 noninferiority trials of cancer drug therapies, 23 (31%; enrolling 21 437 patients) used overall survival as the primary end point. The noninferiority margins for the hazard ratio of overall survival ranged from 1.08 to 1.33. Noninferiority design was justified in 14 trials (61%) but not in 9 (39%). Overall, 18 trials (78%) concluded with a finding of noninferiority. Industry funding was associated with lack of justification for noninferiority design (P = .02, assessed using the Fisher exact test) but not with success in proving noninferiority (P = .80, assessed using the Fisher exact test). When the hazard ratios across the trials were pooled, there was no beneficial or detrimental association with overall survival, with a pooled hazard ratio of 0.97 (95% CI, 0.92-1.02). Conclusions and Relevance The findings suggest that a substantial fraction of noninferiority trials in oncology, most of which are industry funded, lack justification for such a design. Greater attention to the use of noninferiority designs in randomized clinical trials of cancer drugs from local and national regulators is warranted.
Collapse
Affiliation(s)
- Bishal Gyawali
- Division of Cancer Care and Epidemiology, Departments of Medical Oncology and Public Health Sciences, Queen’s University, Kingston, Ontario, Canada
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Frazer A. Tessema
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emily H. Jung
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aaron S. Kesselheim
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
31
|
Onidani K, Shoji H, Kakizaki T, Yoshimoto S, Okaya S, Miura N, Sekikawa S, Furuta K, Lim CT, Shibahara T, Boku N, Kato K, Honda K. Monitoring of cancer patients via next-generation sequencing of patient-derived circulating tumor cells and tumor DNA. Cancer Sci 2019; 110:2590-2599. [PMID: 31169336 PMCID: PMC6676129 DOI: 10.1111/cas.14092] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/21/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Liquid biopsy of circulating tumor cells (CTC) and circulating tumor DNA (ctDNA) is gaining attention as a method for real-time monitoring in cancer patients. Conventional methods based upon epithelial cell adhesion molecule (EpCAM) expression have a risk of missing the most aggressive CTC subpopulations due to epithelial-mesenchymal transition and may, thus, underestimate the total number of actual CTC present in the bloodstream. Techniques utilizing a label-free inertial microfluidics approach (LFIMA) enable efficient capture of CTC without the need for EpCAM expression. In this study, we optimized a method for analyzing genetic alterations using next-generation sequencing (NGS) of extracted ctDNA and CTC enriched using an LFIMA as a first-phase examination of 30 patients with head and neck cancer, esophageal cancer, gastric cancer and colorectal cancer (CRC). Seven patients with advanced CRC were enrolled in the second-phase examination to monitor the emergence of alterations occurring during treatment with epidermal growth factor receptor (EGFR)-specific antibodies. Using LFIMA, we effectively captured CTC (median number of CTC, 14.5 cells/mL) from several types of cancer and detected missense mutations via NGS of CTC and ctDNA. We also detected time-dependent genetic alterations that appeared during anti-EGFR therapy in CTC and ctDNA from CRC patients. The results of NGS analyses indicated that alterations in the genomic profile revealed by the liquid biopsy could be expanded by using a combination of assays with CTC and ctDNA. The study was registered with the University Hospital Medical Information Network Clinical Trials Registry (ID: UMIN000014095).
Collapse
Affiliation(s)
- Kaoru Onidani
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan.,Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo, Japan
| | - Hirokazu Shoji
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan.,Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiko Kakizaki
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Seiichi Yoshimoto
- Department of Head and Neck Surgery, National Cancer Center Central Hospital, Tokyo, Japan
| | - Shinobu Okaya
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Nami Miura
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Shoichi Sekikawa
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo, Japan
| | - Koh Furuta
- Division of Clinical Laboratory, Kanagawa Cancer Center, Kanagawa, Japan
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore.,Biomedical Institute of Global Health Research and Technology, National University of Singapore, Singapore.,Mechanbiology Institute, National University of Singapore, Singapore, Singapore
| | - Takahiko Shibahara
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo, Japan
| | - Narikazu Boku
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Kazufumi Honda
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan.,Japan Agency for Medical Research and Development (AMED) CREST, Tokyo, Japan
| |
Collapse
|
32
|
Clinical Pharmacokinetics and Pharmacodynamics of the Epidermal Growth Factor Receptor Inhibitor Panitumumab in the Treatment of Colorectal Cancer. Clin Pharmacokinet 2019; 57:455-473. [PMID: 28853050 PMCID: PMC5856878 DOI: 10.1007/s40262-017-0590-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite progress in the treatment of metastatic colorectal cancer (mCRC) in the last 15 years, it is still a condition with a relatively low 5-year survival rate. Panitumumab, a fully human monoclonal antibody directed against the epidermal growth factor receptor (EGFR), is able to prolong survival in patients with mCRC. Panitumumab is used in different lines of therapy in combination with chemotherapy, and as monotherapy for the treatment of wild-type (WT) RAS mCRC. It is administered as an intravenous infusion of 6 mg/kg every 2 weeks and has a t½ of approximately 7.5 days. Elimination takes place via two different mechanisms, and immunogenicity rates are low. Only RAS mutations have been confirmed as a negative predictor of efficacy with anti-EGFR antibodies. Panitumumab is generally well tolerated and has a manageable toxicity profile, despite a very high prevalence of dermatologic side effects. This article presents an overview of the clinical pharmacokinetics and pharmacodynamics of panitumumab, including a description of the studies that led to its approval in the different lines of therapy of mCRC.
Collapse
|
33
|
Yoshino T, Arnold D, Taniguchi H, Pentheroudakis G, Yamazaki K, Xu RH, Kim TW, Ismail F, Tan IB, Yeh KH, Grothey A, Zhang S, Ahn JB, Mastura MY, Chong D, Chen LT, Kopetz S, Eguchi-Nakajima T, Ebi H, Ohtsu A, Cervantes A, Muro K, Tabernero J, Minami H, Ciardiello F, Douillard JY. Pan-Asian adapted ESMO consensus guidelines for the management of patients with metastatic colorectal cancer: a JSMO-ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann Oncol 2019; 29:44-70. [PMID: 29155929 DOI: 10.1093/annonc/mdx738] [Citation(s) in RCA: 413] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The most recent version of the European Society for Medical Oncology (ESMO) consensus guidelines for the treatment of patients with metastatic colorectal cancer (mCRC) was published in 2016, identifying both a more strategic approach to the administration of the available systemic therapy choices, and a greater emphasis on the use of ablative techniques, including surgery. At the 2016 ESMO Asia Meeting, in December 2016, it was decided by both ESMO and the Japanese Society of Medical Oncology (JSMO) to convene a special guidelines meeting, endorsed by both ESMO and JSMO, immediately after the JSMO 2017 Annual Meeting. The aim was to adapt the ESMO consensus guidelines to take into account the ethnic differences relating to the toxicity as well as other aspects of certain systemic treatments in patients of Asian ethnicity. These guidelines represent the consensus opinions reached by experts in the treatment of patients with mCRC identified by the Presidents of the oncological societies of Japan (JSMO), China (Chinese Society of Clinical Oncology), Korea (Korean Association for Clinical Oncology), Malaysia (Malaysian Oncological Society), Singapore (Singapore Society of Oncology) and Taiwan (Taiwan Oncology Society). The voting was based on scientific evidence and was independent of both the current treatment practices and the drug availability and reimbursement situations in the individual participating Asian countries.
Collapse
Affiliation(s)
- T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - D Arnold
- CUF Hospitals Cancer Centre, Lisbon, Portugal
| | - H Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - G Pentheroudakis
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece
| | - K Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - R-H Xu
- Department of Medical Oncology, Sun Yat-Sen University (SYSU) Cancer Center, Guangzhou, China
| | - T W Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - F Ismail
- Department of Radiotherapy & Oncology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - I B Tan
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - K-H Yeh
- Department of Oncology, National Taiwan University Hospital, and Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - A Grothey
- Division of Medical Oncology, Mayo Clinic Cancer Center, Rochester, USA
| | - S Zhang
- Cancer Institute, Zhejiang University, Hangzhou, China
| | - J B Ahn
- Division of Oncology, Department of Internal Medicine, Yonsei Cancer Center, Seoul, Korea
| | - M Y Mastura
- Pantai Cancer Institute, Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - D Chong
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - L-T Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - S Kopetz
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Centre, Houston, USA
| | - T Eguchi-Nakajima
- Department of Clinical Oncology, School of Medicine, St. Marianna University, Kanagawa, Japan
| | - H Ebi
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - A Ohtsu
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - A Cervantes
- CIBERONC, Department of Medical Oncology, Institute of Health Research, INCLIVIA, University of Valencia, Valencia, Spain
| | - K Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - J Tabernero
- Medical Oncology Department, Vall d' Hebron University Hospital, Vall d'Hebron Institute of Oncology (V.H.I.O.), Barcelona, Spain
| | - H Minami
- Department of Medical Oncology and Hematology, Kobe University Hospital, Kobe, Japan
| | - F Ciardiello
- Division of Medical Oncology, Seconda Università di Napoli, Naples, Italy
| | | |
Collapse
|
34
|
Nagamine A, Araki T, Nagano D, Miyazaki M, Yamamoto K. L-Lactate dehydrogenase B may be a predictive marker for sensitivity to anti-EGFR monoclonal antibodies in colorectal cancer cell lines. Oncol Lett 2019; 17:4710-4716. [PMID: 30944657 DOI: 10.3892/ol.2019.10075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/31/2019] [Indexed: 01/19/2023] Open
Abstract
Recently, proteins derived from cancer cells have been widely investigated as biomarkers for predicting the efficacy of chemotherapy. In this study, to identify a sensitive biomarker for the efficacy of anti-epidermal growth factor receptor monoclonal antibodies (anti-EGFR mAbs), proteins derived from 6 colorectal cancer (CRC) cell lines with different sensitivities to cetuximab, an anti-EGFR mAb, were analyzed. Cytoplasmic and membrane proteins extracted from each CRC cell line were digested using trypsin and analyzed comprehensively using mass spectrometry. As a result, 148 and 146 peaks from cytoplasmic proteins and 363 and 267 peaks from membrane proteins were extracted as specific peaks for cetuximab-resistant and -sensitive CRC cell lines, respectively. By analyzing the proteins identified from the peptide peaks, cytoplasmic L-lactate dehydrogenase B (LDHB) was detected as a marker of cetuximab sensitivity, and it was confirmed that LDHB expression was increased in cetuximab-resistant CRC cell lines. Furthermore, LDHB expression levels were significantly upregulated with the acquisition of resistance to cetuximab in cetuximab-sensitive CRC cell lines. In conclusion, LDHB was identified as an important factor affecting cetuximab sensitivity using comprehensive proteome analysis for the first time.
Collapse
Affiliation(s)
- Ayumu Nagamine
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Takuya Araki
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Daisuke Nagano
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Mitsue Miyazaki
- Division of Endocrinology Metabolism and Signal Research, Gunma University Initiative for Advanced Research and Institute for Molecular and Cellular Regulation, Maebashi, Gunma 371-8511, Japan
| | - Koujirou Yamamoto
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
35
|
Geldof T, Rawal S, Dyck WV, Huys I. Comparative and combined effectiveness of innovative therapies in cancer: a literature review. J Comp Eff Res 2019; 8:205-216. [PMID: 30616358 DOI: 10.2217/cer-2018-0131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
To achieve therapeutic innovation in oncology, already expensive novel medicines are often concomitantly combined to potentially enhance effectiveness. While this aggravates the pricing problem, comparing effectiveness of novel yet expensive (concomitant) treatments is much needed for healthcare decision-making to deliver effective but affordable treatments. This study reviewed published clinical trials and real-world studies of targeted and immune therapies. In total, 48 studies compared and/or combined multiple novel products on breast, colorectal, lung and melanoma cancers. To a great extent, products evaluated in each study were owned by one manufacturer. However, cross-manufacturer assessments are also needed. Next to costs and intensive market competition, the absence of a regulatory framework enforcing real-world multiproduct studies prevents these from being conducted. Trusted third parties could facilitate such real-world studies, for which appropriate and efficient data access is needed.
Collapse
Affiliation(s)
- Tine Geldof
- Healthcare Management Centre, Vlerick Business School, Reep 1, Ghent 9000, Belgium.,Pharmaceutical Care & Pharmaco-economics, KU Leuven, O&N II, Leuven 3001, Belgium
| | - Smita Rawal
- Department of Pharmaceutical Health Services, Outcomes & Policy, College of Pharmacy, University of Georgia, Athens 30602, GA, USA
| | - Walter Van Dyck
- Healthcare Management Centre, Vlerick Business School, Reep 1, Ghent 9000, Belgium.,Pharmaceutical Care & Pharmaco-economics, KU Leuven, O&N II, Leuven 3001, Belgium
| | - Isabelle Huys
- Pharmaceutical Care & Pharmaco-economics, KU Leuven, O&N II, Leuven 3001, Belgium
| |
Collapse
|
36
|
Buchler T, Chloupkova R, Poprach A, Fiala O, Kiss I, Kopeckova K, Dusek L, Veskrnova V, Slavicek L, Kohoutek M, Finek J, Svoboda M, Petruzelka L, Melichar B. Sequential therapy with bevacizumab and EGFR inhibitors for metastatic colorectal carcinoma: a national registry-based analysis. Cancer Manag Res 2018; 11:359-368. [PMID: 30643461 PMCID: PMC6314050 DOI: 10.2147/cmar.s183093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Although inhibitors of vascular endothelial growth factor and inhibitors of epidermal growth factor receptor (EGFRi) are commonly used for the treatment of metastatic colorectal cancer (mCRC), the optimal sequencing of these agents is currently unclear. Methods A national registry of targeted therapies was used to analyze baseline characteristics and outcomes of patients with mCRC and wild-type KRAS exon 2 status who received bevacizumab and EGFRi (cetuximab or panitumumab) as a part of first- and second-line treatment in either sequence. Results The cohort included 490 patients (181 patients treated with first-line EGFRi and second-line bevacizumab and 309 patients treated with first-line bevacizumab and second-line EGFRi). Median overall survival (OS) from the initiation on first-line therapy was similar for patients treated with either sequence, reaching 31.8 (95% CI 27.5-36.1) vs 31.4 months (95% CI 27.8-35.0) for EGFRi → bevacizumab vs bevacizumab → EGFRi cohort, respectively. Time from first-line initiation to progression on the second-line therapy [progression-free survival (PFS)] was 21.1 (95% CI 19.3-23.0) vs 19.3 months (95% CI 17.3-21.3) for bevacizumab → EGFRi vs EGFRi → bevacizumab cohort, respectively (P=0.016). Conclusion This retrospective analysis of real-world data of patients with wild-type KRAS exon 2 mCRC showed no differences in OS between cohorts treated with bevacizumab → EGFRi vs the reverse sequence while combined PFS favored the bevacizumab → EGFRi sequence.
Collapse
Affiliation(s)
- Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, 140 59 Prague, Czech Republic,
| | - Renata Chloupkova
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Alexandr Poprach
- Department of Comprehensive Cancer Care and Faculty of Medicine, Masaryk Memorial Cancer Institute and Masaryk University, Brno 656 53, Czech Republic
| | - Ondrej Fiala
- Department of Oncology, University Hospital, 304 60 Pilsen, Czech Republic.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Igor Kiss
- Department of Comprehensive Cancer Care and Faculty of Medicine, Masaryk Memorial Cancer Institute and Masaryk University, Brno 656 53, Czech Republic
| | - Katerina Kopeckova
- Department of Oncology, Motol University Hospital and Second Faculty of Medicine, Charles University, 150 00 Prague, Czech Republic
| | - Ladislav Dusek
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Veronika Veskrnova
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, 140 59 Prague, Czech Republic,
| | - Lubomir Slavicek
- Department of Oncology, Jihlava Hospital Comprehensive Cancer Centre, Jihlava, Czech Republic
| | - Milan Kohoutek
- Department of Oncology, T Bata Hospital and Comprehensive Cancer Centre, Zlin, Czech Republic
| | - Jindrich Finek
- Department of Oncology, University Hospital, 304 60 Pilsen, Czech Republic
| | - Marek Svoboda
- Department of Comprehensive Cancer Care and Faculty of Medicine, Masaryk Memorial Cancer Institute and Masaryk University, Brno 656 53, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, General University Hospital and Charles University First Faculty of Medicine, 128 08 Prague, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Palacky University Medical School and Teaching Hospital, 775 20 Olomouc, Czech Republic
| |
Collapse
|
37
|
Tanioka H, Asano M, Yoshida R, Waki N, Uno F, Ishizaki M, Yamashita K, Morishita Y, Nagasaka T. Cetuximab retreatment in patients with metastatic colorectal cancer who exhibited a clinical benefit in response to prior cetuximab: A retrospective study. Oncol Lett 2018; 16:3674-3680. [PMID: 30127977 PMCID: PMC6096178 DOI: 10.3892/ol.2018.9127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/05/2018] [Indexed: 12/13/2022] Open
Abstract
Clinical benefits of cetuximab retreatment in patients with metastatic colorectal (mCRC) have been reported. In the present study, the effect of cetuximab retreatment on predictive markers was investigated by evaluating the clinical benefit of initial cetuximab treatment prior to cetuximab retreatment. Between November 2012 and March 2017, 14 patients with KRAS proto-oncogene GTPase exon 2 wild-type mCRC who exhibited a clinical benefit (confirmed stable disease for at least 6 months or a clinical response) to an initial cetuximab-based regimen, who received multiple lines of chemotherapy following disease progression and ultimately received a second cetuximab and irinotecan regimen, were retrospectively analyzed. For retreatment, patients received bi-weekly irinotecan (120-150 mg/m2) combined with cetuximab (400 mg/m2 as an initial dose, followed by 250 mg/m2, weekly). The median age of the 14 patients (11 males, 3 females) was 68 years (32-77). The median progression-free survival (PFS) following prior cetuximab-based therapy was 6.6 months (range, 4.1-18.4). Initial cetuximab treatment was administered as a first-line treatment in 11 patients, a second-line treatment in 1 patient and a third-line treatment in 2 patients. The median interval time between the last cycle of initial cetuximab-based therapy and the first cycle of cetuximab retreatment was 13.1 months (range, 6.0-37.1). The objective response rate of cetuximab retreatment was 21.4% and the median PFS was 4.4 months (95% confidence interval, 1.4-5.6). The Spearman's correlation coefficient for the PFS following retreatment and duration of initial cetuximab-based regimens demonstrated a more marked correlation compared with that between the PFS following retreatment and the interval time between the two regimens (r=0.45, P=0.11 vs. r=0.08, P=0.79). Cetuximab retreatment may provide clinical benefit to patients with mCRC who were good responders with longer periods of initial cetuximab-based therapy.
Collapse
Affiliation(s)
- Hiroaki Tanioka
- Department of Medical Oncology, Okayama Rosai Hospital, Okayama 702-8055, Japan.,Department of Clinical Oncology, Kawasaki Medical School Hospital, Okayama 701-0192, Japan
| | - Motoi Asano
- Department of Medical Oncology, Okayama Rosai Hospital, Okayama 702-8055, Japan
| | - Ryousuke Yoshida
- Department of Surgery, Okayama Rosai Hospital, Okayama 702-8055, Japan
| | - Naohisa Waki
- Department of Surgery, Okayama Rosai Hospital, Okayama 702-8055, Japan
| | - Futoshi Uno
- Department of Surgery, Okayama Rosai Hospital, Okayama 702-8055, Japan
| | - Masahiro Ishizaki
- Department of Surgery, Okayama Rosai Hospital, Okayama 702-8055, Japan
| | - Kazuki Yamashita
- Department of Surgery, Okayama Rosai Hospital, Okayama 702-8055, Japan
| | - Yuki Morishita
- Department of Pharmacy, Okayama Rosai Hospital, Okayama 702-8055, Japan
| | - Takeshi Nagasaka
- Department of Clinical Oncology, Kawasaki Medical School Hospital, Okayama 701-0192, Japan
| |
Collapse
|
38
|
Pinto C, Normanno N, Orlandi A, Fenizia F, Damato A, Maiello E, Tamburini E, Di Costanzo F, Tonini G, Bilancia D, Corsi D, Pisconti S, Ferrau F, Gori S, Daniele B, Zaniboni A, Soto Parra H, Frassinetti GL, Iaffaioli RV, Cassata A, Zampino MG, Repetto L, Calegari MA, Barone C. Phase III study with FOLFIRI + cetuximab versus FOLFIRI + cetuximab followed by cetuximab alone in RAS and BRAF WT mCRC. Future Oncol 2018; 14:1339-1346. [PMID: 29846100 DOI: 10.2217/fon-2017-0592] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The optimal duration and intensity of first-line therapy in metastatic colorectal cancer patients once they have achieved an objective response is controversial. In a molecularly selected RAS and BRAF wild-type (wt) population, this concern is amplified. Once disease control has been achieved with a combination therapy including an anti-EGFR antibody, further exposure both to cytotoxic drugs and targeted therapy might result only in increased toxicity. In unresectable metastatic RAS and BRAF wt colorectal cancer patients, a deintensified therapy could represent a valuable option that might preserve quality of life. We designed a study to compare FOLFIRI/cetuximab to FOLFIRI/cetuximab for eight cycles followed by cetuximab alone in first-line treatment of RAS and BRAF (wt) metastatic colorectal cancer patients.
Collapse
Affiliation(s)
- Carmine Pinto
- Medical Oncology, Clinical Cancer Center, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Nicola Normanno
- Cell Biology & Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Armando Orlandi
- Oncology Unit, Fondazione Policlinico Universitario "A Gemelli", Roma, Lazio, Italy
| | - Francesca Fenizia
- Cell Biology & Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Angela Damato
- Medical Oncology, Clinical Cancer Center, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Evaristo Maiello
- Oncology Unit, Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Puglia, Italy
| | - Emiliano Tamburini
- Oncology Unit Azienda Unita Sanitaria Locale della Romagna, Rimini, Emilia-Romagna, Italy
| | | | - Giuseppe Tonini
- Oncology Unit, Università Campus Bio-Medico di Roma, Roma, Lazio, Italy
| | | | - Domenico Corsi
- Oncology Unit, San Giovanni Calabita Hospital, Roma, Lazio, Italy
| | | | | | - Stefania Gori
- Medical Oncology, Clinical Cancer Center, Ospedale Sacro Cuore Don Calabria, Negrar, Veneto, Italy
| | - Bruno Daniele
- Oncology Unit, Azienda Ospedaliera Gaetano Rummo, Benevento, Campania, Italy
| | - Alberto Zaniboni
- Oncology Unit, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Lombardia, Italy
| | - Héctor Soto Parra
- Oncology Unit, Università degli Studi di Catania Scuola di Facoltà di Medicina, Catania, Sicilia, Italy
| | - Giovanni Luca Frassinetti
- Oncology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori Meldola, Emilia-Romagna, Italy
| | - Rosario Vincenzo Iaffaioli
- Oncologia Clinica Sperimentale Addome, Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Antonio Cassata
- Oncologia Clinica Sperimentale Addome, Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | | - Lazzaro Repetto
- Oncology Unit, Ospedale Civile "G Borea", Sanremo, Liguria, Italy
| | | | - Carlo Barone
- Oncology Unit, Fondazione Policlinico Universitario "A Gemelli", Roma, Lazio, Italy
| | | |
Collapse
|
39
|
McGregor M, Price TJ. Panitumumab in the treatment of metastatic colorectal cancer, including wild-type RAS, KRAS and NRAS mCRC. Future Oncol 2018; 14:2437-2459. [PMID: 29737864 DOI: 10.2217/fon-2017-0711] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The humanized monoclonal antibody panitumumab, targeted against EGFR, plays an important role in patients with metastatic colorectal cancer. This article reviews the body of evidence for panitumumab which demonstrates significant benefits across multiple lines of therapy in those without an extended RAS mutation. The use of panitumumab with RAS mutations is not beneficial and possibly harmful. Panitumumab is well tolerated with manageable toxicities. The role of panitumumab continues to evolve as understanding of sequencing of therapies grows. There is evidence for use as maintenance therapy and conversion therapy for unresectable liver metastases. Future research is likely to focus on biomarkers for improved patient selection and the development of novel therapeutic strategies to overcome resistance.
Collapse
Affiliation(s)
- Mark McGregor
- Medical Oncology, Adelaide Oncology & Haematology, North Adelaide, Australia.,Medical Oncology, Flinders Medical Centre, Adelaide, Australia
| | - Timothy J Price
- Medical Oncology, Adelaide Oncology & Haematology, North Adelaide, Australia.,Medical Oncology, The Queen Elizabeth Hospital & University of Adelaide, Woodville, Adelaide, Australia
| |
Collapse
|
40
|
Kim TW, Elme A, Park JO, Udrea AA, Kim SY, Ahn JB, Valencia RV, Krishnan S, Manojlovic N, Guan X, Lofton-Day C, Jung AS, Vrdoljak E. Final Analysis of Outcomes and RAS/BRAF Status in a Randomized Phase 3 Study of Panitumumab and Best Supportive Care in Chemorefractory Wild Type KRAS Metastatic Colorectal Cancer. Clin Colorectal Cancer 2018; 17:206-214. [PMID: 29703606 DOI: 10.1016/j.clcc.2018.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/21/2018] [Accepted: 03/14/2018] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Tumor rat sarcoma gene (RAS) status is a negative predictive biomarker for anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer (mCRC). We analyzed outcomes according to RAS and v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutational status, and evaluated early tumor shrinkage (ETS) and depth of response (DpR) for patients with wild type RAS. PATIENTS AND METHODS Patients with confirmed metastatic colon or rectum adenocarcinoma, wild type Kristen rat sarcoma gene tumor exon 2 status, clinical/radiologic disease progression or toxicity during irinotecan or oxaliplatin treatment, and no previous anti-EGFR therapy were randomized 1:1 to receive best supportive care (BSC) with or without panitumumab (6.0 mg/kg, intravenously, on day 1 of each 14-day cycle) in this open-label, multicenter, phase III study (20100007). RAS and BRAF mutation status were determined using Sanger sequencing. ETS was evaluated as maximum percentage change from baseline to week 8; DpR was calculated as the percentage change for tumor shrinkage at nadir versus baseline. RESULTS Overall, 270 patients had RAS wild type mCRC (panitumumab with BSC, n = 142; BSC, n = 128). For patients with wild type RAS tumors, median overall survival (OS; hazard ratio [HR], 0.72; P = .015) and progression-free survival (PFS; HR, 0.45; P < .0001) were improved with panitumumab with BSC versus BSC. Similar improvements were seen for patients with wild type RAS, and wild type BRAF tumors (OS: HR, 0.75; P = .04; PFS: HR, 0.45; P < .0001). Median DpR was 16.9% for the evaluable panitumumab with BSC wild type RAS population. Overall, 69.5% experienced any type of tumor shrinkage at week 8; 38.2% experienced ≥ 20% shrinkage. Similar improvements in OS and PFS were seen with stratification according to ETS. CONCLUSION This analysis showed that panitumumab improved outcomes in wild type RAS mCRC and indicated that ETS and DpR could be used as additional efficacy markers.
Collapse
Affiliation(s)
- Tae Won Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Anneli Elme
- Department of Gastroenterology, Oncology, North Estonia Medical Centre Foundation, Tallinn, Estonia
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Sun Young Kim
- Division of Hemato-Oncology, National Cancer Center, Goyang, Gyeonggi-do, South Korea
| | - Joong Bae Ahn
- Department of Internal Medicine, Yonsei University Health System Severance Hospital, Seoul, South Korea
| | - Ricardo Villalobos Valencia
- Department of Medical Oncology, Hospital de Oncologia, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Srinivasan Krishnan
- Department of Clinical Research, Dr Rai Memorial Medical Centre, Chennai, Tamil Nadu, India
| | - Nebojsa Manojlovic
- Department of Digestive Oncology, Clinic for Gastroenterology and Hepatology of Military Medical Academy of Serbia, Belgrade, Serbia
| | | | | | | | - Eduard Vrdoljak
- Department of Oncology, Center of Oncology, Clinical Hospital Center Split, Split, Croatia
| |
Collapse
|
41
|
A phase II trial to evaluate the efficacy of panitumumab combined with fluorouracil-based chemotherapy for metastatic colorectal cancer: the PF trial. Cancer Chemother Pharmacol 2018; 81:829-838. [DOI: 10.1007/s00280-018-3556-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/28/2018] [Indexed: 12/12/2022]
|
42
|
Fernandes MS, Sanches JM, Seruca R. Targeting the PI3K Signalling as a Therapeutic Strategy in Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1110:35-53. [PMID: 30623365 DOI: 10.1007/978-3-030-02771-1_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer mortality worldwide. Regarded as a heterogeneous disease, a number of biomarkers have been proposed to help in the stratification of CRC patients and to enable the selection of the best therapy for each patient towards personalized therapy. However, although the molecular mechanisms underlying the development of CRC have been elucidated, the therapeutic strategies available for these patients are still quite limited. Thus, over the last few years, a multitude of novel targets and therapeutic strategies have emerged focusing on deregulated molecules and pathways that are implicated in cell growth and survival. Particularly relevant in CRC are the activating mutations in the oncogene PIK3CA that frequently occur in concomitancy with KRAS and BRAF mutations and that lead to deregulation of the major signalling pathways PI3K and MAPK, downstream of EGFR. This review focus on the importance of the PI3K signalling in CRC development, on the current knowledge of PI3K inhibition as a therapeutic approach in CRC and on the implications PI3K signalling molecules may have as potential biomarkers and as new targets for directed therapies in CRC patients.
Collapse
Affiliation(s)
- Maria Sofia Fernandes
- Institute for Systems and Robotics (ISR), Instituto Superior Técnico (IST), Lisboa, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - João Miguel Sanches
- Institute for Systems and Robotics (ISR), Instituto Superior Técnico (IST), Lisboa, Portugal
| | - Raquel Seruca
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal.
- Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
43
|
Lièvre A, Ouine B, Canet J, Cartier A, Amar Y, Cacheux W, Mariani O, Guimbaud R, Selves J, Lecomte T, Guyetant S, Bieche I, Berger F, de Koning L. Protein biomarkers predictive for response to anti-EGFR treatment in RAS wild-type metastatic colorectal carcinoma. Br J Cancer 2017; 117:1819-1827. [PMID: 29024937 PMCID: PMC5729470 DOI: 10.1038/bjc.2017.353] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/02/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Metastatic colorectal cancer (mCRC) patients with mutant KRAS or NRAS are ineligible for anti-epidermal growth factor receptor (anti-EGFR) therapy, as RAS mutations activate downstream pathways independently of EGFR and induce primary resistance. However, even among RAS wild-type (WT) patients, only a fraction responds to anti-EGFR therapy, suggesting that other mechanisms of resistance exist. We hypothesise that different (epi)genetic alterations can lead to primary anti-EGFR resistance and that the crucial end point is the activation of protein signalling pathways. METHODS We analysed the expression and activation of proteins involved in cell signalling, using reverse phase protein arrays, on a multicentre French cohort of RAS WT mCRC treated with anti-EGFR treatment. RESULTS We identify activated EGFR and HER3 as protein biomarkers predictive for better overall survival. Active EGFR signalling and downstream PI3K, but not MAPK, pathway activation are associated with response to anti-EGFR treatment. Left-sided mCRC displays active ErbB2/3 and Wnt pathways and a better response to anti-EGFR therapy compared to right-sided mCRC. CONCLUSIONS We identify active EGFR and PI3K signalling as a key factor for response to anti-EGFR treatment in mCRC and highlight the importance of developing these biomarkers in clinical practice for the selection of RAS WT mCRC patients that would benefit from anti-EGFR treatment.
Collapse
Affiliation(s)
- Astrid Lièvre
- Service des maladies de l’appareil digestif, CHU Pontchaillou, 2 rue Henri Le Guilloux, Rennes cedex 09 35033, France
- Université Rennes 1, Faculté de médecine, 2 Avenue du Prof. Léon Bernard, Rennes 35043, France
- Inserm ER440-Oncogenesis, Stress and Signaling, Rue Bataille Flandres-Dunkerque, Rennes 35042, France
| | - Bérèngere Ouine
- Institut Curie, PSL Research University, Department of Translational Research, 26 rue d’Ulm, Paris 75005, France
| | - Jim Canet
- Institut Curie, PSL Research University, Unit of Biostatistics, 26 rue d’Ulm, Paris 75005, France
| | - Aurélie Cartier
- Institut Curie, PSL Research University, Department of Translational Research, 26 rue d’Ulm, Paris 75005, France
| | - Yael Amar
- Institut Curie, PSL Research University, Unit of Biostatistics, 26 rue d’Ulm, Paris 75005, France
| | - Wulfran Cacheux
- Institut Curie, Department of Medical Oncology, René Huguenin Hospital, 35 rue Dailly, Saint-Cloud 92210, France
- Institut Curie, Unit of Pharmacogenomics, Department of Genetics, 26 rue d’Ulm, Paris 75005, France
| | - Odette Mariani
- Institut Curie, PSL Research University, Biological Resource Center, 26 rue d’Ulm, Paris 75005, France
| | - Rosine Guimbaud
- Centre de Recherche en Cancérologie de Toulouse, Unité Mixte de Recherche, 1037 INSERM—Université Toulouse III, Toulouse 31062, France
- Service d’oncologie médicale, Centre Hospitalier Universitaire de Toulouse, Toulouse 31059, France
| | - Janick Selves
- Centre de Recherche en Cancérologie de Toulouse, Unité Mixte de Recherche, 1037 INSERM—Université Toulouse III, Toulouse 31062, France
- Department of Pathology, Centre Hospitalier Universitaire de Toulouse, Toulouse 31059, France
| | - Thierry Lecomte
- Hôpital Trousseau—CHRU de TOURS, Service d'Hépato-Gastro-Entérologie, Tours 37000, France
- UMR CNRS 7292 (GICC), Université François Rabelais, Tours 37000, France
| | - Serge Guyetant
- Hôpital Trousseau—CHRU de TOURS, Service d'Anatomie et Cytologie Pathologiques—Tumorothèque, Tours 37000, France
| | - Ivan Bieche
- Institut Curie, Unit of Pharmacogenomics, Department of Genetics, 26 rue d’Ulm, Paris 75005, France
| | - Frédérique Berger
- Institut Curie, PSL Research University, Unit of Biostatistics, 26 rue d’Ulm, Paris 75005, France
- Institut Curie, PSL Research University, INSERM U900, 26 rue d’Ulm, Paris 75005, France
| | - Leanne de Koning
- Institut Curie, PSL Research University, Department of Translational Research, 26 rue d’Ulm, Paris 75005, France
| |
Collapse
|
44
|
Matikas A, Margolin S, Hellström M, Johansson H, Bengtsson NO, Karlsson L, Edlund P, Karlsson P, Lidbrink E, Linderholm B, Lindman H, Malmstrom P, Villman K, Foukakis T, Bergh J. Long-term safety and survival outcomes from the Scandinavian Breast Group 2004-1 randomized phase II trial of tailored dose-dense adjuvant chemotherapy for early breast cancer. Breast Cancer Res Treat 2017; 168:349-355. [PMID: 29190004 PMCID: PMC5838137 DOI: 10.1007/s10549-017-4599-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Although adjuvant polychemotherapy improves outcomes for early breast cancer, the significant variability in terms of pharmacokinetics results in differences in efficacy and both short and long-term toxicities. Retrospective studies support the use of dose tailoring according to the hematologic nadirs. METHODS The SBG 2004-1 trial was a randomized feasibility phase II study which assessed tailored dose-dense epirubicin and cyclophosphamide (EC) followed by docetaxel (T) (group A), the same regimen with fixed doses (group B) and the TAC regimen (group C). Women aged 18-65 years, ECOG PS 0-1 with at least one positive axillary lymph node were randomized 1:1:1. The primary endpoint of the study was the safety and feasibility of the treatment. Toxicity was graded according to CTC-AE version 3.0. The design and short-term toxicity have been previously published. Here, we report safety and efficacy data after 10 years of follow-up. RESULTS A total of 124 patients were included in the study. After a median follow-up of 10.3 years, the probability for 10-year survival was 78.5, 75.1, and 63.4% and for relapse free survival 64.1, 71.0, and 59.5% for groups A, B, and C, respectively. There were no cases of clinically diagnosed cardiotoxicity or hematologic malignancies. No patient was lost to follow-up. CONCLUSIONS In this randomized phase II trial, tailored dose adjuvant chemotherapy was feasible, without an increased risk for long-term adverse events after a median follow-up of 10 years.
Collapse
Affiliation(s)
- Alexios Matikas
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden.
| | - Sara Margolin
- Department of Oncology, Stockholm South General Hospital, Stockholm, Sweden
| | - Mats Hellström
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Hemming Johansson
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Lidbrink
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Barbro Linderholm
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Lindman
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Per Malmstrom
- Department of Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | | - Theodoros Foukakis
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
45
|
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common cancers all over the world, but its epidemiology is obviously different in various regions. METHODS The treatment of CRC also has varying characteristics due to differences in economy, geography, disease onset, chemotherapy, and other factors, although international guidelines are used to guide the treatment of CRC in China. RESULTS This paper summarizes the current status of CRC treatment, including surgical therapy, neoadjuvant radiotherapy and chemotherapy, postoperative chemotherapy, targeted therapy, maintenance therapy, and immunotherapy, according to the clinical situation in China, so as to provide better therapy and improve clinical practice for patients with CRC. CONCLUSION This research shows that the treatment of colorectal cancer continues to progress, and the patient's efficacy and quality of life have improved.
Collapse
Affiliation(s)
- Yumei Zhang
- Department of VIP Clinic, Tongji University South Branch of Shanghai East Hospital
| | - Zhiyu Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College
| | - Jin Li
- Department of VIP Clinic, Tongji University South Branch of Shanghai East Hospital
- Department of Oncology, Tongji University Shanghai East Hospital, Shanghai, China
| |
Collapse
|
46
|
Gong J, Cho M, Fakih M. RAS and BRAF in metastatic colorectal cancer management. J Gastrointest Oncol 2016; 7:687-704. [PMID: 27747083 PMCID: PMC5056249 DOI: 10.21037/jgo.2016.06.12] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/17/2016] [Indexed: 12/22/2022] Open
Abstract
The treatment of metastatic colorectal cancer (mCRC) has been further refined with the development of monoclonal antibodies, cetuximab and panitumumab, towards the epidermal growth factor receptor (EGFR). Anti-EGFR therapy has afforded improved survival in those with wild-type RAS mCRC but provides no benefit and even harm in those with RAS-mutant tumors. BRAF mutations have also been shown to predict lack of clinically meaningful benefit to anti-EGFR therapy in mCRC. Mechanisms of resistance to EGFR blockade in wild-type RAS or BRAF metastatic colorectal tumors appear to converge on the mitogen-activated protein kinase (MAPK) signaling pathway. Clinical trials involving combined BRAF, EGFR, and/or MAPK kinase (MEK) inhibition have shown promising activity in BRAF-mutant mCRC. Here, we review pivotal clinical trials that have redefined our treatment approach in mCRC with respect to anti-EGFR therapy based on RAS and BRAF mutation status. Future studies will likely focus on improving efficacy of anti-EGFR-based therapy in mCRC through sustained MAPK pathway inhibition.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - May Cho
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Marwan Fakih
- Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|