1
|
Oliveira VQ, Santos LC, Teixeira SC, Correia TML, Andrade LOSB, Gimenes SNC, Colombini M, Marques LM, Jiménez-Charris E, Freitas-de-Sousa LA, Silva MJB, Magalhães Gusmão ACMD, Ferro EAV, Clissa PB, Melo Rodrigues VD, Lopes DS. Antiangiogenic properties of BthMP, a P-I metalloproteinase from Bothrops moojeni snake venom by VEGF pathway in endothelial cells. Biochem Biophys Res Commun 2024; 706:149748. [PMID: 38460450 DOI: 10.1016/j.bbrc.2024.149748] [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: 11/21/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Angiogenesis is a process that is controlled by a delicate combination of proangiogenic and antiangiogenic molecules and can be disrupted in various illnesses, including cancer. Non-cancerous diseases can also have an abnormal or insufficient vascular growth, inflammation and hypoxia, which exacerbate angiogenesis. These conditions include atherosclerosis, psoriasis, endometriosis, asthma, obesity and AIDS. Based on that, the present work assessed the in vitro and ex vivo antiangiogenic properties stemming from BthMP, a P-I metalloproteinase from Bothrops moojeni snake venom, via the VEGF pathway. BthMP at a concentration of 5 and 40 μg/mL showed no toxicity to endothelial cells (HUVEC) in the MTT assay and was not able to induce necrosis and colony proliferation. Interestingly, BthMP inhibited adhesion, migration and invasion of HUVECs in Matrigel and arrested in vitro angiogenesis by reducing the average number of nodules in toxin-treated cells by 9.6 and 17.32 at 5 and 40 μg/mL, respectively, and the number of tubules by 15.9 at 5 μg/mL and 21.6 at 40 μg/mL in a VEGF-dependent way, an essential proangiogenic property. Furthermore, BthMP inhibited the occurrence of the angiogenic process in an ex vivo aortic ring test by decreasing new vessel formation by 52% at 5 μg/mL and by 66% at 40 μg/mL and by increasing the expression of an antiangiogenic gene, SFLT-1, and decreasing the expression of the proangiogenic genes VEGFA and ANGPT-1. Finally, this toxin reduces the production of nitric oxide, a marker that promotes angiogenesis and VEGF modulation, and decreases the protein expression of VEGFA in the supernatant of the HUVEC culture by about 30 %. These results suggest that BthMP has a promising antiangiogenic property and proves to be a biotechnological mechanism for understanding the antiangiogenic responses induced by snake venom metalloproteinases, which could be applied to a variety of diseases that exhibit an imbalance of angiogenesis mechanisms.
Collapse
Affiliation(s)
- Vinícius Queiroz Oliveira
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | - Luísa Carregosa Santos
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | - Samuel Cota Teixeira
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil.
| | | | | | | | - Mônica Colombini
- Laboratory of Immunopathology, Institute of Butantan, São Paulo, SP, Brazil
| | - Lucas Miranda Marques
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | | | | | - Marcelo José Barbosa Silva
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | | | - Eloisa Amália Vieira Ferro
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | | | - Veridiana de Melo Rodrigues
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia (UFU), Uberlândia-MG, Brazil
| | - Daiana Silva Lopes
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil.
| |
Collapse
|
2
|
Qi S, Deng S, Lian Z, Yu K. Novel Drugs with High Efficacy against Tumor Angiogenesis. Int J Mol Sci 2022; 23:6934. [PMID: 35805939 PMCID: PMC9267017 DOI: 10.3390/ijms23136934] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis is involved in physiological and pathological processes in the body. Tumor angiogenesis is a key factor associated with tumor growth, progression, and metastasis. Therefore, there is great interest in developing antiangiogenic strategies. Hypoxia is the basic initiating factor of tumor angiogenesis, which leads to the increase of vascular endothelial growth factor (VEGF), angiopoietin (Ang), hypoxia-inducible factor (HIF-1), etc. in hypoxic cells. The pathways of VEGF and Ang are considered to be critical steps in tumor angiogenesis. A number of antiangiogenic drugs targeting VEGF/VEGFR (VEGF receptor) or ANG/Tie2, or both, are currently being used for cancer treatment, or are still in various stages of clinical development or preclinical evaluation. This article aims to review the mechanisms of angiogenesis and tumor angiogenesis and to focus on new drugs and strategies for the treatment of antiangiogenesis. However, antitumor angiogenic drugs alone may not be sufficient to eradicate tumors. The molecular chaperone heat shock protein 90 (HSP90) is considered a promising molecular target. The VEGFR system and its downstream signaling molecules depend on the function of HSP90. This article also briefly introduces the role of HSP90 in angiogenesis and some HSP90 inhibitors.
Collapse
Affiliation(s)
- Shiyu Qi
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Shoulong Deng
- National Health Commission (NHC) of China Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China;
| | - Zhengxing Lian
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Kun Yu
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| |
Collapse
|
3
|
Harry JA, Ormiston ML. Novel Pathways for Targeting Tumor Angiogenesis in Metastatic Breast Cancer. Front Oncol 2021; 11:772305. [PMID: 34926282 PMCID: PMC8678517 DOI: 10.3389/fonc.2021.772305] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is the most common cancer affecting women and is the second leading cause of cancer related death worldwide. Angiogenesis, the process of new blood vessel development from pre-existing vasculature, has been implicated in the growth, progression, and metastasis of cancer. Tumor angiogenesis has been explored as a key therapeutic target for decades, as the blockade of this process holds the potential to reduce the oxygen and nutrient supplies that are required for tumor growth. However, many existing anti-angiogenic approaches, such as those targeting Vascular Endothelial Growth Factor, Notch, and Angiopoietin signaling, have been associated with severe side-effects, limited survival advantage, and enhanced cancer regrowth rates. To address these setbacks, alternative pathways involved in the regulation of tumor angiogenesis are being explored, including those involving Bone Morphogenetic Protein-9 signaling, the Sonic Hedgehog pathway, Cyclooxygenase-2, p38-mitogen-activated protein kinase, and Chemokine Ligand 18. This review article will introduce the concept of tumor angiogenesis in the context of breast cancer, followed by an overview of current anti-angiogenic therapies, associated resistance mechanisms and novel therapeutic targets.
Collapse
Affiliation(s)
- Jordan A Harry
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Mark L Ormiston
- Department of Medicine, Queen's University, Kingston, ON, Canada.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.,Department of Surgery, Queen's University, Kingston, ON, Canada
| |
Collapse
|
4
|
Roy S, Banerjee P, Ekser B, Bayless K, Zawieja D, Alpini G, Glaser SS, Chakraborty S. Targeting Lymphangiogenesis and Lymph Node Metastasis in Liver Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2052-2063. [PMID: 34509441 PMCID: PMC8647434 DOI: 10.1016/j.ajpath.2021.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 12/17/2022]
Abstract
Increased lymphangiogenesis and lymph node metastasis, the important prognostic indicators of aggressive hepatobiliary malignancies such as hepatocellular cancer and cholangiocarcinoma, are associated with poor patient outcome. The liver produces 25% to 50% of total lymphatic fluid in the body and has a dense network of lymphatic vessels. The lymphatic system plays critical roles in fluid homeostasis and inflammation and immune response. Yet, lymphatic vessel alterations and function are grossly understudied in the context of liver pathology. Expansion of the lymphatic network has been documented in clinical samples of liver cancer; and although largely overlooked in the liver, tumor-induced lymphangiogenesis is an important player, increasing tumor metastasis in several cancers. This review aims to provide a detailed perspective on the current knowledge of alterations in the hepatic lymphatic system during liver malignancies, as well as various molecular signaling mechanisms and growth factors that may provide future targets for therapeutic intervention. In addition, the review also addresses current mechanisms and bottlenecks for effective therapeutic targeting of tumor-associated lymphangiogenesis.
Collapse
Affiliation(s)
- Sukanya Roy
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Priyanka Banerjee
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kayla Bayless
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - David Zawieja
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University, Indianapolis, Indiana; Richard L Roudebush VA Medical Center, Indianapolis, Indiana
| | - Shannon S Glaser
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas.
| |
Collapse
|
5
|
Molecular Mechanisms of Resistance to Immunotherapy and Antiangiogenic Treatments in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13235981. [PMID: 34885091 PMCID: PMC8656474 DOI: 10.3390/cancers13235981] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype arising from renal cell carcinomas. This tumor is characterized by a predominant angiogenic and immunogenic microenvironment that interplay with stromal, immune cells, and tumoral cells. Despite the obscure prognosis traditionally related to this entity, strategies including angiogenesis inhibition with tyrosine kinase inhibitors (TKIs), as well as the enhancement of the immune system with the inhibition of immune checkpoint proteins, such as PD-1/PDL-1 and CTLA-4, have revolutionized the treatment landscape. This approach has achieved a substantial improvement in life expectancy and quality of life from patients with advanced ccRCC. Unfortunately, not all patients benefit from this success as most patients will finally progress to these therapies and, even worse, approximately 5 to 30% of patients will primarily progress. In the last few years, preclinical and clinical research have been conducted to decode the biological basis underlying the resistance mechanisms regarding angiogenic and immune-based therapy. In this review, we summarize the insights of these molecular alterations to understand the resistance pathways related to the treatment with TKI and immune checkpoint inhibitors (ICIs). Moreover, we include additional information on novel approaches that are currently under research to overcome these resistance alterations in preclinical studies and early phase clinical trials.
Collapse
|
6
|
Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology 2021; 3:e27048. [PMID: 24605265 PMCID: PMC3937194 DOI: 10.4161/onci.27048] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/01/2013] [Indexed: 02/06/2023] Open
Abstract
In 1997, for the first time in history, a monoclonal antibody (mAb), i.e., the chimeric anti-CD20 molecule rituximab, was approved by the US Food and Drug Administration for use in cancer patients. Since then, the panel of mAbs that are approved by international regulatory agencies for the treatment of hematopoietic and solid malignancies has not stopped to expand, nowadays encompassing a stunning amount of 15 distinct molecules. This therapeutic armamentarium includes mAbs that target tumor-associated antigens, as well as molecules that interfere with tumor-stroma interactions or exert direct immunostimulatory effects. These three classes of mAbs exert antineoplastic activity via distinct mechanisms, which may or may not involve immune effectors other than the mAbs themselves. In previous issues of OncoImmunology, we provided a brief scientific background to the use of mAbs, all types confounded, in cancer therapy, and discussed the results of recent clinical trials investigating the safety and efficacy of this approach. Here, we focus on mAbs that primarily target malignant cells or their interactions with stromal components, as opposed to mAbs that mediate antineoplastic effects by activating the immune system. In particular, we discuss relevant clinical findings that have been published during the last 13 months as well as clinical trials that have been launched in the same period to investigate the therapeutic profile of hitherto investigational tumor-targeting mAbs.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|
7
|
Mooi J, Chionh F, Savas P, Da Gama Duarte J, Chong G, Brown S, Wong R, Price TJ, Wann A, Skrinos E, Mariadason JM, Tebbutt NC. Dual Antiangiogenesis Agents Bevacizumab Plus Trebananib, without Chemotherapy, in First-line Treatment of Metastatic Colorectal Cancer: Results of a Phase II Study. Clin Cancer Res 2021; 27:2159-2167. [PMID: 33514526 DOI: 10.1158/1078-0432.ccr-20-2714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/13/2020] [Accepted: 01/27/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess the efficacy and safety of dual antiangiogenesis agents, bevacizumab plus trebananib, without chemotherapy, in first-line treatment of metastatic colorectal cancer (mCRC). PATIENTS AND METHODS This open-label phase II study enrolled patients with unresectable mCRC with no prior systemic treatment. All patients received bevacizumab 7.5 mg/kg 3-weekly and trebananib 15 mg/kg weekly. The primary endpoint was disease control [stable disease, partial response (PR), or complete response (CR)] at 6 months (DC6m). Secondary endpoints included toxicity, overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). Exploratory biomarkers in plasma angiogenesis-related proteins, tumor gene expression, and plasma antibodies to tumor antigens were examined. RESULTS Forty-five patients were enrolled from four Australian sites. DC6m was 63% [95% confidence interval (CI), 47-77]. ORR was 17% (95% CI, 7-32), comprising of seven PRs. Median duration of response was 20 months (range, 10-48 months). Median PFS was 8.4 months and median OS 31.4 months. Grade 1-2 peripheral edema and joint-related symptoms were common. Overall incidence of grade 3-4 adverse events (AE) of any type was 33% (n = 15). Expected AEs of bevacizumab treatment did not appear to be increased by the addition of trebananib. CONCLUSIONS In a first-line mCRC population, the dual antiangiogenic combination, bevacizumab plus trebananib, without chemotherapy, was efficacious with durable responses. The toxicity profile of the combination was manageable and did not exceed that expected with bevacizumab +/- chemotherapy. Exploratory biomarker results raise the hypothesis that the antiangiogenic combination may enable the antitumor immune response in immunotolerant colorectal cancer.
Collapse
Affiliation(s)
- Jennifer Mooi
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Fiona Chionh
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Peter Savas
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jessica Da Gama Duarte
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University, Melbourne, Victoria, Australia
| | - Geoffrey Chong
- University of Melbourne, Melbourne, Victoria, Australia.,Ballarat Regional Integrated Cancer Centre, Ballarat, Victoria, Australia
| | - Stephen Brown
- Ballarat Regional Integrated Cancer Centre, Ballarat, Victoria, Australia
| | - Rachel Wong
- Eastern Health, Box Hill, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | - Timothy J Price
- The Queen Elizabeth Hospital, Adelaide, South Australia, Australia.,University of Adelaide, Adelaide, South Australia, Australia
| | - Alysson Wann
- University of Melbourne, Melbourne, Victoria, Australia
| | - Effie Skrinos
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - John M Mariadason
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University, Melbourne, Victoria, Australia
| | - Niall C Tebbutt
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia. .,University of Melbourne, Melbourne, Victoria, Australia.,Austin Health, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Malapelle U, Rossi A. Emerging angiogenesis inhibitors for non-small cell lung cancer. Expert Opin Emerg Drugs 2019; 24:71-81. [DOI: 10.1080/14728214.2019.1619696] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Antonio Rossi
- Division of Medical Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (Foggia), Italy
| |
Collapse
|
9
|
Jiang W, Ji M. Receptor tyrosine kinases in PI3K signaling: The therapeutic targets in cancer. Semin Cancer Biol 2019; 59:3-22. [PMID: 30943434 DOI: 10.1016/j.semcancer.2019.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 03/09/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway, one of the most commonly activated signaling pathways in human cancers, plays a crucial role in the regulation of cell proliferation, differentiation, and survival. This pathway is usually activated by receptor tyrosine kinases (RTKs), whose constitutive and aberrant activation is via gain-of-function mutations, chromosomal rearrangement, gene amplification and autocrine. Blockage of PI3K pathway by targeted therapy on RTKs with tyrosine kinases inhibitors (TKIs) and monoclonal antibodies (mAbs) has achieved great progress in past decades; however, there still remain big challenges during their clinical application. In this review, we provide an overview about the most frequently encountered alterations in RTKs and focus on current therapeutic agents developed to counteract their aberrant functions, accompanied with discussions of two major challenges to the RTKs-targeted therapy in cancer - resistance and toxicity.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Meiju Ji
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| |
Collapse
|
10
|
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
|
11
|
Evelhoch JL. In vivo MR in the drug pipeline. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2018; 292:117-128. [PMID: 29716831 DOI: 10.1016/j.jmr.2018.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/15/2018] [Accepted: 04/23/2018] [Indexed: 06/08/2023]
Abstract
The application of in vivo magnetic resonance (MR) in drug development has several requirements that differ significantly from most applications primarily because the biopharmaceutical industry must develop new safe and effective drugs more quickly and at lower cost in a highly regulated environment. In vivo MR is recognized as a useful method to provide biomarkers for target engagement, treatment response, safety and mechanism of action that can be translated between animal and clinical studies. Thus, it has the potential to help identify drugs that are more likely to be safe and effective earlier in the process of drug development, which may help reduce the time and money required to get new drugs to patients with an unmet medical need. A brief introduction of how novel drugs are discovered and developed, what drives the biopharmaceutical industry's interest in using biomarkers and what it takes to use MR as a biomarker to support drug development, including regulatory concerns, provides context for understanding what makes the application of in vivo MR in drug development different from most others. Exploration of three programs (trebananib, ZD6126 and axitinib) using dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) in the development of antivascular agents provides insight into how in vivo MR biomarkers impact drug discovery and development and the limitations of biomarkers.
Collapse
Affiliation(s)
- Jeffrey L Evelhoch
- Merck & Co., Inc., 770 Sumneytown Pike, WP 42-211, West Point, PA 19486, USA.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Jiang Y, Fan H, Jiang Y, Song G, Wang F, Li X, Li G. Efficacy and safety of FOLFIRI and biotherapy versus FOLFIRI alone for metastatic colorectal cancer patients: A meta-analysis. Medicine (Baltimore) 2017; 96:e8767. [PMID: 29310351 PMCID: PMC5728752 DOI: 10.1097/md.0000000000008767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Previous randomized controlled trials (RCTs) and meta-analyses have demonstrated the useless of FOLFIRI alone for previously treated patients with metastatic colorectal cancer (mCRC). The role of FOLFIRI regimen combined with biological therapy is unknown. The purpose of this meta-analysis is to evaluate the efficacy and safety of combining biological therapy with chemotherapy in previously treated patients with mCRC. METHODS MEDLINE, EMBASE, Web of Science, Cochrane library, and ClinicalTrials.gov were searched. Eligible studies were RCTs that evaluated the efficacy and safety of the FOLFIRI regimen with or without biological therapy for previously treated patients with mCRC. The hazard ratio (HR) or risk ratio (RR) with 95% confidence interval was estimated. The Chi-squared and I-squared tests were used to assess the statistical heterogeneity. RESULTS The literature search identified 7 RCTs that met the inclusion criteria for the meta-analysis, and 3680 patients with mCRC were included. The meta-analysis showed that combined therapy was associated with a significant improved progression-free survival (PFS) (HR = 0.78, 95% CI = 0.72-0.85, P < .001), overall survival (OS) (HR = 0.84, 95% CI = 0.77-0.92, P < .001), and overall response rate (ORR) (RR = 1.70, 95% CI = 1.25-2.31, P = .001). Sensitivity analysis suggested that combined therapy versus FOLFIRI alone might increase the risk of Grade 3/4 AEs. CONCLUSION The addition of biological therapy to the FOLFIRI regimen improved the PFS, OS, and ORR compared with FOLFIRI alone for previously treated patients with mCRC. Long-term survival outcomes are warranted.
Collapse
Affiliation(s)
| | - Hui Fan
- Department of Radiation Oncology
| | - Yongmei Jiang
- Department of Neurological Rehabilitation, the Second Affiliated Hospital of Dalian Medical University
| | - Guirong Song
- School of Public Health, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Feng Wang
- School of Public Health, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Xiaofeng Li
- School of Public Health, Dalian Medical University, Dalian, Liaoning, P.R. China
| | | |
Collapse
|
15
|
Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 2017; 20:409-426. [PMID: 28660302 DOI: 10.1007/s10456-017-9562-9] [Citation(s) in RCA: 924] [Impact Index Per Article: 132.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/21/2017] [Indexed: 12/27/2022]
Abstract
Tumor blood vessels are a key target for cancer therapeutic management. Tumor cells secrete high levels of pro-angiogenic factors which contribute to the creation of an abnormal vascular network characterized by disorganized, immature and permeable blood vessels, resulting in poorly perfused tumors. The hypoxic microenvironment created by impaired tumor perfusion can promote the selection of more invasive and aggressive tumor cells and can also impede the tumor-killing action of immune cells. Furthermore, abnormal tumor perfusion also reduces the diffusion of chemotherapeutic drugs and radiotherapy efficiency. To fight against this defective phenotype, the normalization of the tumor vasculature has emerged as a new therapeutic strategy. Vascular normalization, by restoring proper tumor perfusion and oxygenation, could limit tumor cell invasiveness and improve the effectiveness of anticancer treatments. In this review, we investigate the mechanisms involved in tumor angiogenesis and describe strategies used to achieve vascular normalization.
Collapse
|
16
|
Ronca R, Benkheil M, Mitola S, Struyf S, Liekens S. Tumor angiogenesis revisited: Regulators and clinical implications. Med Res Rev 2017. [PMID: 28643862 DOI: 10.1002/med.21452] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since Judah Folkman hypothesized in 1971 that angiogenesis is required for solid tumor growth, numerous studies have been conducted to unravel the angiogenesis process, analyze its role in primary tumor growth, metastasis and angiogenic diseases, and to develop inhibitors of proangiogenic factors. These studies have led in 2004 to the approval of the first antiangiogenic agent (bevacizumab, a humanized antibody targeting vascular endothelial growth factor) for the treatment of patients with metastatic colorectal cancer. This approval launched great expectations for the use of antiangiogenic therapy for malignant diseases. However, these expectations have not been met and, as knowledge of blood vessel formation accumulates, many of the original paradigms no longer hold. Therefore, the regulators and clinical implications of angiogenesis need to be revisited. In this review, we discuss recently identified angiogenesis mediators and pathways, new concepts that have emerged over the past 10 years, tumor resistance and toxicity associated with the use of currently available antiangiogenic treatment and potentially new targets and/or approaches for malignant and nonmalignant neovascular diseases.
Collapse
Affiliation(s)
- Roberto Ronca
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mohammed Benkheil
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Leuven, Belgium
| | - Stefania Mitola
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Leuven, Belgium
| | - Sandra Liekens
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Leuven, Belgium
| |
Collapse
|
17
|
Abou-Alfa GK, Blanc JF, Miles S, Ganten T, Trojan J, Cebon J, Liem AK, Lipton L, Gupta C, Wu B, Bass M, Hollywood E, Ma J, Bradley M, Litten J, Saltz LB. Phase II Study of First-Line Trebananib Plus Sorafenib in Patients with Advanced Hepatocellular Carcinoma. Oncologist 2017; 22:780-e65. [PMID: 28592620 PMCID: PMC5507650 DOI: 10.1634/theoncologist.2017-0058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/05/2017] [Indexed: 12/22/2022] Open
Abstract
Lessons Learned. Trebananib leveraging anti‐angiogenic mechanism that is distinct from the classic sorafenib anti‐vascular endothelial growth factor inhibition did not demonstrate improved progression‐free survival at 4 months in patients with advanced hepatocellular carcinoma (HCC). In support of previously reported high Ang‐2 levels’ association with poor outcome in HCC for patients, trebananib treatment with lower baseline Ang‐2 at study entry was associated with improved overall survival to 22 months and may suggest future studies to be performed within the context of low baseline Ang‐2.
Background. Ang‐1 and Ang‐2 are angiopoietins thought to promote neovascularization via activation of the Tie‐2 angiopoietin receptor. Trebananib sequesters Ang‐1 and Ang‐2, preventing interaction with the Tie‐2 receptor. Trebananib plus sorafenib combination has acceptable toxicity. Elevated Ang‐2 levels are associated with poor prognosis in hepatocellular carcinoma (HCC). Methods. Patients with HCC, Eastern Cooperative Oncology Group ≤2, and Childs‐Pugh A received IV trebananib at 10 mg/kg or 15 mg/kg weekly plus sorafenib 400 mg orally twice daily. The study was planned for ≥78% progression‐free survival (PFS) rate at 4 months relative to 62% for sorafenib historical control (power = 80% α = 0.20). Secondary endpoints included safety, tolerability, overall survival (OS), and multiple biomarkers, including serum Ang‐2. Results. Thirty patients were enrolled sequentially in each of the two nonrandomized cohorts. Demographics were comparable between the two arms and the historical controls. PFS rates at 4 months were 57% and 54% on the 10 mg/kg and 15 mg/kg trebananib cohorts, respectively. Median OS was 17 and 11 months, respectively. Grade 3 and above events noted in ≥10% of patients included fatigue, hypertension, diarrhea, liver failure, palmar‐plantar erythrodysesthesia syndrome, dyspnea, and hypophosphatemia. One death was due to hepatic failure. Serum Ang‐2 dichotomized at the median was associated with improved OS in both cohorts. Conclusion. There was no improvement in PFS rate at 4 months in either cohort, when compared with sorafenib historical control.
Collapse
Affiliation(s)
- Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | | | - Steven Miles
- Cedars Sinai Hospital, Los Angeles, California, USA
| | - Tom Ganten
- University of Heidelberg, Heidelberg, Germany
| | - Jörg Trojan
- Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Jonathan Cebon
- Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Victoria, Australia
| | - Andre K Liem
- Translational Oncology Research International, Long Beach, California, USA
| | - Lara Lipton
- Western Hospital, Footscray, Victoria, Australia
| | | | | | | | - Ellen Hollywood
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jennifer Ma
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | - Leonard B Saltz
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
18
|
Pecqueux M, Liebetrau I, Werft W, Dienemann H, Muley T, Pfannschmidt J, Müssle B, Rahbari N, Schölch S, Büchler MW, Weitz J, Reissfelder C, Kahlert C. A Comprehensive MicroRNA Expression Profile of Liver and Lung Metastases of Colorectal Cancer with Their Corresponding Host Tissue and Its Prognostic Impact on Survival. Int J Mol Sci 2016; 17:ijms17101755. [PMID: 27775664 PMCID: PMC5085780 DOI: 10.3390/ijms17101755] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/04/2016] [Accepted: 10/12/2016] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs are small non-coding RNAs with a length of 18–25 nucleotides. They can regulate tumor invasion and metastasis by changing the expression and translation of their target mRNAs. Their expression is substantially altered in colorectal cancer cells as well as in the adjacent tumor-associated stroma. Both of these compartments have a mutual influence on tumor progression. In the development of metastases, cancer cells initially interact with the host tissue. Therefore, compartment-specific expression signatures of these three locations—tumor, associated stroma, and host tissue—can provide new insights into the complex tumor biology of colorectal cancer. Frozen tissue samples of colorectal liver (n = 25) and lung metastases (n = 24) were laser microdissected to separate tumor cells and the adjacent tumor-associated stroma cells. Additionally, normal lung and liver tissue was collected from the same patients. We performed a microarray analysis in four randomly selected liver metastases and four randomly selected lung metastases, analyzing a total of 939 human miRNAs. miRNAs with a significant change >2-fold between the tumor, tumor stroma, and host tissue were analyzed in all samples using RT-qPCR (11 miRNAs) and correlated with the clinical data. We found a differential expression of several miRNAs between the tumor, the tumor-associated stroma, and the host tissue compartment. When comparing liver and lung metastases, miR-194 showed a 1.5-fold; miR-125, miR-127, and miR-192 showed a 2.5-fold; miR-19 and miR-215 a 3-fold; miR-145, miR-199-3, and miR-429 a 5-fold; miR-21 a 7-fold; and, finally, miR-199-5 a 12.5-fold downregulation in liver metastases compared to lung metastases. Furthermore miR-19, miR-125, miR-127, miR-192, miR-194, miR-199-5, and miR-215 showed a significant upregulation in the normal liver tissue compared to the normal lung tissue. Univariate analysis identified an association of poor survival with the expression of miR-125 (p = 0.05), miR-127 (p = 0.001), miR-145 (p = 0.005), miR-192 (p = 0.015), miR-194 (0.003), miR-199-5 (p = 0.008), miR-215 (p < 0.001), and miR-429 (p = 0.03) in the host liver tissue of the liver metastases. Colorectal liver and lung metastases have a unique miRNA expression profile. miRNA expression in the host tissue of colorectal liver metastases seems to be able to influence tumor progression and survival. These findings can be used in the development of tailored therapies.
Collapse
Affiliation(s)
- Mathieu Pecqueux
- Department of General, Visceral and Thoracic Surgery, University of Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | | | - Wiebke Werft
- German Cancer Research Center, Division of Biostatistics, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Hendrik Dienemann
- Department of Thoracic Surgery, Thoraxklinik am Universitätsklinikum Heidelberg, Amalienstrasse 5, 69126 Heidelberg, Germany.
| | - Thomas Muley
- Department of Thoracic Surgery, Thoraxklinik am Universitätsklinikum Heidelberg, Amalienstrasse 5, 69126 Heidelberg, Germany.
| | - Joachim Pfannschmidt
- Department of Thoracic Surgery, Helios Clinic Emil von Behring, Walterhöferstraße 11, 14165 Berlin, Germany.
| | - Benjamin Müssle
- Department of General, Visceral and Thoracic Surgery, University of Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Nuh Rahbari
- Department of General, Visceral and Thoracic Surgery, University of Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Sebastian Schölch
- Department of General, Visceral and Thoracic Surgery, University of Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Markus W Büchler
- Department of Surgery at Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Jürgen Weitz
- Department of General, Visceral and Thoracic Surgery, University of Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Christoph Reissfelder
- Department of General, Visceral and Thoracic Surgery, University of Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Christoph Kahlert
- Department of General, Visceral and Thoracic Surgery, University of Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| |
Collapse
|
19
|
Tabit CE, Chen P, Kim GH, Fedson SE, Sayer G, Coplan MJ, Jeevanandam V, Uriel N, Liao JK. Elevated Angiopoietin-2 Level in Patients With Continuous-Flow Left Ventricular Assist Devices Leads to Altered Angiogenesis and Is Associated With Higher Nonsurgical Bleeding. Circulation 2016; 134:141-52. [PMID: 27354285 DOI: 10.1161/circulationaha.115.019692] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 05/18/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND Nonsurgical bleeding is the most common adverse event in patients with continuous-flow left ventricular assist devices (LVADs) and is caused by arteriovenous malformations. We hypothesized that deregulation of an angiogenic factor, angiopoietin-2 (Ang-2), in patients with LVADs leads to increased angiogenesis and higher nonsurgical bleeding. METHODS Ang-2 and thrombin levels were measured by ELISA and Western blotting, respectively, in blood samples from 101 patients with heart failure, LVAD, or orthotopic heart transplantation. Ang-2 expression in endothelial biopsy was quantified by immunofluorescence. Angiogenesis was determined by in vitro tube formation from serum from each patient with or without Ang-2-blocking antibody. Ang-2 gene expression was measured by reverse transcription-polymerase chain reaction in endothelial cells incubated with plasma from each patient with or without the thrombin receptor blocker vorapaxar. RESULTS Compared with patients with heart failure or those with orthotopic heart transplantation, serum levels and endothelial expression of Ang-2 were higher in LVAD patients (P=0.001 and P<0.001, respectively). This corresponded to an increased angiogenic potential of serum from patients with LVADs (P<0.001), which was normalized with Ang-2 blockade. Furthermore, plasma from LVAD patients contained higher amounts of thrombin (P=0.003), which was associated with activation of the contact coagulation system. Plasma from LVAD patients induced more Ang-2 gene expression in endothelial cells (P<0.001), which was reduced with thrombin receptor blockade (P=0.013). LVAD patients with Ang-2 levels above the mean (12.32 ng/mL) had more nonsurgical bleeding events compared with patients with Ang-2 levels below the mean (P=0.003). CONCLUSIONS Our findings indicate that thrombin-induced Ang-2 expression in LVAD patients leads to increased angiogenesis in vitro and may be associated with higher nonsurgical bleeding events. Ang-2 therefore may contribute to arteriovenous malformation formation and subsequent bleeding in LVAD patients.
Collapse
Affiliation(s)
- Corey E Tabit
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - Phetcharat Chen
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - Gene H Kim
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - Savitri E Fedson
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - Gabriel Sayer
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - Mitchell J Coplan
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - Valluvan Jeevanandam
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - Nir Uriel
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.)
| | - James K Liao
- From the Department of Medicine, Section of Cardiology (C.E.T., P.C., G.H.K., G.S., M.J.C., N.U., J.K.L.) and Department of Surgery, Section of Cardiac and Thoracic Surgery (V.T.), University of Chicago, IL; and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX (S.E.F.).
| |
Collapse
|
20
|
Stachura J, Wachowska M, Kilarski WW, Güç E, Golab J, Muchowicz A. The dual role of tumor lymphatic vessels in dissemination of metastases and immune response development. Oncoimmunology 2016; 5:e1182278. [PMID: 27622039 PMCID: PMC5006909 DOI: 10.1080/2162402x.2016.1182278] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/16/2016] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
Lymphatic vasculature plays a crucial role in the immune response, enabling transport of dendritic cells (DCs) and antigens (Ags) into the lymph nodes. Unfortunately, the lymphatic system has also a negative role in the progression of cancer diseases, by facilitating the metastatic spread of many carcinomas to the draining lymph nodes. The lymphatics can promote antitumor immune response as well as tumor tolerance. Here, we review the role of lymphatic endothelial cells (LECs) in tumor progression and immunity and mechanism of action in the newest anti-lymphatic therapies, including photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Joanna Stachura
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland; Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Wachowska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland; Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland
| | - Witold W Kilarski
- Institute for Molecular Engineering, University of Chicago , Chicago, IL, USA
| | - Esra Güç
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh , Edinburgh, UK
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw , Warsaw, Poland
| | | |
Collapse
|
21
|
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
|
22
|
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
|
23
|
A phase I trial of ANG1/2-Tie2 inhibitor trebaninib (AMG386) and temsirolimus in advanced solid tumors (PJC008/NCI♯9041). Invest New Drugs 2015; 34:104-11. [PMID: 26686201 PMCID: PMC4718956 DOI: 10.1007/s10637-015-0313-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/03/2015] [Indexed: 11/22/2022]
Abstract
Background There is crosstalk between the ANG-Tie2 and the PI3K/Akt/mTOR pathways. Combined ANG1/2 and mTOR blockade may have additive anti-cancer activity. The combination of trebananib, an inhibitor of ANG1/2-Tie2 interaction, with temsirolimus was evaluated in patients with advanced solid tumors to determine tolerability, maximum tolerated dose (MTD), and preliminary antitumor activity. Methods Patients were enrolled using 3 + 3 design, and were given intravenous trebananib and temsirolimus on Day 1, 8, 15 and 22 of a 28-day cycle. Dose limiting toxicities (DLTs) were evaluated during cycle 1. Peripheral blood was collected for evaluation of Tie2-expressing monocytes (TEMs) and thymidine phosphorylase (TP). Sparse pharmacokinetic (PK) sampling for trebananib drug levels was performed on Day 1 and 8 of cycle 2. Results Twenty-one patients were enrolled, 6 at dose level (DL) 1, 7 at DL −1, and 8 at DL −2. No effect of temsirolimus on trebananib PK was observed. The most common treatment-related adverse events (AEs) were: fatigue (81 %), edema (62 %), anorexia (57 %), nausea (52 %), rash (43 %) and mucositis (43 %). The most common grade ≥ 3 AEs included lymphopenia (28 %) and fatigue (28 %). The MTD was exceeded at DL-2. Of 18 response evaluable patients, 1 partial response was observed (ER+/HER2−/PIK3CA mutant breast cancer) and 4 patients had prolonged SD ≥ 24 weeks. No correlation with clinical benefit was observed with change in number TEMs or TP expression in TEMs with treatment. Conclusions The MTD was exceeded at trebananib 10 mg/kg weekly and temsirolimus 20 mg weekly, with frequent overlapping toxicities including fatigue, edema, and anorexia.
Collapse
|
24
|
Paller CJ, Bradbury PA, Ivy SP, Seymour L, LoRusso PM, Baker L, Rubinstein L, Huang E, Collyar D, Groshen S, Reeves S, Ellis LM, Sargent DJ, Rosner GL, LeBlanc ML, Ratain MJ. Design of phase I combination trials: recommendations of the Clinical Trial Design Task Force of the NCI Investigational Drug Steering Committee. Clin Cancer Res 2015; 20:4210-7. [PMID: 25125258 DOI: 10.1158/1078-0432.ccr-14-0521] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Anticancer drugs are combined in an effort to treat a heterogeneous tumor or to maximize the pharmacodynamic effect. The development of combination regimens, while desirable, poses unique challenges. These include the selection of agents for combination therapy that may lead to improved efficacy while maintaining acceptable toxicity, the design of clinical trials that provide informative results for individual agents and combinations, and logistic and regulatory challenges. The phase I trial is often the initial step in the clinical evaluation of a combination regimen. In view of the importance of combination regimens and the challenges associated with developing them, the Clinical Trial Design (CTD) Task Force of the National Cancer Institute Investigational Drug Steering Committee developed a set of recommendations for the phase I development of a combination regimen. The first two recommendations focus on the scientific rationale and development plans for the combination regimen; subsequent recommendations encompass clinical design aspects. The CTD Task Force recommends that selection of the proposed regimens be based on a biologic or pharmacologic rationale supported by clinical and/or robust and validated preclinical evidence, and accompanied by a plan for subsequent development of the combination. The design of the phase I clinical trial should take into consideration the potential pharmacokinetic and pharmacodynamic interactions as well as overlapping toxicity. Depending on the specific hypothesized interaction, the primary endpoint may be dose optimization, pharmacokinetics, and/or pharmacodynamics (i.e., biomarker).
Collapse
Affiliation(s)
- Channing J Paller
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore
| | | | - S Percy Ivy
- National Cancer Institute, Bethesda, Maryland
| | - Lesley Seymour
- NCIC Clinical Trials Group, Queen's University, Kingston, Ontario, Canada
| | | | | | | | - Erich Huang
- National Cancer Institute, Bethesda, Maryland
| | | | - Susan Groshen
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | | | - Lee M Ellis
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Gary L Rosner
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore
| | - Michael L LeBlanc
- Fred Hutchinson Cancer Research Center, Cancer Research and Biostatistics, Seattle, Washington
| | - Mark J Ratain
- The University of Chicago, Department of Medicine, Chicago, Illinois; and
| |
Collapse
|
25
|
Cañadas I, Taus Á, Villanueva X, Arpí O, Pijuan L, Rodríguez Y, Menéndez S, Mojal S, Rojo F, Albanell J, Rovira A, Arriola E. Angiopoietin-2 is a negative prognostic marker in small cell lung cancer. Lung Cancer 2015; 90:302-6. [PMID: 26428740 DOI: 10.1016/j.lungcan.2015.09.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/14/2015] [Accepted: 09/22/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is a highly lethal disease due to its chemorefractory nature after initial treatment. Angiogenesis plays an important role in tumor growth, metastasis and chemoresistance. We hypothesized that angiogenesis could predict chemoresistance in SCLC patients and be potentially a therapeutic target in this disease. METHODS Serum samples from forty-three SCLC patients were prospectively obtained at diagnosis, response evaluation and progression. Angiogenesis-related cytokines (Angiopoietin-2, VEGF-A, C and D) were simultaneously quantified by Luminex Technology. Clinical data were prospectively recorder. RESULTS Significantly higher concentration of angiogenesis-related cytokines were found in SCLC patients at diagnosis compared to healthy volunteers. High baseline serum concentration of Angiopoietin-2 (sAngiopoietin-2) were associated with a worse overall survival (p=0.006) and remained independently associated with survival in the multivariate analysis (p=0.008). In addition, sAngiopoietin-2 significantly increased at progression when compared to baseline. CONCLUSION These data provide novel evidence on a role of sAngiopoietin-2 in the adverse clinical behavior of SCLC and could be a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Israel Cañadas
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain.
| | - Álvaro Taus
- Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | - Oriol Arpí
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
| | - Lara Pijuan
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Yara Rodríguez
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
| | - Silvia Menéndez
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
| | - Sergi Mojal
- Consulting Service on Methodology for Biomedical Research, IMIM, Barcelona, Spain
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain; Oncology Department, Hospital del Mar, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain; Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Edurne Arriola
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain; Oncology Department, Hospital del Mar, Barcelona, Spain
| |
Collapse
|
26
|
Hong DS, Kurzrock R, Mulay M, Rasmussen E, Wu BM, Bass MB, Zhong ZD, Friberg G, Rosen LS. A phase 1b, open-label study of trebananib plus bevacizumab or motesanib in patients with solid tumours. Oncotarget 2015; 5:11154-67. [PMID: 25525888 PMCID: PMC4294348 DOI: 10.18632/oncotarget.2568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/02/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND To examine the angiopoietin pathway inhibitor trebananib IV plus the anti-VEGF agents bevacizumab or motesanib in advanced solid tumours. METHODS In this open-label phase 1b study, patients received IV trebananib 3 mg kg-1 QW plus bevacizumab 15 mg kg-1 Q3W (cohort 1) or motesanib orally 75 mg (cohort 2); or trebananib 10 mg kg-1 plus bevacizumab 15 mg kg-1 (cohort 3) or motesanib 125 mg (cohort 4). If <33% of patients had dose-limiting toxicities (DLTs), dose escalation occurred. Endpoints were treatment-related adverse events (AEs) incidence and pharmacokinetics (primary); anti-trebananib antibodies, biomarkers, and tumour response (secondary). RESULTS Thirty-six patients received ≥ 1 dose of trebananib (cohorts 1, 2, 3, 4; n = 6, 8, 19, 3). DLT of G3 intestinal perforation and G3 tumor haemorrhage occurred in cohorts 2 and 3, respectively (both n = 1). Across both trebananib plus bevacizumab cohorts, the most common AEs included fatigue (n = 8), diarrhoea (n =4), constipation (n = 3), nausea (n = 3), and epistaxis (n = 3). Three patients across those cohorts had grade ≥ 3 AEs. Across the trebananib plus motesanib cohorts, the most common AEs included hypertension (n = 4), diarrhoea (n = 4), nausea (n = 3), fatigue (n = 3), vomiting (n = 2), and decreased appetite (n = 2). Two patients had grade ≥ 3 AEs. Trebananib did not markedly affect motesanib pharmacokinetics. Across the trebananib plus bevacizumab cohorts, two patients had a partial response; 11 patients had stable disease lasting >6 months. Across the trebananib plus motesanib cohorts, one patient had a partial response; five patients had stable disease lasting >6 months. CONCLUSION Trebananib IV 3 mg kg-1 or 10 mg kg-1 plus bevacizumab or motesanib in advanced solid tumours may be associated with less severe toxicities relative to those emerging when combining two anti-VEGF agents.
Collapse
Affiliation(s)
- David S Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77230-1402, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and CTO, Division of Hematology and Oncology, UC San Diego Moores Cancer Center, La Jolla, CA 92093-0658, USA
| | - Marilyn Mulay
- Mulay Educational and Clinical Consulting Associates, Los Angeles, CA 90025, USA
| | - Erik Rasmussen
- Department of Biostatistics, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Benjamin M Wu
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Michael B Bass
- Department of Molecular Sciences and Computational Biology, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Zhandong D Zhong
- Department of Clinical Immunology and Biological Sample Management, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Greg Friberg
- Department of Early Development, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Lee S Rosen
- Department of Medicine, Division of Hematology and Oncology, UCLA, Santa Monica, CA 90404, USA
| |
Collapse
|
27
|
Atkins MB, Gravis G, Drosik K, Demkow T, Tomczak P, Wong SS, Michaelson MD, Choueiri TK, Wu B, Navale L, Warner D, Ravaud A. Trebananib (AMG 386) in Combination With Sunitinib in Patients With Metastatic Renal Cell Cancer: An Open-Label, Multicenter, Phase II Study. J Clin Oncol 2015; 33:3431-8. [PMID: 26304872 DOI: 10.1200/jco.2014.60.6012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Trebananib, an investigational recombinant peptide-Fc fusion protein, neutralizes the receptor-ligand interaction between Tie2 and angiopoietin-1/2. This phase II study was conducted to evaluate trebananib plus sunitinib, a vascular endothelial growth factor receptor inhibitor, in patients with metastatic clear cell renal cell carcinoma. PATIENTS AND METHODS Adults with metastatic renal cell carcinoma were enrolled sequentially onto two cohorts that received sunitinib 50 mg once per day for 4 weeks on and 2 weeks off and intravenous trebananib once per week at a dose of 10 mg/kg in cohort A or 15 mg/kg in cohort B. The primary end points were incidences of adverse events (AEs) and dose interruptions of sunitinib during the first 12 weeks of treatment. Secondary end points included objective response rate and progression-free survival. RESULTS Eighty-five patients were enrolled: 43 in cohort A, and 42 in cohort B. During the first 12 weeks of treatment, 58% and 57% of patients in cohorts A and B, respectively, had sunitinib dose interruptions (dose decrease, withholding, or withdrawal). The most frequent AEs were diarrhea (cohort A, 74%; cohort B, 67%), mucosal inflammation (cohort A, 49%; cohort B, 60%), and hypertension (cohort A, 52%; cohort B, 45%). AEs of grade 3 or greater occurred in 58% of patients in cohort A and in 69% of patients in cohort B. The objective response rate was 58% and 63% in cohorts A and B, respectively. The median progression-free survival time was 13.9 months (95% CI, 10.4 to 19.2) and 16.3 months (95% CI, 13.1 to 21.4) in cohorts A and B, respectively. The median overall survival time was 36 months (95% CI, 25.2 to not estimable) in cohort A and was not estimable (median follow-up, 25 months) in cohort B. CONCLUSION Trebananib plus sunitinib seemed to increase toxicity at the tested doses. Efficacy results suggest a potential benefit for the addition of trebananib to sunitinib.
Collapse
Affiliation(s)
- Michael B Atkins
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA.
| | - Gwenaelle Gravis
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Kazimierz Drosik
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Tomasz Demkow
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Piotr Tomczak
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Shirley S Wong
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - M Dror Michaelson
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Toni K Choueiri
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Benjamin Wu
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Lynn Navale
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Douglas Warner
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| | - Alain Ravaud
- Michael B. Atkins, Georgetown University, Washington, DC; Gwenaelle Gravis, Institut Paoli Calmettes, Marseille; Alain Ravaud, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint André, Bordeaux, France; Kazimierz Drosik, Regional Cancer Center, Opole; Tomasz Demkow, Maria Skłodowska-Curie Memorial Cancer Center, Warsaw; Piotr Tomczak, University of Medical Sciences, Poznan, Poland; Shirley S. Wong, Western Hospital, Footscray, Victoria, Australia; M. Dror Michaelson, Massachusetts General Hospital; Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA; and Benjamin Wu, Lynn Navale, and Douglas Warner, Amgen, Thousand Oaks, CA
| |
Collapse
|
28
|
Marech I, Leporini C, Ammendola M, Porcelli M, Gadaleta CD, Russo E, De Sarro G, Ranieri G. Classical and non-classical proangiogenic factors as a target of antiangiogenic therapy in tumor microenvironment. Cancer Lett 2015; 380:216-26. [PMID: 26238184 DOI: 10.1016/j.canlet.2015.07.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 12/21/2022]
Abstract
Angiogenesis is sustained by classical and non-classical proangiogenic factors (PFs) acting in tumor microenvironment and these factors are also potential targets of antiangiogenic therapies. All PFs induce the overexpression of several signaling pathways that lead to migration and proliferation of endothelial cells contributing to tumor angiogenesis and survival of cancer cells. In this review, we have analyzed each PF with its specific receptor/s and we have summarized the available antiangiogenic drugs (e.g. monoclonal antibodies) targeting these PFs, some of these agents have already been approved, others are currently in development for the treatment of several human malignancies.
Collapse
Affiliation(s)
- Ilaria Marech
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Christian Leporini
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Michele Ammendola
- Department of Medical and Surgery Sciences, Clinical Surgery Unit, University "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Mariangela Porcelli
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Cosmo Damiano Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Emilio Russo
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Giovambattista De Sarro
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy
| | - Girolamo Ranieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy.
| |
Collapse
|
29
|
Hong DS, Rosen P, Lockhart AC, Fu S, Janku F, Kurzrock R, Khan R, Amore B, Caudillo I, Deng H, Hwang YC, Loberg R, Ngarmchamnanrith G, Beaupre DM, Lee P. A first-in-human study of AMG 208, an oral MET inhibitor, in adult patients with advanced solid tumors. Oncotarget 2015; 6:18693-706. [PMID: 26155941 PMCID: PMC4621921 DOI: 10.18632/oncotarget.4472] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/30/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This first-in-human study evaluated AMG 208, a small-molecule MET inhibitor, in patients with advanced solid tumors. METHODS Three to nine patients were enrolled into one of seven AMG 208 dose cohorts (25, 50, 100, 150, 200, 300, and 400 mg). Patients received AMG 208 orally on days 1 and days 4-28 once daily. The primary objectives were to evaluate the safety, tolerability, pharmacokinetics, and maximum tolerated dose (MTD) of AMG 208. RESULTS Fifty-four patients were enrolled. Six dose-limiting toxicities were observed: grade 3 increased aspartate aminotransferase (200 mg), grade 3 thrombocytopenia (200 mg), grade 4 acute myocardial infarction (300 mg), grade 3 prolonged QT (300 mg), and two cases of grade 3 hypertension (400 mg). The MTD was not reached. The most frequent grade ≥3 treatment-related adverse event was anemia (n = 3) followed by hypertension, prolonged QT, and thrombocytopenia (two patients each). AMG 208 exposure increased linearly with dose; mean plasma half-life estimates were 21.4-68.7 hours. One complete response (prostate cancer) and three partial responses (two in prostate cancer, one in kidney cancer) were observed. CONCLUSIONS In this study, AMG 208 had manageable toxicities and showed evidence of antitumor activity, particularly in prostate cancer.
Collapse
Affiliation(s)
| | - Peter Rosen
- Tower Cancer Research Foundation, Beverly Hills, CA, USA
| | | | - Siqing Fu
- MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Rabia Khan
- MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | - Peter Lee
- Tower Cancer Research Foundation, Beverly Hills, CA, USA
| |
Collapse
|
30
|
Marchetti C, Ledermann JA, Benedetti Panici P. An overview of early investigational therapies for chemoresistant ovarian cancer. Expert Opin Investig Drugs 2015. [DOI: 10.1517/13543784.2015.1072168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
31
|
Azevedo R, Ferreira JA, Peixoto A, Neves M, Sousa N, Lima A, Santos LL. Emerging antibody-based therapeutic strategies for bladder cancer: A systematic review. J Control Release 2015. [PMID: 26196222 DOI: 10.1016/j.jconrel.2015.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bladder cancer is the most common malignancy of the urinary tract, presents the highest recurrence rate among solid tumors and is the second leading cause of death in genitourinary cancers. Despite recent advances in understanding of pathophysiology of the disease, the management of bladder cancer patients remains a clinically challenging problem. Particularly, bladder tumors invading the muscularis propria and disseminated disease are often not responsive to currently available therapeutic approaches, which include surgery and conventional chemotherapy. Antibody-based therapeutic strategies have become an established treatment option for over a decade in several types of cancer. However, bladder cancer has remained mostly an "orphan disease" regarding the introduction of these novel therapeutics, which has been translated in few improvements in patients overall survival. In order to shift this paradigm, several clinical studies involving antibody-based therapeutic strategies targeting the most prominent bladder cancer-related biomolecular pathways and immunological mediators are ongoing. This systematic review explores antibody-based therapeutics for bladder cancer undergoing clinical trial and discusses the future perspectives in this field, envisaging the development of more effective guided therapeutics.
Collapse
Affiliation(s)
- Rita Azevedo
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Manuel Neves
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Nuno Sousa
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Aurea Lima
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (Iinfacts), Department of Pharmaceutical Sciences, Rua Central de Gandra 1317, 4585-116, Gandra-PRD, Portugal; Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Lucio Lara Santos
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Health School of University of Fernando Pessoa, Praça 9 de Abril 349, 4249-004 Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
| |
Collapse
|
32
|
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
|
33
|
Gayed BA, Gillen J, Christie A, Peña-Llopis S, Xie XJ, Yan J, Karam JA, Raj G, Sagalowsky AI, Lotan Y, Margulis V, Brugarolas J. Prospective evaluation of plasma levels of ANGPT2, TuM2PK, and VEGF in patients with renal cell carcinoma. BMC Urol 2015; 15:24. [PMID: 25885592 PMCID: PMC4411704 DOI: 10.1186/s12894-015-0019-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/17/2015] [Indexed: 12/15/2022] Open
Abstract
Background To assess pathological correlations and temporal trends of Angiopoietin-2 (ANGPT2), vascular endothelial growth factor (VEGF) and M2 Pyruvate kinase (TuM2PK), markers of tumor vascular development and metabolism, in patients with renal cell carcinoma (RCC). Methods We prospectively collected plasma samples from 89 patients who underwent surgical/ablative therapy for RCC and 38 patients with benign disease (nephrolithiasis, hematuria without apparent neoplastic origin, or renal cysts). In RCC patients, marker levels were compared between at least 1 preoperative and 1 postoperative time point generally 3 weeks after surgery. Marker temporal trends were assessed using the Wilcoxon sign-rank test. Plasma VEGF, ANGPT2, and TuM2PK levels were determined by ELISA and tested for association with pathological variables. Results Median age was comparable between groups. 83/89 (93%) of the cohort underwent surgical extirpation. 82% of the tumors were organ confined (T ≤2, N0). Only ANGPT2 exhibited significantly elevated preoperative levels in patients with RCC compared to benign disease (p = 0.046). Elevated preoperative levels of ANGPT2 and TuM2PK significantly correlated with increased tumor size and advanced grade (p < 0.05). Chromophobe RCC exhibited higher levels of ANGPT2 compared to other histologies (p < 0.05). A decline in marker level after surgery was not observed, likely due to the timing of the analyses. Conclusion Our results suggest that ANGPT2 is a marker of RCC. Additionally, ANGPT2 and TuM2PK significantly correlated with several adverse pathological features. Further studies are needed to determine clinical applicability.
Collapse
Affiliation(s)
- Bishoy A Gayed
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Jessica Gillen
- Department Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Alana Christie
- Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Samuel Peña-Llopis
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Xian-Jin Xie
- Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Jingsheng Yan
- Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Jose A Karam
- Department of Urology, MD Anderson Cancer Center, Houston, Texas, USA.
| | - Ganesh Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Arthur I Sagalowsky
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA. .,Department of Urology, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd., Dallas, 75390-9110, Texas, USA.
| | - James Brugarolas
- Department Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA. .,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
34
|
Diéras V, Wildiers H, Jassem J, Dirix LY, Guastalla JP, Bono P, Hurvitz SA, Gonçalves A, Romieu G, Limentani SA, Jerusalem G, Lakshmaiah KC, Roché H, Sánchez-Rovira P, Pienkowski T, Seguí Palmer MÁ, Li A, Sun YN, Pickett CA, Slamon DJ. Trebananib (AMG 386) plus weekly paclitaxel with or without bevacizumab as first-line therapy for HER2-negative locally recurrent or metastatic breast cancer: A phase 2 randomized study. Breast 2015; 24:182-90. [PMID: 25747197 DOI: 10.1016/j.breast.2014.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/31/2014] [Accepted: 11/05/2014] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION This phase 2 randomized study evaluated trebananib (AMG 386), a peptide-Fc fusion protein that inhibits angiogenesis by neutralizing the interaction of angiopoietin-1 and -2 with Tie2, in combination with paclitaxel with or without bevacizumab in previously untreated patients with HER2-negative locally recurrent/metastatic breast cancer. METHODS Patients received paclitaxel 90 mg/m(2) once weekly (3-weeks-on/1-week-off) and were randomly assigned 1:1:1:1 to also receive blinded bevacizumab 10 mg/kg once every 2 weeks plus either trebananib 10 mg/kg once weekly (Arm A) or 3 mg/kg once weekly (Arm B), or placebo (Arm C); or open-label trebananib 10 mg/kg once a week (Arm D). Progression-free survival was the primary endpoint. RESULTS In total, 228 patients were randomized. Median estimated progression-free survival for Arms A, B, C, and D was 11.3, 9.2, 12.2, and 10 months, respectively. Hazard ratios (95% CI) for Arms A, B, and D versus Arm C were 0.98 (0.61-1.59), 1.12 (0.70-1.80), and 1.28 (0.79-2.09), respectively. The objective response rate was 71% in Arm A, 51% in Arm B, 60% in Arm C, and 46% in Arm D. The incidence of grade 3/4/5 adverse events was 71/9/4%, 61/14/5%, 62/16/3%, and 52/4/7% in Arms A/B/C/D. In Arm D, median progression-free survival was 12.8 and 7.4 months for those with high and low trebananib exposure (AUCss ≥ 8.4 versus < 8.4 mg·h/mL), respectively. CONCLUSIONS There was no apparent prolongation of estimated progression-free survival with the addition of trebananib to paclitaxel and bevacizumab at the doses tested. Toxicity was manageable. Exposure-response analyses support evaluation of combinations incorporating trebananib at doses > 10 mg/kg in this setting. TRIAL REGISTRATION ClinicalTrials.gov, NCT00511459.
Collapse
Affiliation(s)
| | | | | | - Luc Y Dirix
- General Hospital Sint-Augustinus, Antwerp, Belgium.
| | | | - Petri Bono
- Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | | | | | | - K C Lakshmaiah
- Kidwai Memorial Institute of Oncology, Bangalore, India.
| | | | | | | | | | - Ai Li
- Amgen Inc., Thousand Oaks, CA, USA.
| | | | | | | |
Collapse
|
35
|
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
|
36
|
Biomarker in Cisplatin-Based Chemotherapy for Urinary Bladder Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 867:293-316. [PMID: 26530373 DOI: 10.1007/978-94-017-7215-0_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The treatment of metastasized bladder cancer has been evolving during recent years. Cisplatin based chemotherapy combinations are still gold standard in the treatment of advanced and metastasized bladder cancer. But new therapies are approaching. Based to this fact biological markers will become more important for decisions in bladder cancer treatment. A systematic MEDLINE search of the key words "cisplatin", "bladder cancer", "DNA marker", "protein marker", "methylation biomarker", "predictive marker", "prognostic marker" has been made. This review aims to highlight the most relevant clinical and experimental studies investigating markers for metastasized transitional carcinoma of the urothelium treated by cisplatin based regimens.
Collapse
|
37
|
Sonpavde G, Jones BS, Bellmunt J, Choueiri TK, Sternberg CN. Future directions and targeted therapies in bladder cancer. Hematol Oncol Clin North Am 2014; 29:361-76, x. [PMID: 25836940 DOI: 10.1016/j.hoc.2014.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
There are substantial unmet needs for patients with metastatic urothelial carcinoma (UC). First-line cisplatin-based chemotherapy regimens yield a median survival of 12 to 15 months and long-term survival in 5% to 15%. Salvage systemic therapy yields a median survival of 6 to 8 months. Hence, the discovery of novel therapeutic targets is of paramount importance. Recent molecular analyses have provided insights regarding molecular tumor tissue alterations on multiple platforms. A multidisciplinary effort using innovative clinical trial designs and exploiting preclinical signals of robust activity guided by predictive biomarkers may provide much needed clinical advances in therapy for advanced UC.
Collapse
Affiliation(s)
- Guru Sonpavde
- University of Alabama at Birmingham (UAB) Comprehensive Cancer Center, 1720 2nd Ave. S., Birmingham, AL 35294, USA
| | - Benjamin S Jones
- University of Alabama at Birmingham (UAB) Comprehensive Cancer Center, 1720 2nd Ave. S., Birmingham, AL 35294, USA
| | - Joaquim Bellmunt
- Bladder Cancer Institute, Dana Farber Cancer Institute, Dana-Farber/Brigham and Women's Cancer Center, Boston, 450, Brookline Ave, MA 02215, USA
| | - Toni K Choueiri
- Bladder Cancer Institute, Dana Farber Cancer Institute, Dana-Farber/Brigham and Women's Cancer Center, Boston, 450, Brookline Ave, MA 02215, USA
| | - Cora N Sternberg
- Department of Medical Oncology, San Camillo Forlanini Hospital, Padiglioni Flajani, 1st Floor, Circonvallazione Gianicolense 87, Rome 00152, Italy.
| |
Collapse
|
38
|
Blank S, Deck C, Dreikhausen L, Weichert W, Giese N, Falk C, Schmidt T, Ott K. Angiogenic and growth factors in gastric cancer. J Surg Res 2014; 194:420-429. [PMID: 25577146 DOI: 10.1016/j.jss.2014.11.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/10/2014] [Accepted: 11/19/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Antiangiogenic treatment is at the horizon in the palliative treatment of gastric cancer (GC), but data on proangiogenic biomarkers are still limited. The aim of this study was to analyze five proteins with a function in tumor angiogenesis: vascular endothelial growth factor (VEGF), angiopoietin-2 (Ang-2), follistatin, leptin, and platelet endothelial cell adhesion molecule 1 (CD31) in peripheral blood and corresponding tumor tissue. MATERIAL AND METHODS From 2008-2010, tumor tissue (n = 76) and corresponding preoperative serum (n = 69) of patients with localized GC were collected; 45 had perioperative chemotherapy. Protein serum or tumor lysate levels of these factors were measured by an angiogenesis multiplex immunoassay and correlated with response and survival. RESULTS Serum Ang-2 had prognostic relevance in the whole study population (P = 0.027). In subgroup analysis, serum VEGF and Ang-2 had prognostic relevance in primarily resected patients (P = 0.028; P = 0.048) but no association was found in neoadjuvantly treated patients. Follistatin concentration in the tumor tissue was associated with prognosis in all patients (P = 0.019). Tumor VEGF concentrations were correlated with histopathologic response (P = 0.011), with patients showing >50% remaining tumor having higher VEGF concentrations. The tissue Ang-2/VEGF ratio was significantly correlated with both clinical and histopathologic response (P = 0.029, P = 0.009). Additionally, the level of leptin in the tissue was associated with clinical response: nonresponding patients had higher leptin levels than those of responding patients (P = 0.032). CONCLUSIONS Our results show the importance of angiogenetic factors in serum and tumor tissue in GC for prognosis and treatment response. Further trials in larger patient populations are warranted for a further evaluation of proangiogenetic factors as biomarkers in gastrointestinal cancer.
Collapse
Affiliation(s)
- Susanne Blank
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany.
| | - Catrin Deck
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Lena Dreikhausen
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Natalia Giese
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Christine Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Thomas Schmidt
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Katja Ott
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
39
|
Antagonism of Ang-Tie2 and Dll4-Notch signaling has opposing effects on tumor endothelial cell proliferation, evidenced by a new flow cytometry method. J Transl Med 2014; 94:1296-308. [PMID: 25243900 DOI: 10.1038/labinvest.2014.116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 11/08/2022] Open
Abstract
Sustained angiogenesis is essential for tumor growth as it provides the tumor with a network of blood vessels that supply both oxygen and essential nutrients. Limiting tumor-associated angiogenesis is a proven strategy for the treatment of human cancer. To date, the rapid detection and quantitation of tumor-associated endothelial cell (TAEC) proliferation has been challenging, largely due to the low frequency of endothelial cells (ECs) within the tumor microenvironment. In this report, we address this problem using a new multiparametric flow cytometry method capable of rapid and precise quantitation of proliferation by measuring bromodeoxyuridine (BrdUrd) uptake in mouse TAECs from established human tumor xenografts. We determined the basal proliferation labeling index of TAECs in two human tumor xenografts representing two distinct histologies, COLO 205 (colorectal cancer) and U-87 (glioblastoma). We then investigated the effects of two large-molecule antiangiogenic agents targeting different biochemical pathways. Blocking angiopoietin-Tie2 signaling with the peptide-Fc fusion protein, trebananib (AMG 386), inhibited proliferation of TAECs, whereas blocking Dll4-Notch signaling with an anti-Dll4-specific antibody induced hyperproliferation of TAECs. These pharmacodynamic studies highlight the sensitivity and utility of this flow cytometry-based method and demonstrate the value of this assay to rapidly assess the in vivo proliferative effects of angiogenesis-targeted agents on both the tumor and the associated vasculature.
Collapse
|
40
|
Liontos M, Lykka M, Dimopoulos MA, Bamias A. Profile of trebananib (AMG386) and its potential in the treatment of ovarian cancer. Onco Targets Ther 2014; 7:1837-45. [PMID: 25336975 PMCID: PMC4199819 DOI: 10.2147/ott.s65522] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Angiogenesis has been implicated in ovarian cancer pathogenesis. Bevacizumab, an anti-vascular endothelial growth factor monoclonal antibody, has recently been incorporated in ovarian cancer treatment in combination with chemotherapy both in a frontline setting and in disease recurrence. However, resistance eventually develops and treatment with bevacizumab is associated with increased risk for toxicities such as thromboembolic and hemorrhagic events, gastrointestinal perforation, and impaired wound healing, suggesting the need for new therapeutic approaches. Targeting of the angiopoietins/Tie2 pathway has gained accumulating interest during the last few years as a strategy to overcome bevacizumab resistance and toxicities. Trebananib is a first-in-class peptibody that inhibits angiopoietin 1 and 2 interaction with their receptor Tie2. The molecular profile of this agent, the preclinical data, and clinical studies demonstrating its efficacy in ovarian cancer are discussed in this review.
Collapse
Affiliation(s)
- Michalis Liontos
- Oncology Department, Therapeutics Clinic, Medical School, University of Athens, Athens, Greece
| | - Maria Lykka
- Oncology Department, Therapeutics Clinic, Medical School, University of Athens, Athens, Greece
| | | | - Aristotle Bamias
- Oncology Department, Therapeutics Clinic, Medical School, University of Athens, Athens, Greece
| |
Collapse
|
41
|
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
|
42
|
Vergote I. Novel therapies, including enzastaurin, in the treatment of ovarian cancer. Expert Opin Investig Drugs 2014; 23:579-98. [DOI: 10.1517/13543784.2014.900542] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Cereda V, Formica V, Massimiani G, Tosetto L, Roselli M. Targeting metastatic castration-resistant prostate cancer: mechanisms of progression and novel early therapeutic approaches. Expert Opin Investig Drugs 2014; 23:469-87. [PMID: 24490883 DOI: 10.1517/13543784.2014.885950] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Advances in clinical research have led to official approval of several new treatments for metastatic prostate cancer in the last three years: sipuleucel-T, cabazitaxel, abiraterone acetate, radium-223 and enzalutamide. Although these agents have all been shown to improve overall survival in randomized Phase III trials, metastatic castration-resistant prostate cancer (mCRPC) remains incurable. AREAS COVERED First, the review summarizes the current literature on the biology of mCRPC. The emerging data are increasing our understanding of the mechanisms that underlie the pathogenesis of castrate resistance and where future treatment might be headed. In the second part of the review, the authors assess the future directions in disease therapy. Indeed, novel selected therapeutic approaches, including novel agents and combinatorial therapies, are showing promising early results. EXPERT OPINION Targeting different molecular pathways in combination with immunotherapy can be a promising direction in metastatic castration prostate cancer treatment. However, several challenges still exist including elucidating the optimal use and sequencing of these new agents. There are also challenges in both the design and the interpretation of the results from clinical trials.
Collapse
Affiliation(s)
- Vittore Cereda
- University of Rome 'Tor Vergata', Tor Vergata Clinical Center, Department of Systems Medicine, Medical Oncology , V.le Oxford 81, 00133, Rome , Italy +390 620 908 190 ; +390 620 904 576 ;
| | | | | | | | | |
Collapse
|
44
|
Tumour-site-dependent expression profile of angiogenic factors in tumour-associated stroma of primary colorectal cancer and metastases. Br J Cancer 2013; 110:441-9. [PMID: 24292449 PMCID: PMC3899771 DOI: 10.1038/bjc.2013.745] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 09/29/2013] [Accepted: 11/05/2013] [Indexed: 01/29/2023] Open
Abstract
Background: Tumour-associated stroma has a critical role in tumour proliferation. Our aim was to determine a specific protein expression profile of stromal angiogenic cytokines and matrix metalloproteinases (MMPs) to identify potential biomarkers or new therapy targets. Methods: Frozen tissue of primary colorectal cancer (n=25), liver (n=25) and lung metastases (n=23) was laser-microdissected to obtain tumour epithelial cells and adjacent tumour-associated stroma. Protein expression of nine angiogenic cytokines and eight MMPs was analysed using a multiplex-based protein assay. Results: We found a differential expression of several MMPs and angiogenic cytokines in tumour cells compared with adjacent tumour stroma. Cluster analysis displayed a tumour-site-dependent stromal expression of MMPs and angiogenic cytokines. Univariate analysis identified stromal MMP-2 and MMP-3 in primary colorectal cancer, stromal MMP-1, -2, -3 and Angiopoietin-2 in lung metastases and stromal MMP-12 and VEGF in liver metastases as prognostic markers (P>0.05, respectively). Furthermore, stroma-derived Angiopoietin-2 proved to be an independent prognostic marker in colorectal lung metastases. Conclusion: Expression of MMPs and angiogenic cytokines in tumour cells and adjacent tumour stroma is dependent on the tumour site. Stroma-derived MMPs and angiogenic cytokines may be useful prognostic biomarkers. These data can be helpful to identify new agents for a targeted therapy in patients with colorectal cancer.
Collapse
|
45
|
Wu B, Sun YN. Pharmacokinetics of Peptide-Fc fusion proteins. J Pharm Sci 2013; 103:53-64. [PMID: 24285510 DOI: 10.1002/jps.23783] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 01/11/2023]
Abstract
Peptide-Fc fusion proteins (or peptibodies) are chimeric proteins generated by fusing a biologically active peptide with the Fc-domain of immunoglobulin G. In this review, we describe recent studies that have evaluated the absorption, distribution, metabolism, and excretion characteristics of peptibodies. Key features of the pharmacokinetics of peptibodies include their extended half-life due to recycling by the neonatal Fc receptor (FcRn), a substantial contribution by renal excretion to total clearance and, for certain peptibodies, target-mediated drug disposition. The prolonged half-life of peptibodies permits less-frequent dose administration compared with small therapeutic peptides, thereby supporting patient convenience and compliance. Hence, a considerable number of peptibodies are currently in preclinical and clinical development. Investigation of the metabolism (biotransformation) of biologics is an evolving area of research: ligand-binding mass spectrometry techniques have been employed for the characterization of the peptibody romiplostim, providing a new approach to evaluation of the degradation products of biologics. Pharmacokinetic/pharmacodynamic modeling and simulation techniques have been used to predict the pharmacokinetics of peptibodies which can inform clinical decision-making, particularly selection of dosing regimens. This integrated review highlights the distinct pharmacokinetic characteristics of peptibodies and their influence on the drug development process for this emerging family of therapeutics.
Collapse
Affiliation(s)
- Benjamin Wu
- Department of Pharmacokinetics and Drug Metabolism, Quantitative Pharmacology Group, Amgen Inc, Thousand Oaks, California, 91320
| | | |
Collapse
|
46
|
Incidence and management of edema associated with trebananib (AMG 386). Gynecol Oncol 2013; 130:636-41. [DOI: 10.1016/j.ygyno.2013.05.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 05/09/2013] [Accepted: 05/16/2013] [Indexed: 01/24/2023]
|
47
|
Posadas EM, Limvorasak S, Sharma S, Figlin RA. Targeting angiogenesis in renal cell carcinoma. Expert Opin Pharmacother 2013; 14:2221-36. [DOI: 10.1517/14656566.2013.832202] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
48
|
Development of a biosensor-based immunogenicity assay capable of blocking soluble drug target interference. J Immunol Methods 2013; 396:44-55. [PMID: 23933325 DOI: 10.1016/j.jim.2013.07.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 07/23/2013] [Accepted: 07/23/2013] [Indexed: 01/13/2023]
Abstract
As with other protein therapeutics, trebananib (AMG 386), an investigational peptide Fc-fusion protein ("peptibody") that inhibits angiogenesis by neutralizing the interaction of angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) with the Tie2 receptor, has the potential to trigger an immune response in cancer patients treated with the therapeutic. An electrochemiluminescence bridging anti-drug antibody (ADA) assay that was utilized to support early-phase clinical trials in the development of trebananib was found to lack adequate sensitivity and drug tolerance in later-phase clinical studies when higher doses of trebananib were administered. Therefore, we developed a surface plasmon resonance (SPR) immunoassay method utilizing a secondary confirmatory detector antibody (goat anti-human IgG F[ab']2) known to cross-react with human IgG and IgM to better assess the potential impact of immunogenicity on the pharmacokinetics, pharmacodynamics, and toxicity of trebananib. The SPR method was more sensitive than the electrochemiluminescence bridging assay because of signal amplification from the confirmatory binding of the detector antibody; drug tolerance was improved since antibody binding avidity does not affect detection on this platform. Despite the inability of the confirmatory detector antibody to bind angiopoietins in protein-free buffer, false-positive ADA results were generated from patient serum samples containing Ang1 and Ang2 through an apparently specific binding between the angiopoietins and the confirmatory detector antibody, likely mediated by the interaction of the angiopoietins with serum immunoglobulins. Addition to the sample diluent of a human antibody that specifically binds to Ang1 and Ang2 with high affinity resulted in a complete block of angiopoietin interference without affecting ADA detection. This biosensor-based assay provides a reliable method for assessing immunogenicity in phase 3 clinical trials.
Collapse
|
49
|
Buehler D, Rush P, Hasenstein JR, Rice SR, Hafez GR, Longley BJ, Kozak KR. Expression of angiopoietin-TIE system components in angiosarcoma. Mod Pathol 2013; 26:1032-40. [PMID: 23558570 PMCID: PMC3706492 DOI: 10.1038/modpathol.2013.43] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 01/08/2023]
Abstract
Angiosarcoma is an aggressive malignancy of endothelial differentiation. Potential roles of the endothelial angiopoietin-tunica interna endothelial cell kinase (ANGPT-TIE) system in angiosarcoma diagnosis, pathogenesis, prognosis and treatment are undefined. To examine the expression and prognostic significance of angiopoietin-1, angiopoietin-2, TIE1 and TEK (TIE2) proteins in angiosarcoma, we immunohistochemically evaluated clinically annotated human angiosarcoma samples. Correlations of protein expression with overall survival and pathological features were explored. The cohort included 51 patients diagnosed with angiosarcoma at the age of 30-86 years (median 67). The 5-year overall survival was 45% with a median of 26 months. Moderate to strong expression of angiopoietin-1, TIE1 and TEK (TIE2) was identified in the majority of angiosarcomas and moderate to strong expression of angiopoietin-2 was observed in 42% of angiosarcomas. Increased angiopoietin-1 expression correlated with improved survival. Non-significant trends toward longer survival were also observed with increased TIE1 and TEK (TIE2) expression. Increased expression of angiopoietin-2, TIE1 and TEK (TIE2) was associated with vasoformative architecture. No differences in expression of these proteins were observed when patients were segregated by age, gender, presence or absence of metastases at diagnosis, primary tumor location, radiation association or the presence of necrosis. We conclude that components of the ANGPT-TIE system are commonly expressed in angiosarcomas. Reduced expression of these proteins is associated with non-vasoformative and clinically more aggressive lesions.
Collapse
Affiliation(s)
- Darya Buehler
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Patrick Rush
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jason R. Hasenstein
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Stephanie R. Rice
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Gholam Reza Hafez
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - B. Jack Longley
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kevin R Kozak
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
50
|
Eroglu Z, Stein CA, Pal SK. Targeting angiopoietin-2 signaling in cancer therapy. Expert Opin Investig Drugs 2013; 22:813-25. [DOI: 10.1517/13543784.2013.793306] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|