1
|
Zhang J, Su J, Zhou Y, Lu J. Evaluating the efficacy and safety of trebananib in treating ovarian cancer and non-ovarian cancer patients: a meta-analysis and systematic review. Expert Rev Anticancer Ther 2024; 24:881-891. [PMID: 38970210 DOI: 10.1080/14737140.2024.2377793] [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: 05/04/2024] [Accepted: 06/21/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVES Due to its anti-angiogenic properties, trebananib is frequently employed in the treatment of cancer patients, particularly those with ovarian cancer. We conducted a meta-analysis to assess the efficacy and safety profile of trebananib in combination with other drugs for treating both ovarian and non-ovarian cancer patients. METHODS Our search encompassed PubMed, Medline, Cochrane, and Embase databases, with a focus on evaluating study quality. Data extraction was conducted from randomized controlled trials (RCTs), and RevMan 5.3 facilitated result analysis. RESULTS Combining trebananib with other drugs extended progression-free survival (PFS) [HR 0.81, (95%CI: 0.65, 0.99), p = 0.04] and overall survival (OS) [HR 0.88, (95%CI: 0.79, 1.00), p = 0.04] in ovarian cancer patients. Ovarian cancer patients exhibited a higher objective response rate (ORR) with trebananib compared to non-ovarian cancer cohorts. Moreover, the incorporation of trebananib into the standard treatment regimen for malignant tumors did not significantly elevate drug-related adverse events [RR 1.05, (95% CI: 1.00, 1.11), p = 0.05]. CONCLUSION Trebananib plus other drugs can improve the PFS, OS and ORR in patients with cancer, especially ovarian cancer. Our recommendation is to use trebananib plus other drugs to treat advanced cancer, and to continuously monitor and manage drug-related adverse events. REGISTRATION PROSPERO (No. CRD42023466988).
Collapse
Affiliation(s)
- Jialin Zhang
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Jingyang Su
- Department of General internal medicine, Tongde Hospital Affiliated to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, China
| | - Yeyue Zhou
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Jinhua Lu
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
2
|
Vergote I, Scambia G, O'Malley DM, Van Calster B, Park SY, Del Campo JM, Meier W, Bamias A, Colombo N, Wenham RM, Covens A, Marth C, Raza Mirza M, Kroep JR, Ma H, Pickett CA, Monk BJ. Trebananib or placebo plus carboplatin and paclitaxel as first-line treatment for advanced ovarian cancer (TRINOVA-3/ENGOT-ov2/GOG-3001): a randomised, double-blind, phase 3 trial. Lancet Oncol 2019; 20:862-876. [PMID: 31076365 DOI: 10.1016/s1470-2045(19)30178-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/19/2019] [Accepted: 02/21/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Angiopoietin 1 and 2 regulate angiogenesis and vascular remodelling by interacting with the tyrosine kinase receptor Tie2, and inhibition of angiogenesis has shown promise in the treatment of ovarian cancer. We aimed to assess whether trebananib, a peptibody that inhibits binding of angiopoietin 1 and 2 to Tie2, improved progression-free survival when added to carboplatin and paclitaxel as first-line therapy in advanced epithelial ovarian, primary fallopian tube, or peritoneal cancer in a phase 3 clinical trial. METHODS TRINOVA-3, a multicentre, multinational, phase 3, double-blind study, was done at 206 investigational sites (hospitals and cancer centres) in 14 countries. Eligible patients were aged 18 years or older with biopsy-confirmed International Federation of Gynecology and Obstetrics (FIGO) stage III to IV epithelial ovarian, primary peritoneal, or fallopian tube cancers, and an ECOG performance status of 0 or 1. Eligible patients were randomly assigned (2:1) using a permuted block method (block size of six patients) to receive six cycles of paclitaxel (175 mg/m2) and carboplatin (area under the serum concentration-time curve 5 or 6) every 3 weeks, plus weekly intravenous trebananib 15 mg/kg or placebo. Maintenance therapy with trebananib or placebo continued for up to 18 additional months. The primary endpoint was progression-free survival, as assessed by the investigators, in the intention-to-treat population. Safety analyses included patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, number NCT01493505, and is complete. FINDINGS Between Jan 30, 2012, and Feb 25, 2014, 1164 patients were screened and 1015 eligible patients were randomly allocated to treatment (678 to trebananib and 337 to placebo). After a median follow-up of 27·4 months (IQR 17·7-34·2), 626 patients had progression-free survival events (405 [60%] of 678 in the trebananib group and 221 [66%] of 337 in the placebo group). Median progression-free survival did not differ between the trebananib group (15·9 months [15·0-17·6]) and the placebo group (15·0 months [12·6-16·1]) groups (hazard ratio 0·93 [95% CI 0·79-1·09]; p=0·36). 512 (76%) of 675 patients in the trebananib group and 237 (71%) of 336 in the placebo group had grade 3 or worse treatment-emergent adverse events; of which the most common events were neutropenia (trebananib 238 [35%] vs placebo 126 [38%]) anaemia (76 [11%] vs 40 [12%]), and leucopenia (81 [12%] vs 35 [10%]). 269 (40%) patients in the trebananib group and 104 (31%) in the placebo group had serious adverse events. Two fatal adverse events in the trebananib group were considered related to trebananib, paclitaxel, and carboplatin (lung infection and neutropenic colitis); two were considered to be related to paclitaxel and carboplatin (general physical health deterioration and platelet count decreased). No treatment-related fatal adverse events occurred in the placebo group. INTERPRETATION Trebananib plus carboplatin and paclitaxel did not improve progression-free survival as first-line treatment for advanced ovarian cancer. The combination of trebananib plus carboplatin and paclitaxel did not produce new safety signals. These results show that trebananib in combination with carboplatin and paclitaxel is minimally effective in this patient population. FUNDING Amgen.
Collapse
Affiliation(s)
- Ignace Vergote
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Leuven, Belgium; European Network of Gynaecological Oncological Trial groups (ENGOT), Divison of Gynecologic Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium.
| | - Giovanni Scambia
- Multicenter Italian Trials in Ovarian Cancer Society (MITO), Rome, Italy; Fondazione Policlinico Universitario A Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica, Rome, Italy
| | - David M O'Malley
- Gynecologic Oncology Group, James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Ben Van Calster
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | | | - Josep M Del Campo
- Grupo Español de Investigación en Cáncer de Ovario (GEICO), Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Werner Meier
- Arbeitsgemeinschaft Gynaekologische Onkologie Study Group (AGO), Department of Gynecology and Obstetrics, AGO-Germany and Evangelic Hospital Düsseldorf, Düsseldorf, Germany
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, Alexandra Hospital, National & Kapodistrian University of Athens, Athens, Greece; Hellenic Cooperative Oncology Group (HECOG), Athens, Greece
| | - Nicoletta Colombo
- Mario Negri Gynecologic Oncology Group (MANGO), European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; Università Milano Bicocca, Milan, Italy
| | - Robert M Wenham
- Gynecologic Oncology Group, Department of Gynecologic Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Al Covens
- Division of Gynecologic Oncology, Toronto Sunnybrook Regional Cancer Center, University of Toronto, Toronto, ON, Canada
| | - Christian Marth
- Arbeitsgemeinschaft Gynaekologische Onkologie Study Group (AGO)-Austria, Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Mansoor Raza Mirza
- Nordic Society of Gynaecological Oncology (NSGO), Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Judith R Kroep
- Dutch Gynecological Oncology Group (DCOG), Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Haijun Ma
- Global Development Oncology, Amgen, Thousand Oaks, CA, USA
| | | | - Bradley J Monk
- Gynecologic Oncology Group, Arizona Oncology (US Oncology Network), University of Arizona, Phoenix, AZ, USA; Creighton University, Phoenix, AZ, USA
| | | |
Collapse
|
3
|
Phase 1b Study of Trebananib Plus Paclitaxel and Trastuzumab in Patients With HER2-Positive Locally Recurrent or Metastatic Breast Cancer. Clin Breast Cancer 2018; 19:47-57. [PMID: 30420181 DOI: 10.1016/j.clbc.2018.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/21/2018] [Accepted: 09/29/2018] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Trebananib, a peptide-Fc fusion protein, blocks angiogenesis by inhibiting binding of angiopoietin-1/2 to the receptor tyrosine kinase Tie2. Trebananib plus trastuzumab and paclitaxel was evaluated in human epidermal growth factor receptor 2-positive breast cancer in an open-label phase 1b clinical study. PATIENTS AND METHODS Women with human epidermal growth factor receptor 2-positive breast cancer received weekly paclitaxel (80 mg/m2), trastuzumab (8 mg/m2 then 6 mg/kg every 3 weeks), and intravenous trebananib (10 mg/kg or 30 mg/kg weekly) beginning week 2. The primary end point was the incidence of dose-limiting toxicities. Secondary end points included incidence of adverse events (AEs), pharmacokinetics, and tumor response (objective response and duration of response). RESULTS Forty women were enrolled; 2 experienced dose-limiting toxicities (grade 3 ocular transient ischemic attack [10 mg/kg cohort] and grade 3 elevation in γ-glutamyl transferase [30 mg/kg cohort]). The most common treatment-emergent AEs were peripheral edema (n = 28), diarrhea (n = 27), alopecia (n = 26), fatigue (n = 24), and nausea (n = 24). Maximum observed concentration and area under the concentration-time curve increased proportionally with the trebananib dose. Objective response was confirmed in 31 patients. In the 10 mg/kg cohort, 16 patients (80%) experienced partial response, and none experienced complete response. In the 30 mg/kg cohort, 12 patients (71%) experienced partial response and 3 (18%) experienced complete response. Median (95% confidence interval) duration of response in the 10 and 30 mg/kg cohorts was 12.6 (4.3-20.2) and 16.6 (8.2-not estimable) months, respectively. CONCLUSION This phase 1b study showed that trebananib was tolerated with manageable AEs at a dose up to 30 mg/kg weekly. Trebananib demonstrated anticancer activity, as indicated by objective response and duration of response.
Collapse
|
4
|
Jiang Y, Sun X, Kong B, Jiang J. Antiangiogenesis therapy in ovarian cancer patients: An updated meta-analysis for 15 randomized controlled trials. Medicine (Baltimore) 2018; 97:e11920. [PMID: 30142803 PMCID: PMC6112884 DOI: 10.1097/md.0000000000011920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Antiangiogenesis therapy has been demonstrated to prolong the free survival with tolerable toxicity. However the efficacy of these drugs in overall survival (OS) remains controversial. This study was designed to assess the overall performance of antiangiogenesis therapy in improving the survival of ovary cancer (OC) patients. METHODS Electronic database of PubMed, Embase, MEDLINE, and the Cochrane Central Register of Controlled Trials were searched to identify relevant clinical randomized control trial (RCTs) assessing the therapeutic value of antiangiogenesis therapy in OC patients during 2011 to 2017. Additionally, abstracts of annual meetings were also conducted. Only English articles were considered. Progression free survival (PFS), OS, and objective response rate (ORR) were obtained from eligible RCTs. The HRs for time-to-event variables and ORs for dichotomous outcomes with their 95% CIs were used for this meta-analysis. All the statistical analyses were carried out by Stata 11.0 software using a fixed or random-models according to heterogeneity. RESULTS A total of 15 RCTs including 9359 patients were recruited into this meta-analysis. Addition of antiangiogenic agents improved PFS (HR = 0.71, 95% CI 0.62-0.81, P < .001), OS (HR = 0.92, 95% CI 0.86-0.98, P = .008) and ORR (OR = 1.74, 95% CI 1.27-2.39, P = .001) compared to placebo or chemotherapy alone in overall analysis. Antiangiogenic agents prolonged both PFS (HR = 0.58, 95% CI 0.52-0.65, P = .000) and OS (HR=0.84, 95% CI 0.76-0.92, P = .000) in recurrent settings but only PFS in primary settings (HR = 0.88, 95% CI 0.79-0.98, P = .020), longer PFS and OS in both platinum-sensitive recurrent patients (HR = 0.56, 95% CI 0.48-0.64, P = .000, PFS; HR = 0.86, 95% CI 0.76-0.98, P = .027, OS) as well as platinum-resistant recurrent cases (HR = 0.54, 95% CI 0.41-0.71, P = .000, PFS; HR = 0.84, 95% CI 0.71-0.98, P = .029, OS). Throughout therapy improved PFS (HR = 0.66, 95% CI 0.57-0.76, P < .001) and OS (HR = 0.89, 95% CI 0.83-0.96, P = .001). However the maintenance therapy of antiangiogenic agents was irrelevant to a longer PFS or OS. CONCLUSION Based on the available studies, antiangiogenic agents play an important role in the survival of OC patients. More randomized controlled trials are needed to reach more convinced conclusion.
Collapse
Affiliation(s)
- Yanyan Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
| | - Xiaomei Sun
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
- Key Laboratory of Gynecologic Oncology of Shandong Province, PR China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
- Key Laboratory of Gynecologic Oncology of Shandong Province, PR China
| | - Jie Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University
| |
Collapse
|
5
|
He YM, Yu C, Li WB, Li ZP, Xu N. Evaluation of short-term effectiveness of eight targeted agents combined with chemotherapy for treating esophageal-gastric junction adenocarcinoma: A network meta-analysis. J Cell Biochem 2018; 119:1183-1192. [PMID: 28708307 DOI: 10.1002/jcb.26288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022]
Abstract
This study aimed to evaluate the short-term effectiveness of eight targeted agents (ramucirumab, bevacizumab, rilotumumab, panitumumab, cetuximab, trebananib, trastuzumab, matuzumab) plus chemotherapy in esophageal-gastric junction adenocarcinoma (EGJA) by a network meta-analysis (NMA). PubMed, Embase, and Cochrane Library databases were systematically retrieved for randomized clinical trials (RCTs) concerning targeted agents plus chemotherapy in the treatment of EGJA. This NMA combined both direct and indirect evidence to evaluate odds ratio (OR) and to draw the surface under the cumulative ranking curve (SUCRA). In total 11 RCTs with 3649 EGJA patients (1907 patients treated with targeted agents plus chemotherapy were regarded as the case group, and 1742 patients with placebo plus chemotherapy were assigned into the control group) were enrolled in this study. Targeted agents in terms of stable disease (SD), partial response (PR), disease control rate (DCR), and overall response ratio (ORR) with the SUCRA values of 0.838, 0.807, 0.934, and 0.793, respectively. Cetuximab and trastuzumab, with the SUCRA values of 0.884 and 0.758, came on top as the best outcomes for treating EGJA in terms of progressive disease (PD) and complete response (CR). Cluster analysis results indicated that ramucirumab plus chemotherapy might be the optimal treatment for EGJA. Our findings indicated that ramucirumab plus chemotherapy might be the optimal treatment for EGJA amongst the nine treatment regimens, which provided clinical guidance for clinicians in the treatment of EGJA.
Collapse
Affiliation(s)
- Yong-Ming He
- Department of Integrated Traditional Chinese and Western Medicine, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Chen Yu
- Department of Integrated Traditional Chinese and Western Medicine, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Wei-Bing Li
- Department of Integrated Traditional Chinese and Western Medicine, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Zhi-Peng Li
- Department of Integrated Traditional Chinese and Western Medicine, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Nan Xu
- Department of Integrated Traditional Chinese and Western Medicine, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| |
Collapse
|
6
|
Cavaco M, Castanho MARB, Neves V. Peptibodies: An elegant solution for a long-standing problem. Biopolymers 2017; 110. [PMID: 29266205 DOI: 10.1002/bip.23095] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/20/2017] [Accepted: 11/24/2017] [Indexed: 01/09/2023]
Abstract
Chimeric proteins composed of a biologically active peptide and a fragment crystallizable (Fc) domain of immunoglobulin G (IgG) are known as peptibodies. They present an extended half-life due to neonatal Fc receptor (FcRn) salvage pathway, a decreased renal clearance rate owing to its increased size (≈70 kDa) and, depending on the peptide used in the design of the peptibody, an active-targeting moiety. Also, the peptides therapeutic activity is boosted by the number of peptides in the fusion protein (at least two peptides) and to some peptides' alterations. Peptibodies are mainly obtained through recombinant DNA technology. However, to improve peptide properties, "unnatural" changes have been introduced to the original peptides' sequence, for instance, the incorporation of D- or non-natural amino acid residues or even cyclization thus, limiting the application of genetic engineering in the production of peptibodies, since these peptides must be obtained via chemical synthesis. This constrains prompted the development of new methods for conjugation of peptides to Fc domains. Another challenge, subject of intense research, relates to the large-scale production of such peptibodies using these new techniques, which can be minimized by their proved value. To date, two peptibodies, romiplostim and dulaglutide, have been approved and stay as the standard of care in their areas of action. Furthermore, a considerable number of peptibodies are currently in preclinical and clinical development.
Collapse
Affiliation(s)
- Marco Cavaco
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
| | - Miguel A R B Castanho
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
| | - Vera Neves
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
| |
Collapse
|
7
|
Reardon DA, Lassman AB, Schiff D, Yunus SA, Gerstner ER, Cloughesy TF, Lee EQ, Gaffey SC, Barrs J, Bruno J, Muzikansky A, Duda DG, Jain RK, Wen PY. Phase 2 and biomarker study of trebananib, an angiopoietin-blocking peptibody, with and without bevacizumab for patients with recurrent glioblastoma. Cancer 2017; 124:1438-1448. [PMID: 29266174 DOI: 10.1002/cncr.31172] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/22/2017] [Accepted: 11/02/2017] [Indexed: 11/09/2022]
Abstract
BACKGROUND Angiopoietins contribute to tumor angiogenesis and may be upregulated as a compensatory factor after vascular endothelial growth factor (VEGF) blockade. The authors performed a phase 2 and biomarker study to evaluate trebananib, an angiopoietin 1 and angiopoietin 2 blocking peptibody, with and without bevacizumab in patients with recurrent glioblastoma. METHODS Forty-eight patients who had bevacizumab-naive, recurrent glioblastoma were treated with trebananib (30 mg/kg weekly) as single agent (n = 11) or combined with bevacizumab (n = 37). The primary endpoint was 6-month progression-free survival rate as determined by investigator review. Circulating biomarker levels were assessed before and after study therapy. RESULTS Trebananib was well tolerated as monotherapy and did not enhance bevacizumab-associated toxicity. Trebananib had no single-agent activity, and all treated patients exhibited progressive disease within 2 months. The 6-month progression-free survival rate for trebananib plus bevacizumab was 24.3% (95% confidence interval [CI], 12.1%-38.8%); whereas the median overall survival was 9.5 months (95% CI, 7.5-4.7 months), and the 12-month overall survival rate was 37.8% (95% CI, 22.6%-53.0%). Baseline and post-treatment changes in circulating vascular VEGF and interleukin-8 levels were correlated with survival among patients who received trebananib plus bevacizumab. CONCLUSIONS Angiopoietin 1 and angiopoietin 2 inhibition with trebananib was ineffective as monotherapy and did not enhance the ability of VEGF blockade with bevacizumab to improve the outcomes of patients with recurrent glioblastoma. Cancer 2018;124:1438-48. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Andrew B Lassman
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - David Schiff
- Department of Neurology, University of Virginia Medical Center, Charlottesville, Virginia
| | - Shakeeb A Yunus
- Hematology/Oncology, University of Massachusetts Memorial Medical Center, Worchester, Massachusetts
| | - Elizabeth R Gerstner
- Department of Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Timothy F Cloughesy
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | - Eudocia Quant Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah C Gaffey
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jennifer Barrs
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jennifer Bruno
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alona Muzikansky
- Biostatistics, Massachusetts General Hospital, Boston, Massachusetts
| | - Daniel G Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
8
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
9
|
Monk BJ, Poveda A, Vergote I, Raspagliesi F, Fujiwara K, Bae DS, Oaknin A, Ray-Coquard I, Provencher DM, Karlan BY, Lhommé C, Richardson G, Rincón DG, Coleman RL, Marth C, Brize A, Fabbro M, Redondo A, Bamias A, Ma H, Vogl FD, Bach BA, Oza AM. Final results of a phase 3 study of trebananib plus weekly paclitaxel in recurrent ovarian cancer (TRINOVA-1): Long-term survival, impact of ascites, and progression-free survival-2. Gynecol Oncol 2016; 143:27-34. [PMID: 27546885 DOI: 10.1016/j.ygyno.2016.07.112] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/12/2016] [Accepted: 07/23/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Trebananib, a peptibody that blocks binding of angiopoietin-1 and -2 to Tie2, significantly prolonged progression-free survival (PFS) in patients with recurrent epithelial ovarian cancer in the phase 3 TRINOVA-1 study. We report overall survival (OS) in the intent-to-treat population and clinically relevant subgroups and time to second disease progression (PFS-2). PATIENTS AND METHODS Women with recurrent disease (platinum-free interval<12months) were randomized to receive intravenous paclitaxel 80mg/m(2) (3weeks on/1week off) plus intravenous trebananib 15mg/kg or placebo, weekly. OS in the intent-to-treat population was a key secondary endpoint. Exploratory analysis of PFS-2 was conducted according to guidance by the European Medicines Agency. RESULTS Median OS was not significantly improved with trebananib compared with placebo (19.3 versus 18.3months; HR, 0.95; 95% CI, 0.81-1.11; P=0.52) in the intent-to-treat population (n=919). In subgroup analysis, trebananib improved median OS compared with placebo (14.5 versus 12.3months; HR, 0.72; 95% CI, 0.55-0.93; P=0.011) in patients with ascites at baseline (n=295). In the intent-to-treat population, trebananib significantly improved median PFS-2 compared with placebo (12.5 versus 10.9months; HR, 0.85; 95% CI, 0.74-0.98; P=0.024). The incidence and type of adverse events in this updated analysis was consistent with that described in the primary analysis; no new safety signals were detected. CONCLUSIONS OS was not significantly longer in the intent-to-treat population, although there was an improvement in OS in patients with ascites receiving trebananib. PFS-2 confirmed that the PFS benefit associated with trebananib was maintained through the second disease progression independent of the choice of subsequent therapy.
Collapse
Affiliation(s)
- Bradley J Monk
- Department of Obstetrics and Gynecology, University of Arizona Cancer Center at Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| | - Andrés Poveda
- Area Clinica de Oncologia Ginecológica, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Ignace Vergote
- Department of Obstetrics and Gynecology, University Hospital Leuven, Leuven Cancer Institute, KU Leuven, European Union, Belgium
| | - Francesco Raspagliesi
- Gynecologic Oncology Unit, Fondazione IRCCS, Istituto Nazionale per la Cura e lo Studio dei Tumori, Milano, Italy
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka-Shi, Japan
| | - Duk-Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, South Korea
| | - Ana Oaknin
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Diane M Provencher
- Division of Gynecologic Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Catherine Lhommé
- Department of Medicine, Institut Gustave Roussy, Villejuif, France
| | - Gary Richardson
- Academic Haematology and Oncology, Cabrini Hospital, Malvern, VIC, Australia
| | | | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christian Marth
- Universitätsklinik für Gynäkologie und Geburtshilfe, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Arija Brize
- Latvian Oncology Center, Riga Eastern Clinical University Hospital, Riga, Latvia
| | - Michel Fabbro
- Regional Cancer Institute Montpellier, Montpellier, France
| | | | - Aristotelis Bamias
- Alexandra Hospital, Department of Clinical Therapeutics, National & Kapodistrian University of Athens, Athens, Greece
| | - Haijun Ma
- Global Biostatistical Science, Amgen Inc., Thousand Oaks, CA, USA
| | - Florian D Vogl
- Global Development Oncology, Amgen Inc., Thousand Oaks, CA, USA
| | - Bruce A Bach
- Global Development Oncology, Amgen Inc., Thousand Oaks, CA, USA
| | - Amit M Oza
- Department of Medicine, Princess Margaret Hospital, University of Toronto, ON, Canada
| |
Collapse
|
10
|
Health-related quality of life in women with recurrent ovarian cancer receiving paclitaxel plus trebananib or placebo (TRINOVA-1). Ann Oncol 2016; 27:1006-1013. [DOI: 10.1093/annonc/mdw147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/17/2016] [Indexed: 11/14/2022] Open
|
11
|
Dowlati A, Vlahovic G, Natale RB, Rasmussen E, Singh I, Hwang YC, Rossi J, Bass MB, Friberg G, Pickett CA. A Phase I, First-in-Human Study of AMG 780, an Angiopoietin-1 and -2 Inhibitor, in Patients with Advanced Solid Tumors. Clin Cancer Res 2016; 22:4574-84. [PMID: 27076631 DOI: 10.1158/1078-0432.ccr-15-2145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 03/18/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess the toxicity, pharmacokinetics, tumor vascular response, tumor response, and pharmacodynamics of AMG 780, a mAb designed to inhibit the interaction between angiopoietin-1 and -2 and the Tie2 receptor. EXPERIMENTAL DESIGN This was a phase I dose-escalation study of patients with advanced solid tumors refractory to standard treatment without previous antiangiogenic treatment. AMG 780 was administered by intravenous infusion every 2 weeks in doses from 0.1 to 30 mg/kg. The primary endpoints were incidences of dose-limiting toxicity (DLT) and adverse events (AE), and pharmacokinetics. Secondary endpoints included tumor response, changes in tumor volume and vascularity, and anti-AMG 780 antibody formation. RESULTS Forty-five patients were enrolled across nine dose cohorts. Three patients had DLTs (0.6, 10, and 30 mg/kg), none of which prevented dose escalation. At 30 mg/kg, no MTD was reached. Pharmacokinetics of AMG 780 were dose proportional; median terminal elimination half-life was 8 to 13 days. No anti-AMG 780 antibodies were detected. At week 5, 6 of 16 evaluable patients had a >20% decrease in volume transfer constant (K(trans)), suggesting reduced capillary blood flow/permeability. The most frequent AEs were hypoalbuminemia (33%), peripheral edema (29%), decreased appetite (27%), and fatigue (27%). Among 35 evaluable patients, none had an objective response; 8 achieved stable disease. CONCLUSIONS AMG 780 could be administered at doses up to 30 mg/kg every 2 weeks in patients with advanced solid tumors. AMG 780 treatment resulted in tumor vascular effects in some patients. AEs were in line with toxicity associated with antiangiopoietin treatment. Clin Cancer Res; 22(18); 4574-84. ©2016 AACR.
Collapse
Affiliation(s)
- Afshin Dowlati
- Case Western Reserve University and University Hospitals Seidman Cancer Center, Cleveland, Ohio.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Monk BJ, Minion LE, Coleman RL. Anti-angiogenic agents in ovarian cancer: past, present, and future. Ann Oncol 2016; 27 Suppl 1:i33-i39. [PMID: 27141068 PMCID: PMC6283356 DOI: 10.1093/annonc/mdw093] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Angiogenesis plays a pivotal role in normal ovarian physiology as well as in the progression of ovarian cancer through ascites formation and metastatic spread. Bevacizumab (Avastin(®), Genentech; South San Francisco, CA, USA), a humanized anti-vascular endothelial growth factor (VEGF) monoclonal antibody, is the most widely studied anti-angiogenesis agent both across tumor types and specifically in epithelial ovarian cancer. In 2005, single-agent bevacizumab at 15 mg/kg (IV) every 3 weeks was first reported to be active in a case of recurrent high-grade serous ovarian cancer after failing 11th line cytotoxic treatment. Since then, many case series, phase II and phase III trials have confirmed these results leading to regulatory approval in most countries including the US Food and Drug Administration in 2014. Guidelines now give clear recommendations as to when and how bevacizumab should be integrated into the ovarian cancer treatment paradigm. Other anti-VEGF agents such as the VEGF receptor (VEGFR) tyrosine kinase inhibitors have not shown increased activity or reduced toxicity relative to bevacizumab. However, anti-angiogenics other than anti-VEGF/VEGFR agents such as those targeting Angiopoietin-1 and -2 are in development as well as novel combinations with vascular disrupting agents (VDAs), PARP inhibitors and immune checkpoint inhibitors. Clearly, the benefits of anti-angiogenic agents such as bevacizumab must be carefully weighed against the cost and associated toxicities. Although almost all patients with ovarian cancer will receive an anti-angiogenic compound, cures are not increased. Predictive biomarkers are an urgent unmet need.
Collapse
Affiliation(s)
- B J Monk
- Division of Gynecologic Oncology, The University of Arizona Cancer Center, Creighton University School of Medicine at St Joseph's Hospital and Medical Center, Phoenix
| | - L E Minion
- Division of Gynecologic Oncology, The University of Arizona Cancer Center, Creighton University School of Medicine at St Joseph's Hospital and Medical Center, Phoenix
| | - R L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
13
|
Ye W. The Complexity of Translating Anti-angiogenesis Therapy from Basic Science to the Clinic. Dev Cell 2016; 37:114-25. [DOI: 10.1016/j.devcel.2016.03.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/11/2016] [Accepted: 03/21/2016] [Indexed: 12/24/2022]
|
14
|
Al Wadi K, Ghatage P. Efficacy of trebananib (AMG 386) in treating epithelial ovarian cancer. Expert Opin Pharmacother 2016; 17:853-60. [PMID: 26933765 DOI: 10.1517/14656566.2016.1161027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Epithelial ovarian cancer (EOC) is the leading cause of death among gynecologic cancers. The majority of women are diagnosed with advanced stage disease. It is considered a chemosensitive cancer with a high initial response rate to first-line platinum and taxane-based chemotherapy. However, most patients with advanced EOC will relapse with subsequent resistance to conventional chemotherapy and ultimately succumb to their disease. Therefore, new therapeutic agents and strategies are desperately needed to improve the outcomes in patients with advanced EOC. AREAS COVERED This review focuses on the use of Trebananib (a non-VEGF-dependent angiogenesis pathway inhibitor) in EOC. Angiogenesis has been recognized as an important process promoting EOC growth and metastasis. Targeting angiogenesis in EOC have been developed and studied with demonstrated clinical efficacy. Bevacizumab, a humanized monoclonal antibody, that targets vascular endothelial growth factor A (VEGF-A), has been the most well evaluated molecular targeted therapy in the treatment of advanced and recurrent EOC with proven clinical efficacy. However, VEGF-dependent angiogenesis pathway inhibitors are often associated with serious toxicities and drug resistance ultimately develops. Hence, new therapeutic approach targeting the angiopoietin-Tie-2 complex pathway (a non-VEGF-dependent angiogenesis pathway) has gained interest over the past few years as an alternative strategy to overcome VEGF-dependent anti-angiogenesis-related toxicity and resistance. EXPERT OPINION Targeting angiopoietin-Tie-2 pathway represents a promising alternative approach to tumor anti-angiogenesis with a distinct toxicity profile from the VEGF-dependent pathway inhibitors. However, there are still many questions to be answered regarding the optimal treatment schedules, maintenance regimens, duration of maintenance therapy, and the best combination strategy. Currently there is no reliable surrogate molecular, cellular, or genetic marker that would definitively predict response to anti-angiogenic therapy. Identification of certain relevant and predictive biomarkers in the future may optimize treatment's efficacy by distinguishing the subset group of patients with EOC that would derive the most benefit from existing antiangiogenic treatment regimens.
Collapse
Affiliation(s)
- Khalid Al Wadi
- a Division of Gynecologic Oncology , Tom Baker Cancer Centre , Calgary , AB , Canada.,b Women's Specialized Hospital, King Fahad Medical City , Riyadh , Saudi Arabia
| | - Prafull Ghatage
- a Division of Gynecologic Oncology , Tom Baker Cancer Centre , Calgary , AB , Canada
| |
Collapse
|
15
|
Gadducci A, Lanfredini N, Sergiampietri C. Antiangiogenic agents in gynecological cancer: State of art and perspectives of clinical research. Crit Rev Oncol Hematol 2015; 96:113-28. [PMID: 26126494 DOI: 10.1016/j.critrevonc.2015.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/08/2015] [Accepted: 05/12/2015] [Indexed: 12/27/2022] Open
Abstract
Vascular endothelial growth factor [VEGF] pathway, which plays a key role in angiogenesis, may be blocked by either extracellular interference with VEGF itself (bevacizumab [BEV] or aflibercept), or intracytoplasmic inhibition of VEGF receptor (pazopanib, nintedanib, cediranid, sunitinib and sorafenib). An alternative approach is represented by trebananib, a fusion protein that prevents the interaction of angiopoietin [Ang]-1 and Ang-2 with Tie2 receptor on vascular endothelium. The combination of antiangiogenic agents, especially BEV, and chemotherapy is a rational therapeutic option for primary or recurrent ovarian carcinoma. However, it will be difficult to accept that it represents the new standard treatment, until biological characterization of ovarian carcinoma has not identified subsets of tumors with different responsiveness to BEV. Anti-angiogenesis is an interesting target also for recurrent cervical or endometrial cancer, but nowadays the use of anti-angiogenic agents in these malignancies should be reserved to patients enrolled in clinical trials.
Collapse
Affiliation(s)
- Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy.
| | - Nora Lanfredini
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy
| | - Claudia Sergiampietri
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy
| |
Collapse
|
16
|
Diamond JR, Wu B, Agarwal N, Bowles DW, Lam ET, Werner TL, Rasmussen E, Gamelin E, Soto F, Friberg G, Sun YN, Sharma S. Pharmacokinetic drug-drug interaction study of the angiopoietin-1/angiopoietin-2-inhibiting peptibody trebananib (AMG 386) and paclitaxel in patients with advanced solid tumors. Invest New Drugs 2015; 33:691-9. [PMID: 25895965 DOI: 10.1007/s10637-015-0236-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/25/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Trebananib is an anti-angiogenic peptibody under investigation in patients with advanced cancer. This study evaluated the pharmacokinetic (PK) drug-drug interaction of paclitaxel and trebananib. PATIENTS AND METHODS Patients with advanced solid tumors received weekly 80 mg/m(2) intravenous (IV) paclitaxel (3 weeks on/1 week off) with weekly 15 mg/kg IV trebananib starting at Week 2. Blood samples for PK analysis were collected at Week 1 (paclitaxel alone), Week 6 (paclitaxel and trebananib), and Week 8 (trebananib alone). An absence of interaction was to be concluded if the 90 % confidence intervals (CI) for the differences in paclitaxel exposure fell within the 0.80-1.25 interval. RESULTS The primary study was conducted between 7/2012 and 10/2013. Thirty-five patients were enrolled and 34 received both treatments. Most patients were white (91 %) and female (59 %); mean age was 61 years. The most common tumor types were ovarian (32 %) and bladder (27 %), 71 % of patients had stage IV disease, and all had Eastern Cooperative Oncology Group (ECOG) scores of 0 or 1. PK parameter analysis was done on patients with evaluable PK data at both assessments (with and without concomitant therapy; n = 28). The geometric least squares mean (GLSM) ratio (90 % CI) of paclitaxel AUCinf with and without trebananib was 1.17 (1.10, 1.25). The GLSM ratio (90 % CI) of trebananib AUCtau,ss with and without paclitaxel was 0.92 (0.87, 0.97). The most common adverse events were fatigue, local edema, peripheral edema, and nausea. CONCLUSIONS This study showed no evidence of clinically meaningful PK interaction between paclitaxel and trebananib.
Collapse
Affiliation(s)
- Jennifer R Diamond
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO, 80045, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Marchetti C, Gasparri ML, Ruscito I, Palaia I, Perniola G, Carrone A, Farooqi AA, Pecorini F, Muzii L, Panici PB. Advances in anti-angiogenic agents for ovarian cancer treatment: The role of trebananib (AMG 386). Crit Rev Oncol Hematol 2015; 94:302-10. [PMID: 25783620 DOI: 10.1016/j.critrevonc.2015.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 11/16/2014] [Accepted: 02/04/2015] [Indexed: 01/01/2023] Open
Abstract
Ovarian cancer is a multifaceted and genomically complex disease and has emerged as leading cause of death among gynecological malignancies. Gold-standard treatment consisted of cytoreductive surgery and paclitaxel-carboplatin chemotherapy. Recently, promising results of randomized trials have definitively confirmed the importance of angiogenesis in oncogenesis and ovarian cancer behavior, by showing a significant prolongation of progression-free survival with the addiction of an angiogenesis inhibitor to standard treatment in the first and second line setting. Research over the years has sequentially provided a rapidly broadening signaling landscape and many drugs targeting different signaling pathways of angiogenesis have been developed and investigated. Recently accumulating scientific evidence has started to shed light on the efficacy of AMG 386, a new peptibody reported to neutralize the interaction between angiopoietins (Ang1/2) and their Tie2 receptors, thus representing a promising alternative, both in terms of efficacy and toxicity profile and is considerably under investigation. The aim of this review is to summarize the recent researches and clinical progresses of AMG 386 as a novel target agent in ovarian cancer.
Collapse
Affiliation(s)
- Claudia Marchetti
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy.
| | - Maria Luisa Gasparri
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ilary Ruscito
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Innocenza Palaia
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Giorgia Perniola
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Angela Carrone
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | - Francesco Pecorini
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ludovico Muzii
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Pierluigi Benedetti Panici
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
18
|
Yang X, Shen F, Hu W, Coleman RL, Sood AK. New ways to successfully target tumor vasculature in ovarian cancer. Curr Opin Obstet Gynecol 2015; 27:58-65. [PMID: 25502429 PMCID: PMC4529067 DOI: 10.1097/gco.0000000000000136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The aim of this article was to review the recent literature on potential therapeutic strategies for overcoming resistance to antivascular endothelial growth factor drugs in ovarian cancer. RECENT FINDINGS Although clinical benefits of antivascular endothelial growth factor therapy were observed in ovarian cancer treatment trials, this use yielded only modest improvement in progression-free survival and, with the exception of cediranib, no effect on overall survival. Adaptive resistance and escape from antiangiogenesis therapy is likely a multifactorial process, including induction of hypoxia, vascular modulators, and immune response. New drugs targeting the tumor vasculature or other components of the surrounding microenvironment have shown promising results. SUMMARY When to start and end antiangiogenesis therapy and the choice of optimal treatment combinations remain controversial. Further evaluation of personalized novel angiogenesis-based therapy is warranted. Defining the critical interaction of these agents and pathways and the appropriate predictive markers will become an increasingly important objective for effective treatment.
Collapse
Affiliation(s)
- Xiaoyun Yang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Fangrong Shen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert L. Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
19
|
Suh DH, Kim HS, Kim B, Song YS. Metabolic orchestration between cancer cells and tumor microenvironment as a co-evolutionary source of chemoresistance in ovarian cancer: a therapeutic implication. Biochem Pharmacol 2014; 92:43-54. [PMID: 25168677 DOI: 10.1016/j.bcp.2014.08.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/15/2014] [Accepted: 08/18/2014] [Indexed: 12/12/2022]
Abstract
Our group reported a significant association between hexokinase II overexpression and chemoresistance in ovarian cancer, suggesting that aerobic glycolysis in the so-called Warburg effect might contribute to cancer progression. However, a growing body of evidence indicates contradictory findings with regard to the Warburg effect, such as high mitochondrial activity in highly invasive tumors and low ATP contribution of glycolysis in ovarian cancer. As a solution for the dilemma of the Warburg effect, the "reverse Warburg effect" was proposed in which aerobic glycolysis might occur in the stromal compartment of the tumor rather than in the cancer cells, indicating that the glycolytic tumor stroma feed the cancer cells through a type of symbiotic relationship. The reverse Warburg effect acting on the relationship between cancer cells and cancer-associated fibroblasts has evolved into dynamic interplay between cancer cells and multiple tumor stromal compartments, including cancer-associated fibroblasts, the extracellular matrix, endothelial cells, mesenchymal stem cells, adipocytes, and tumor-associated macrophages. Peritoneal cavities including ascites and the omentum also form a unique environment that is highly receptive for carcinomatosis in the advanced stages of ovarian cancer. The complicated but ingeniously orchestrated stroma-mediated cancer metabolism in ovarian cancer provides great heterogeneity in tumors with chemoresistance, which makes the disease thus far difficult to cure by single stromal-targeting agents. This review will discuss the experimental and clinical evidence of the cross-talk between cancer cells and various components of tumor stroma in terms of heterogeneous chemoresistance with focal points for therapeutic intervention in ovarian cancer.
Collapse
Affiliation(s)
- Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea
| | - Boyun Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea.
| |
Collapse
|
20
|
Vergote I, Oaknin A, Baurain JF, Ananda S, Wong S, Su X, Wu B, Zhong Z, Warner D, Casado A. A phase 1b, open-label study of trebananib in combination with paclitaxel and carboplatin in patients with ovarian cancer receiving interval or primary debulking surgery. Eur J Cancer 2014; 50:2408-16. [PMID: 25037684 DOI: 10.1016/j.ejca.2014.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/10/2014] [Accepted: 06/13/2014] [Indexed: 01/29/2023]
Abstract
AIM To evaluate the tolerability, pharmacokinetics and tumour response of first-line trebananib plus paclitaxel and carboplatin followed by trebananib maintenance in high-risk or advanced ovarian cancer. METHODS In this open-label phase 1b study, patients received intravenous (IV) trebananib 15 mg/kg administered weekly (QW) plus paclitaxel 175 mg/m(2) once every 3 weeks (Q3W) and carboplatin 6 mg/mL · min Q3W followed by trebananib 15 mg/kg QW monotherapy for 18 months. End-points were dose-limiting toxicities (DLTs; primary); treatment-emergent adverse events (AEs), anti-trebananib antibodies, pharmacokinetics and tumour response (secondary). RESULTS Twenty seven patients (interval debulking surgery [IDS], n=13) were enrolled. No DLTs occurred. During the combination therapy phase, AEs (>50%) in patients with IDS were nausea, diarrhoea, fatigue, decreased appetite and thrombocytopenia. In patients with primary debulking surgery (PDS), they were nausea, diarrhoea, fatigue and localised oedema. Grade 4 AEs were neutropenia (IDS, PDS; all n=3) and thrombocytopenia (IDS, PDS; all n=1). No deaths occurred. Toxicity results pertaining to trebananib maintenance were immature. The treatment combination did not markedly affect the pharmacokinetics across agents. In patients with IDS (n=14 after one patient was reassigned from PDS to IDS), 12 patients had a partial response (PR), two patients had stable disease. In patients with PDS (n=4), three patients had a complete response, one patient had a PR. CONCLUSIONS In women with ovarian cancer receiving IDS or PDS, IV trebananib 15 mg/kg QW plus paclitaxel and carboplatin appears tolerable. Results suggest that the treatment combination followed by trebananib 15 mg/kg monotherapy is associated with antitumour activity.
Collapse
Affiliation(s)
- I Vergote
- University Hospitals-KU Leuven, Leuven Cancer Institute, Department of Obstetrics and Gynecology, Herestraat 49, B-3000 Leuven, Belgium.
| | - A Oaknin
- Vall d'Hebron University Hospital, Medical Oncology Department, and Vall d'Hebron Institute of Oncology (VHIO), Head, Neck, and Gynecological Tumors Group, P. Vall d'Hebron 119-129, Barcelona 08035, Spain.
| | - J-F Baurain
- Université Catholique de Louvain, Centre du Cancer, Service d'Oncologie Médicale des Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, Bruxelles 1200, Belgium.
| | - S Ananda
- Royal Women's Hospital, Oncology Unit, 20 Flemington Road, Parkville 3052, VIC, Australia.
| | - S Wong
- Western Hospital, Department of Medical Oncology, Oncology Research Level 2 South, Gordon Street, Footscray 3011, VIC, Australia.
| | - X Su
- Amgen Inc., Department of Biostatistics, One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA.
| | - B Wu
- Amgen Inc., Department of Pharmacokinetics and Drug Metabolism, One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA.
| | - Z Zhong
- Amgen Inc., Department of Clinical Immunology and Biological Sample Management, One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA.
| | - D Warner
- Amgen Inc., Department of Clinical Development, One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA.
| | - A Casado
- Hospital Universitario Clínico San Carlos, Servicio de Oncologia Medica, Calle del Professor Martín Lagos s/n, Madrid 28040, Spain.
| |
Collapse
|
21
|
Monk BJ, Poveda A, Vergote I, Raspagliesi F, Fujiwara K, Bae DS, Oaknin A, Ray-Coquard I, Provencher DM, Karlan BY, Lhommé C, Richardson G, Rincón DG, Coleman RL, Herzog TJ, Marth C, Brize A, Fabbro M, Redondo A, Bamias A, Tassoudji M, Navale L, Warner DJ, Oza AM. Anti-angiopoietin therapy with trebananib for recurrent ovarian cancer (TRINOVA-1): a randomised, multicentre, double-blind, placebo-controlled phase 3 trial. Lancet Oncol 2014; 15:799-808. [PMID: 24950985 DOI: 10.1016/s1470-2045(14)70244-x] [Citation(s) in RCA: 220] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Angiogenesis is a valid target in the treatment of epithelial ovarian cancer. Trebananib inhibits the binding of angiopoietins 1 and 2 to the Tie2 receptor, and thereby inhibits angiogenesis. We aimed to assess whether the addition of trebananib to single-agent weekly paclitaxel in patients with recurrent epithelial ovarian cancer improved progression-free survival. METHODS For this randomised, double-blind phase 3 study undertaken between Nov 10, 2010, and Nov 19, 2012, we enrolled women with recurrent epithelial ovarian cancer from 32 countries. Patient eligibility criteria included having been treated with three or fewer previous regimens, and a platinum-free interval of less than 12 months. We enrolled patients with a computerised interactive voice response system, and patients were randomly assigned using a permuted block method (block size of four) in a 1:1 ratio to receive weekly intravenous paclitaxel (80 mg/m(2)) plus either weekly masked intravenous placebo or trebananib (15 mg/kg). Patients were stratified on the basis of platinum-free interval (≥0 and ≤6 months vs >6 and ≤12 months), presence or absence of measurable disease, and region (North America, western Europe and Australia, or rest of world). The sponsor, investigators, site staff, and patients were masked to the treatment assignment. The primary endpoint was progression-free survival assessed in the intention-to-treat population. The trial is registered with ClinicalTrials.gov, NCT01204749, and is no longer accruing patients. FINDINGS 919 patients were enrolled, of whom 461 were randomly assigned to the trebananib group and 458 to the placebo group. Median progression-free survival was significantly longer in the trebananib group than in the placebo group (7·2 months [5·8-7·4] vs 5·4 months [95% CI 4·3-5·5], respectively, hazard ratio 0·66, 95% CI 0·57-0·77, p<0·0001). Incidence of grade 3 or higher adverse events was similar between treatment groups (244 [54%] of 452 patients in the placebo group vs 258 [56%] of 461 patients in the trebananib group). Trebananib was associated with more adverse event-related treatment discontinuations than was placebo (77 [17%] patients vs 27 [6%], respectively) and higher incidences of oedema (294 [64%] patients had any-grade oedema in the trebananib group vs 127 [28%] patients in the placebo group). Grade 3 or higher adverse events included ascites (34 [8%] in the placebo group vs 52 [11%] in the trebananib group), neutropenia (40 [9%] vs 26 [6%]), and abdominal pain (21 [5%] vs 22 [5%]). We recorded serious adverse events in 125 (28%) patients in the placebo group and 159 (34%) patients in the trebananib group. There was a difference of 2% or less in class-specific adverse events associated with anti-VEGF therapy (hypertension, proteinuria, wound-healing complications, thrombotic events, gastrointestinal perforations), except bleeding, which was more common in the placebo group than in the trebananib group (75 [17%] vs 46 [10%]). INTERPRETATION Inhibition of angiopoietins 1 and 2 with trebananib provided a clinically meaningful prolongation in progression-free survival. This non-VEGF anti-angiogenesis option for women with recurrent epithelial ovarian cancer should be investigated in other settings and in combination with additional agents. Although oedema was increased, typical anti-VEGF associated adverse events were not prominent. FUNDING Amgen.
Collapse
Affiliation(s)
- Bradley J Monk
- Creighton University School of Medicine and University of Arizona Cancer Center at St Joseph's Hospital, Phoenix, AZ, USA.
| | - Andrés Poveda
- Fundacion Instituto Valenciano de Oncología, Valencia, Spain
| | | | | | - Keiichi Fujiwara
- Saitama Medical University International Medical Center, Hidaka-Shi, Japan
| | | | - Ana Oaknin
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | - Arija Brize
- Riga Eastern Clinical University Hospital, Riga, Latvia
| | - Michel Fabbro
- Regional Cancer Institute Montpellier, Montpellier, France
| | | | - Aristotelis Bamias
- Alexandra Hospital, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | - Amit M Oza
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|