1
|
Concannon KF, Glisson BS, Doebele RC, Huang C, Marotti M, Camidge DR, Heymach JV. A Phase I Open-Label Study of Cediranib Plus Etoposide and Cisplatin as First-Line Therapy for Patients With Extensive-Stage Small-Cell Lung Cancer or Metastatic Neuroendocrine Non-Small-Cell Lung Cancer. Clin Lung Cancer 2024; 25:601-611. [PMID: 39307607 DOI: 10.1016/j.cllc.2024.08.015] [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: 07/22/2024] [Accepted: 08/27/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Small cell lung cancer (SCLC) is known to express high levels of the proangiogenic factor vascular endothelial growth factor (VEGF). We assessed the safety and tolerability of cediranib, an oral inhibitor of VEGF receptor tyrosine kinases, in combination with etoposide and cisplatin as first-line therapy for extensive-stage (ES) SCLC or metastatic lung neuroendocrine cancer (NEC). METHODS Patients received up to six 21-day cycles of etoposide (100 mg/m2, days 1-3) and cisplatin (80 mg/m2, day 1) with once-daily cediranib until disease progression or unacceptable toxicity. Cediranib dosing started at 30 mg with de-escalation cohorts planned based on cycle 1 dose-limiting toxicities (DLTs). An expansion cohort of 12 patients was enrolled at the recommended phase II dose. RESULTS Twenty-two patients (18 with ES SCLC, 4 with NEC) received treatment. Only 4 patients were enrolled at the 30 mg cediranib dose before other studies established 20 mg/day as the recommended dose with chemotherapy. Among the 18 patients enrolled at the 20-mg dose, common adverse events included nausea/vomiting, neutropenia, and diarrhea; 8 patients (44%) had grade 1 or 2 hypertension, and 2 (11%) had grade 3 hemoptysis. For all 18 patients, the objective response rate and median progression-free survival duration were 67% and 7.9 months. Plasma levels of VEGF were significantly higher, and those of soluble VEGFR2 were significantly lower, on day 22 than at baseline but were not correlated with tumor shrinkage. CONCLUSIONS Cediranib (20 mg) plus etoposide and cisplatin is well tolerated and has promising clinical activity.
Collapse
Affiliation(s)
- Kyle F Concannon
- Department of Hematology/Onscology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bonnie S Glisson
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado Cancer Center, Aurora, CO
| | - Chao Huang
- Department of Medical Oncology, University of Kansas, Kansas City, KS
| | | | - D Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado Cancer Center, Aurora, CO
| | - John V Heymach
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; AstraZeneca, Macclesfield, United Kingdom.
| |
Collapse
|
2
|
Satora M, Kułak K, Zaremba B, Grunwald A, Świechowska-Starek P, Tarkowski R. New hopes and promises in the treatment of ovarian cancer focusing on targeted treatment-a narrative review. Front Pharmacol 2024; 15:1416555. [PMID: 38948462 PMCID: PMC11212463 DOI: 10.3389/fphar.2024.1416555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
Unfortunately, ovarian cancer is still diagnosed most often only in an advanced stage and is also the most lethal gynecological cancer. Another problem is the fact that treated patients have a high risk of disease recurrence. Moreover, ovarian cancer is very diverse in terms of molecular, histological features and mutations. Many patients may also develop platinum resistance, resulting in poor response to subsequent lines of treatment. To improve the prognosis of patients with ovarian cancer, it is expected to make better existing and implement new, promising treatment methods. Targeted therapies seem very promising. Currently, bevacizumab - a VEGF inhibitor and therapy with olaparib - a polyADP-ribose polymerase inhibitor are approved. Other methods worth considering in the future include: folate receptor α, immune checkpoints or other immunotherapy methods. To improve the treatment of ovarian cancer, it is also important to ameliorate the determination of molecular features to describe and understand which group of patients will benefit most from a given treatment method. This is important because a larger group of patients treated for ovarian cancer can have a greater chance of surviving longer without recurrence.
Collapse
Affiliation(s)
- Małgorzata Satora
- 1st Chair and Department of Oncological Gynecology and Gynecology, Students’ Scientific Association, Medical University of Lublin, Lublin, Poland
| | - Krzysztof Kułak
- 1st Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin, Poland
| | - Bartłomiej Zaremba
- 1st Chair and Department of Oncological Gynecology and Gynecology, Students’ Scientific Association, Medical University of Lublin, Lublin, Poland
| | - Arkadiusz Grunwald
- 1st Chair and Department of Oncological Gynecology and Gynecology, Students’ Scientific Association, Medical University of Lublin, Lublin, Poland
| | | | - Rafał Tarkowski
- 1st Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
3
|
Alhadeethi A, Adel Awwad S, Abed M, Amin AM, Aboelkhier MM, Yassin MNA, Morsi MH, Kashbour MO. Nintedanib in Combination With Chemotherapy in the Treatment of Non-small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e53812. [PMID: 38465177 PMCID: PMC10924634 DOI: 10.7759/cureus.53812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
Lung cancer remains a major global health challenge, contributing to substantial morbidity and mortality rates. Nintedanib, a tyrosine kinase inhibitor, has demonstrated potential as a treatment for lung cancer. We aim to evaluate nintedanib's efficacy in treating patients with non-small cell lung cancer (NSCLC), depending on the available evidence. Our search for relevant articles was conducted on PubMed, Cochrane Library, Scopus, and Web of Science for randomized controlled trials (RCTs) that involved adult patients with NSCLC up to August 15, 2023. These trials compared the combination of nintedanib and chemotherapy to either placebo plus chemotherapy or chemotherapy alone. Our main outcomes include progression-free survival (PFS) and overall survival (OS). We utilized the Review Manager Software V.5.4 (The Cochrane Collaboration) to analyze all relevant data. Three identified trials, which included 2270 patients, fulfilled the inclusion criteria. Our analysis showed significantly improved PFS (hazard ratio (HR) = 0.79; 95% confidence interval (CI) 0.71-0.88, P < 0.0001) in patients receiving nintedanib compared to placebo. However, OS was not statistically significant (HR = 0.96; 95% CI 0.88-1.05, P = 0.35). In conclusion, a combination of nintedanib and chemotherapy in treating patients with NSCLC was associated with improved PFS than chemotherapy alone but not with improved OS. Further clinical trials assessing nintedanib in the setting of NSCLC are necessary before any further recommendations can be made.
Collapse
Affiliation(s)
- Abdulhameed Alhadeethi
- Department of General Medicine, Medical Research Group of Egypt, Negida Academy LCC, Arlington, USA
- Department of General Medicine, Al-Salam Teaching Hospital, Mosul, IRQ
| | - Sara Adel Awwad
- College of Medicine, Jordan University of Science and Technology, Irbid, JOR
| | - Mohamed Abed
- Department of Internal Medicine, Faculty of Medicine, University of Tripoli, Tripoli, LBY
| | - Ahmed Mostafa Amin
- Department of Internal Medicine, Faculty of Medicine, Al-Azhar University, Cairo, EGY
| | - Menna M Aboelkhier
- Department of Internal Medicine, Faculty of Science, Cairo University, Cairo, EGY
| | | | - Maha H Morsi
- Department of Oncology, Medical Research Group of Egypt, Negida Academy LLC, Arlington, USA
- Department of Chemical Pathology, Misr University for Science and Technology, Giza, EGY
| | - Muataz Omar Kashbour
- Department of Diagnostic Radiology, National Cancer Institute, Misrata, LBY
- Department of Radiology, Medical Research Group of Libya, Negida Research Academy, Arlington, USA
| |
Collapse
|
4
|
Pang LL, Gan JD, Huang YH, Liao J, Zhuang WT, Ali WAS, Hong SD, Zhang L, Fang WF. Role of antiangiogenic agents in first-line treatment for advanced NSCLC in the era of immunotherapy. BMC Cancer 2023; 23:72. [PMID: 36670414 PMCID: PMC9862794 DOI: 10.1186/s12885-022-10446-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 12/12/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND & OBJECTIVE "Anti-angiogenetic drugs plus chemotherapy" (anti-angio-chemo) and "immune checkpoint inhibitors plus chemotherapy" (ICI-chemo) are superior to traditional chemotherapy in the first-line treatment of patients with advanced non-small-cell lung cancer (NSCLC). However, in the absence of a direct comparison of ICI-chemo with anti-angio-chemo, the superior one between them has not been decided, and the benefit of adding anti-angiogenetic agents to ICI-chemo remains controversial. This study aimed to investigate the role of antiangiogenic agents for advanced NSCLC in the era of immunotherapy. METHODS Eligible randomized controlled trials (RCTs) comparing chemotherapy versus therapeutic regimens involving ICIs or anti-angiogenetic drugs were included. Outcomes included progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and rate of grade 3-4 toxicity assessment. R-4.3.1 was utilized to perform the analysis. RESULTS A total of 54 studies with a sample size of 25,046 were finally enrolled. "Atezolizumab + Bevacizumab + Chemotherapy" significantly improved the ORR compared with "Atezolizumab + Chemotherapy" (Odds ratio (OR) = 2.73, 95% confidence interval (CI): 1.27-5.87). The trend also favored "Atezolizumab + Bevacizumab + Chemotherapy" in PFS and OS (hazard ratio (HR) = 0.71, 95% CI: 0.39-1.31; HR = 0.94, 95% CI: 0.77-1.16, respectively). In addition, "Pembrolizumab + Chemotherapy" and "Camrelizumab + Chemotherapy" significantly prolonged the PFS compared to "Bevacizumab + Chemotherapy" (HR = 0.65, 95% CI: 0.46-0.92; HR = 0.63, 95% CI: 0.41-0.97; respectively). Meanwhile, "Pembrolizumab + Chemotherapy" and "Sintilimab + Chemotherapy" yielded more OS benefits than "Bevacizumab + Chemotherapy" (HR = 0.69, 95% CI: 0.56-0.83; HR = 0.64, 95%CI: 0.46-0.91; respectively). Scheme between "Atezolizumab + Bevacizumab + Chemotherapy" and "Atezolizumab + Chemotherapy" made no significant difference (OR = 1.18, 95%CI: 0.56-2.42) concerning the rate of grade 3-4 toxicity. It seemed that ICI-chemo yielded more improvement in quality-adjusted life-year (QALY) than "Bevacizumab + Chemotherapy" in cost-effectiveness analysis. CONCLUSION Our results suggest that ICI-chemo is associated with potentially longer survival, better cost-effectiveness outcomes, and comparable safety profiles than anti-angio-chemo. Also, adding bevacizumab to ICI-chemo seemed to provide additional therapeutic benefits without adding treatment burden. Our findings would supplement the current standard of care and help the design of future clinical trials for the first-line treatment of patients with advanced NSCLC.
Collapse
Affiliation(s)
- Lan-Lan Pang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Jia-Di Gan
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Yi-Hua Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Jun Liao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Wei-Tao Zhuang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Wael-Abdullah-Sultan Ali
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Shao-Dong Hong
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Wen-Feng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| |
Collapse
|
5
|
Lung Cancer Clinical Trials with a Seamless Phase II/III Design: Systematic Review. J Clin Med 2022; 11:jcm11237176. [PMID: 36498749 PMCID: PMC9739886 DOI: 10.3390/jcm11237176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Current lung cancer clinical research focuses on biomarkers and personalized treatment strategies. Adaptive clinical trial designs have gained significant ground due to their increased flexibility, compared to the conventional model of drug development from phase I to phase IV trials. One such adaptive approach is the seamless phase II/III design, which has been used to reduce the total sample size and drug development time. In this context, an algorithmic systematic search was conducted in MEDLINE (PUBMED), SCOPUS, EMBASE and Cochrane Central Register of Controlled Trials until 31 June 2022 in order to identify lung cancer trials of systematic treatments that have employed the seamless phase II/III methodology and to describe their characteristics. The search strategy yielded a total of 1420 records that were screened through their title and abstract; 28 eligible trials were included in the systematic review. Based on the study endpoints, the most common subtype included phase II/III trials with inefficacy/futility analyses (61%; 17/28), followed by dose escalation phase II/III trials (18%; 5/28), one multi-arm multi stage trial and 5 trials with other design (18%). Most eligible trials were open-label (71%; 20/27), included patients with non-small cell lung cancer (82%; 23/28), evaluated targeted therapies and/or immunotherapies (82%; 23/28) and recruited patients with advanced disease (89.3%; 25/28). In conclusion, the seamless phase II/III design is a feasible and suitable approach in lung cancer research, with distinct design subcategories according to study endpoints.
Collapse
|
6
|
Zhang K, Wang W, Zhang T, Liang L. Efficacy and treatment-related adverse events of multi-targeted tyrosine kinase inhibitors in advanced non-small-cell lung cancer: a meta-analysis of randomized controlled trials. Int J Clin Pharm 2022; 44:1232-1246. [PMID: 35951217 DOI: 10.1007/s11096-022-01465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/27/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Multitargeted tyrosine kinase inhibitors (TKIs) are used to treat advanced non-small cell lung cancer (NSCLC). Their efficacy and safety have been studied in randomized controlled trials. AIM This meta-analysis aimed to summarize the most up-to-date evidence regarding the efficacy and adverse events of TKIs in NSCLC treatment. METHOD Randomized controlled trials were searched from PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials. The intervention arm was the TKI-containing group, and the control arm was the TKI-free group. Objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival, and adverse events were extracted and synthesized. The last search was performed in April 2022. Two researchers independently screened articles, extracted data, and evaluated the quality of the included studies. The Cochrane risk-of-bias tool was used to assess the quality of each study. Random or fixed-effect models were used in statistical methods. I2 statistics were used to assess heterogeneity. RESULTS Thirty-one studies (12,517 patients) were included. Compared to the control group, the TKI group had significantly higher ORR (relative risk RR 1.52, 95% confidence interval, CI [1.29, 1.80], P < 0.05), DCR (RR 1.34, 95%CI [1.19, 1.51], P < 0.05), and prolonged PFS (hazard ratio HR 0.67, 95%CI [0.59, 0.77], P < 0.05). The TKI group showed a higher rate of adverse events (RR 1.70, 95%CI [1.34, 2.16], P < 0.05) and grade 3-5 adverse events (RR 1.59, 95% CI [1.35, 1.88], P < 0.05). CONCLUSION TKIs could increase ORR and DCR and prolong PFS for advanced NSCLC. Adverse events should be closely monitored.
Collapse
Affiliation(s)
- Keqian Zhang
- Department of Oncology, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Wenwei Wang
- Department of Respiratory, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Ting Zhang
- Department of Respiratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Liang
- Department of Respiratory, The First Hospital Affiliated to Army Medical University, Chongqing, China.
| |
Collapse
|
7
|
Anti-Angiogenic Therapy in ALK Rearranged Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2022; 23:ijms23168863. [PMID: 36012123 PMCID: PMC9407780 DOI: 10.3390/ijms23168863] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
The management of advanced lung cancer has been transformed with the identification of targetable oncogenic driver alterations. This includes anaplastic lymphoma kinase (ALK) gene rearrangements. ALK tyrosine kinase inhibitors (TKI) are established first-line treatment options in advanced ALK rearranged non-small cell lung cancer (NSCLC), with several next-generation ALK TKIs (alectinib, brigatinib, ensartinib and lorlatinib) demonstrating survival benefit compared with the first-generation ALK TKI crizotinib. Still, despite high objective response rates and durable progression-free survival, drug resistance inevitably ensues, and treatment options beyond ALK TKI are predominantly limited to cytotoxic chemotherapy. Anti-angiogenic therapy targeting the vascular endothelial growth factor (VEGF) signaling pathway has shown efficacy in combination with platinum-doublet chemotherapy in advanced NSCLC without a driver alteration, and with EGFR TKI in advanced EGFR mutated NSCLC. The role for anti-angiogenic therapy in ALK rearranged NSCLC, however, remains to be elucidated. This review will discuss the pre-clinical rationale, clinical trial evidence to date, and future directions to evaluate anti-angiogenic therapy in ALK rearranged NSCLC.
Collapse
|
8
|
Hubbard JM, Yin J, Schenk EL, Qin R, Reid J, Strand C, Fiskum J, Menefee M, Lin G, Doyle LA, Ivy P, Erlichman C, Adjei A, Haluska P, Costello BA. Phase I study of cediranib, an oral VEGFR inhibitor, in combination with selumetinib, an oral MEK inhibitor, in patients with advanced solid malignancies. Invest New Drugs 2022; 40:115-123. [PMID: 34515877 PMCID: PMC8766914 DOI: 10.1007/s10637-021-01175-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Targeting the vascular endothelial growth factor (VEGF) pathway improves progression free survival in multiple advanced malignancies but durable responses are uncommon. Inhibition of the VEGF pathway at multiple levels of signal transduction may improve clinical outcomes. Preclinical data with cediranib, an inhibitor of all 3 VEGF receptors, in combination with selumetinib, an inhibitor of MEK 1/2, demonstrated improved tumor control experimentally. This phase I trial was designed to test the two agents in combination to evaluate the tolerability, safety and assess disease response. METHODS Patients with advanced solid malignancies were enrolled into this phase I trial. Cediranib and selumetinib were dosed using a toxicity-adaptive isotonic design for the dose escalation/de-escalation of each agent. Both cediranib and selumetinib were administered daily and continuously. Cycles were 28 days in length. RESULTS Eighteen patients were enrolled. At all dose levels, dose limiting toxicities (DLT) were observed, which limited dose escalation and further evaluation. The maximum tolerated dose of cediranib and selumetinib in combination could not be determined. The best response of stable disease was observed in eight patients. CONCLUSIONS Cediranib and selumetinib in combination on a continuous schedule was not tolerable, with patients experiencing cardiovascular and other DLTs. Intermittent schedules may be needed to establish a safe and tolerable combination of cediranib and selumetinib.
Collapse
Affiliation(s)
- Joleen M. Hubbard
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, United States
| | - Jun Yin
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, United States
| | - Erin L. Schenk
- Division of Medical Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Rui Qin
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, United States
| | - Joel Reid
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, United States
| | - Carrie Strand
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, United States
| | - Jack Fiskum
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, United States
| | | | - Grace Lin
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, United States
| | - L. Austin Doyle
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Percy Ivy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Charles Erlichman
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, United States
| | - Alex Adjei
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, United States
| | - Paul Haluska
- Bristol Myers Squibb, Lawrenceville, NJ 08648, United States
| | - Brian A. Costello
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
9
|
Guo X, Qian X, Jin Y, Kong X, Qi Z, Cai T, Zhang L, Wu C, Li W. Hypertension Induced by Combination Therapy of Cancer: A Systematic Review and Meta-Analysis of Global Clinical Trials. Front Pharmacol 2021; 12:712995. [PMID: 34552487 PMCID: PMC8451955 DOI: 10.3389/fphar.2021.712995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Nowadays, due to the limitation of single therapy, combination therapy for cancer treatments has become important strategy. With the advancement of research on cardiotoxicities induced by anti-cancer treatment, among which cancer treatment-induced hypertension is the most frequent case. However, due to the small sample size and the absence of comparison (single-arm study alone), these studies have limitations to produce a feasible conclusion. Therefore, it is necessary to carry out a meta-analysis focusing on hypertension caused by cancer combination therapy. Methods: We systematically searched PubMed, Embase, Cochrane Library, Web of Science, and CNKI, from database inception to November 31, 2020, with randomized controlled trials (RCTs) associated with hypertension induced by cancer combination drugs. The main endpoint of which was to assess the difference in the incidence of hypertension in cancer patients with monotherapy or combination therapy. We calculated the corresponding 95% confidence interval (95% CIs) according to the random effect model and evaluated the heterogeneity between different groups. Results: According to the preset specific inclusion and exclusion criteria, a total of 23 eligible RCTs have been included in the present meta-analysis, including 6,241 patients (Among them, 2872 patients were the control group and 3369 patients were the experimental group). The results showed that cancer patients with combination therapy led to a higher risk of hypertension (All-grade: RR 2.85, 95% CI 2.52∼3.22; 1∼2 grade: RR 2.43, 95% CI 2.10∼2.81; 3∼4 grade: RR 4.37, 95% CI 3.33∼5.72). Furthermore, compared with the control group who received or did not receive a placebo, there was a higher risk of grade 3-4 hypertension caused by cancer combination treatment. Conclusion: The present meta-analysis carries out a comprehensive analysis on the risk of patients suffering from hypertension in the process of multiple cancer combination therapies. Findings in our study support that the risk of hypertension may increase significantly in cancer patients with multiple cancer combination therapies. The outcomes of this meta-analysis may provide a reference value for clinical practice and may supply insights in reducing the incidence of hypertension caused by cancer combined treatment.
Collapse
Affiliation(s)
- Xiaodan Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xiaoyu Qian
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Ying Jin
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihong Qi
- Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Tie Cai
- State Key Laboratory of Coal Resources and Safe Mining, School of Chemical and Environmental Engineering, China University of Mining and Technology, Beijing, China
| | - Lin Zhang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Weihua Li
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
10
|
Implications of inhibition of Rev1 interaction with Y family DNA polymerases for cisplatin chemotherapy. Genes Dev 2021; 35:1256-1270. [PMID: 34385260 PMCID: PMC8415319 DOI: 10.1101/gad.348662.121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/15/2021] [Indexed: 11/25/2022]
Abstract
Chemotherapy with cisplatin becomes limiting due to toxicity and secondary malignancies. In principle, therapeutics could be improved by targeting translesion synthesis (TLS) polymerases (Pols) that promote replication through intrastrand cross-links, the major cisplatin-induced DNA adduct. However, to specifically target malignancies with minimal adverse effects on normal cells, a good understanding of TLS mechanisms in normal versus cancer cells is paramount. We show that in normal cells, TLS through cisplatin intrastrand cross-links is promoted by Polη- or Polι-dependent pathways, both of which require Rev1 as a scaffolding component. In contrast, cancer cells require Rev1-Polζ. Our findings that a recently identified Rev1 inhibitor, JH-RE-06, purported to specifically disrupt Rev1 interaction with Polζ to block TLS through cisplatin adducts in cancer cells, abrogates Rev1's ability to function with Y family Pols as well, implying that by inactivating Rev1-dependent TLS in normal cells, this inhibitor will exacerbate the toxicity and tumorigenicity of chemotherapeutics with cisplatin.
Collapse
|
11
|
The real-world efficacy and safety of anlotinib in advanced non-small cell lung cancer. J Cancer Res Clin Oncol 2021; 148:1721-1735. [PMID: 34357411 PMCID: PMC8343360 DOI: 10.1007/s00432-021-03752-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/01/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Anlotinib is an anti-angiogenetic multi-targeted tyrosine kinase inhibitor. This study aimed to evaluate the efficacy and safety of anlotinib in advanced non-small cell lung cancer (aNSCLC) in the real world. METHODS Patients with aNSCLC receiving anlotinib were enrolled in two cohorts (treatment naive and previously treated). The endpoints included progression-free survival (PFS), overall survival (OS) and anlotinib-related adverse events (ar-AEs). RESULTS 203 patients accrued in the study. In the treatment-naïve cohort (n = 80), the PFS was 7.4 (95% confidence interval [CI] 4.1-10.7) and OS was 10.8 (95% CI 5.8-15.8) months of monotherapy group (immature survival for combination group). In previously treated cohort (n = 123), the PFS was 8.0 months (95% CI 6.1-9.9) in the combination group and 4.3 months (95% CI 2.1-6.6) in the monotherapy group (hazard ratio [HR] 0.49; 95% CI 0.29-0.83; p = 0.007), respectively. The OS was 18.5 months (95% CI 10.5-26.6) in the combination group and 7.8 months (95% CI 7.1-8.4) in the monotherapy group (HR 0.38; 95% CI 0.22-0.66; p = 0.001), respectively. The ar-AEs of grade ≥ 3 in the monotherapy and the combination groups were hypertension (9.0 and 8.7%), fatigue (8.1 and 7.6%), hand-foot syndrome (8.1 and 6.5%), diarrhea (5.4 and 8.7%), proteinuria (5.4 and 5.4%), and mucositis oral (6.3 and 8.7%). CONCLUSION In aNSCLC, anlotinib monotherapy has a promising efficacy in the first-line setting. It may be an option for those who are ineligible for chemotherapy; anlotinib combination therapy in a ≥ second-line setting showed manageable toxicities and encouraging efficacy, indicating a good application prospect. TRIAL REGISTRATION This study was retrospectively registered with ISRCTN Registry (ID ISRCTN35543977) on January 26th, 2021 and Chinese Clinical Trial Register (ChiCTR2000032265) on April 4th, 2020.
Collapse
|
12
|
Paludan-Müller AS, Créquit P, Boutron I. Reporting of harms in oncological clinical study reports submitted to the European Medicines Agency compared to trial registries and publications-a methodological review. BMC Med 2021; 19:88. [PMID: 33827569 PMCID: PMC8028762 DOI: 10.1186/s12916-021-01955-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/01/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND An accurate and comprehensive assessment of harms is a fundamental part of an accurate weighing of benefits and harms of an intervention when making treatment decisions; however, harms are known to be underreported in journal publications. Therefore, we sought to compare the completeness of reporting of harm data, discrepancies in harm data reported, and the delay to access results of oncological clinical trials between three sources: clinical study reports (CSRs), clinical trial registries and journal publications. METHODS We used the EMA clinical data website to identify all trials submitted to the EMA between 2015 and 2018. We retrieved all CSRs and included all phase II, II/III or III randomised controlled trials (RCTs) assessing targeted therapy and immunotherapy for cancer. We then identified related records in clinical trial registries and journals. We extracted harms data for eight pre-specified variables and determined the completeness of reporting of harm data in each of the three sources. RESULTS We identified 42 RCTs evaluating 13 different drugs. Results were available on the EMA website in CSRs for 37 (88%) RCTs, ClinicalTrials.gov for 36 (86%), the European Clinical Trials Register (EUCTR) for 20 (48%) and in journal publications for 32 (76%). Harms reporting was more complete in CSRs than other sources. We identified marked discrepancies in harms data between sources, e.g. the number of patients discontinuing due to adverse events differed in CSRs and clinical trial registers for 88% of trials with data in both sources. For CSRs and publications, the corresponding number was 90%. The median (interquartile range) delay between the primary trial completion date and access to results was 4.34 (3.09-7.22) years for CSRs, 2.94 (1.16-4.52) years for ClinicalTrials.gov, 5.39 (4.18-7.33) years for EUCTR and 2.15 (0.64-5.04) years for publications. CONCLUSIONS Harms of recently approved oncological drugs were reported more frequently and in more detail in CSRs than in trial registries and journal publications. Systematic reviews seeking to address harms of oncological treatments should ideally use CSRs as the primary source of data; however, due to problems with access, this is currently not feasible.
Collapse
Affiliation(s)
- Asger S Paludan-Müller
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark , Department of Clinical Research, University of Southern Denmark, JB Winsløwsvej 9b, 3rd Floor, 5000, Odence C, Denmark. .,Open Patient data Exploratory Network (OPEN) , Odense University Hospital , Odense, Denmark.
| | - Perrine Créquit
- Direction de la recherche Clinique, Hôpital Foch, Suresnes, France.,Université de Paris, CRESS, INSERM, INRA, F-75004, Paris, France.,Cochrane France, Paris, France
| | - Isabelle Boutron
- Université de Paris, CRESS, INSERM, INRA, F-75004, Paris, France.,Cochrane France, Paris, France.,Centre d'Epidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
| |
Collapse
|
13
|
Ledermann JA, Embleton-Thirsk AC, Perren TJ, Jayson GC, Rustin GJS, Kaye SB, Hirte H, Oza A, Vaughan M, Friedlander M, González-Martín A, Deane E, Popoola B, Farrelly L, Swart AM, Kaplan RS, Parmar MKB. Cediranib in addition to chemotherapy for women with relapsed platinum-sensitive ovarian cancer (ICON6): overall survival results of a phase III randomised trial. ESMO Open 2021; 6:100043. [PMID: 33610123 PMCID: PMC7903311 DOI: 10.1016/j.esmoop.2020.100043] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Cediranib, an oral anti-angiogenic VEGFR 1-3 inhibitor, was studied at a daily dose of 20 mg in combination with platinum-based chemotherapy and as maintenance in a randomised trial in patients with first relapse of 'platinum-sensitive' ovarian cancer and has been shown to improve progression-free survival (PFS). PATIENTS AND METHODS ICON6 (NCT00532194) was an international three-arm, double-blind, placebo-controlled randomised trial. Between December 2007 and December 2011, 456 women were randomised, using stratification, to receive either chemotherapy with placebo throughout (arm A, reference); chemotherapy with concurrent cediranib, followed by maintenance placebo (arm B, concurrent); or chemotherapy with concurrent cediranib, followed by maintenance cediranib (arm C, maintenance). Due to an enforced redesign of the trial in September 2011, the primary endpoint became PFS between arms A and C which we have previously published, and the overall survival (OS) was defined as a secondary endpoint, which is reported here. RESULTS After a median follow-up of 25.6 months, strong evidence of an effect of concurrent plus maintenance cediranib on PFS was observed [hazard ratio (HR) 0.56, 95% confidence interval (CI) 0.44-0.72, P < 0.0001]. In this final update of the survival analysis, 90% of patients have died. There was a 7.4-month difference in median survival and an HR of 0.86 (95% CI: 0.67-1.11, P = 0.24) in favour of arm C. There was strong evidence of a departure from the assumption of non-proportionality using the Grambsch-Therneau test (P = 0.0031), making the HR difficult to interpret. Consequently, the restricted mean survival time (RMST) was used and the estimated difference over 6 years by the RMST was 4.8 months (95% CI: -0.09 to 9.74 months). CONCLUSIONS Although a statistically significant difference in time to progression was seen, the enforced curtailment in recruitment meant that the secondary analysis of OS was underpowered. The relative reduction in the risk of death of 14% risk of death was not conventionally statistically significant, but this improvement and the increase in the mean survival time in this analysis suggest that cediranib may have worthwhile activity in the treatment of recurrent ovarian cancer and that further research should be undertaken.
Collapse
Affiliation(s)
- J A Ledermann
- UCL Cancer Institute, Cancer Research UK & UCL Trials Centre, London, UK.
| | | | - T J Perren
- Leeds Institute of Medical Research at St James's, Leeds, UK
| | - G C Jayson
- Christie Hospital and University of Manchester, Manchester, UK
| | | | - S B Kaye
- Royal Marsden Hospital, London, UK
| | - H Hirte
- Juravinski Cancer Centre, Hamilton, Canada
| | - A Oza
- Princess Margaret Cancer Centre, Toronto, Canada
| | - M Vaughan
- Christchurch Hospital, Christchurch, New Zealand
| | - M Friedlander
- Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | | | - E Deane
- UCL Comprehensive Clinical Trials Unit, London, UK
| | - B Popoola
- Medical Research Council Clinical Trials Unit at UCL, London, UK
| | - L Farrelly
- UCL Cancer Institute, Cancer Research UK & UCL Trials Centre, London, UK
| | - A M Swart
- University of East Anglia, Norwich, UK
| | - R S Kaplan
- Medical Research Council Clinical Trials Unit at UCL, London, UK
| | - M K B Parmar
- Medical Research Council Clinical Trials Unit at UCL, London, UK
| |
Collapse
|
14
|
Spagnuolo A, Palazzolo G, Sementa C, Gridelli C. Vascular endothelial growth factor receptor tyrosine kinase inhibitors for the treatment of advanced non-small cell lung cancer. Expert Opin Pharmacother 2020; 21:491-506. [DOI: 10.1080/14656566.2020.1713092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- A Spagnuolo
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - G Palazzolo
- Division of Medical Oncology, “ULSS 15 Cittadella”, Cittadella, Padova, Italy
| | - C Sementa
- Division of Legal Medicine, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - C Gridelli
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| |
Collapse
|
15
|
Embleton-Thirsk A, Deane E, Townsend S, Farrelly L, Popoola B, Parker J, Rustin G, Sydes M, Parmar M, Ledermann J, Kaplan R. Impact of retrospective data verification to prepare the ICON6 trial for use in a marketing authorization application. Clin Trials 2019; 16:502-511. [PMID: 31347385 PMCID: PMC6801797 DOI: 10.1177/1740774519862528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background: The ICON6 trial (ISRCTN68510403) is a phase III academic-led, international,
randomized, three-arm, double-blind, placebo-controlled trial of the
addition of cediranib to chemotherapy in recurrent ovarian cancer. It
investigated the use of placebo during chemotherapy and maintenance (arm A),
cediranib alongside chemotherapy followed by placebo maintenance (arm B) and
cediranib throughout both periods (arm C). Results of the primary comparison
showed a meaningful gain in progression-free survival (time to progression
or death from any cause) when comparing arm A (placebo) with arm C
(cediranib). As a consequence of the positive results, AstraZeneca was
engaged with the Medical Research Council trials unit to discuss regulatory
submission using ICON6 as the single pivotal trial. Methods: A relatively limited level of on-site monitoring, single data entry and
investigator’s local evaluation of progression were used on trial. In order
to submit a license application, it was decided that (a) extensive
retrospective source data verification of medical records against case
report forms should be performed, (b) further quality control checks for
accuracy of data entry should be performed and (c) blinded independent
central review of images used to define progression should be undertaken. To
assess the value of these extra activities, we summarize the impact on both
efficacy and safety outcomes. Results: Data point changes were minimal; those key to the primary results had a 0.47%
error rate (36/7686), and supporting data points had a 0.18% error rate
(109/59,261). The impact of the source data verification and quality control
processes were analyzed jointly. The conclusion drawn for the primary
outcome measure of progression-free survival between arm A and arm C was
unchanged. The log-rank test p-value changed only at the sixth decimal
place, the hazard ratio does not change from 0.57 with the exception of a
marginal change in its upper bound (0.74–0.73) and the median
progression-free survival benefit from arm C remained at 2.4 months.
Separately, the blinded independent central review of progression scans was
performed as a sensitivity analysis. Estimates and p values varied slightly
but overall demonstrated a difference in arms, which is consistent with the
initial result. Some increases in toxicity were observed, though these were
generally minor, with the exception of hypertension. However, none of these
increases were systematically biased toward one arm. Conclusion: The conduct of this pragmatic, academic-sponsored trial was sufficient given
the robustness of the results, shown by the results remaining largely
unchanged following retrospective verification despite not being designed
for use in a marketing authorization. The burden of such comprehensive
retrospective effort required to ensure the results of ICON6 were acceptable
to regulators is difficult to justify.
Collapse
Affiliation(s)
| | - Elizabeth Deane
- Comprehensive Clinical Trials Unit, University College London, London, UK
| | | | | | | | - Judith Parker
- Edinburgh Clinical Trials Unit, The University of Edinburgh, Edinburgh, UK
| | | | - Matthew Sydes
- MRC Clinical Trials Unit, University College London, London, UK
| | - Mahesh Parmar
- MRC Clinical Trials Unit, University College London, London, UK
| | | | - Richard Kaplan
- MRC Clinical Trials Unit, University College London, London, UK
| |
Collapse
|
16
|
Lv WW, Zhang JJ, Zhou XL, Song Z, Wei CM. Safety of combining vascular endothelial growth factor receptor tyrosine-kinase inhibitors with chemotherapy in patients with advanced non-small-cell lung cancer: A PRISMA-compliant meta-analysis. Medicine (Baltimore) 2019; 98:e15806. [PMID: 31169681 PMCID: PMC6571213 DOI: 10.1097/md.0000000000015806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) have been developed for targeted therapies in non-small-cell lung cancer (NSCLC); moreover, some drug-related toxic reactions among cancer patients have been reported. A meta-analysis of randomized controlled trials (RCTs) to definite the incidence and the risk of grade ≥3 adverse events (AEs), serious and fatal AEs (SAEs and FAEs), with VEGFR-TKIs in advanced/metastatic NSCLC patients was performed. METHODS A comprehensive literature search was conducted for the clinical trials published up to December 2017. Qualified studies allotted patients with advanced/metastatic NSCLC to receive either chemotherapy alone or in combination with VEGFR-TKIs. Data were extracted by 2 authors. RESULTS Eighteen RCTs of VEGFR-TKIs plus chemotherapy, involving 8461 advanced NSCLC patients were included. The proportion of patients with grade ≥3 AEs was increased with the addition of VEGFR-TKIs (relative risk, 1.35; 95% confidence interval [CI] 1.19-1.52; incidence, 68.1% vs 50.1%; P < .001). The most common grade ≥3 AEs was neutropenia (24.9% vs 15.4%, P < .001). Addition of VEGFR-TKIs was also related to the increased risk of SAEs (relative risk, 1.34; 95% CI 1.14-1.56; incidence, 37.8% vs 27.9%; P < .001) and FAEs (relative risk, 2.16, 95% CI 1.47-3.19; incidence, 3.4% vs 1.8%). Subgroup analysis suggested there was no difference in the rates of SAEs and FAEs in the second-line settings. No evidence of bias was found between the literatures. The study was registered with PROSPERO (CRD42018099654). CONCLUSIONS In comparison with chemotherapy alone, the addition of VEGFR-TKIs in advanced NSCLC patients was related to the increased risk of grades ≥3 AEs, SAEs, and FAEs, especially in the first-line settings. Physicians should be aware of some specific grade ≥3 adverse effect, especially haematologic adverse events, and it is also necessary to monitor cancer patients receiving VEGFR-TKIs.
Collapse
|
17
|
Ding F, Liu B, Wang Y. Risk of hand-foot skin reaction associated with vascular endothelial growth factor-tyrosine kinase inhibitors: A meta-analysis of 57 randomized controlled trials involving 24,956 patients. J Am Acad Dermatol 2019; 83:788-796. [PMID: 30991119 DOI: 10.1016/j.jaad.2019.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multiple randomized controlled trials have assessed hand-foot skin reaction (HFSR) caused by vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs). OBJECTIVE We performed a meta-analysis to determine the incidence and the relative risk (RR) of HFSR associated with these agents. METHODS Databases were searched for relevant studies. Statistical analyses were conducted to calculate the summary incidences, RR, and 95% confidence intervals (CIs) by using random-effects or fixed-effects models according to the heterogeneity of the included studies. RESULTS A total of 24,956 patients from 57 studies were included. The overall incidence of all-grade and high-grade HFSR associated with VEGFR-TKIs was 35.0% (95% CI, 28.6%-41.6%) and 9.7% (95% CI, 7.3%-12.3%), respectively. The use of VEGFR-TKIs significantly increased the risk of developing all-grade (RR, 5.09; 95% CI, 3.52-7.35; P < .001) and high-grade (RR, 9.42; 95% CI, 5.59-15.90; P < .001) HFSR. Subgroup analyses revealed that the risk of HFSR was significantly increased according to tumor type, VEGFR-TKI, trial phase, treatment regimen, and control therapy. No evidence of publication bias was observed. LIMITATION High heterogeneity in most studies. CONCLUSION High risk of HFSR is prone to develop in cancer patients receiving VEGFR-TKIs.
Collapse
Affiliation(s)
- Fengxia Ding
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
| | - Bo Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yaping Wang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| |
Collapse
|
18
|
Qin S, Li A, Yi M, Yu S, Zhang M, Wu K. Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. J Hematol Oncol 2019; 12:27. [PMID: 30866992 PMCID: PMC6417086 DOI: 10.1186/s13045-019-0718-5] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/05/2019] [Indexed: 02/08/2023] Open
Abstract
Angiogenesis has always been the topic of major scientific interest in the field of malignant tumors. Nowadays, targeting angiogenesis has achieved success in various carcinomas by several mechanisms, including the use of anti-angiogenic small molecule receptor tyrosine kinase inhibitors (TKIs). The development of TKIs targeting pro-angiogenic receptors, mainly vascular endothelial growth factor receptor (VEGFR) family, have significantly improved the outcome of certain types of cancers, like renal cell carcinoma, hepatocellular carcinoma, and colorectal carcinoma. However, the general response rate is not very satisfactory. The particular toxicity profile and resistance to anti-angiogenic targeted agents are unavoidable, and no specific marker is available to screen responsive patients to TKIs for precision therapy. To date, about 11 anti-angiogenic TKIs with different binding capacities to angiogenic receptor tyrosine kinase have been approved for the treatment of patients with advanced cancers. This review presents all approved anti-angiogenic small molecule receptor TKIs so far with an emphasis on their indications and clinical efficacy. We also discuss the combination between TKIs and immune checkpoint blockade inhibitors based on the most recent exciting outcome in immunotherapy.
Collapse
Affiliation(s)
- Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Anping Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Mingsheng Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
19
|
Liu L, Zhang Y, Wei J, Chen Z, Yu J. VEGFR-TKIs combined with chemotherapy for advanced non-small cell lung cancer: A systematic review. J Cancer 2019; 10:799-809. [PMID: 30854085 PMCID: PMC6400799 DOI: 10.7150/jca.29643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 01/09/2019] [Indexed: 12/18/2022] Open
Abstract
Introduction: To estimate the efficacy and safety of vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) in combination with chemotherapy for patients with advanced non-small cell lung cancer (NSCLC). Methods: We searched PubMed, PMC database, EMBASE, EBSCO-Medline, Cochrane Central Register of Controlled Trials (CENTRAL), American Society of Clinical Oncology (ASCO), International Association for the Study of Lung Cancer (IASLC) and the European Society of Medical Oncology (ESMO), http://www.clinicaltrials.gov/, CNKI, and Wanfang databases to identify primary research reporting the survival outcomes and safety of VEGFR-TKIs in patients with advanced NSCLC. A meta-analysis was conducted to generate combined hazard ratios (HRs) with 95% confidence intervals (CI) for overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and risk ratios (RRs) with 95% CI for adverse events (AEs). Results: A total of 20 RCTs (8,366 participants) were included. The VEGFR-TKIs resulted in improved PFS (HR 0.82, 95% CI 0.78-0.87), ORR (HR 1.72, 95% CI 1.34-2.22), and DCR (1.45, 1.26-1.67) in patients with advanced NSCLC, but had no impact on OS (HR 0.94, 95% CI 0.89-1.00). The incidence of some high grade (≥ 3) AEs increased, such as hemorrhage, hypertension and neutropenia. Conclusions: Our study demonstrated that regimens with VEGFR-TKIs combined with chemotherapy improved PFS, ORR and DCR in patients with advanced NSCLC, but had no impact on OS. VEGFR-TKIs induced more frequent and serious AEs compared with control therapies.
Collapse
Affiliation(s)
- Lian Liu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Yue Zhang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Jia Wei
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Zhaoxin Chen
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| |
Collapse
|
20
|
Wang F, Molina J, Satele D, Yin J, Lim VS, Adjei AA. A phase I study of the vascular endothelial growth factor inhibitor Vatalanib in combination with Pemetrexed disodium in patients with advanced solid tumors. Invest New Drugs 2018; 37:658-665. [PMID: 30382439 DOI: 10.1007/s10637-018-0690-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/25/2018] [Indexed: 02/07/2023]
Abstract
Introduction Vatalanib is an oral receptor tyrosine kinase inhibitor that blocks all known VEGF, PDGF, and c-Kit receptors. This phase I study evaluated the safety, tolerability, and biologic activity of the combination of vatalanib with pemetrexed disodium in patients with advanced solid tumors. Methods Patients were administered escalating twice daily doses of vatalanib in combination with pemetrexed disodium in 21-day cycles. A dose expansion cohort was enrolled to further define the maximum tolerated dose (MTD) and further evaluate efficacy. Results A total of 29 patients were enrolled in the study (dose escalation, 9; dose expansion, 20). Dose-limiting toxicities included grade 4 thrombocytopenia (6.9%) and febrile neutropenia, anorexia, constipation, and dehydration. Other common adverse events were fatigue (75%), nausea (66%), vomiting (48%), oral mucositis (31%) and diarrhea (28%). The majority of these toxicities were Grade 1-2. The MTD was reached at vatalanib 250 mg twice daily continuously combined with pemetrexed disodium 500 mg/m2 day 1. Overall, 2 patients (6.9%) had partial responses, 8 (27.6%) had stable disease for at least 4 cycles, 5 had progressive disease (17.2%) and 5 went off study before disease assessment. Conclusion The combination of vatalanib with pemetrexed disodium was feasible, but not well tolerated. The modest efficacy results are consistent with other results obtained from combinations of chemotherapy and a large number of VEGF tyrosine kinase inhibitors. This combination should not be developed further unless predictive biomarkers can be identified.
Collapse
Affiliation(s)
- Fen Wang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Julian Molina
- Department of Oncology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, United States
| | - Daniel Satele
- Department of Health Sciences Research, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, United States
| | - Jun Yin
- Department of Health Sciences Research, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, United States
| | - Vun-Sin Lim
- Department of Oncology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, United States
| | - Alex A Adjei
- Department of Oncology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, United States.
| |
Collapse
|
21
|
Li J, Gu J. Cardiovascular Toxicities with Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitors in Cancer Patients: A Meta-Analysis of 77 Randomized Controlled Trials. Clin Drug Investig 2018; 38:1109-1123. [DOI: 10.1007/s40261-018-0709-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
22
|
Arasada RR, Shilo K, Yamada T, Zhang J, Yano S, Ghanem R, Wang W, Takeuchi S, Fukuda K, Katakami N, Tomii K, Ogushi F, Nishioka Y, Talabere T, Misra S, Duan W, Fadda P, Rahman MA, Nana-Sinkam P, Evans J, Amann J, Tchekneva EE, Dikov MM, Carbone DP. Notch3-dependent β-catenin signaling mediates EGFR TKI drug persistence in EGFR mutant NSCLC. Nat Commun 2018; 9:3198. [PMID: 30097569 PMCID: PMC6090531 DOI: 10.1038/s41467-018-05626-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/02/2018] [Indexed: 12/29/2022] Open
Abstract
EGFR tyrosine kinase inhibitors cause dramatic responses in EGFR-mutant lung cancer, but resistance universally develops. The involvement of β-catenin in EGFR TKI resistance has been previously reported, however, the precise mechanism by which β-catenin activation contributes to EGFR TKI resistance is not clear. Here, we show that EGFR inhibition results in the activation of β-catenin signaling in a Notch3-dependent manner, which facilitates the survival of a subset of cells that we call "adaptive persisters". We previously reported that EGFR-TKI treatment rapidly activates Notch3, and here we describe the physical association of Notch3 with β-catenin, leading to increased stability and activation of β-catenin. We demonstrate that the combination of EGFR-TKI and a β-catenin inhibitor inhibits the development of these adaptive persisters, decreases tumor burden, improves recurrence free survival, and overall survival in xenograft models. These results supports combined EGFR-TKI and β-catenin inhibition in patients with EGFR mutant lung cancer.
Collapse
Affiliation(s)
- Rajeswara Rao Arasada
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA.
| | - Konstantin Shilo
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Tadaaki Yamada
- Division of Medical Oncology, Kanazawa University Cancer Research Institute, Kanazawa, 920-0934, Japan
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Seiji Yano
- Division of Medical Oncology, Kanazawa University Cancer Research Institute, Kanazawa, 920-0934, Japan
| | - Rashelle Ghanem
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Walter Wang
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Shinji Takeuchi
- Division of Medical Oncology, Kanazawa University Cancer Research Institute, Kanazawa, 920-0934, Japan
| | - Koji Fukuda
- Division of Medical Oncology, Kanazawa University Cancer Research Institute, Kanazawa, 920-0934, Japan
| | - Nobuyuki Katakami
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation, Kobe, 650-0047, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, 650-0047, Japan
| | - Fumitaka Ogushi
- Division of Pulmonary Medicine, National Hospital Organization National Kochi Hospital, Kochi, 780-8077, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, 770-8503, Japan
| | - Tiffany Talabere
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Shrilekha Misra
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Wenrui Duan
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Paolo Fadda
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Mohammad A Rahman
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and the Center for Critical Care Medicine, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Patrick Nana-Sinkam
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and the Center for Critical Care Medicine, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Jason Evans
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Joseph Amann
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Elena E Tchekneva
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Mikhail M Dikov
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - David P Carbone
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
23
|
Abstract
Unlike for adenocarcinomas of the lung, no molecular targeted therapies have yet been developed for squamous cell lung cancers, because targetable oncogenic aberrations are scarce in this tumor type. Recent discoveries have established that the fibroblast growth factor (FGF) signaling pathway plays a fundamental role in cancer development by supporting tumor angiogenesis and cancer cell proliferation via different mechanisms. Through comprehensive genomic studies, aberrations in the FGF pathway have been identified in various tumor types, including squamous cell lung cancer, making FGF receptor (FGFR) a potentially druggable target in this malignancy. Several multi-targeted tyrosine kinase inhibitors include FGFR in their target spectrum and a number of these compounds have been approved for clinical use in different cancers. Novel agents selectively targeting FGFRs have been developed and are currently under investigation in clinical trials, showing promising results. This article reviews FGFR aberrations and the clinical data involving selective and multikinase FGFR inhibitors in squamous cell lung cancer.
Collapse
|
24
|
Abstract
Angiogenesis is frequent in non-small-cell lung cancer (nsclc) and is associated with more aggressive disease. Many clinical trials have evaluated the addition of antiangiogenic therapy to standard therapies for patients with nsclc. Bevacizumab, a monoclonal antibody directed against serum vascular endothelial growth factor, in combination with carboplatin-paclitaxel chemotherapy, has been shown to improve survival for patients with nsclc. However, bevacizumab-based therapy is not suitable for many nsclc patients, including those with squamous histology, poor performance status, brain metastases, and the presence of bleeding or thrombotic disorders. Similar efficacy has also been seen with carboplatin-pemetrexed followed by maintenance pemetrexed chemotherapy. In the second-line setting, the addition of ramucirumab to docetaxel-or the addition of bevacizumab to paclitaxel-has resulted in a modest improvement in efficacy, although the clinical importance of those findings is questionable. Many trials in nsclc have also evaluated oral antiangiogenic compounds, both in the first line in combination with chemotherapy and upon disease progression either as combination or single-agent therapy. No clear improvements in overall survival have been observed, although a subgroup analysis of a trial evaluating the addition of nintedanib to docetaxel showed improved survival that was limited to patients with adenocarcinoma. Those findings require validation, however. All of the oral antiangiogenic agents result in added toxicities. Some agents have resulted in an increased risk of death, limiting their development. Available evidence supports a limited number of antiangiogenic therapies for patients with nsclc, but no biomarkers to help in patient selection are currently available, and additional translational research is needed to identify predictive biomarkers for antiangiogenic therapy.
Collapse
Affiliation(s)
- A. Alshangiti
- Department of Oncology, McMaster University, Juravinski Cancer Centre, Hamilton, ON
| | - G. Chandhoke
- Department of Oncology, McMaster University, Juravinski Cancer Centre, Hamilton, ON
| | - P.M. Ellis
- Department of Oncology, McMaster University, Juravinski Cancer Centre, Hamilton, ON
| |
Collapse
|
25
|
Varricchi G, Ameri P, Cadeddu C, Ghigo A, Madonna R, Marone G, Mercurio V, Monte I, Novo G, Parrella P, Pirozzi F, Pecoraro A, Spallarossa P, Zito C, Mercuro G, Pagliaro P, Tocchetti CG. Antineoplastic Drug-Induced Cardiotoxicity: A Redox Perspective. Front Physiol 2018; 9:167. [PMID: 29563880 PMCID: PMC5846016 DOI: 10.3389/fphys.2018.00167] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Antineoplastic drugs can be associated with several side effects, including cardiovascular toxicity (CTX). Biochemical studies have identified multiple mechanisms of CTX. Chemoterapeutic agents can alter redox homeostasis by increasing the production of reactive oxygen species (ROS) and reactive nitrogen species RNS. Cellular sources of ROS/RNS are cardiomyocytes, endothelial cells, stromal and inflammatory cells in the heart. Mitochondria, peroxisomes and other subcellular components are central hubs that control redox homeostasis. Mitochondria are central targets for antineoplastic drug-induced CTX. Understanding the mechanisms of CTX is fundamental for effective cardioprotection, without compromising the efficacy of anticancer treatments. Type 1 CTX is associated with irreversible cardiac cell injury and is typically caused by anthracyclines and conventional chemotherapeutic agents. Type 2 CTX, associated with reversible myocardial dysfunction, is generally caused by biologicals and targeted drugs. Although oxidative/nitrosative reactions play a central role in CTX caused by different antineoplastic drugs, additional mechanisms involving directly and indirectly cardiomyocytes and inflammatory cells play a role in cardiovascular toxicities. Identification of cardiologic risk factors and an integrated approach using molecular, imaging, and clinical data may allow the selection of patients at risk of developing chemotherapy-related CTX. Although the last decade has witnessed intense research related to the molecular and biochemical mechanisms of CTX of antineoplastic drugs, experimental and clinical studies are urgently needed to balance safety and efficacy of novel cancer therapies.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- Institute of Cardiology, Center of Excellence on Aging, Università degli Studi “G. d'Annunzio” Chieti – Pescara, Chieti, Italy
- Department of Internal Medicine, Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center, Houston, TX, United States
| | - Giancarlo Marone
- Section of Hygiene, Department of Public Health, University of Naples Federico II, Naples, Italy
- Monaldi Hospital Pharmacy, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Parrella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Concetta Zito
- Division of Clinical and Experimental Cardiology, Department of Medicine and Pharmacology, Policlinico “G. Martino” University of Messina, Messina, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
26
|
Liu B, Ding F, Liu Y, Xiong G, Lin T, He D, Zhang Y, Zhang D, Wei G. Incidence and risk of hypertension associated with vascular endothelial growth factor receptor tyrosine kinase inhibitors in cancer patients: a comprehensive network meta-analysis of 72 randomized controlled trials involving 30013 patients. Oncotarget 2018; 7:67661-67673. [PMID: 27602778 PMCID: PMC5341903 DOI: 10.18632/oncotarget.11813] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/23/2016] [Indexed: 01/12/2023] Open
Abstract
Background Tyrosine kinase inhibitors (TKIs) have been developed during the last decade that target the vascular endothelial growth factor receptor (VEGFR) are currently being evaluated as treatments for malignant tumors. The increased application of VEGFR-TKIs means that the probability of hypertension is a serious concern. However, the reported incidence varies markedly between clinical trials. Here, we undertook an up-to-date, comprehensive meta-analysis on clinical works to build the incidence of hypertension along with VEGFR-TKIs. The goal was to understand better of the overall venture of cancer patients’ hypertension treated with these drugs. Methods Databases (EMBASE, PubMed, and Cochrane library) and the abstracts of the American Society of Clinical Oncology annual meeting and European Society of Medical Oncology were searched to identify related studies. 95% confidence intervals (CIs), summary incidences, and relative risk (RR) were calculated utilizing either fixed-effects models on the basis of the heterogeneity of the included studies or random-effects. Results Seventy-two randomized controlled trials (including 30013 patients) were involved. The total incidence of high-grade and all-grade hypertensive events along with VEGFR-TKIs was 23.0% (95% CI, 20.1–26.0%) and 4.4% (95% CI, 3.7–5.0%), respectively. The use of VEGFR-TKIs remarkably enhanced the venture of developing high-grade (RR, 4.60; 95% CI, 3.92–5.40; P < 0.001) and all-grade (RR, 3.85; 95% CI, 3.37–4.40; P < 0.001) hypertensive events. Subgroup analyses revealed that the risk of a hypertensive event varied significantly in accordance with tumor type, VEGFR-TKI, trial phase, VEGFR-TKIs-based regimen, control therapy, and chemotherapy regimen. Conclusions Patients with cancer that receive VEGFR-TKIs are at a remarkable venture of developing hypertension. Therefore, suitable treatment and monitoring should be introduced to avoid cardiovascular complications.
Collapse
Affiliation(s)
- Bo Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Fengxia Ding
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Yang Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Geng Xiong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Deying Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
Denisenko TV, Budkevich IN, Zhivotovsky B. Cell death-based treatment of lung adenocarcinoma. Cell Death Dis 2018; 9:117. [PMID: 29371589 PMCID: PMC5833343 DOI: 10.1038/s41419-017-0063-y] [Citation(s) in RCA: 446] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/18/2017] [Accepted: 09/13/2017] [Indexed: 12/13/2022]
Abstract
The most common type of lung cancer is adenocarcinoma (ADC), comprising around 40% of all lung cancer cases. In spite of achievements in understanding the pathogenesis of this disease and the development of new approaches in its treatment, unfortunately, lung ADC is still one of the most aggressive and rapidly fatal tumor types with overall survival less than 5 years. Lung ADC is often diagnosed at advanced stages involving disseminated metastatic tumors. This is particularly important for the successful development of new approaches in cancer therapy. The high resistance of lung ADC to conventional radiotherapies and chemotherapies represents a major challenge for treatment effectiveness. Here we discuss recent advances in understanding the molecular pathways driving tumor progression and related targeted therapies in lung ADCs. In addition, the cell death mechanisms induced by different treatment strategies and their contribution to therapy resistance are analyzed. The focus is on approaches to overcoming drug resistance in order to improve future treatment decisions.
Collapse
Affiliation(s)
- Tatiana V Denisenko
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Inna N Budkevich
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Boris Zhivotovsky
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia. .,Institute of Environmental Medicine, Division of Toxicology, Karolinska Institutet, Box 210, Stockholm, SE-171 77, Sweden.
| |
Collapse
|
28
|
Qu J, Zhang Y, Chen X, Yang H, Zhou C, Yang N. Newly developed anti-angiogenic therapy in non-small cell lung cancer. Oncotarget 2017. [PMID: 29515799 PMCID: PMC5839380 DOI: 10.18632/oncotarget.23755] [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] [Indexed: 01/17/2023] Open
Abstract
Angiogenesis and its role in the growth and development of non-small cell lung cancer (NSCLC) metastases has become an increasing clinical problem. Vascular endothelial growth factor (VEGF) plays a key role in advanced NSCLC. To some extent, anti-angiogenic therapies acquired some efficacy in combination with chemotherapy, target therapy and immunotherapy. However, the reliable clinical benefit obtained with these drugs is still questionable and often quantitatively limited. In this review, the authors highlight the data obtained from first-line, second-line, epidermal growth factor receptor tyrosine kinase inhibitor(EGFR-TKI) target therapy and immunotherapy in NSCLC patients who are treated with anti-angiogenic molecules in advanced NSCLC. The purpose of this study is to help us truly understand how to best use angiogenesis therapy in advanced NSCLC.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Lung Cancer and Gastrointestinal Oncology Medicine, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
| | - Yongchang Zhang
- Department of Lung Cancer and Gastrointestinal Oncology Medicine, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
| | - Xue Chen
- Department of Lung Cancer and Gastrointestinal Oncology Medicine, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
| | - Haiyan Yang
- Department of Lung Cancer and Gastrointestinal Oncology Medicine, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
| | - Chunhua Zhou
- Department of Lung Cancer and Gastrointestinal Oncology Medicine, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
| | - Nong Yang
- Department of Lung Cancer and Gastrointestinal Oncology Medicine, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
| |
Collapse
|
29
|
Does local delivery of bisphosphonates influence the osseointegration of titanium implants? A systematic review. Int J Oral Maxillofac Surg 2017; 46:1429-1436. [DOI: 10.1016/j.ijom.2017.04.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/21/2017] [Accepted: 04/18/2017] [Indexed: 11/22/2022]
|
30
|
Gu L, Deng ZJ, Roy S, Hammond PT. A Combination RNAi-Chemotherapy Layer-by-Layer Nanoparticle for Systemic Targeting of KRAS/P53 with Cisplatin to Treat Non-Small Cell Lung Cancer. Clin Cancer Res 2017; 23:7312-7323. [PMID: 28912139 DOI: 10.1158/1078-0432.ccr-16-2186] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 06/22/2017] [Accepted: 09/07/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Mutation of the Kirsten ras sarcoma viral oncogene homolog (KRAS) and loss of p53 function are commonly seen in patients with non-small cell lung cancer (NSCLC). Combining therapeutics targeting these tumor-defensive pathways with cisplatin in a single-nanoparticle platform are rarely developed in clinic.Experimental Design: Cisplatin was encapsulated in liposomes, which multiple polyelectrolyte layers, including siKRAS and miR-34a were built on to generate multifunctional layer-by-layer nanoparticle. Structure, size, and surface charge were characterized, in addition to in vitro toxicity studies. In vivo tumor targeting and therapy was investigated in an orthotopic lung cancer model by microCT, fluorescence imaging, and immunohistochemistry.Results: The singular nanoscale formulation, incorporating oncogene siKRAS, tumor-suppressor stimulating miR-34a, and cisplatin, has shown enhanced toxicity against lung cancer cell line, KP cell. In vivo, systemic delivery of the nanoparticles indicated a preferential uptake in lung of the tumor-bearing mice. Efficacy studies indicated prolonged survival of mice from the combination treatment.Conclusions: The combination RNA-chemotherapy in an LbL formulation provides an enhanced treatment efficacy against NSCLC, indicating promising potential in clinic. Clin Cancer Res; 23(23); 7312-23. ©2017 AACR.
Collapse
Affiliation(s)
- Li Gu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Zhou J Deng
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sweta Roy
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts. .,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
31
|
Strategies targeting angiogenesis in advanced non-small cell lung cancer. Oncotarget 2017; 8:53854-53872. [PMID: 28881856 PMCID: PMC5581155 DOI: 10.18632/oncotarget.17957] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/27/2017] [Indexed: 12/26/2022] Open
Abstract
Tumor angiogenesis is a frequent event in the development and progression of non-small cell lung cancer (NSCLC) and has been identified as a promising therapeutic target. The vascular endothelial growth factor (VEGF) family and other angiogenic factors, including fibroblast growth factor and platelet-derived growth factor, promote the growth of newly formed vessels from preexisting vessels and change the tumor microenvironment. To date, two antiangiogenic monoclonal antibodies, bevacizumab and ramucirumab, which target VEGF-A and its receptor VEGF receptor-2, respectively, have been approved for the treatment of locally advanced or metastatic NSCLC when added to first-line standard chemotherapy. Numerous oral multitargeting angiogenic small molecule tyrosine kinase inhibitors (TKIs) have been widely evaluated in advanced NSCLC, but only nintedanib in combination with platinum-based doublet chemotherapy has demonstrated a survival benefit in the second-line setting. Additionally, small-molecule TKIs remain the standard of care for patients with mutated EGFR, ALK or ROS1. Moreover, immune checkpoint inhibitors that target the programmed cell death protein 1 (PD-1) and programmed cell death protein ligand 1 (PD-L1) are changing the current strategy in the treatment of advanced NSCLC without driver gene mutations. The potential synergistic activity of antiangiogenic agents and TKIs or immunotherapy is an interesting topic of research. This review will summarize the novel antiangiogenic agents, antiangiogenic monotherapy, as well as potential combination therapeutic strategies for the clinical management of advanced NSCLC.
Collapse
|
32
|
Rijavec E, Genova C, Barletta G, Biello F, Rossi G, Tagliamento M, Dal Bello MG, Coco S, Vanni I, Boccardo S, Alama A, Grossi F. Investigational drugs targeting fibroblast growth factor receptor in the treatment of non-small cell lung cancer. Expert Opin Investig Drugs 2017; 26:551-561. [PMID: 28388262 DOI: 10.1080/13543784.2017.1316714] [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: 10/19/2022]
Abstract
INTRODUCTION Fibroblast growth factor receptor (FGFR) due to its central role in regulating cell survival, is a promising target for cancer therapeutics. Dysregulation of the FGFR pathway has been observed in several malignancies, including non-small cell lung cancer (NSCLC) particularly in patients with squamous histology. Areas covered: The aim of this article is to review the most relevant findings of clinical trials investigating drugs targeting FGFR pathway: such as FGFR tyrosine kinase inhibitors (TKIs), FGFR monoclonal antibodies and FGF ligand traps in NSCLC patients. Expert opinion: At present, clinical activity of drugs targeting FGFR in NSCLC is disappointing. Further studies are needed in order to better identify patients who might benefit from these drugs and to clarify the mechanisms of resistance to these compounds.
Collapse
Affiliation(s)
- Erika Rijavec
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Carlo Genova
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy.,b Dipartimento di Medicina Interna e Specialità Mediche (DIMI) , Università di Genova , Genova , Italy
| | - Giulia Barletta
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Federica Biello
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Giovanni Rossi
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Marco Tagliamento
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Maria Giovanna Dal Bello
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Simona Coco
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Irene Vanni
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Simona Boccardo
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Angela Alama
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| | - Francesco Grossi
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST- Istituto Nazionale per la Ricerca sul Cancro , Genova , Italy
| |
Collapse
|
33
|
Kurzrock R, Stewart DJ. Exploring the Benefit/Risk Associated with Antiangiogenic Agents for the Treatment of Non-Small Cell Lung Cancer Patients. Clin Cancer Res 2017; 23:1137-1148. [PMID: 27940520 DOI: 10.1158/1078-0432.ccr-16-1968] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 11/16/2022]
Abstract
Following the approval of bevacizumab, an antibody targeting VEGF-A, for advanced non-squamous non-small cell lung cancer (NSCLC) in 2006, intensive efforts were put into the clinical development of antiangiogenic agents for NSCLC. Currently, the other antiangiogenic agents approved for NSCLC are ramucirumab, a VEGF receptor-2 (VEGFR-2)-targeting antibody indicated for both squamous and non-squamous NSCLC in the United States, and nintedanib, an anti-VEGFR-1/2/3, platelet-derived growth factor receptor-α/β, fibroblast growth factor receptor-1/2/3 angiokinase inhibitor indicated for adenocarcinoma of the lung in the European Union. Many other antiangiogenic agents are being evaluated in phase III trials for NSCLC, including aflibercept, sunitinib, sorafenib, cediranib, and vandetanib. Although many of the same signaling pathways are targeted by these novel agents, mixed efficacy results have been observed in these trials. Moreover, safety issues have raised concerns about using antiangiogenic agents in this patient population, and fatal bleeding events have been reported. Importantly, although no biomarker has yet been validated for antiangiogenic agents in NSCLC, biomarkers that show potential include circulating levels of short VEGF-A isoforms, expression of neuropilin-1 and VEGFR-1 in tumors and plasma, genetic variants in VEGF-A and VEGFR, and tumor protein p53 mutations (with the latter having been shown to correlate with increased levels of VEGF-A transcripts). This review provides an overview of the clinical benefit and risk associated with the use of antiangiogenic agents for NSCLC, and summarizes the research to date on the identification of predictive biomarkers for antiangiogenic therapies. Clin Cancer Res; 23(5); 1137-48. ©2016 AACR.
Collapse
MESH Headings
- Angiogenesis Inhibitors/adverse effects
- Angiogenesis Inhibitors/therapeutic use
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Bevacizumab/therapeutic use
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/epidemiology
- Carcinoma, Non-Small-Cell Lung/pathology
- Humans
- Indoles/adverse effects
- Indoles/therapeutic use
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/epidemiology
- Neovascularization, Pathologic/pathology
- Pyrroles/adverse effects
- Pyrroles/therapeutic use
- Receptors, Vascular Endothelial Growth Factor/therapeutic use
- Recombinant Fusion Proteins/adverse effects
- Recombinant Fusion Proteins/therapeutic use
- Risk Assessment
- Sunitinib
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Ramucirumab
Collapse
Affiliation(s)
- Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology & Oncology, University of California San Diego Moores Cancer Center, San Diego, California.
| | - David J Stewart
- Division of Medical Oncology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
34
|
Targeting Neovasculature with Multitargeted Antiangiogenesis Tyrosine Kinase Inhibitors in Non-small Cell Lung Cancer. BioDrugs 2017; 30:421-439. [PMID: 27670779 DOI: 10.1007/s40259-016-0194-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chemotherapy has reached a plateau in the efforts for survival improvement in non-small cell lung cancer (NSCLC). The growing knowledge of NSCLC molecular pathobiology has led to the development of new treatments that target specific tumor functions. Angiogenesis is a tumor function leading to the formation of new tumor vessels that are crucial for its survival. Although vascular endothelial growth factor (VEGF) plays a primary role in angiogenesis, the inhibition of the VEGF pathway with VEGF-receptor (VEGFR) tyrosine kinase inhibitors (TKIs) is associated with a modest survival benefit due to the development of resistance by the tumor that has been mainly attributed to the up-regulation of other stimulators of angiogenesis. Thus, the use of multitargeted antiangiogenesis TKIs (MATKIs) for simultaneous inhibition of multiple angiogenic pathways has been proposed. This review summarizes data about novel treatment strategies incorporating the inhibition of angiogenesis with MATKIs in NSCLC. The data from all relevant studies shows that MATKIs do not offer additional survival benefit to currently available chemotherapeutic options in unselected NSCLC patients. However, the diversity in disease response to MATKI-containing regimens implies that specific patient subgroups may benefit from or be harmed by these agents. In this context, most studies agree that the VEGFR-targeting MATKIs are harmful in squamous NSCLC while specific MATKIs (i.e., motesanib, vandetanib and nintedanib) are associated with improved progression free survival in non-squamous NSCLC. However, overall survival benefit was found only in adenocarcinoma and Asian non-squamous NSCLC patients with the use of nintedanib and motesanib, respectively.
Collapse
|
35
|
Raphael J, Chan K, Karim S, Kerbel R, Lam H, Santos KD, Saluja R, Verma S. Antiangiogenic Therapy in Advanced Non-small-cell Lung Cancer: A Meta-analysis of Phase III Randomized Trials. Clin Lung Cancer 2017; 18:345-353.e5. [PMID: 28188101 DOI: 10.1016/j.cllc.2017.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 12/26/2016] [Accepted: 01/03/2017] [Indexed: 11/25/2022]
Abstract
We conducted a meta-analysis to evaluate the efficacy of adding any antiangiogenic therapy (AT) to the standard of care in advanced non-small-cell lung cancer (NSCLC). The electronic databases Ovid PubMed, Cochrane Central Register of Controlled Trials, and Embase were searched to identify eligible trials. We included all phase III randomized trials with any line and type of treatment, histology. and AT dose. Pooled hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS), and pooled odds ratio (OR) for overall response rates (RR) were calculated. We divided the population into 2 subgroups based on the bevacizumab dose. Data of 19,098 patients from 25 phase III trials were analyzed. Compared with the standard of care, the addition of AT did not prolong OS (HR 0.98; 95% confidence interval [CI], 0.96-1.00; P = .1 and HR 0.97; 95% CI, 0.94-1.00; P = .06 for groups 1 and 2, respectively). However, there was a significant improvement in PFS with the addition of AT (HR 0.85; 95% CI, 0.79-0.91; P < .00001 and HR 0.81; 95% CI, 0.75-0.88; P < .00001 for groups 1 and 2, respectively) and overall RR (OR 1.61; 95% CI, 1.30-2.01; P < .0001 and OR 1.72; 95% CI, 1.39-2.14; P < .00001 for groups 1 and 2, respectively). This is the first meta-analysis including only all phase III trials with AT in NSCLC showing no significant effect on OS and an improvement in PFS and RR only. The role of AT in advanced NSCLC is still questionable; strong validated biomarkers are eagerly needed to predict which subgroup might benefit the most from such therapy.
Collapse
Affiliation(s)
- Jacques Raphael
- Medical Oncology Division, Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada.
| | - Kelvin Chan
- Medical Oncology Division, Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | - Safiya Karim
- Medical Oncology Division, Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | - Robert Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Henry Lam
- Medical Oncology Division, Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | - Keemo Delos Santos
- Medical Oncology Division, Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | - Ronak Saluja
- Medical Oncology Division, Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | - Sunil Verma
- Medical Oncology Division, Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada; Department of Oncology, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| |
Collapse
|
36
|
Li BT, Barnes TA, Chan DL, Naidoo J, Lee A, Khasraw M, Marx GM, Kris MG, Clarke SJ, Drilon A, Rudin CM, Pavlakis N. The addition of anti-angiogenic tyrosine kinase inhibitors to chemotherapy for patients with advanced non-small-cell lung cancers: A meta-analysis of randomized trials. Lung Cancer 2016; 102:21-27. [PMID: 27987583 DOI: 10.1016/j.lungcan.2016.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/16/2016] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The role of anti-angiogenic tyrosine kinase inhibitors (AATKI) for patients with non-small-cell lung cancers (NSCLC) is uncertain. We conducted a comprehensive meta-analysis to assess the overall utility of adding AATKI to chemotherapy. MATERIALS AND METHODS We included 15 randomized controlled trials (RCTs) of AATKI plus chemotherapy versus chemotherapy involving 7997 patients with advanced NSCLC. Meta-analysis was performed to obtain pooled hazard ratios (HR) for OS and PFS, and pooled odds ratios (OR) for objective response rate (ORR) and grade 3 or greater toxicity. Pre-specified subgroup analyses were performed according to line of chemotherapy, chemotherapeutic regimen and histology. RESULTS The addition of AATKI to chemotherapy significantly increased progression-free survival (PFS) (HR 0.83, 95% CI 0.79, 0.87; P<0.00001) and ORR [OR 1.63, 95% CI 1.45, 1.84; P<0.00001], but not overall survival (OS) (HR 0.96, 95% CI 0.91, 1.01; P=0.14). OS benefit was seen in the subset of patients with adenocarcinomas (HR 0.86; 95% CI 0.79, 0.95; P=0.002), especially in the second line setting (HR 0.85; 95% CI 0.76, 0.96; P=0.008). However, both grade ≥3 toxicity (HR 2.08, 95% CI 1.59, 2.73; P<0.00001) and treatment-related deaths (OR 2.37, 95% CI 1.58, 3.56; P<0.0001) were significantly higher with the addition of AATKI. CONCLUSION The addition of AATKI to chemotherapy in patients with advanced NSCLC significantly increased PFS and ORR but not OS, and did so at the expense of increased toxicity and treatment-related deaths. Preclinical and translational research in predictive biomarkers are essential for the clinical development of this class of drugs.
Collapse
Affiliation(s)
- Bob T Li
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, 300 East 66th Street, New York, NY 10065, USA; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Tristan A Barnes
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - David L Chan
- Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Jarushka Naidoo
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 301 Building Suite 4500, Baltimore, MD 21224, USA
| | - Adrian Lee
- Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Mustafa Khasraw
- Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Gavin M Marx
- Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; SAN Integrated Cancer Centre, Sydney Adventist Hospital, 185 Fox Valley Rd, Wahroonga, NSW 2076, Australia
| | - Mark G Kris
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, 300 East 66th Street, New York, NY 10065, USA
| | - Stephen J Clarke
- Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, 300 East 66th Street, New York, NY 10065, USA
| | - Charles M Rudin
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, 300 East 66th Street, New York, NY 10065, USA
| | - Nick Pavlakis
- Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW 2065, Australia.
| |
Collapse
|
37
|
Bordinhão ALR, Evangelista AF, Oliveira RJS, Macedo T, Silveira HC, Reis RM, Marques MM. MicroRNA profiling in human breast cancer cell lines exposed to the anti-neoplastic drug cediranib. Oncol Rep 2016; 36:3197-3206. [PMID: 27748845 DOI: 10.3892/or.2016.5153] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/15/2016] [Indexed: 11/06/2022] Open
Abstract
Cediranib, a pan-tyrosine kinase inhibitor is showing promising results for the treatment of several solid tumours. In breast cancer, its effects remain unclear, and there are no predictive biomarkers. Several studies have examined the expression profiles of microRNAs (miRNAs) in response to different chemotherapy treatments and found that the expression patterns may be associated with the treatment response. Therefore, our aim was to evaluate the cellular behaviour and differential expression profiles of miRNAs in breast cancer cell lines exposed to cediranib. The biological effect of this drug was measured by viability, migration, invasion and cell death in in vitro assays. Signaling pathways were assessed using a human phospho-receptor tyrosine kinase array. Furthermore, using a miRNA array and quantitative real-time PCR (qRT‑PCR), we assessed the relative expression of miRNAs following cediranib treatment. The breast cancer cell lines exhibited a distinct cytotoxic response to cediranib treatment. Cediranib exposure resulted in a decrease in the cell migration and invasion of all the breast cancer cell lines. Treatment with cediranib appeared to be able to modulate the activation of several RTKs that are targets of cediranib such as EGFR and a new potential target ROR2. Furthermore, this drug was able to modulate the expression profile of different microRNAs such as miR-494, miR-923, miR-449a, miR-449b and miR-886-3 in breast cancer cell lines. These miRNAs are reported to regulate genes involved in important molecular processes, according to bioinformatics prediction tools.
Collapse
Affiliation(s)
- A L R Bordinhão
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - A F Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - R J S Oliveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - T Macedo
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - H C Silveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - R M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - M M Marques
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| |
Collapse
|
38
|
Tew WP. Ovarian cancer in the older woman. J Geriatr Oncol 2016; 7:354-61. [PMID: 27499341 DOI: 10.1016/j.jgo.2016.07.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/02/2016] [Accepted: 07/18/2016] [Indexed: 02/08/2023]
Abstract
Ovarian cancer is the seventh most common cancer in women worldwide and accounts for nearly 4% of all new cases of cancer in women. Almost half of all patients with ovarian cancer are over the age of 65 at diagnosis, and over 70% of deaths from ovarian cancer occur in this same age group. As the population ages, the number of older women with ovarian cancer is increasing. Compared to younger women, older women with ovarian cancer receive less surgery and chemotherapy, develop worse toxicity, and have poorer outcomes. They are also significantly under-represented in clinical trials and thus application of standard treatment regimens can be challenging. Performance status alone has been shown to be an inadequate tool to predict toxicity of older patients from chemotherapy. Use of formal geriatric assessment tools is a promising direction for stratifying older patients on trials. Elderly-specific trials, adjustments to the eligibility criteria, modified treatment regimens, and interventions to decrease morbidities in the vulnerable older population should be encouraged.
Collapse
|
39
|
Ethier JL, Lheureux S, Oza A. The role of cediranib in ovarian cancer: current status and further investigation. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1196130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
40
|
Incorporation of Antiangiogenic Therapy Into the Non-Small-Cell Lung Cancer Paradigm. Clin Lung Cancer 2016; 17:493-506. [PMID: 27381269 DOI: 10.1016/j.cllc.2016.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/29/2016] [Accepted: 05/31/2016] [Indexed: 12/18/2022]
Abstract
Although molecular targeted agents have improved the treatment of lung cancer, their use has largely been restricted to limited subsets of the overall population that carry specific mutations. Angiogenesis, the formation of new blood vessels from existing networks, is an attractive, more general process for the development of targeted anticancer therapies, because it is critical for the growth of solid tumors, including non-small-cell lung cancer. Growing tissues require a vascular supply within a few millimeters. Therefore, solid tumors create a proangiogenic microenvironment to facilitate the development of new tumor-associated blood vessels, thus providing an adequate vascular supply for continued tumor growth. Antiangiogenic agents can specifically target the vascular endothelial growth factor (VEGF) signaling pathways, broadly inhibit multiple tyrosine kinases, or interfere with other angiogenic processes, such as disruption of existing tumor vasculature. The present report provides an overview of antiangiogenic therapy for non-small-cell lung cancer, including both currently approved antiangiogenic therapies (bevacizumab [anti-VEGF] and ramucirumab [anti-VEGF receptor 2] monoclonal antibodies), and a variety of promising novel agents in development. Although recent data have demonstrated promising efficacy for some novel agents, the overall development of antiangiogenic therapy has been hampered by redundancy in signaling pathways and the highly heterogeneous nature of tumors. An improved understanding of the molecular basis of angiogenesis will guide the development of new antiangiogenic therapies and the identification of biomarkers to predict which patients with lung cancer are most likely to benefit from antiangiogenic therapy.
Collapse
|
41
|
Are VEGFR-TKIs effective or safe for patients with advanced non-small cell lung cancer? Oncotarget 2016; 6:18206-23. [PMID: 26156021 PMCID: PMC4627246 DOI: 10.18632/oncotarget.4524] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 06/05/2015] [Indexed: 12/25/2022] Open
Abstract
Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) might be new therapeutic strategies for advanced non-small cell lung cancer (NSCLC). Here a total of 12,520 patients from 23 randomized controlled trials (RCTs) were enrolled to evaluate the efficacy and safety of VEGFR-TKIs quantitatively in advanced NSCLC. Compared with non-VEGFR-TKIs, VEGFR-TKIs regimen significantly improved progression-free survival (PFS) [hazard ratio (HR): 0.839, 95% confidence interval (CI): 0.805-0.874, P < 0.001], objective response rates (ORR) [relative risk (RR): 1.374, 95% CI: 1.193-1.583, P < 0.001] and disease control rates (DCR) (RR: 1.113, 95% CI: 1.027-1.206, P = 0.009), but not overall survival (OS) (HR: 0.960, 95% CI: 0.921-1.002, P = 0.060) for NSCLC patients. The RR of all-grade neutropenia, thrombocytopenia, hypertension, hemorrhage, fatigue, anorexia, stomatitis, diarrhea, rash, hand-foot skin reaction (HFSR) were increased in patients received VEGFR-TKIs. As for high-grade (≥ 3) adverse events (AEs), VEGFR-TKIs were associated with higher RR of neutropenia, thrombocytopenia, hypertension, fatigue, stomatitis, diarrhea, rash and HFSR. This study demonstrates VEGFR-TKIs improve PFS, ORR and DCR, but not OS in advanced NSCLC patients. VEGFR-TKIs induce more frequent and serious AEs compared with control therapies.
Collapse
|
42
|
Ellis PM. Anti-angiogenesis in Personalized Therapy of Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 893:91-126. [PMID: 26667340 DOI: 10.1007/978-3-319-24223-1_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Upregulation of angiogenesis is a frequent occurrence in lung cancer and is reported to represent a negative prognostic factor. This provides a rationale for the development and evaluation of anti-angiogenic agents. To date bevacizumab, a monoclonal antibody directed against serum VEGF, is the only anti-angiogenic agent that has demonstrated improved overall survival for patients with lung cancer. Meta-analysis of trials of bevacizumab in combination with platinum-based chemotherapy for NSCLC, show a 10% reduction in the risk of death (HR 0.90, 95% CI 0.81-0.99). However, therapy with bevacizumab is limited to NSCLC patients with non-squamous histology, good performance status, no brain metastases and the absence of bleeding or thrombotic disorders. More recently, similar survival was observed in a non bevacizumab containing regimen of carboplatin, pemetrexed and maintenance pemetrexed. Multiple oral anti-angiogenic compounds have been evaluated in NSCLC, both in first-line therapy, or upon disease progression. The majority of agents have shown some evidence of activity, but none have clearly demonstrated improvements in overall survival. Increased toxicities have been observed, including an increased risk of death for some agents, limiting their development. Promising data exist for sunitinib in patients with heavily pre-treated NSCLC, and nintedanib in combination with docetaxel, as second-line therapy for NSCLC. However, these findings require validation. Currently, there is no established role for anti-angiogenic therapy in SCLC, although there is some promise for sunitinib as maintenance therapy following platinum and etoposide chemotherapy. The challenge for anti-angiogenic therapy is to understand whether treatment effects in a subpopulation, are lost among a larger unselected population of patients. There is a need for additional translational research to identify predictive biomarkers for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Peter M Ellis
- Department of Oncology, McMaster University, Hamilton, ON, Canada. .,Juravinski Cancer Centre, Hamilton, ON, Canada.
| |
Collapse
|
43
|
Ledermann JA, Embleton AC, Raja F, Perren TJ, Jayson GC, Rustin GJS, Kaye SB, Hirte H, Eisenhauer E, Vaughan M, Friedlander M, González-Martín A, Stark D, Clark E, Farrelly L, Swart AM, Cook A, Kaplan RS, Parmar MKB. Cediranib in patients with relapsed platinum-sensitive ovarian cancer (ICON6): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2016; 387:1066-1074. [PMID: 27025186 DOI: 10.1016/s0140-6736(15)01167-8] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Angiogenesis is a validated clinical target in advanced epithelial ovarian cancer. Cediranib is an oral antiangiogenic vascular endothelial growth factor receptor 1-3 inhibitor that has shown antitumour activity in recurrent ovarian cancer. We assessed efficacy and safety of cediranib in combination with platinum-based chemotherapy and as continued maintenance treatment in patients with first relapse of platinum-sensitive ovarian cancer. METHODS In this randomised, three-arm, double-blind, placebo-controlled phase 3 trial, we randomly assigned patients aged 18 years or older with relapsed platinum-sensitive ovarian cancer at 63 centres in Australia, Canada, New Zealand, Spain, and the UK. Participants received up to six cycles of platinum-based chemotherapy (once every 3 weeks) then entered a maintenance phase. Participants were randomly allocated (2:3:3), with five stratification factors and in alternating blocks, to receive placebo alongside chemotherapy and then placebo only maintenance (arm A; reference), cediranib 20 mg once-daily alongside chemotherapy then placebo only maintenance (arm B; concurrent), or cediranib 20 mg once-daily alongside chemotherapy then cediranib 20 mg once-daily maintenance (arm C; maintenance). Patients continued treatment to progression or excessive toxic effects. The primary efficacy endpoint was progression-free survival between arms A and C. Efficacy analysis was by intention to treat. Safety was assessed in all patients who received the allocated study drug. This trial is registered with ClinicalTrials.gov, number NCT00532194; the ISRCTN registry, number ISRCTN68510403; and ANZ Clinical Trials Registry, number ACTRN1261000016003. FINDINGS We randomly assigned 486 [corrected] women between Nov 13, 2007, and Dec 23, 2011; results presented are for 456 patients randomly assigned subsequent to the 30mg safety phase. During a median of 19·5 months (IQR 14-26) follow-up, 113 (96%) of 118 women assigned to arm A and 141 (86%) of 164 assigned to arm C had disease progression. Median progression-free survival was 11·0 months (95% CI 10·4-11·7) in arm C and 8·7 months (7·7-9·4) in arm A (hazard ratio 0·56, 0·44-0·72, p<0·0001). 156 (90%) of 174 patients in arm B had disease progression, and median progression-free survival was 9·9 months (95% CI 9·4-10·5). Diarrhoea, neutropenia, hypertension, and voice changes were significantly more common, during chemotherapy with cediranib, and diarrhoea, hypothyroidism and voice changes were more common during maintenance. Poor compliance with cediranib was noted during maintenance treatment with toxic effects being the most common cause for discontinuation. INTERPRETATION Cediranib, when given orally with chemotherapy and continued as maintenance, yielded a meaningful improvement [corrected] in progression-free survival in women with recurrent platinum-sensitive ovarian cancer, albeit with added toxic effects. The positive results in ICON6 could provide women with a new therapeutic option for recurrent ovarian cancer. Assessment of the secondary endpoint of overall survival will need longer follow-up. FUNDING Medical Research Council, Cancer Research UK, Canadian Cancer Society Research Institute, Cancer Australia, National Gynecological Cancer Centre, and AstraZeneca.
Collapse
Affiliation(s)
- Jonathan A Ledermann
- Cancer Research UK and UCL Cancer Trials Centre, University College London and UCL Hospitals, London, UK.
| | - Andrew C Embleton
- Medical Research Council Clinical Trials Unit at UCL, University College London, London, UK
| | - Fharat Raja
- University College London Hospitals, London, UK
| | - Timothy J Perren
- St James's Institute of Oncology and Leeds Institute of Cancer Medicine and Pathology, Leeds, UK
| | - Gordon C Jayson
- Christie Hospital and University of Manchester, Manchester, UK
| | | | | | - Hal Hirte
- Juravinski Cancer Centre and McMaster University, Hamilton, ON, Canada
| | - Elizabeth Eisenhauer
- Canadian Cancer Trials Group (CCTG) and Queen's University, Kingston, ON, Canada
| | | | - Michael Friedlander
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Antonio González-Martín
- Spanish Group for Investigation in Ovarian Cancer (GEICO) and MD Anderson Cancer Center Madrid, Madrid, Spain
| | | | - Elizabeth Clark
- Medical Research Council Clinical Trials Unit at UCL, University College London, London, UK
| | - Laura Farrelly
- Medical Research Council Clinical Trials Unit at UCL, University College London, London, UK
| | | | - Adrian Cook
- Medical Research Council Clinical Trials Unit at UCL, University College London, London, UK
| | - Richard S Kaplan
- Medical Research Council Clinical Trials Unit at UCL, University College London, London, UK
| | - Mahesh K B Parmar
- Medical Research Council Clinical Trials Unit at UCL, University College London, London, UK
| |
Collapse
|
44
|
Cuffe S, Azad AK, Qiu X, Qiu X, Brhane Y, Kuang Q, Marsh S, Savas S, Chen Z, Cheng D, Leighl NB, Goss G, Laurie SA, Seymour L, Bradbury PA, Shepherd FA, Tsao MS, Chen BE, Xu W, Liu G. ABCC2 polymorphisms and survival in the Princess Margaret cohort study and the NCIC clinical trials group BR.24 trial of platinum-treated advanced stage non-small cell lung cancer patients. Cancer Epidemiol 2016; 41:50-6. [PMID: 26816351 DOI: 10.1016/j.canep.2015.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 12/20/2015] [Accepted: 12/31/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND The drug transporter ABCC2 is upregulated in non-small cell lung cancer (NSCLC) and implicated in platinum resistance. We evaluated the association between germline polymorphisms in the ABCC2 gene and survival outcomes of platinum-treated advanced NSCLC patients. MATERIAL AND METHODS Ten candidate and tagging germline polymorphisms in the ABCC2 gene were genotyped in a discovery cohort of 170 platinum-treated stage IV NSCLC patients from the Princess Margaret Cancer Centre. Associations with overall survival were assessed using multivariate Cox proportional hazard models adjusted for prognostic variables. To validate our results, we analyzed the association of the two top polymorphisms in the ABCC2 gene on survival outcomes of 219 stage IIIB-IV NSCLC patients enrolled on the NCIC Clinical Trials Group BR.24 clinical trial. RESULTS Only one polymorphism was validated across both cohorts for an association with overall survival: the A allele of the ABCC2 polymorphism, rs8187710 (4544G>A), was associated with adverse overall survival (adjusted hazard ratio [aHR] 2.22; 95% CI: 1.2-4.0; p=0.009) among our stage IV NSCLC patients. A significant association with overall survival (aHR 1.73; 95% CI: 1.0-2.9; p=0.036) was observed for the same ABCC2 polymorphism in the BR.24 validation cohort. No other ABCC2 polymorphisms were associated with outcome. CONCLUSION The ABCC2 polymorphism, rs8187710 (4544G>A), is associated with overall survival in platinum-treated advanced NSCLC patients. Additional studies are needed to evaluate the predictive versus prognostic nature of this relationship, and to explore the functional effect of this polymorphism on the pharmacokinetics of platinum drugs.
Collapse
Affiliation(s)
- Sinead Cuffe
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada; HOPE Directorate, St James's Hospital, Dublin 8, Ireland.
| | - Abul Kalam Azad
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| | - Xiaoping Qiu
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| | - Xin Qiu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Yonathan Brhane
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Qin Kuang
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| | - Sharon Marsh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Sevtap Savas
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada; Discipline of Genetics, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Zhuo Chen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| | - Dangxiao Cheng
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| | - Natasha B Leighl
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| | - Glenwood Goss
- Division of Medical Oncology, Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| | - Scott A Laurie
- Division of Medical Oncology, Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| | - Lesley Seymour
- NCIC Clinical Trials Group, Queens University, Kingston, ON, Canada
| | - Penelope A Bradbury
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada; NCIC Clinical Trials Group, Queens University, Kingston, ON, Canada
| | - Frances A Shepherd
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| | - Ming Sound Tsao
- Department of Pathology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Bingshu E Chen
- NCIC Clinical Trials Group, Queens University, Kingston, ON, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Geoffrey Liu
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, and Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
45
|
Bruce JY, LoRusso PM, Goncalves PH, Heath EI, Sadowski E, Shalinsky DR, Zhang Y, Traynor AM, Breazna A, Ricart AD, Tortorici M, Liu G. A pharmacodynamically guided dose selection of PF-00337210 in a phase I study in patients with advanced solid tumors. Cancer Chemother Pharmacol 2016; 77:527-38. [PMID: 26791870 DOI: 10.1007/s00280-016-2958-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/05/2016] [Indexed: 12/29/2022]
Abstract
PURPOSE PF-00337210 is an oral, highly selective vascular endothelial growth factor receptor (VEGFR) inhibitor. We evaluated a composite of biomarkers in real time to identify the recommended phase 2 dose (RP2D) and preliminary anticancer activity of PF-00337210. PATIENTS AND METHODS Patients (Pts) with advanced cancers were treated once (QD) or twice daily (BID) with escalating doses. Acute effects on tumor perfusion and vascularity were assessed using DCE-MRI, weekly BP readings, soluble VEGFR-2, and hemoglobin levels. RESULTS Forty-six pts were treated with 0.67-9 mg QD and 4-6 mg BID of PF-00337210. Nineteen pts (41%) previously received VEGF/VEGFR inhibitors. Two pts had dose-limiting toxicity (DLT) at 9 mg QD (troponin I increase and hypertension). The MTD at QD dose was 8 mg. Common drug-related adverse events were hypertension, fatigue, proteinuria, and nausea. Hypertension incidence and intensity corresponded with dose, but was well controlled with medication. Two confirmed partial responses and minor regressions (>10 to <30% reduction in target lesions) were noted. Complete DCE-MRI was acquired in 21 pts (20 evaluable for vascular response). Ten pts were vascular responders, including 5/6 pts at BID doses. Greatest modulation of soluble VEGFR-2 was at 6 mg BID. The maximum change from baseline in diastolic BP was higher at BID doses. There were no significant differences for systolic BP and hemoglobin levels. CONCLUSIONS PF-00337210 has profound VEGFR inhibition effects at well-tolerated doses. Antitumor activity and VEGF inhibition effects were observed across BID doses. The RP2D was 6 mg BID.
Collapse
Affiliation(s)
- Justine Yang Bruce
- Wisconsin Institute for Medical Research, University of Wisconsin Carbone Cancer Center, Room 7105, 1111 Highland Avenue, Madison, WI, 53705, USA.
| | - Patricia M LoRusso
- Karmanos Cancer Institute, Wayne State University, 4100 John R Street, Detroit, MI, 48201, USA
| | - Priscila H Goncalves
- Karmanos Cancer Institute, Wayne State University, 4100 John R Street, Detroit, MI, 48201, USA
| | - Elisabeth I Heath
- Karmanos Cancer Institute, Wayne State University, 4100 John R Street, Detroit, MI, 48201, USA
| | - Elizabeth Sadowski
- E3/366 Clinical Science Center, Department of Radiology, University of Wisconsin SMPH, 600 Highland Avenue, Madison, WI, 53792-3252, USA
| | - David R Shalinsky
- Department of Pharmacology, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Yanwei Zhang
- Department of Statistics, Pfizer Inc., 610 Main Street, Cambridge, MA, 02139, USA
| | - Anne M Traynor
- Wisconsin Institute for Medical Research, University of Wisconsin Carbone Cancer Center, Room 3103, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Aurora Breazna
- Department of Biostatistics, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Alejandro D Ricart
- Department of Biotechnology and Oncology Research, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Michael Tortorici
- Department of Clinical Pharmacology, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Glenn Liu
- Wisconsin Institute for Medical Research, University of Wisconsin Carbone Cancer Center, Room 7105, 1111 Highland Avenue, Madison, WI, 53705, USA
| |
Collapse
|
46
|
Hall RD, Le TM, Haggstrom DE, Gentzler RD. Angiogenesis inhibition as a therapeutic strategy in non-small cell lung cancer (NSCLC). Transl Lung Cancer Res 2015; 4:515-23. [PMID: 26629420 DOI: 10.3978/j.issn.2218-6751.2015.06.09] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In many cancers, including non-small cell lung cancer (NSCLC), tumor angiogenesis pathways have been identified as important therapeutic targets. Angiogenesis is essential in the process of primary tumor growth, proliferation and metastasis. One of the best characterized group of protein factors for angiogenesis include the members of the vascular endothelial growth factor (VEGF) family, consisting of VEGF-(A-D), and placenta growth factor (PIGF). Targeting tumor angiogenesis has been approached through two primary methods, monoclonal antibodies that block VEGF-vascular endothelial growth factor receptor (VEGFR) binding or small molecule tyrosine kinase inhibitors (TKIs) that inhibit the downstream VEGFR mediated signaling. Many TKIs inhibit multiple pro-angiogenic and pro-proliferative pathways such as the mitogen activated protein (MAP) kinase pathway. Bevacizumab and ramucirumab, monoclonal antibodies targeting VEGF and the VEGFR, respectively, have each led to improvements in overall survival (OS) for NSCLC when added to standard first and second line chemotherapy, respectively. Small incremental gains seen with both bevacizumab and ramucirumab may be further improved upon by incorporating novel agents and treatment strategies, and many additional trials are ongoing.
Collapse
Affiliation(s)
- Richard D Hall
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| | - Tri M Le
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| | - Daniel E Haggstrom
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| | - Ryan D Gentzler
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| |
Collapse
|
47
|
Bar J, Ding K, Zhao H, Han L, Laurie SA, Seymour L, Addison CL, Shepherd FA, Goss GD, Dimitroulakos J, Bradbury PA. Angiotensin-Converting Enzyme and Aldosterone Serum Levels as Prognostic and Predictive Biomarkers for Cediranib in NCIC Clinical Trials Group Study BR.24. Clin Lung Cancer 2015; 16:e189-201. [DOI: 10.1016/j.cllc.2015.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/01/2015] [Accepted: 05/05/2015] [Indexed: 11/26/2022]
|
48
|
Analysis of serum protein levels of angiogenic factors and their soluble receptors as markers of response to cediranib in the NCIC CTG BR.24 clinical trial. Lung Cancer 2015; 90:288-95. [DOI: 10.1016/j.lungcan.2015.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/02/2015] [Accepted: 09/05/2015] [Indexed: 01/10/2023]
|
49
|
Wick W, Platten M, Wick A, Hertenstein A, Radbruch A, Bendszus M, Winkler F. Current status and future directions of anti-angiogenic therapy for gliomas. Neuro Oncol 2015; 18:315-28. [PMID: 26459812 DOI: 10.1093/neuonc/nov180] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/03/2015] [Indexed: 12/24/2022] Open
Abstract
Molecular targets for the pathological vasculature are the vascular endothelial growth factor (VEGF)/VEGF receptor axis, integrins, angiopoietins, and platelet-derived growth factor receptor (PDGFR), as well as several intracellular or downstream effectors like protein kinase C beta and mammalian target of rapamycin (mTOR). Besides hypoxic damage or tumor cell starvation, preclinical models imply vessel independent tumor regression and suggest differential effects of anti-angiogenic treatments on tumorous and nontumorous precursor cells or the immune system. Despite compelling preclinical data and positive data in other cancers, the outcomes of clinical trials with anti-angiogenic agents in gliomas by and large have been disappointing and include VEGF blockage with bevacizumab, integrin inhibition with cilengitide, VEGF receptor inhibition with sunitinib or cediranib, PDGFR inhibition with imatinib or dasatinib, protein kinase C inhibition with enzastaurin, and mTOR inhibition with sirolimus, everolimus, or temsirolimus. Importantly, there is a lack of real understanding for this negative data. Anti-angiogenic therapies have stimulated the development of standardized imaging assessment and the integration of functional MRI sequences into daily practice. Here, we delineate directions in the identification of molecularly or image-based defined subgroups, anti-angiogenic cotreatment for immunotherapy, and the potential of ongoing trials or modified targets to change the game.
Collapse
Affiliation(s)
- Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany (W.W., M.P., A.W., A.H., F.W.); Department of Neuroradiology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany (A.R., M.B.)
| | - Michael Platten
- Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany (W.W., M.P., A.W., A.H., F.W.); Department of Neuroradiology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany (A.R., M.B.)
| | - Antje Wick
- Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany (W.W., M.P., A.W., A.H., F.W.); Department of Neuroradiology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany (A.R., M.B.)
| | - Anne Hertenstein
- Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany (W.W., M.P., A.W., A.H., F.W.); Department of Neuroradiology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany (A.R., M.B.)
| | - Alexander Radbruch
- Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany (W.W., M.P., A.W., A.H., F.W.); Department of Neuroradiology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany (A.R., M.B.)
| | - Martin Bendszus
- Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany (W.W., M.P., A.W., A.H., F.W.); Department of Neuroradiology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany (A.R., M.B.)
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany (W.W., M.P., A.W., A.H., F.W.); Department of Neuroradiology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany (A.R., M.B.)
| |
Collapse
|
50
|
Koller M, Warncke S, Hjermstad MJ, Arraras J, Pompili C, Harle A, Johnson CD, Chie WC, Schulz C, Zeman F, van Meerbeeck JP, Kuliś D, Bottomley A. Use of the lung cancer-specific Quality of Life Questionnaire EORTC QLQ-LC13 in clinical trials: A systematic review of the literature 20 years after its development. Cancer 2015; 121:4300-23. [PMID: 26451520 DOI: 10.1002/cncr.29682] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/24/2015] [Accepted: 07/02/2015] [Indexed: 12/19/2022]
Abstract
The European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire-Lung Cancer 13 (QLQ-LC13) covers 13 typical symptoms of lung cancer patients and was the first module developed in conjunction with the EORTC core quality-of-life (QL) questionnaire. This review investigates how the module has been used and reported in cancer clinical trials in the 20 years since its publication. Thirty-six databases were searched with a prespecified algorithm. This search plus an additional hand search generated 770 hits, 240 of which were clinical studies. Two raters extracted data using a coding scheme. Analyses focused on the randomized controlled trials (RCTs). Of the 240 clinical studies that were identified using the LC13, 109 (45%) were RCTs. More than half of the RCTs were phase 3 trials (n = 58). Twenty RCTs considered QL as the primary endpoint, and 68 considered it as a secondary endpoint. QL results were addressed in the results section of the article (n = 89) or in the abstract (n = 92); and, in half of the articles, QL results were presented in the form of tables (n = 53) or figures (n = 43). Furthermore, QL results had an impact on the evaluation of the therapy that could be clearly demonstrated in the 47 RCTs that yielded QL differences between treatment and control groups. The EORTC QLQ-LC13 fulfilled its mission to be used as a standard instrument in lung cancer clinical trials. An update of the LC13 is underway to keep up with new therapeutic trends and to ensure optimized and relevant QL assessment in future trials.
Collapse
Affiliation(s)
- Michael Koller
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Sophie Warncke
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Marianne J Hjermstad
- Regional Centre for Excellence in Palliative Care, Department of Oncology, Oslo University Hospital and European Palliative Care Research Centre, Department of Cancer and Molecular Medicine, Norwegian University of Science and Technology, Norway
| | - Juan Arraras
- Oncology Departments, Navarra Hospital Complex, Pamplona, Spain
| | - Cecilia Pompili
- Division of Thoracic Surgery, St. James's University Hospital, Leeds, United Kingdom
| | - Amelie Harle
- The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Colin D Johnson
- University Surgical Unit, University Hospital Southampton, Hampshire, United Kingdom
| | - Wei-Chu Chie
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Christian Schulz
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Florian Zeman
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | | | | | | | | | | |
Collapse
|