51
|
Herzog J, Ehrlich SM, Pfitzer L, Liebl J, Fröhlich T, Arnold GJ, Mikulits W, Haider C, Vollmar AM, Zahler S. Cyclin-dependent kinase 5 stabilizes hypoxia-inducible factor-1α: a novel approach for inhibiting angiogenesis in hepatocellular carcinoma. Oncotarget 2017; 7:27108-21. [PMID: 27027353 PMCID: PMC5053636 DOI: 10.18632/oncotarget.8342] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 03/16/2016] [Indexed: 01/25/2023] Open
Abstract
We recently introduced CDK5 as target in HCC, regulating DNA damage response. Based on this and on our previous knowledge about vascular effects of CDK5, we investigated the role of CDK5 in angiogenesis in HCC, one of the most vascularized tumors. We put a special focus on the transcription factor HIF-1α, a master regulator of tumor angiogenesis. The interaction of CDK5 with HIF-1α was tested by Western blot, PCR, reporter gene assay, immunohistochemistry, kinase assay, co-immunoprecipitation, mass spectrometry, and mutation studies. In vivo, different murine HCC models, were either induced by diethylnitrosamine or subcutaneous injection of HUH7 or HepG2 cells. The correlation of vascular density and CDK5 was assessed by immunostaining of a microarray of liver tissues from HCC patients. Inhibition of CDK5 in endothelial or HCC cells reduced HIF-1α levels in vitro and in vivo, and transcription of HIF-1α target genes (VEGFA, VEGFR1, EphrinA1). Mass spectrometry and site directed mutagenesis revealed a stabilizing phosphorylation of HIF-1α at Ser687 by CDK5. Vascular density was decreased in murine HCC models by CDK5 inhibition. In conclusion, inhibiting CDK5 is a multi-modal systemic approach to treat HCC, hitting angiogenesis, as well as the tumor cells themselves.
Collapse
Affiliation(s)
- Julia Herzog
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Munich, Germany
| | - Sandra M Ehrlich
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Munich, Germany
| | - Lisa Pfitzer
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Munich, Germany
| | - Johanna Liebl
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center Munich, University of Munich, Munich, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center Munich, University of Munich, Munich, Germany
| | - Wolfgang Mikulits
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Christine Haider
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Munich, Germany
| | - Stefan Zahler
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Munich, Germany
| |
Collapse
|
52
|
Pak RW, Hadjiabadi DH, Senarathna J, Agarwal S, Thakor NV, Pillai JJ, Pathak AP. Implications of neurovascular uncoupling in functional magnetic resonance imaging (fMRI) of brain tumors. J Cereb Blood Flow Metab 2017; 37:3475-3487. [PMID: 28492341 PMCID: PMC5669348 DOI: 10.1177/0271678x17707398] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Functional magnetic resonance imaging (fMRI) serves as a critical tool for presurgical mapping of eloquent cortex and changes in neurological function in patients diagnosed with brain tumors. However, the blood-oxygen-level-dependent (BOLD) contrast mechanism underlying fMRI assumes that neurovascular coupling remains intact during brain tumor progression, and that measured changes in cerebral blood flow (CBF) are correlated with neuronal function. Recent preclinical and clinical studies have demonstrated that even low-grade brain tumors can exhibit neurovascular uncoupling (NVU), which can confound interpretation of fMRI data. Therefore, to avoid neurosurgical complications, it is crucial to understand the biophysical basis of NVU and its impact on fMRI. Here we review the physiology of the neurovascular unit, how it is remodeled, and functionally altered by brain cancer cells. We first discuss the latest findings about the components of the neurovascular unit. Next, we synthesize results from preclinical and clinical studies to illustrate how brain tumor induced NVU affects fMRI data interpretation. We examine advances in functional imaging methods that permit the clinical evaluation of brain tumors with NVU. Finally, we discuss how the suppression of anomalous tumor blood vessel formation with antiangiogenic therapies can "normalize" the brain tumor vasculature, and potentially restore neurovascular coupling.
Collapse
Affiliation(s)
- Rebecca W Pak
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Darian H Hadjiabadi
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Janaka Senarathna
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Shruti Agarwal
- 2 Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Nitish V Thakor
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jay J Pillai
- 2 Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Arvind P Pathak
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA.,2 Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, USA.,3 Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
53
|
Vennepureddy A, Singh P, Rastogi R, Atallah JP, Terjanian T. Evolution of ramucirumab in the treatment of cancer - A review of literature. J Oncol Pharm Pract 2017; 23:525-539. [PMID: 27306885 DOI: 10.1177/1078155216655474] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ramucirumab is a recombinant human monoclonal antibody and is used in the treatment of advanced malignancies. Its mechanism of action is by inhibiting angiogenesis in tumor cells by targeting the vascular endothelial growth factor receptor 2. United States Food and Drug Administration (FDA) approved it initially in 2014 for the treatment of advanced gastric or gastro-esophageal junction adenocarcinoma and metastatic non-small cell lung carcinoma. It was approved by FDA in 2015 for the treatment of advanced colorectal cancer. This manuscript consolidates pre-clinical trials to phase I, II, and III trial data indicating the effects of ramucirumab on different cancer types, which led to its approval. By comparing these clinical trials alongside each other, we can more easily examine the studies that have already been completed, along with currently ongoing studies and potential further areas of interest for this newly approved treatment. This approach makes it convenient to compare dosages, overall survival, adverse events, as well as possible routes for combination therapy with ramucirumab. By compiling results for various oncological malignancies, we can differentiate between treatments that are effective and have the highest incidence of stable disease, and those that do not seem promising. Ramucirumab has been effective in the treatment of various carcinomas and this article outlines other tumors in which this treatment option may be successful.
Collapse
Affiliation(s)
- A Vennepureddy
- 1 Department of Internal Medicine, Staten Island University Hospital, NY, USA
| | - P Singh
- 1 Department of Internal Medicine, Staten Island University Hospital, NY, USA
| | - R Rastogi
- 1 Department of Internal Medicine, Staten Island University Hospital, NY, USA
| | - J P Atallah
- 2 Division of Hematology and Oncology, Staten Island University Hospital, NY, USA
| | - T Terjanian
- 2 Division of Hematology and Oncology, Staten Island University Hospital, NY, USA
| |
Collapse
|
54
|
Ramnefjell M, Aamelfot C, Aziz S, Helgeland L, Akslen LA. Microvascular proliferation is associated with aggressive tumour features and reduced survival in lung adenocarcinoma. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2017; 3:249-257. [PMID: 29085665 PMCID: PMC5653928 DOI: 10.1002/cjp2.78] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/21/2017] [Accepted: 06/29/2017] [Indexed: 01/15/2023]
Abstract
Despite new treatment options in lung cancer, there is still a need for better biomarkers to assist in therapy decisions. Angiogenesis has been associated with tumour growth and dissemination, and the vascular proliferation index (VPI) is a valuable prognostic marker in other tumours. Nestin, a marker of immature endothelium, was previously applied in combination with Ki67 for proliferating endothelium as a novel marker (Nestin‐Ki67) of ongoing angiogenesis. Here, the prevalence and prognostic impact of vascular proliferation on lung cancer‐specific survival (LCSS) in lung adenocarcinomas was studied. Selected tumour slides from a cohort of 210 patients treated surgically for adenocarcinoma at Haukeland University Hospital (Norway) from 1993 to 2010 were stained for Nestin‐Ki67. VPI, the ratio between the density of proliferating vessels and the overall microvessel density were used, and the cut‐off value was set at 4.4% (upper quartile). High VPI was associated with the presence of blood vessel invasion (p = 0.007) and tumour necrosis (p = 0.007). Further, high VPI was significantly associated with reduced LCSS (p = 0.020). By multivariate analysis, VPI remained an independent prognostic factor for reduced LCSS (HR 1.7; 95% CI 1.04–2.68; p = 0.033) when adjusted for other prognostic clinico‐pathological features. In conclusion, microvessel proliferation assessed using the VPI was associated with aggressive tumour features such as blood vessel invasion and tumour necrosis and, independently, decreased LCSS. This marker should be further explored in separate cohorts, and in trials of anti‐angiogenesis therapy.
Collapse
Affiliation(s)
- Maria Ramnefjell
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for PathologyUniversity of BergenBergenNorway
| | - Christina Aamelfot
- Department of Thoracic MedicineHaukeland University HospitalBergenNorway
| | - Sura Aziz
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Lars Helgeland
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for PathologyUniversity of BergenBergenNorway.,Department of PathologyHaukeland University HospitalBergenNorway
| |
Collapse
|
55
|
Di Salvatore M, Pietrantonio F, Orlandi A, Del Re M, Berenato R, Rossi E, Caporale M, Guarino D, Martinetti A, Basso M, Mennitto R, Santonocito C, Mennitto A, Schinzari G, Bossi I, Capoluongo E, Danesi R, de Braud F, Barone C. IL-8 and eNOS polymorphisms predict bevacizumab-based first line treatment outcomes in RAS mutant metastatic colorectal cancer patients. Oncotarget 2017; 8:16887-16898. [PMID: 28129643 PMCID: PMC5370008 DOI: 10.18632/oncotarget.14810] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/02/2016] [Indexed: 12/19/2022] Open
Abstract
Background Predictive biomarkers of efficacy and toxicity of bevacizumab have not yet been validated. This study assessed the influence of IL-8, eNOS and VEGF-A polymorphisms in RAS mutated metastatic colorectal cancer patients receiving bevacizumab-based chemotherapy. Methods 120 patients treated with first-line combination FOLFOX6 plus bevacizumab were included. A historical cohort of 112 RAS mutated colorectal cancer patients treated with FOLFOX6 alone served as control group. The following SNPs were analyzed: IL-8 c.-251T>A; eNOS c.-786T>C and c.-894G>T; VEGF-A c.936C>T, c.958T>C, c.1154A>G and c.2578C>A. Correlation of SNPs, baseline IL-8 serum levels and bevacizumab-efficacy was done. Results In the bevacizumab group, carriers of the IL-8 alleles c.-251TA+AA showed a shorter PFS (P=0.002) and OS (P=0.03) compared to TT alleles. Patients with pre-treatment IL-8 < 18.25 pg/ml showed significantly longer median PFS and OS (PFS: 10.9 vs 7.6 months, P=0.005; OS: 30.7 vs 18.2 months, P<0.001) compared to patients with IL-8 higher levels (>18,25 pg/ml). IL-8 c.-251TA+AA carriers had significantly higher IL-8 levels (P<0.0001). Multivariate analysis confirmed association of IL-8 polymorphism with PFS, and of IL-8 baseline levels with both PFS and OS. IL-8 SNP did not affect the outcome in the control group. The eNOS polymorphism c.-894G>T was found associated with higher severe toxicity (P=0.0002) in patients carrying the c.-894TT genotype. Conclusions Although our data need prospective validation, IL-8 and eNOS SNPs may be have a role as predictive biomarkers for bevacizumab efficacy and toxicity.
Collapse
Affiliation(s)
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Armando Orlandi
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Rosa Berenato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Ernesto Rossi
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marta Caporale
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Donatella Guarino
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Michele Basso
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Roberta Mennitto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Concetta Santonocito
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alessia Mennitto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Giovanni Schinzari
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ilaria Bossi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Ettore Capoluongo
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Carlo Barone
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
56
|
A Review of Anti-Angiogenic Targets for Monoclonal Antibody Cancer Therapy. Int J Mol Sci 2017; 18:ijms18081786. [PMID: 28817103 PMCID: PMC5578174 DOI: 10.3390/ijms18081786] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor angiogenesis is a key event that governs tumor progression and metastasis. It is controlled by the complicated and coordinated actions of pro-angiogenic factors and their receptors that become upregulated during tumorigenesis. Over the past several decades, vascular endothelial growth factor (VEGF) signaling has been identified as a central axis in tumor angiogenesis. The remarkable advent of recombinant antibody technology has led to the development of bevacizumab, a humanized antibody that targets VEGF and is a leading clinical therapy to suppress tumor angiogenesis. However, despite the clinical efficacy of bevacizumab, its significant side effects and drug resistance have raised concerns necessitating the identification of novel drug targets and development of novel therapeutics to combat tumor angiogenesis. This review will highlight the role and relevance of VEGF and other potential therapeutic targets and their receptors in angiogenesis. Simultaneously, we will also cover the current status of monoclonal antibodies being developed to target these candidates for cancer therapy.
Collapse
|
57
|
Qiu C, Zhang D, Chi Y, Chen Q, Xu L, Xie Q. Clinical significance of 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester-labeled microspheres for detecting endothelial progenitor cells in human peripheral blood. Exp Ther Med 2017; 14:1659-1664. [PMID: 28810633 DOI: 10.3892/etm.2017.4657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/14/2017] [Indexed: 11/06/2022] Open
Abstract
The aims of the present study were to establish a single-platform flow cytometry method using 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled microspheres as the reference for determining endothelial progenitor cell (EPC) number and to evaluate the efficacy of this detection method. Single-platform flow cytometry was used to count cell numbers using CFSE-stained fluorescent microspheres as the internal reference and the EPC numbers in specimens using this novel method were compared with an in vitro clonogenic counting assay. The results of the two counting methods were consistent and compared with the in vitro clonogenic counting assay, the time and cost of the novel method was markedly reduced, as were the corresponding technical requirements. The present findings indicated that single-platform flow cytometry, with CFSE-labeled microspheres as the reference, provides faster and improved detection of EPCs in human peripheral blood specimens, with reduced time and cost, making it more suitable for routine clinical application.
Collapse
Affiliation(s)
- Chaolin Qiu
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Denghai Zhang
- Central Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Yongbin Chi
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Qing Chen
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Limin Xu
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Qiuhua Xie
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| |
Collapse
|
58
|
Wang B, Wang W, Yu Y, Zhang Y, Zhang J, Yuan Z. The study of angiogenesis stimulated by multivalent peptide ligand-modified alginate. Colloids Surf B Biointerfaces 2017; 154:383-390. [DOI: 10.1016/j.colsurfb.2017.03.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 01/13/2023]
|
59
|
El Bairi K, Amrani M, Kandhro AH, Afqir S. Prediction of therapy response in ovarian cancer: Where are we now? Crit Rev Clin Lab Sci 2017; 54:233-266. [PMID: 28443762 DOI: 10.1080/10408363.2017.1313190] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Therapy resistance is a major challenge in the management of ovarian cancer (OC). Advances in detection and new technology validation have led to the emergence of biomarkers that can predict responses to available therapies. It is important to identify predictive biomarkers to select resistant and sensitive patients in order to reduce important toxicities, to reduce costs and to increase survival. The discovery of predictive and prognostic biomarkers for monitoring therapy is a developing field and provides promising perspectives in the era of personalized medicine. This review article will discuss the biology of OC with a focus on targetable pathways; current therapies; mechanisms of resistance; predictive biomarkers for chemotherapy, antiangiogenic and DNA-targeted therapies, and optimal cytoreductive surgery; and the emergence of liquid biopsy using recent studies from the Medline database and ClinicalTrials.gov.
Collapse
Affiliation(s)
- Khalid El Bairi
- a Faculty of Medicine and Pharmacy , Mohamed Ist University , Oujda , Morocco
| | - Mariam Amrani
- b Equipe de Recherche ONCOGYMA, Faculty of Medicine, Pathology Department , National Institute of Oncology, Université Mohamed V , Rabat , Morocco
| | - Abdul Hafeez Kandhro
- c Department of Biochemistry , Healthcare Molecular and Diagnostic Laboratory , Hyderabad , Pakistan
| | - Said Afqir
- d Department of Medical Oncology , Mohamed VI University Hospital , Oujda , Morocco
| |
Collapse
|
60
|
Chen XW, Sun JG, Zhang LP, Liao XY, Liao RX. Recruitment of CD11b +Ly6C + monocytes in non-small cell lung cancer xenografts challenged by anti-VEGF antibody. Oncol Lett 2017; 14:615-622. [PMID: 28693213 PMCID: PMC5494733 DOI: 10.3892/ol.2017.6236] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 03/03/2017] [Indexed: 12/30/2022] Open
Abstract
A series of antibodies against vascular endothelial growth factor (VEGF) have been developed for the treatment of various types of cancer, including non-small cell lung cancer (NSCLC) in recent years. However, tumors frequently demonstrate resistance to these strategies of VEGF inhibition. Efforts to better understand the mechanism underlying the acquired resistance to anti-VEGF antibodies are warranted. In the present study, in order to develop a xenograft model of acquired resistance to anti-VEGF antibody, xenografts of human adenocarcinoma A549 cells were generated through the successive inoculation of tumor tissue explants into first (F1), second (F2) and third (F3) generations of mice treated with the anti-VEGF antibody B20. Tumor growth rate and vessel-forming ability, assessed via cluster of differentiation (CD) 31 staining, were significantly lower in the F1, F2 and F3 groups compared with in the F0 control group (P<0.01), suggesting that drug resistance was not successfully acquired. The percentages of CD11b+ myeloid-derived suppressor cells and lymphocyte antigen 6C (Ly6C)+ subsets were significantly smaller in F1, F2 and F3 groups compared with in F0 (P<0.01). However, the ratio of Ly6C+ to CD11b+ cells was significantly higher in the F3 group compared with in F0 and F1 groups (P<0.01), indicating increasing recruitment of the Ly6C+ subset with successive challenges with the anti-VEGF antibody. In conclusion, the recruitment of CD11b+Ly6C+ monocytes increased with successive generations of NSCLC-xenografted mice challenged by B20, an anti-VEGF agent.
Collapse
Affiliation(s)
- Xie-Wan Chen
- Medical English Department, College of Basic Medicine, Third Military Medical University, Chongqing 400038, P.R. China.,Cancer Institute of People's Liberation Army, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Jian-Guo Sun
- Cancer Institute of People's Liberation Army, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Lu-Ping Zhang
- Cancer Institute of People's Liberation Army, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Xing-Yun Liao
- Cancer Institute of People's Liberation Army, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Rong-Xia Liao
- Medical English Department, College of Basic Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
61
|
Masoud V, Pagès G. Targeted therapies in breast cancer: New challenges to fight against resistance. World J Clin Oncol 2017; 8:120-134. [PMID: 28439493 PMCID: PMC5385433 DOI: 10.5306/wjco.v8.i2.120] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/16/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common type of cancer found in women and today represents a significant challenge to public health. With the latest breakthroughs in molecular biology and immunotherapy, very specific targeted therapies have been tailored to the specific pathophysiology of different types of breast cancers. These recent developments have contributed to a more efficient and specific treatment protocol in breast cancer patients. However, the main challenge to be further investigated still remains the emergence of therapeutic resistance mechanisms, which develop soon after the onset of therapy and need urgent attention and further elucidation. What are the recent emerging molecular resistance mechanisms in breast cancer targeted therapy and what are the best strategies to apply in order to circumvent this important obstacle? The main scope of this review is to provide a thorough update of recent developments in the field and discuss future prospects for preventing resistance mechanisms in the quest to increase overall survival of patients suffering from the disease.
Collapse
|
62
|
Dufies M, Giuliano S, Ambrosetti D, Claren A, Ndiaye PD, Mastri M, Moghrabi W, Cooley LS, Ettaiche M, Chamorey E, Parola J, Vial V, Lupu-Plesu M, Bernhard JC, Ravaud A, Borchiellini D, Ferrero JM, Bikfalvi A, Ebos JM, Khabar KS, Grépin R, Pagès G. Sunitinib Stimulates Expression of VEGFC by Tumor Cells and Promotes Lymphangiogenesis in Clear Cell Renal Cell Carcinomas. Cancer Res 2017; 77:1212-1226. [PMID: 28087600 DOI: 10.1158/0008-5472.can-16-3088] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 11/16/2022]
Abstract
Sunitinib is an antiangiogenic therapy given as a first-line treatment for renal cell carcinoma (RCC). While treatment improves progression-free survival, most patients relapse. We hypothesized that patient relapse can stem from the development of a lymphatic network driven by the production of the main growth factor for lymphatic endothelial cells, VEGFC. In this study, we found that sunitinib can stimulate vegfc gene transcription and increase VEGFC mRNA half-life. In addition, sunitinib activated p38 MAPK, which resulted in the upregulation/activity of HuR and inactivation of tristetraprolin, two AU-rich element-binding proteins. Sunitinib stimulated a VEGFC-dependent development of lymphatic vessels in experimental tumors. This may explain our findings of increased lymph node invasion and new metastatic sites in 30% of sunitinib-treated patients and increased lymphatic vessels found in 70% of neoadjuvant treated patients. In summary, a therapy dedicated to destroying tumor blood vessels induced the development of lymphatic vessels, which may have contributed to the treatment failure. Cancer Res; 77(5); 1212-26. ©2017 AACR.
Collapse
Affiliation(s)
- Maeva Dufies
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Sandy Giuliano
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
- Biomedical Department, Centre Scientifique de Monaco, Monaco, Principality of Monaco
| | - Damien Ambrosetti
- Central Laboratory of Pathology, Centre Hospitalier Universitaire (CHU) de Nice, Hôpital Pasteur, Nice, France
| | - Audrey Claren
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
- Radiotherapy Department, Centre Antoine Lacassagne, Nice, France
| | - Papa Diogop Ndiaye
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Michalis Mastri
- Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, New York
| | - Walid Moghrabi
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Marc Ettaiche
- Statistics Department, Centre Antoine Lacassagne, Nice, France
| | | | - Julien Parola
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Valerie Vial
- Biomedical Department, Centre Scientifique de Monaco, Monaco, Principality of Monaco
| | - Marilena Lupu-Plesu
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | | | - Alain Ravaud
- Service d'Oncologie Médicale, Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | | | | | | | - John M Ebos
- Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, New York
| | - Khalid Saad Khabar
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Renaud Grépin
- Biomedical Department, Centre Scientifique de Monaco, Monaco, Principality of Monaco
| | - Gilles Pagès
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France.
| |
Collapse
|
63
|
Jahanban-Esfahlan R, Seidi K, Zarghami N. Tumor vascular infarction: prospects and challenges. Int J Hematol 2017; 105:244-256. [DOI: 10.1007/s12185-016-2171-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 12/21/2022]
|
64
|
Martin AR, Ronco C, Demange L, Benhida R. Hypoxia inducible factor down-regulation, cancer and cancer stem cells (CSCs): ongoing success stories. MEDCHEMCOMM 2017; 8:21-52. [PMID: 30108689 PMCID: PMC6071925 DOI: 10.1039/c6md00432f] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022]
Abstract
In cancers, hypoxia inducible factor 1 (HIF-1) is an over-expressed transcription factor, which regulates a large set of genes involved in tumour vascularization, metastases, and cancer stem cells (CSCs) formation and self-renewal. This protein has been identified as a relevant target in oncology and several HIF-1 modulators are now marketed or in advanced clinical trials. The purpose of this review is to summarize the advances in the understanding of its regulation and its inhibition, from the medicinal chemist point of view. To this end, we selected in the recent literature relevant examples of "hit" compounds, including small-sized organic molecules, pseudopeptides and nano-drugs, exhibiting in vitro and/or in vivo both anti-HIF-1 and anti-tumour activities. Whenever possible, a particular emphasis has been dedicated to compounds that selectively target CSCs.
Collapse
Affiliation(s)
- Anthony R Martin
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
| | - Cyril Ronco
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
| | - Luc Demange
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
- UFR des Sciences Pharmaceutiques , Université Paris Descartes , Sorbonne Paris Cité , 4 avenue de l'Observatoire , Paris Fr-75006 , France
- UFR Biomédicale des Saints Pères , 45 rue des Saints Pères , Paris Fr-75006 , France
| | - Rachid Benhida
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
| |
Collapse
|
65
|
Daudigeos-Dubus E, Le Dret L, Bawa O, Opolon P, Vievard A, Villa I, Bosq J, Vassal G, Geoerger B. Dual inhibition using cabozantinib overcomes HGF/MET signaling mediated resistance to pan-VEGFR inhibition in orthotopic and metastatic neuroblastoma tumors. Int J Oncol 2016; 50:203-211. [PMID: 27922668 DOI: 10.3892/ijo.2016.3792] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/06/2016] [Indexed: 11/06/2022] Open
Abstract
MET is expressed on neuroblastoma cells and may trigger tumor growth, neoangiogenesis and metastasis. MET upregulation further represents an escape mechanism to various anticancer treatments including VEGF signaling inhibitors. We developed in vitro a resistance model to pan-VEGFR inhibition and explored the simultaneous inhibition of VEGFR and MET in neuroblastoma models in vitro and in vivo using cabozantinib, an inhibitor of the tyrosine kinases including VEGFR2, MET, AXL and RET. Resistance in IGR-N91-Luc neuroblastoma cells under continuous in vitro exposure pressure to VEGFR1-3 inhibition using axitinib was associated with HGF and p-ERK overexpression. Cabozantinib exhibited anti-proliferative effects in neuroblastoma cells and reduced cell migration in vitro as measured by phase-contrast with IncuCyte system. In vivo, an enhanced number of animals with IGR-N91-Luc metastases was noted following axitinib treatment as compared to control animals. Orally administered cabozantinib per gavage at 30 and 60 mg/kg/day significantly inhibited tumor growth of orthotopic adrenal IGR-N91-Luc and metastatic IMR-32-Luc xenografts. Antitumor activity was associated with decreased vascularization, inhibition of p-SRC and induction of apoptotic cell death. Activation of the HGF-mediated MET pathway is involved in escape to selective VEGFR inhibition in neuroblastoma suggesting combined inhibition of MET and VEGFR signaling to reduce secondary resistance and enhanced invasiveness.
Collapse
Affiliation(s)
- Estelle Daudigeos-Dubus
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ludivine Le Dret
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Olivia Bawa
- Preclinical Evaluation Platform, Gustave Roussy, Villejuif, France
| | - Paule Opolon
- Preclinical Evaluation Platform, Gustave Roussy, Villejuif, France
| | | | - Irène Villa
- Pathology Laboratory, Gustave Roussy, Villejuif, France
| | - Jacques Bosq
- Pathology Laboratory, Gustave Roussy, Villejuif, France
| | - Gilles Vassal
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Birgit Geoerger
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
66
|
孙 鹿, 史 健. [Advance in Research of Angiotensin II and Its Receptor and Malignant Tumor]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 19:615-9. [PMID: 27666553 PMCID: PMC5972952 DOI: 10.3779/j.issn.1009-3419.2016.09.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/26/2015] [Accepted: 12/28/2015] [Indexed: 11/05/2022]
Abstract
Angiotensin AngII, a linear small peptide,which is composed of eight amino acids, is the main effectors of renin-angiotensin systen (Renin-angiotensin system, RAS). AngII, a main biopolypeptide of the RAS, has important pathophysiologic in effects participating in cardiac hypertrophy, vascular cell proproliferation, inflammation and tissue remodeling through G-protein-coupled receptors. In recent years, Ang II can promote tumor cell proliferation, tumor vessel formation and inhibit the differentiation of the tumor cells. This suggests that inhibit the production of AngII or block its effect is expected to become a new measure for the treatment of malignant tumors. This article reviews the advances in research on the relationship between AngII and its receptor and malignant tumor in recent years.
Collapse
Affiliation(s)
- 鹿璐 孙
- 050000 石家庄,河北医科大学Graduate Student of Hebei Medical University, Shijiazhuang 050000, China
| | - 健 史
- 050000 石家庄,河北医科大学Graduate Student of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
67
|
Giuliano S, Cormerais Y, Dufies M, Grépin R, Colosetti P, Belaid A, Parola J, Martin A, Lacas-Gervais S, Mazure NM, Benhida R, Auberger P, Mograbi B, Pagès G. Resistance to sunitinib in renal clear cell carcinoma results from sequestration in lysosomes and inhibition of the autophagic flux. Autophagy 2016; 11:1891-904. [PMID: 26312386 DOI: 10.1080/15548627.2015.1085742] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Metastatic renal cell carcinomas (mRCC) are highly vascularized tumors that are a paradigm for the treatment with antiangiogenesis drugs targeting the vascular endothelial growth factor (VEGF) pathway. The available drugs increase the time to progression but are not curative and the patients eventually relapse. In this study we have focused our attention on the molecular mechanisms leading to resistance to sunitinib, the first line treatment of mRCC. Because of the anarchic vascularization of tumors the core of mRCC tumors receives only suboptimal concentrations of the drug. To mimic this in vivo situation, which is encountered in a neoadjuvant setting, we exposed sunitinib-sensitive mRCC cells to concentrations of sunitinib below the concentration of the drug that gives 50% inhibition of cell proliferation (IC50). At these concentrations, sunitinib accumulated in lysosomes, which downregulated the activity of the lysosomal protease CTSB (cathepsin B) and led to incomplete autophagic flux. Amino acid deprivation initiates autophagy enhanced sunitinib resistance through the amplification of autolysosome formation. Sunitinib stimulated the expression of ABCB1 (ATP-binding cassette, sub-family B [MDR/TAP], member 1), which participates in the accumulation of the drug in autolysosomes and favor its cellular efflux. Inhibition of this transporter by elacridar or the permeabilization of lysosome membranes with Leu-Leu-O-methyl (LLOM) resensitized mRCC cells that were resistant to concentrations of sunitinib superior to the IC50. Proteasome inhibitors also induced the death of resistant cells suggesting that the ubiquitin-proteasome system compensates inhibition of autophagy to maintain a cellular homeostasis. Based on our results we propose a new therapeutic approach combining sunitinib with molecules that prevent lysosomal accumulation or inhibit the proteasome.
Collapse
Affiliation(s)
- Sandy Giuliano
- a University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice; UMR CNRS 7284; INSERM ; Nice , France
| | - Yann Cormerais
- b Centre Scientifique de Monaco Biomedical Department, Monaco, Principality of Monaco
| | - Maeva Dufies
- a University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice; UMR CNRS 7284; INSERM ; Nice , France
| | - Renaud Grépin
- b Centre Scientifique de Monaco Biomedical Department, Monaco, Principality of Monaco
| | - Pascal Colosetti
- c University of Nice Sophia Antipolis; Center Méditerranéen de Médecine Moléculaire; INSERM ; Nice , France
| | - Amine Belaid
- a University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice; UMR CNRS 7284; INSERM ; Nice , France
| | | | - Anthony Martin
- e University of Nice Sophia Antipolis; Institut de Chimie de Nice; UMR CNRS 7272 ; Nice , France
| | - Sandra Lacas-Gervais
- f University of Nice Sophia Antipolis; Center de Microscopie Appliquée ; Nice , France
| | - Nathalie M Mazure
- a University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice; UMR CNRS 7284; INSERM ; Nice , France
| | - Rachid Benhida
- e University of Nice Sophia Antipolis; Institut de Chimie de Nice; UMR CNRS 7272 ; Nice , France
| | - Patrick Auberger
- c University of Nice Sophia Antipolis; Center Méditerranéen de Médecine Moléculaire; INSERM ; Nice , France
| | - Baharia Mograbi
- a University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice; UMR CNRS 7284; INSERM ; Nice , France
| | - Gilles Pagès
- a University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice; UMR CNRS 7284; INSERM ; Nice , France
| |
Collapse
|
68
|
Carpenter RL, Paw I, Zhu H, Sirkisoon S, Xing F, Watabe K, Debinski W, Lo HW. The gain-of-function GLI1 transcription factor TGLI1 enhances expression of VEGF-C and TEM7 to promote glioblastoma angiogenesis. Oncotarget 2016; 6:22653-65. [PMID: 26093087 PMCID: PMC4673189 DOI: 10.18632/oncotarget.4248] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/21/2015] [Indexed: 11/25/2022] Open
Abstract
We recently discovered that truncated glioma-associated oncogene homolog 1 (TGLI1) is highly expressed in glioblastoma (GBM) and linked to increased GBM vascularity. The mechanisms underlying TGLI1-mediated angiogenesis are unclear. In this study, we compared TGLI1- with GLI1-expressing GBM xenografts for the expression profile of 84 angiogenesis-associated genes. The results showed that expression of six genes were upregulated and five were down-regulated in TGLI1-carrying tumors compared to those with GLI1. Vascular endothelial growth factor-C (VEGF-C) and tumor endothelial marker 7 (TEM7) were selected for further investigations because of their significant correlations with high vascularity in 135 patient GBMs. TGLI1 bound to both VEGF-C and TEM7 gene promoters. Conditioned medium from TGLI1-expressing GBM cells strongly induced tubule formation of brain microvascular endothelial cells, and the induction was prevented by VEGF-C/TEM7 knockdown. Immunohistochemical analysis of 122 gliomas showed that TGLI1 expression was positively correlated with VEGF-C, TEM7 and microvessel density. Analysis of NCBI Gene Expression Omnibus datasets with 161 malignant gliomas showed an inverse relationship between tumoral VEGF-C, TEM7 or microvessel density and patient survival. Together, our findings support an important role that TGLI1 plays in GBM angiogenesis and identify VEGF-C and TEM7 as novel TGLI1 target genes of importance to GBM vascularity.
Collapse
Affiliation(s)
- Richard L Carpenter
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Ivy Paw
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Hu Zhu
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sherona Sirkisoon
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Fei Xing
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Waldemar Debinski
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
69
|
Gu Y, Lu H, Boisson-Vidal C, Li H, Bousquet G, Janin A, Di Benedetto M. [Resistance to anti-angiogenic therapy: a clinical and scientific current issue]. Med Sci (Paris) 2016; 32:370-7. [PMID: 27137694 DOI: 10.1051/medsci/20163204015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although the use of anti-angiogenic agents has been considered a promising strategy to block tumor growth and improve the bioavailability of drugs into the tumor, the use of most of them in clinical trials is limited. The development of resistance to some anti-angiogenic agents and their high toxicity are currently under investigations. However, the approach is still valid since this therapeutic tool has lengthened survival of patients with colon, breast, kidney, lungs and liver cancers. The identification of biomarkers in response to this family of drugs is an important area of investigation.
Collapse
Affiliation(s)
- Yuchen Gu
- Inserm UMR-S1165, université Paris Diderot, Sorbonne Paris Cité, 1, avenue Claude Vellefaux, 75010 Paris, France
| | - He Lu
- Inserm UMR-S1165, université Paris Diderot, Sorbonne Paris Cité, 1, avenue Claude Vellefaux, 75010 Paris, France
| | | | - Hong Li
- Microenvironnement et renouvellement cellulaire intégré (MERCI - EA 3829), faculté de médecine et de pharmacie, université de Rouen, France
| | - Guilhem Bousquet
- Inserm UMR-S1165, université Paris Diderot, Sorbonne Paris Cité, 1, avenue Claude Vellefaux, 75010 Paris, France
| | - Anne Janin
- Inserm UMR-S1165, université Paris Diderot, Sorbonne Paris Cité, 1, avenue Claude Vellefaux, 75010 Paris, France
| | - Mélanie Di Benedetto
- Inserm UMR-S1165, université Paris Diderot, Sorbonne Paris Cité, 1, avenue Claude Vellefaux, 75010 Paris, France - Université Paris 13, avenue Jean-Baptiste Clément, 93430 Villetaneuse, France
| |
Collapse
|
70
|
Fleetwood F, Güler R, Gordon E, Ståhl S, Claesson-Welsh L, Löfblom J. Novel affinity binders for neutralization of vascular endothelial growth factor (VEGF) signaling. Cell Mol Life Sci 2016; 73:1671-83. [PMID: 26552422 PMCID: PMC11108507 DOI: 10.1007/s00018-015-2088-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/19/2015] [Accepted: 11/03/2015] [Indexed: 02/06/2023]
Abstract
Angiogenesis denotes the formation of new blood vessels from pre-existing vasculature. Progression of diseases such as cancer and several ophthalmological disorders may be promoted by excess angiogenesis. Novel therapeutics to inhibit angiogenesis and diagnostic tools for monitoring angiogenesis during therapy, hold great potential for improving treatment of such diseases. We have previously generated so-called biparatopic Affibody constructs with high affinity for the vascular endothelial growth factor receptor-2 (VEGFR2), which recognize two non-overlapping epitopes in the ligand-binding site on the receptor. Affibody molecules have previously been demonstrated suitable for imaging purposes. Their small size also makes them attractive for applications where an alternative route of administration is beneficial, such as topical delivery using eye drops. In this study, we show that decreasing linker length between the two Affibody domains resulted in even slower dissociation from the receptor. The new variants of the biparatopic Affibody bound to VEGFR2-expressing cells, blocked VEGFA binding, and inhibited VEGFA-induced signaling of VEGFR2 over expressing cells. Moreover, the biparatopic Affibody inhibited sprout formation of endothelial cells in an in vitro angiogenesis assay with similar potency as the bivalent monoclonal antibody ramucirumab. This study demonstrates that the biparatopic Affibody constructs show promise for future therapeutic as well as in vivo imaging applications.
Collapse
Affiliation(s)
- Filippa Fleetwood
- Division of Protein Technology, School of Biotechnology, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91, Stockholm, Sweden
| | - Rezan Güler
- Division of Protein Technology, School of Biotechnology, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91, Stockholm, Sweden
| | - Emma Gordon
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv. 20, Uppsala, Sweden
| | - Stefan Ståhl
- Division of Protein Technology, School of Biotechnology, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91, Stockholm, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv. 20, Uppsala, Sweden
| | - John Löfblom
- Division of Protein Technology, School of Biotechnology, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91, Stockholm, Sweden.
| |
Collapse
|
71
|
Eccles SA, Court W, Patterson L. In Vitro Assays for Endothelial Cell Functions Required for Angiogenesis: Proliferation, Motility, Tubular Differentiation, and Matrix Proteolysis. Methods Mol Biol 2016; 1430:121-147. [PMID: 27172950 DOI: 10.1007/978-1-4939-3628-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
This chapter deconstructs the process of angiogenesis into its component parts in order to provide simple assays to measure discrete endothelial cell functions. The techniques described will be suitable for studying stimulators and/or inhibitors of angiogenesis and determining which aspect of the process is modulated. The assays are designed to be robust and straightforward, using human umbilical vein endothelial cells, but with an option to use other sources such as microvascular endothelial cells from various tissues or lymphatic endothelial cells. It must be appreciated that such reductionist approaches cannot cover the complexity of the angiogenic process as a whole, incorporating as it does a myriad of positive and negative signals, three-dimensional interactions with host tissues and many accessory cells including fibroblasts, macrophages, pericytes and platelets. The extent to which in vitro assays predict physiological or pathological processes in vivo (e.g., wound healing, tumor angiogenesis) or surrogate techniques such as the use of Matrigel™ plugs, sponge implants, corneal assays etc remains to be determined.
Collapse
Affiliation(s)
- Suzanne A Eccles
- Cancer Research UK Cancer Therapeutics Unit, Centre for Cancer Imaging, The Institute of Cancer Research, Cotswold Rd., Sutton, Surrey, SM2 5NG, UK.
| | - William Court
- Cancer Research UK Cancer Therapeutics Unit, Centre for Cancer Imaging, The Institute of Cancer Research, Cotswold Rd., Sutton, Surrey, SM2 5NG, UK
| | - Lisa Patterson
- Cancer Research UK Cancer Therapeutics Unit, Centre for Cancer Imaging, The Institute of Cancer Research, Cotswold Rd., Sutton, Surrey, SM2 5NG, UK
| |
Collapse
|
72
|
Wang Y, Yan P, Liu Z, Yang X, Wang Y, Shen Z, Bai H, Wang J, Wang Z. MEK inhibitor can reverse the resistance to bevacizumab in A549 cells harboring Kirsten rat sarcoma oncogene homolog mutation. Thorac Cancer 2015; 7:279-87. [PMID: 27148412 PMCID: PMC4846615 DOI: 10.1111/1759-7714.12325] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/10/2015] [Indexed: 01/28/2023] Open
Abstract
Background Bevacizumab (BV) is broadly used to treat a number of cancers; however, BV resistance mechanisms and strategies to overcome this resistance are yet to be determined. Methods We established xenograft mice models harboring Kirsten rat sarcoma oncogene homolog (KRAS) mutations based on the A549 cell line, and tested the responses of xenograft tumors to a series of drugs in ex vivo and in vivo experiments. Changes in transcriptive level were analyzed by ribonucleic acid (RNA) sequencing and the expressions of connexins were determined by immunohistochemistry staining. Results A549 cell mutation type (KRAS G12S) was confirmed by sequencing. After treating the xenograft tumors with BV, the median interval time from BV administration to tumor volume more than 2.5‐fold of the original was 37 days, compared with 21 days in the control (P = 0.025). A549 cells showed resistantance to selumitinib (MEK inhibitor) but were sensitive to selumitinib plus BEZ235 (phosphoinositide 3‐kinase/mammalian target of rapamycin dual inhibitor). However, selumitinib could effectively reverse the resistance to BV in in vivo experiments. RNA sequencing showed that mouse genes, but not human genes, activated the mitogen‐activated protein kinase signaling pathway, accompanied by activation of the Wnt and Hedgehog pathways. Connexin43 (S261) was phosphorylated before and during BV treatment, and subsequently transitioned to negative phosphorylated‐connexin 43‐S261 after resistance to BV. Conclusion Combining an MEK inhibitor with BV was a potential strategy to reverse initial BV resistance. Phosphorylated‐connexin 43 might be associated with the response to BV.
Collapse
Affiliation(s)
- Yuan Wang
- The Department of Ophthalmology National Key Discipline of Pediatrics (Capital Medical University) Ministry of Education Beijing Children's Hospital Capital Medical University Beijing China
| | - Ping Yan
- The Department of Emergency Medicine Qingdao Municipal Hospital Qingdao China
| | - Zhentao Liu
- The Department of Thoracic Medical Oncology Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Peking University Cancer Hospital & Beijing Institute for Cancer Research Beijing China
| | - Xiaodan Yang
- The Department of Thoracic Medical Oncology Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Peking University Cancer Hospital & Beijing Institute for Cancer Research Beijing China
| | - Yaping Wang
- The University of Texas School of Public Health Houston Texas USA
| | - Zhirong Shen
- National Institute of Biological Sciences Zhongguancun Life Science Park Beijing China
| | - Hua Bai
- The Department of Medical Oncology Cancer Hospital Chinese Academy of Medical Sciences Beijing China
| | - Jie Wang
- The Department of Medical Oncology Cancer Hospital Chinese Academy of Medical Sciences Beijing China
| | - Zhijie Wang
- The Department of Thoracic Medical Oncology Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Peking University Cancer Hospital & Beijing Institute for Cancer Research Beijing China
| |
Collapse
|
73
|
Vitale MG, Cartenì G. Clinical management of metastatic kidney cancer: the role of new molecular drugs. Future Oncol 2015; 12:83-93. [PMID: 26617188 DOI: 10.2217/fon.15.283] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Over the last few years, the most recent advances of the molecular mechanisms involved in renal cell carcinoma have led to the use of new drugs targeting VEGF, such as bevacizumab plus interferon, sorafenib, sunitinib, pazopanib, and axitinib, or the mTOR, such as temsirolimus and everolimus. The purpose of this review is to analyze the results of Phase III trial with these targeted agents, and on the management of the treatment and, in particular, when to start and to stop therapy and the use of alternative schedule of sunitinib. Recent developments in immunotherapy are also discussed.
Collapse
Affiliation(s)
- Maria Giuseppa Vitale
- UOSC Oncologia Medica, Azienda Ospedaliera di Rilievo Nazionale 'Antonio Cardarelli', 80131 Naples, Italy
| | - Giacomo Cartenì
- UOSC Oncologia Medica, Azienda Ospedaliera di Rilievo Nazionale 'Antonio Cardarelli', 80131 Naples, Italy
| |
Collapse
|
74
|
Do K, Cao L, Kang Z, Turkbey B, Lindenberg ML, Larkins E, Holkova B, Steinberg SM, Raffeld M, Peer CJ, Figg WD, Eugeni M, Jacobs P, Choyke P, Wright JJ, Doroshow JH, Kummar S. A Phase II Study of Sorafenib Combined With Cetuximab in EGFR-Expressing, KRAS-Mutated Metastatic Colorectal Cancer. Clin Colorectal Cancer 2015; 14:154-61. [PMID: 25861837 PMCID: PMC6657351 DOI: 10.1016/j.clcc.2015.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/19/2015] [Accepted: 02/26/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mutations in the KRAS gene predict for resistance to anti-epidermal growth factor receptor (EGFR) therapies, including cetuximab. Upregulation of vascular endothelial growth factor (VEGF)-A has been implicated in resistance to anti-EGFR treatment. Abrogation of the VEGF and RAS/RAF/MEK/ERK pathways has the potential to restore cetuximab sensitivity. PATIENTS AND METHODS Adult patients with histologically documented, measurable, EGFR-expressing, KRAS-mutated metastatic colorectal cancer (mCRC) that had progressed after 5-fluorouracil-based regimens were treated with sorafenib 400 mg orally twice daily and intravenous cetuximab weekly in 28-day cycles. The primary endpoint was the response rate (complete response, partial response, and stable disease at 4 cycles). The secondary endpoints included plasma biomarker analysis of angiogenic cytokines and correlative imaging studies with dynamic contrast-enhanced magnetic resonance imaging and zirconium 89-panitumumab. RESULTS Of the 30 patients enrolled, 26 were evaluable for response. Of the 26 patients evaluated, 4 had stable disease at 4 cycles and 1 had stable disease at 8 cycles. The median progression-free survival was 1.84 months. The common toxicities were rash, diarrhea, and liver enzyme elevations. Of the angiogenic cytokines evaluated, only the placental growth factor increased significantly with treatment (P < .0001). No pharmacodynamic parameters were associated with the treatment response. CONCLUSION We report the results of a trial that combined cetuximab and sorafenib for the treatment of KRAS-mutated mCRC, with correlative imaging studies and pharmacodynamic angiogenic cytokine profiling as downstream markers of EGFR and VEGF receptor (VEGFR) signaling. No objective responses were observed. Additional development of biomarkers for patient selection is needed to evaluate combined EGFR and VEGFR blockade as a therapeutic option in KRAS-mutated CRC.
Collapse
Affiliation(s)
- Khanh Do
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Liang Cao
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Zhigang Kang
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Baris Turkbey
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | | | - Erin Larkins
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Beata Holkova
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Seth M Steinberg
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Mark Raffeld
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Cody J Peer
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - William D Figg
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Michelle Eugeni
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Paula Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Peter Choyke
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - John J Wright
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD.
| |
Collapse
|
75
|
CHEN DARREN, LIN CHE, WANG YUFEN. Window of opportunity: A new insight into sequential bevacizumab and paclitaxel in two cases of metastatic triple-negative breast cancer. Exp Ther Med 2015; 10:885-888. [PMID: 26622409 PMCID: PMC4533205 DOI: 10.3892/etm.2015.2586] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 06/05/2015] [Indexed: 01/23/2023] Open
Abstract
Bevacizumab, an antiangiogenic monoclonal antibody against vascular endothelial growth factor, was designed to normalize tumor vasculature and reduce intratumoral pressure. It can create a 'normalization window' during which the cancer can be attacked the most effectively, and the effects of chemotherapeutic drugs are enhanced. Representative trials (E2100, AVADO, RIBBON-1, RIBBON-2 and TURANDOT) have shown that the addition of bevacizumab to chemotherapy has significant benefits on progression-free survival for metastatic breast cancer, but not on overall survival. The present study describes two patients with metastatic triple-negative breast cancer who received 6 courses of bevacizumab-containing chemotherapy. Each course comprised 5-7.5 mg/kg bevacizumab administered on days 1 and 15, and 20-24 h after bevacizumab delivery, 80 mg/m2 paclitaxel was administered for 3 weeks on days 2, 9 and 16, followed by 1 week of rest. Following sequential treatment with bevacizumab and paclitaxel, the results of computed tomography showed that the tumors were rapidly reduced in size. Based on the imaging findings from three-dimension power Doppler ultrasonography in one of the breast cancer patients who received neoadjuvant chemotherapy with bevacizumab, the possible timing of the normalization window was 20-24 h after the administration of bevacizumab. The normalization window may provide an opportunity to enhance the effect of chemotherapy with the aid of bevacizumab.
Collapse
Affiliation(s)
- DAR-REN CHEN
- Comprehensive Breast Cancer Center, Changhua Christian Hospital, Changhua 50006, Taiwan, R.O.C
- Cancer Research Center, Changhua Christian Hospital, Changhua 50006, Taiwan, R.O.C
| | - CHE LIN
- Comprehensive Breast Cancer Center, Changhua Christian Hospital, Changhua 50006, Taiwan, R.O.C
- Department of Environmental Engineering, National Chung-Hsing University, Taichung 40227, Taiwan, R.O.C
| | - YU-FEN WANG
- Cancer Research Center, Changhua Christian Hospital, Changhua 50006, Taiwan, R.O.C
| |
Collapse
|
76
|
Lee JA, Shinn P, Jaken S, Oliver S, Willard FS, Heidler S, Peery RB, Oler J, Chu S, Southall N, Dexheimer TS, Smallwood J, Huang R, Guha R, Jadhav A, Cox K, Austin CP, Simeonov A, Sittampalam GS, Husain S, Franklin N, Wild DJ, Yang JJ, Sutherland JJ, Thomas CJ. Novel Phenotypic Outcomes Identified for a Public Collection of Approved Drugs from a Publicly Accessible Panel of Assays. PLoS One 2015; 10:e0130796. [PMID: 26177200 PMCID: PMC4503722 DOI: 10.1371/journal.pone.0130796] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
Phenotypic assays have a proven track record for generating leads that become first-in-class therapies. Whole cell assays that inform on a phenotype or mechanism also possess great potential in drug repositioning studies by illuminating new activities for the existing pharmacopeia. The National Center for Advancing Translational Sciences (NCATS) pharmaceutical collection (NPC) is the largest reported collection of approved small molecule therapeutics that is available for screening in a high-throughput setting. Via a wide-ranging collaborative effort, this library was analyzed in the Open Innovation Drug Discovery (OIDD) phenotypic assay modules publicly offered by Lilly. The results of these tests are publically available online at www.ncats.nih.gov/expertise/preclinical/pd2 and via the PubChem Database (https://pubchem.ncbi.nlm.nih.gov/) (AID 1117321). Phenotypic outcomes for numerous drugs were confirmed, including sulfonylureas as insulin secretagogues and the anti-angiogenesis actions of multikinase inhibitors sorafenib, axitinib and pazopanib. Several novel outcomes were also noted including the Wnt potentiating activities of rotenone and the antifolate class of drugs, and the anti-angiogenic activity of cetaben.
Collapse
Affiliation(s)
- Jonathan A. Lee
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Paul Shinn
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Susan Jaken
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Sarah Oliver
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Francis S. Willard
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Steven Heidler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Robert B. Peery
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Jennifer Oler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Shaoyou Chu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Noel Southall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas S. Dexheimer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey Smallwood
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ajit Jadhav
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Karen Cox
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Christopher P. Austin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - G. Sitta Sittampalam
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Saba Husain
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Natalie Franklin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - David J. Wild
- Indiana University School of Informatics and Computing, Bloomington, Indiana, United States of America
| | - Jeremy J. Yang
- Indiana University School of Informatics and Computing, Bloomington, Indiana, United States of America
| | - Jeffrey J. Sutherland
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
- * E-mail: (JJS); (CJT)
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JJS); (CJT)
| |
Collapse
|
77
|
Vergote I, Leamon CP. Vintafolide: a novel targeted therapy for the treatment of folate receptor expressing tumors. Ther Adv Med Oncol 2015; 7:206-18. [PMID: 26136852 DOI: 10.1177/1758834015584763] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Despite advances in the development of molecularly targeted therapies, limited improvements in overall survival have been noted among many cancer patients with solid tumors, primarily due to development of drug resistance. Accordingly, there is an unmet need for new targeted therapies and treatment approaches for cancer, especially for overcoming resistance. Expression of the folate receptor is upregulated in many tumor types and thus represents an ideal target for cancer treatment. Several folate receptor targeted therapies are in development, including the small molecule drug conjugate vintafolide, the monoclonal antibody farletuzumab, and the antibody-drug conjugate IMGN853. The role of the folate receptor as a target in cancer progression and resistance as well as emerging preclinical and clinical data from studies on those folate receptor targeted agents that are in development with a focus on vintafolide are reviewed. The folate receptor has several unique properties, such as high expression in several tumor types, that make it a rational target for cancer treatment, and allow for selective delivery of folate receptor targeted agents. Early-stage clinical data in lung and ovarian cancer suggest that vintafolide has the potential for combination with other standard approved agents.
Collapse
|
78
|
Yang F, Gong L, Jin H, Pi J, Bai H, Wang H, Cai H, Yang P, Cai J. Chrysin-organogermanium (IV) complex induced Colo205 cell apoptosis-associated mitochondrial function and anti-angiogenesis. SCANNING 2015; 37:246-257. [PMID: 25914235 DOI: 10.1002/sca.21205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/16/2015] [Accepted: 02/24/2015] [Indexed: 06/04/2023]
Abstract
Colorectal cancer, a kind of malignant cancer, has more than 1 million new patients and results in 0.5 million deaths every year globally based on the estimation of Globocan in 2008. One of the most important issues against colon cancer is tumor metastasis. Anti-angiogenesis, a form of targeted therapy uses drugs or other substances to prevent the new blood vessel formation, which is critical for tumor metastasis. In our previous studies, we have demonstrated a simple method to synthesize Chry-Ge complex through the reaction between chrysin and triphenylgermanium bromide. In this work, we investigated the mechanism of Chry-Ge induced Colo205 cell apoptosis. We found that Chry-Ge could induce apoptosis in Colo205 cells in mitochondrial-dependent pathway, cause the reorganization of cytoskeleton and induce the damage of nucleus in Colo205 cells. Besides, Chry-Ge was also found to induce membrane ultrastructural changes in Colo205 cells by AFM. Further, we found that Chry-Ge can inhibit tube formation of human umbilical vascular endothelial cell in vitro. Chry-Ge was also tested in vivo in the chicken chorioallantoic membrane (CAM) assay and found to inhibit bFGF-treated CAMs development. These results suggested that Chry-Ge could induce Colo205 cell apoptosis by mitochondrial pathway and anti-angiogenesis, highlighting the use of organic germanium agents for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Fen Yang
- Department of Chemistry of Jinan University, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Longcai Gong
- Department of Bioengineering, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Hua Jin
- Department of Chemistry of Jinan University, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jiang Pi
- Department of Chemistry of Jinan University, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Haihua Bai
- Department of Chemistry of Jinan University, Guangzhou, China
| | - Hong Wang
- Department of Bioengineering, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Huaihong Cai
- Department of Chemistry of Jinan University, Guangzhou, China
| | - Peihui Yang
- Department of Chemistry of Jinan University, Guangzhou, China
| | - Jiye Cai
- Department of Chemistry of Jinan University, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| |
Collapse
|
79
|
AECHL-1, a novel triterpenoid, targets tumor neo-vasculature and impairs the endothelial cell cytoskeleton. Angiogenesis 2015; 18:283-99. [PMID: 25952529 PMCID: PMC4472952 DOI: 10.1007/s10456-015-9466-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/29/2015] [Indexed: 11/26/2022]
Abstract
Tumor angiogenesis is characterized by abnormal vessel morphology leading to erratic and insufficient delivery of chemotherapeutics and oxygen, making the tumor core not only highly hypoxic but also unresponsive toward treatment. Such hypoxic conditions promote tumor aggressiveness, leading to the establishment of metastatic disease. Most anti-angiogenic treatments aim toward the destruction of tumor vasculature, which proves countereffective by further increasing its aggressive nature. Hence, developing drugs which target or regulate these processes might lead to a better delivery of chemotherapeutics resulting in tumor shrinkage. Plant-derived natural compounds having a bioactive ingredient, especially triterpenoids, have been known to possess anticancer properties. AECHL-1, a recently isolated novel triterpenoid with proven anticancer potential, is seemingly noncytotoxic toward HEK 293 and HUVECs. Also, cytotoxicity was absent during in vivo studies involving intraperitoneal injections with 5 µg/kg body weight AECHL-1 on SCID mice. When used at subtoxic doses, it was found to be effective in suppression of neo-vessel formation as demonstrated in the chick chorioallantoic membrane, rat aortic rings, Matrigel plugs and xenograft tumors implanted in SCID mice. Tumor vasculature from AECHL-1-treated mice showed greater mural cell coverage and relatively normalized architecture. Investigations into the molecular mechanisms responsible for these observations revealed an effect on the actin cytoskeleton of stimulated HUVECs as well as the VEGFR2-mediated MAPK pathway. AECHL-1 could effectively distinguish between stimulated and nonstimulated endothelial cells. AECHL-1 could also downregulate HIF-1α expression and VEGF secretion under hypoxic conditions, thus reducing the fears of unnecessarily aggravating tumor metastasis as a result of anti-angiogenic therapy. Results obtained from the aforementioned studies make it clear that though AECHL-1 shows promise in discouraging and pruning neo-vasculature, it may not affect existing vasculature as the doses used for the assays are significantly lower than the ones causing endothelial cell death and has potential to be considered as a candidate for therapeutic drug development.
Collapse
|
80
|
The anticancer effect of Huaier (Review). Oncol Rep 2015; 34:12-21. [PMID: 25955759 DOI: 10.3892/or.2015.3950] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/24/2015] [Indexed: 12/18/2022] Open
Abstract
Trametes robiniophila Murr. (Huaier) is a sandy beige mushroom found on the trunks of trees and has been widely used in traditional Chinese medicine (TCM) for ~1,600 years. The anticancer effects of Huaier have attracted increasing worldwide interest in recent years. Accumulating evidence suggests that the anticancer mechanism of Huaier may be associated with various biological activities, such as inhibition of cell proliferation, anti-metastasis, interference with tumor angiogenesis and tumor-specific immunomodulatory effect. Animal and experimental studies suggest that Huaier is a promising anticancer agent. Further clinical research is warranted to illustrate the untapped chemopreventive and therapeutic potential of Huaier either alone or in conjunction with existing therapies.
Collapse
|
81
|
Wentink MQ, Huijbers EJM, de Gruijl TD, Verheul HMW, Olsson AK, Griffioen AW. Vaccination approach to anti-angiogenic treatment of cancer. Biochim Biophys Acta Rev Cancer 2015; 1855:155-71. [PMID: 25641676 DOI: 10.1016/j.bbcan.2015.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/16/2015] [Accepted: 01/22/2015] [Indexed: 02/07/2023]
Abstract
Improvement of patient survival by anti-angiogenic therapy has proven limited. A vaccination approach inducing an immune response against the tumor vasculature combines the benefits of immunotherapy and anti-angiogenesis, and may overcome the limitations of current anti-angiogenic drugs. Strategies to use whole endothelial cell vaccines and DNA- or protein vaccines against key players in the VEGF signaling axis, as well as specific markers of tumor endothelial cells, have been tested in preclinical studies. Current clinical trials are now testing the promise of this specific anti-cancer vaccination approach. This review will highlight the state-of-the-art in this exciting field of cancer research.
Collapse
Affiliation(s)
- Madelon Q Wentink
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Henk M W Verheul
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
82
|
Dynamic contrast-enhanced micro-computed tomography correlates with 3-dimensional fluorescence ultramicroscopy in antiangiogenic therapy of breast cancer xenografts. Invest Radiol 2015; 49:445-56. [PMID: 24598441 DOI: 10.1097/rli.0000000000000038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Dynamic contrast-enhanced (DCE) micro-computed tomography (micro-CT) has emerged as a valuable imaging tool to noninvasively obtain quantitative physiological biomarkers of drug effect in preclinical studies of antiangiogenic compounds. In this study, we explored the ability of DCE micro-CT to assess the antiangiogenic treatment response in breast cancer xenografts and correlated the results to the structural vessel response obtained from 3-dimensional (3D) fluorescence ultramicroscopy (UM). MATERIAL AND METHODS Two groups of tumor-bearing mice (KPL-4) underwent DCE micro-CT imaging using a fast preclinical dual-source micro-CT system (TomoScope Synergy Twin, CT Imaging GmbH, Erlangen, Germany). Mice were treated with either a monoclonal antibody against the vascular endothelial growth factor or an unspecific control antibody. Changes in vascular physiology were assessed measuring the mean value of the relative blood volume (rBV) and the permeability-surface area product (PS) in different tumor regions of interest (tumor center, tumor periphery, and total tumor tissue). Parametric maps of rBV were calculated of the tumor volume to assess the intratumoral vascular heterogeneity. Isotropic 3D UM vessel scans were performed from excised tumor tissue, and automated 3D segmentation algorithms were used to determine the microvessel density (MVD), relative vessel volume, and vessel diameters. In addition, the accumulation of coinjected fluorescence-labeled trastuzumab was quantified in the UM tissue scans to obtain an indirect measure of vessel permeability. Results of the DCE micro-CT were compared with corresponding results obtained by ex vivo UM. For validation, DCE micro-CT and UM parameters were compared with conventional histology and tumor volume. RESULTS Examination of the parametric rBV maps revealed significantly different patterns of intratumoral blood supply between treated and control tumors. Whereas control tumors showed a characteristic vascular rim pattern with considerably elevated rBV values in the tumor periphery, treated tumors showed a widely homogeneous blood supply. Compared with UM, the physiological rBV maps showed excellent agreement with the spatial morphology of the intratumoral vascular architecture. Regional assessment of mean physiological values exhibited a significant decrease in rBV (P < 0.01) and PS (P < 0.05) in the tumor periphery after anti-vascular endothelial growth factor treatment. Structural validation with UM showed a significant reduction in reduction of relative vessel volume (rVV) (P < 0.01) and MVD (P < 0.01) in the corresponding tumor region. The reduction in rBV correlated well with the rVV (R = 0.73 for single values and R = 0.95 for mean values). Spatial maps of antibody penetration showed a significantly reduced antibody accumulation (P < 0.01) in the tumor tissue after treatment and agreed well with the physiological change of PS. Examination of vessel diameters revealed a size-dependent antiangiogenic treatment effect, which showed a significant reduction in MVD (P < 0.001) for vessels with diameters smaller than 25 μm. No treatment effect was observed by tumor volume. CONCLUSIONS Noninvasive DCE micro-CT provides valuable physiological information of antiangiogenic drug effect in the intact animal and correlates with ex vivo structural analysis of 3D UM. The combined use of DCE micro-CT with UM constitutes a complementary imaging toolset that can help to enhance our understanding of antiangiogenic drug mechanisms of action in preclinical drug research.
Collapse
|
83
|
Mackey JR, Ramos-Vazquez M, Lipatov O, McCarthy N, Krasnozhon D, Semiglazov V, Manikhas A, Gelmon KA, Konecny GE, Webster M, Hegg R, Verma S, Gorbunova V, Abi Gerges D, Thireau F, Fung H, Simms L, Buyse M, Ibrahim A, Martin M. Primary results of ROSE/TRIO-12, a randomized placebo-controlled phase III trial evaluating the addition of ramucirumab to first-line docetaxel chemotherapy in metastatic breast cancer. J Clin Oncol 2015; 33:141-8. [PMID: 25185099 DOI: 10.1200/jco.2014.57.1513] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Currently, antiangiogenic strategies in metastatic breast cancer have demonstrated modest improvements in progression-free survival (PFS) but not improved quality or duration of survival, warranting evaluation of new agents in a placebo-controlled setting. Ramucirumab is a human immunoglobulin G1 antibody that binds vascular endothelial growth factor receptor-2 and blocks ligand-stimulated activation. The ROSE/TRIO-012 trial evaluated ramucirumab with docetaxel in unresectable, locally recurrent, or metastatic breast cancer. PATIENTS AND METHODS In this double-blind, placebo-controlled, randomized, multinational phase III trial, 1,144 patients with human epidermal growth factor receptor 2 (HER2) -negative breast cancer who had not received cytotoxic chemotherapy in the advanced setting were randomly assigned at a two-to-one ratio to receive docetaxel 75 mg/m(2) plus ramucirumab 10 mg/kg or docetaxel 75 mg/m(2) plus placebo once every 3 weeks. Treatment continued until disease progression, unacceptable toxicity, or other withdrawal criteria. Patients were stratified by previous taxane therapy, visceral metastasis, hormone receptor status, and geographic region. An independent data monitoring committee oversaw the trial. The primary end point was investigator-assessed PFS. RESULTS Median PFS in patients treated with ramucirumab plus docetaxel was 9.5 months, compared with 8.2 months in patients who received placebo plus docetaxel (hazard ratio [HR], 0.88; P = .077). Median overall survival was 27.3 months in patients who received ramucirumab plus docetaxel, compared with 27.2 months in patients who received placebo plus docetaxel (HR, 1.01; P = .915). Toxicities seen at significantly higher rates in patients receiving ramucirumab included fatigue, hypertension, febrile neutropenia, palmar-plantar erythrodysesthesia syndrome, and stomatitis. CONCLUSION Addition of ramucirumab to docetaxel in HER2-negative advanced breast cancer did not meaningfully improve important clinical outcomes.
Collapse
Affiliation(s)
- John R Mackey
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ.
| | - Manuel Ramos-Vazquez
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Oleg Lipatov
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Nicole McCarthy
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Dmitriy Krasnozhon
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Vladimir Semiglazov
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Alexey Manikhas
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Karen A Gelmon
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Gottfried E Konecny
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Marc Webster
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Roberto Hegg
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Sunil Verma
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Vera Gorbunova
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Dany Abi Gerges
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Francois Thireau
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Helena Fung
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Lorinda Simms
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Marc Buyse
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Ayman Ibrahim
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| | - Miguel Martin
- John R. Mackey, Cross Cancer Institute; Francois Thireau and Helena Fung, Translational Research in Oncology, Edmonton; Marc Webster, Tom Baker Cancer Centre, Calgary, Alberta; Karen A. Gelmon, British Columbia Cancer Agency, Vancouver, British Columbia; Sunil Verma, Sunnybrook Health Sciences Center; Lorinda Simms, Eli Lilly, Toronto, Ontario, Canada; Manuel Ramos-Vazquez, Centro Oncológico de Galicia "José Antonio Quiroga y Piñeiro," A Coruña; Miguel Martin, Instituto de Investigación Sanitaria Gregorio Marañon, Universidad Complutense, Madrid, Spain; Oleg Lipatov, Republican Clinical Oncology Dispensary, Bashkortostan Republic Ministry of Health, Ufa; Dmitriy Krasnozhon, Leningrad Regional Oncology Dispensary, Leningrad; Vladimir Semiglazov, Institute of Oncology N.N. Petrov; Alexey Manikhas, City Clinical Oncology Dispensary, St Petersburg; Vera Gorbunova, N.N. Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia; Nicole McCarthy, ICON Cancer Care Wesley, Brisbane, Queensland, Australia; Gottfried E. Konecny, University of California Los Angeles, Los Angeles, CA; Roberto Hegg, Hospital Pérola Byigton Centro de Referência da Saúde da Mulher, Sao Paulo, Brazil; Dany Abi Gerges, Middle East Institute of Health, Bsalim, Lebanon; Marc Buyse, International Drug Development Institute, Louvain-la-Neuve, Belgium; and Ayman Ibrahim, ImClone Systems, Bridgewater, NJ
| |
Collapse
|
84
|
|
85
|
Huang L, Hu C, DI Benedetto M, Varin R, Liu J, Jin J, Wang L, Vannier JP, Janin A, Lu H, Li H. Cross-drug resistance to sunitinib induced by doxorubicin in endothelial cells. Oncol Lett 2014; 9:1287-1292. [PMID: 25663899 PMCID: PMC4315062 DOI: 10.3892/ol.2014.2819] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 10/15/2014] [Indexed: 01/08/2023] Open
Abstract
Multiple drug resistance remains an unsolved problem in cancer therapy. A previous study has demonstrated that the chemotherapeutic drug doxorubicin (Dox) induced upregulation of P-glycoprotein in endothelial cells, resulting in a 20-fold increase in drug resistance and reduced efficiency of doxorubicin treatment in a mouse tumor model. In the present study, the cross-resistance and sensitivity of HMECd1 and HMECd2 established cell lines to anti-angiogenic drugs, particularly sunitinib, was explored. The results revealed that Dox treatment induced a significant increase in the breast cancer resistance protein (ABCG2) gene transcription and protein expression. This increase gave rise to a 4- to 5-fold increase in the half maximal inhibitory concentration of the HMECd1 and HMECd2 cells in response to sunitinib treatment in vitro. Functionally, the role of ABCG2 in the resistance to sunitinib was confirmed by the use of the ABCG2 inhibitors fumitremorgin C and diethylstilbestrol, which blocked cell resistance. The present study indicates that endothelial cells exhibit cross-resistance between cytotoxic drugs and anti-angiogenic drugs. This suggests that multiple drug resistance induced by chemotherapy in endothelial cells may affect the efficiency of anti-angiogenic drugs.
Collapse
Affiliation(s)
- Limin Huang
- Department of Oncology, People's Hospital of Guizhou Province, Guiyang, Guizhou 550000, P.R. China
| | - Chaoquan Hu
- Department of Surgery, Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Mélanie DI Benedetto
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France
| | - Rémi Varin
- Laboratory of MERCI (EA 3829), Faculty of Medicine and Pharmacy, University of Rouen, Rouen 76183, France
| | - Jielin Liu
- Department of Surgery, Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China ; French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France
| | - Jian Jin
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France ; School of Medicine and Pharmaceutics, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Li Wang
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France
| | - Jean-Pierre Vannier
- Laboratory of MERCI (EA 3829), Faculty of Medicine and Pharmacy, University of Rouen, Rouen 76183, France
| | - Anne Janin
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France ; Laboratory of Pathology, Paris Diderot University, Sorbonne Paris Cité, UMR-S 1165, France ; Saint-Louis Hospital, Laboratory of Pathology, Paris 75010, France
| | - He Lu
- French Institute of Health and Medical Research, UMR-S 1165, University Institute of Hematology, Saint Louis Hospital, Paris 75010, France ; Laboratory of Pathology, Paris Diderot University, Sorbonne Paris Cité, UMR-S 1165, France
| | - Hong Li
- Laboratory of MERCI (EA 3829), Faculty of Medicine and Pharmacy, University of Rouen, Rouen 76183, France
| |
Collapse
|
86
|
Huang L, Hu C, Di Benedetto M, Varin R, Liu J, Wang L, Vannier JP, Jin J, Janin A, Lu H, Li H. Induction of multiple drug resistance in HMEC-1 endothelial cells after long-term exposure to sunitinib. Onco Targets Ther 2014; 7:2249-55. [PMID: 25587220 PMCID: PMC4262216 DOI: 10.2147/ott.s67251] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Multiple drug resistance is still an unsolved problem in cancer therapy. Our previous study demonstrated that the chemotherapeutic drug doxorubicin (Dox) induced upregulation of P-glycoprotein (P-gp) in endothelial cells, resulting in a 20-fold increase in drug resistance and reduced efficiency of Dox treatment in a mice tumor model. In this study, we exposed human microvascular endothelial cells (HMEC-1) to sunitinib, a tyrosine kinase receptor inhibitor, to induce drug resistance. The results show that sunitinib treatment induced multiple drug resistance in these cells. They became resistant not only to sunitinib but also to Dox, paclitaxel, and vinblastine. Significant increases in P-gp (9.3-fold), ABCG2 (breast cancer resistance protein, 1.9-fold), and multidrug resistance-associated protein 1 (2.7-fold) gene transcription were found by quantitative polymerase chain reaction quantification, and their protein expression was confirmed by Western blot. These increases gave rise to an approximately five-fold increase in half maximal inhibitory concentration in these cells in response to sunitinib treatment in vitro. The inhibitors of adenosine triphosphate-binding cassette transporters did not reverse the drug resistance in sunitinib-resistant HMEC-1 cells, assumedly because of a blockage of the pump function caused by sunitinib. Our study indicates that the antiangiogenic drug sunitinib induces multiple drug resistance in endothelial cells. The induction of adenosine triphosphate-binding cassette transporters seems not to be responsible for observed multiple drug resistance, and the underlying mechanisms remain unknown.
Collapse
Affiliation(s)
- Limin Huang
- Department of Oncology, People's Hospital of Guizhou Province, Guiyang, People's Republic of China
| | - Chaoquan Hu
- Department of Surgery, Affiliated Hospital, Guiyang Medical University, Guiyang, People's Republic of China
| | | | - Rémi Varin
- MERCI (EA 3829), Faculté de Médecine et de Pharmacie, Université de Rouen, Rouen, France
| | - Jielin Liu
- Department of Surgery, Affiliated Hospital, Guiyang Medical University, Guiyang, People's Republic of China ; INSERM UMR-S 1165, Paris, France
| | - Li Wang
- INSERM UMR-S 1165, Paris, France
| | - Jean-Pierre Vannier
- MERCI (EA 3829), Faculté de Médecine et de Pharmacie, Université de Rouen, Rouen, France
| | - Jian Jin
- INSERM UMR-S 1165, Paris, France ; School of Medicine and Pharmaceutics, Jiangnan University, Wuxi, People's Republic of China
| | - Anne Janin
- INSERM UMR-S 1165, Paris, France ; Université Paris Diderot, Laboratoire de Pathologie, Paris, France ; AP-HP-Hôpital Saint-Louis, Laboratoire de Pathologie, Paris, France
| | - He Lu
- INSERM UMR-S 1165, Paris, France ; Université Paris Diderot, Laboratoire de Pathologie, Paris, France
| | - Hong Li
- MERCI (EA 3829), Faculté de Médecine et de Pharmacie, Université de Rouen, Rouen, France
| |
Collapse
|
87
|
Zhang LL, Wu J, Liu Q, Zhang Y, Sun ZL, Jing H. MiR-886-5p inhibition inhibits growth and induces apoptosis of MCF7 cells. Asian Pac J Cancer Prev 2014; 15:1511-5. [PMID: 24641359 DOI: 10.7314/apjcp.2014.15.4.1511] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND AND AIMS To explore the molecular mechanisms of miR-886-5p in breast cancer., we examined roles in inhibiting growth and migration of MCF-7 cells. METHODS MiR-886-5p mimics and inhibitors were used to express or inhibit MiR-886-5p, respectively, and MTT and clone formation assays were used to determine the survival and proliferation. Hoechst 33342/ PI double staining was applied to detect apoptosis. The expression of caspase-3, caspase-8, caspase-9, MT1-MMP, VEGF-C and VEGF-D was detected by Western blotting, and the levels of MMP2 and MMP9 secreted from MCF-7 cells were assessed by ELISA. MCF-7 cell migration was determined by wound healing and Transwell assays. RESULTS We found that the growth of MCF-7 cells was inhibited upon decreasing miR-886-5p levels. Inhibiting miR-866-5p also significantly induced apoptosis and decreased the migratory capacity of these cells. The expression of VEGF-C, VEGF-D, MT1-MMP, MMP2, and MMP9 was also found to be decreased as compared to controls. CONCLUSIONS Our data show that downregulation of miR-886-5p expression in MCF-7 cells could significantly inhibit cell growth and migration. This might imply that inhibiting miR-886-5p could be a therapeutic strategy in breast cancer.
Collapse
Affiliation(s)
- Lei-Lei Zhang
- Department of General Pathology, Huaihe Hospital, Henan University, Kai Feng, China E-mail :
| | | | | | | | | | | |
Collapse
|
88
|
Voelkel NF, Gomez-Arroyo J. The Role of Vascular Endothelial Growth Factor in Pulmonary Arterial Hypertension. The Angiogenesis Paradox. Am J Respir Cell Mol Biol 2014; 51:474-84. [DOI: 10.1165/rcmb.2014-0045tr] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
89
|
Menhofer MH, Bartel D, Liebl J, Kubisch R, Busse J, Wagner E, Müller R, Vollmar AM, Zahler S. In vitro and in vivo characterization of the actin polymerizing compound chondramide as an angiogenic inhibitor. Cardiovasc Res 2014; 104:303-14. [PMID: 25239826 DOI: 10.1093/cvr/cvu210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIMS Inhibiting angiogenesis is a major approach in tumour therapy. To combat angiogenesis, the tubulin cytoskeleton has emerged as an interesting target in many pre- and clinical studies. Contrarily, the actin cytoskeleton has been largely neglected as a potential drug target in angiogenesis. However, due to the development of drug resistances, new therapeutic strategies are always needed in tumour treatment. Therefore, the therapeutic potential of actin-binding small molecules is of particular interest. METHODS AND RESULTS We investigate the impact of chondramide (Ch), an actin polymerizing myxobacterial compound, on angiogenesis and underlying signalling. Chondramide treatment not only reduces the migration of endothelial cells but also the maturation of endothelial tube networks on matrigel. These observations can partly be explained by a disintegration of stress fibres due to aggregation and subsequent accumulation of actin in cellular structures known as 'aggresomes'. Chondramide treatment impairs the maturation of focal adhesions and reduces the amount of active β1 integrin at the cell surface. Accordingly, signalling events downstream of focal adhesions are reduced. Thus, we observed that the activity of Src and downstream factors Rho-GTPases Rac1 and Rho is reduced upon Ch treatment. In vivo, Ch was well tolerated in mice and vascularization of a tumour xenograft as well as of the developing retina was significantly reduced. CONCLUSION Chondramide diminishes angiogenesis via two ways: (i) the disintegration of stress fibres and (ii) the reduction of promigratory signals. Our findings highlight Ch as a novel class of therapeutic lead compound with anti-angiogenic potential.
Collapse
Affiliation(s)
- Magdalena H Menhofer
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Dominik Bartel
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Johanna Liebl
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Rebekka Kubisch
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Johanna Busse
- Department of Pharmacy, Pharmaceutical Biotechnology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Ernst Wagner
- Department of Pharmacy, Pharmaceutical Biotechnology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Stefan Zahler
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| |
Collapse
|
90
|
Serio RN. Wnt of the Two Horizons: Putting Stem Cell Self-Renewal and Cell Fate Determination into Context. Stem Cells Dev 2014; 23:1975-90. [DOI: 10.1089/scd.2014.0055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Ryan N. Serio
- Graduate School of Pharmacology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
91
|
Sounni NE, Cimino J, Blacher S, Primac I, Truong A, Mazzucchelli G, Paye A, Calligaris D, Debois D, De Tullio P, Mari B, De Pauw E, Noel A. Blocking lipid synthesis overcomes tumor regrowth and metastasis after antiangiogenic therapy withdrawal. Cell Metab 2014; 20:280-94. [PMID: 25017943 DOI: 10.1016/j.cmet.2014.05.022] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 04/01/2014] [Accepted: 05/15/2014] [Indexed: 01/21/2023]
Abstract
The molecular mechanisms responsible for the failure of antiangiogenic therapies and how tumors adapt to these therapies are unclear. Here, we applied transcriptomic, proteomic, and metabolomic approaches to preclinical models and provide evidence for tumor adaptation to vascular endothelial growth factor blockade through a metabolic shift toward carbohydrate and lipid metabolism in tumors. During sunitinib or sorafenib treatment, tumor growth was inhibited and tumors were hypoxic and glycolytic. In sharp contrast, treatment withdrawal led to tumor regrowth, angiogenesis restoration, moderate lactate production, and enhanced lipid synthesis. This metabolic shift was associated with a drastic increase in metastatic dissemination. Interestingly, pharmacological lipogenesis inhibition with orlistat or fatty acid synthase downregulation with shRNA inhibited tumor regrowth and metastases after sunitinib treatment withdrawal. Our data shed light on metabolic alterations that result in cancer adaptation to antiangiogenic treatments and identify key molecules involved in lipid metabolism as putative therapeutic targets.
Collapse
Affiliation(s)
- Nor Eddine Sounni
- Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium.
| | - Jonathan Cimino
- Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium; Mass Spectrometry Laboratory, GIGA-R, Department of Chemistry, University of Liege, 4000 Liege, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium
| | - Irina Primac
- Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium
| | - Alice Truong
- Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium
| | - Gabriel Mazzucchelli
- Mass Spectrometry Laboratory, GIGA-R, Department of Chemistry, University of Liege, 4000 Liege, Belgium
| | - Alexandra Paye
- Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium
| | - David Calligaris
- Mass Spectrometry Laboratory, GIGA-R, Department of Chemistry, University of Liege, 4000 Liege, Belgium
| | - Delphine Debois
- Mass Spectrometry Laboratory, GIGA-R, Department of Chemistry, University of Liege, 4000 Liege, Belgium
| | - Pascal De Tullio
- Laboratory of Drug Research Center, University of Liege, 4000 Liege, Belgium
| | - Bernard Mari
- UMR-7275 CNRS, University of Nice Sophia-Antipolis, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France
| | - Edwin De Pauw
- Mass Spectrometry Laboratory, GIGA-R, Department of Chemistry, University of Liege, 4000 Liege, Belgium
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium
| |
Collapse
|
92
|
The roles of CD73 in cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:460654. [PMID: 25126561 PMCID: PMC4121992 DOI: 10.1155/2014/460654] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 02/06/2023]
Abstract
Purinergic signaling has emerged as an important player in cancer progression and is regulated by a series of nucleotidases. Among the enzyme cascade, CD73, which catelyzes AMP breakdown to adenosine, has been found to be overexpressed in many types of cancer. Various factors and mechanisms are employed to regulate expression of CD73. Accumulating studies have shown that CD73 is a key regulatory molecule of cancer cells proliferation, migration and invasion in vitro, tumor angiogenesis, and tumor immune escape in vivo. With such important roles in cancer, CD73 has become an appealing therapy target. Recent evidences in mice models demonstrated that targeted blockade of CD73 could be a favorable therapeutic approach for cancer patients in the future. In this review, we will summarize the multiple roles of CD73 in cancer development, including its clinical significance, its promotive effects on tumor growth, metastasis, and angiogenesis, and its suppressive effects on immune response, regulatory mechanisms of CD73 expression, and current situation of anti-CD73 cancer therapy.
Collapse
|
93
|
Microenvironment, oncoantigens, and antitumor vaccination: lessons learned from BALB-neuT mice. BIOMED RESEARCH INTERNATIONAL 2014; 2014:534969. [PMID: 25136593 PMCID: PMC4065702 DOI: 10.1155/2014/534969] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/12/2014] [Indexed: 12/20/2022]
Abstract
The tyrosine kinase human epidermal growth factor receptor 2 (HER2) gene is amplified in approximately 20% of human breast cancers and is associated with an aggressive clinical course and the early development of metastasis. Its crucial role in tumor growth and progression makes HER2 a prototypic oncoantigen, the targeting of which may be critical for the development of effective anticancer therapies. The setup of anti-HER2 targeting strategies has revolutionized the clinical outcome of HER2+ breast cancer. However, their initial success has been overshadowed by the onset of pharmacological resistance that renders them ineffective. Since the tumor microenvironment (TME) plays a crucial role in drug resistance, the design of more effective anticancer therapies should depend on the targeting of both cancer cells and their TME as a whole. In this review, starting from the successful know-how obtained with a HER2+ mouse model of mammary carcinogenesis, the BALB-neuT mice, we discuss the role of TME in mammary tumor development. Indeed, a deeper knowledge of antigens critical for cancer outbreak and progression and of the mechanisms that regulate the interplay between cancer and stromal cell populations could advise promising ways for the development of the best anticancer strategy.
Collapse
|
94
|
Bartolotti M, Franceschi E, Poggi R, Tosoni A, Battista MD, Brandes AA. Resistance to antiangiogenic therapies. Future Oncol 2014; 10:1417-25. [DOI: 10.2217/fon.14.57] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT: Angiogenesis is a key process for tumoral growth, which has become a main target for anticancer treatments. A wide number of agents targeting both VEGF and its receptor have recently become standard treatments for different tumor types. Unfortunately, most of the tumors become resistant to these agents after few months of treatment. Different mechanisms of resistance to antiangiogenic drugs have been proposed and investigated; some agents demonstrated to be able to restore sensitivity to antiangiogenic drugs by blocking pathways or molecules involved in the resistance in preclinical models. Biomarkers for the prediction of response or resistance to antiangiogenic agents are under evaluation.
Collapse
Affiliation(s)
- Marco Bartolotti
- Department of Medical Oncology, Azienda Unità Sanitaria Locale, Bologna, Italy
| | - Enrico Franceschi
- Department of Medical Oncology, Azienda Unità Sanitaria Locale, Bologna, Italy
| | - Rosalba Poggi
- Department of Medical Oncology, Azienda Unità Sanitaria Locale, Bologna, Italy
| | - Alicia Tosoni
- Department of Medical Oncology, Azienda Unità Sanitaria Locale, Bologna, Italy
| | - Monica Di Battista
- Department of Medical Oncology, Azienda Unità Sanitaria Locale, Bologna, Italy
| | - Alba A Brandes
- Department of Medical Oncology, Azienda Unità Sanitaria Locale, Bologna, Italy
| |
Collapse
|
95
|
Lucena SE, Romo K, Suntravat M, Sánchez EE. Anti-angiogenic activities of two recombinant disintegrins derived from the Mohave and Prairie rattlesnakes. Toxicon 2013; 78:10-7. [PMID: 24269784 DOI: 10.1016/j.toxicon.2013.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 11/06/2013] [Accepted: 11/12/2013] [Indexed: 01/20/2023]
Abstract
Angiogenesis plays a crucial role in the growth and spread of cancer. New vascularization nourishes cancer cells with oxygen and nutrients, allowing these cells to grow, invade nearby tissue, spread to other parts of the body, and form new colonies of cancer cells. Tumor angiogenesis consists of endothelial cell proliferation, migration, and tube formation into the tumor mass. The study of natural and synthetic angiogenesis inhibitors is a promising area for therapeutics since tumors cannot grow or spread without the formation of new blood vessels. Anti-angiogenic activities have been identified in peptides known as disintegrins. Disintegrins are a family of small proteins (45-84 amino acids in length), many which are found in snake venom that function as potent inhibitors of both platelet aggregation and integrin-dependent cell adhesion. This study reports two recombinant disintegrins (r-mojastin 1 and r-viridistatin 2) inhibiting, with similar effectiveness, distinct steps in angiogenesis such as proliferation, adhesion to fibronectin, migration, and tube formation in vitro and in vivo. Both recombinant disintegrins bind to α(v)β₃ and α(v)β₅ receptors that are upregulated in tumor endothelial cells, having a higher binding activity to α(v)β₃ integrin.
Collapse
Affiliation(s)
- Sara E Lucena
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, MSC 224, 975 West Avenue B, Kingsville, TX 78363, USA
| | - Karen Romo
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, MSC 224, 975 West Avenue B, Kingsville, TX 78363, USA
| | - Montamas Suntravat
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, MSC 224, 975 West Avenue B, Kingsville, TX 78363, USA
| | - Elda E Sánchez
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, MSC 224, 975 West Avenue B, Kingsville, TX 78363, USA; Department of Chemistry, Texas A&M University-Kingsville, MSC 161, Kingsville, TX 78363, USA.
| |
Collapse
|
96
|
Cheng YD, Yang H, Chen GQ, Zhang ZC. Molecularly targeted drugs for metastatic colorectal cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:1315-22. [PMID: 24204124 PMCID: PMC3817019 DOI: 10.2147/dddt.s52485] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The survival rate of patients with metastatic colorectal cancer (mCRC) has significantly improved with applications of molecularly targeted drugs, such as bevacizumab, and led to a substantial improvement in the overall survival rate. These drugs are capable of specifically targeting the inherent abnormal pathways in cancer cells, which are potentially less toxic than traditional nonselective chemotherapeutics. In this review, the recent clinical information about molecularly targeted therapy for mCRC is summarized, with specific focus on several of the US Food and Drug Administration-approved molecularly targeted drugs for the treatment of mCRC in the clinic. Progression-free and overall survival in patients with mCRC was improved greatly by the addition of bevacizumab and/or cetuximab to standard chemotherapy, in either first- or second-line treatment. Aflibercept has been used in combination with folinic acid (leucovorin)-fluorouracil-irinotecan (FOLFIRI) chemotherapy in mCRC patients and among patients with mCRC with wild-type KRAS, the outcomes were significantly improved by panitumumab in combination with folinic acid (leucovorin)-fluorouracil-oxaliplatin (FOLFOX) or FOLFIRI. Because of the new preliminary studies, it has been recommended that regorafenib be used with FOLFOX or FOLFIRI as first- or second-line treatment of mCRC chemotherapy. In summary, an era of new opportunities has been opened for treatment of mCRC and/or other malignancies, resulting from the discovery of new selective targeting drugs.
Collapse
Affiliation(s)
- Ying-dong Cheng
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | | | | | | |
Collapse
|
97
|
Synergistic inhibition of angiogenesis by artesunate and captopril in vitro and in vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:454783. [PMID: 24223058 PMCID: PMC3816047 DOI: 10.1155/2013/454783] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/15/2013] [Accepted: 08/20/2013] [Indexed: 02/02/2023]
Abstract
Inhibition of angiogenesis represents one major strategy of cancer chemotherapy. In the present investigation, we investigated the synergism of artesunate and captopril to inhibit angiogenesis. Artesunate is an antimalarial derivative of artemisinin from the Chinese medicinal plant, Artemisia annua L., which also reveals profound anticancer activity in vitro and in vivo. Captopril is an angiotensin I-converting (ACE) inhibitor, which is well established in Western academic medicine. Both compounds inhibited migration of human umbilical vein endothelial cells (HUVECs) in vitro. The combination of both drugs resulted in synergistically inhibited migration. Whereas artesunate inhibited HUVEC growth in the XTT assay, captopril did not, indicating independent modes of action. We established a chorioallantoic membrane (CAM) assay of quail embryos (Coturnix coturnix L.) and a computer-based evaluation routine for quantitative studies on vascularization processes in vivo. Artesunate and captopril inhibited blood vessel formation and growth. For the first time, we demonstrated that both drugs revealed synergistic effects when combined. These results may also have clinical impact, since cardiovascular diseases and cancer frequently occur together in older cancer patients. Therefore, comorbid patients may take advantage, if they take captopril to treat cardiovascular symptoms and artesunate to treat cancer.
Collapse
|
98
|
Lavie M, Struyf S, Stroh-Dege A, Rommelaere J, Van Damme J, Dinsart C. Capacity of wild-type and chemokine-armed parvovirus H-1PV for inhibiting neo-angiogenesis. Virology 2013; 447:221-32. [PMID: 24210118 DOI: 10.1016/j.virol.2013.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 07/30/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
Abstract
Anti-angiogenic therapy has been recognized as a powerful potential strategy for impeding the growth of various tumors. However no major therapeutic effects have been observed to date, mainly because of the emergence of several resistance mechanisms. Among novel strategies to target tumor vasculature, some oncolytic viruses open up new prospects. In this context, we addressed the question whether the rodent parvovirus H-1PV can target endothelial cells. We show that cultures of human normal (HUVEC) and immortalized (KS-IMM) endothelial cells sustain an abortive viral cycle upon infection with H-1PV and are sensitive to H-1PV cytotoxicity. H-1PV significantly inhibits infected KS-IMM tumor growth. This effect may be traced back by the virus ability to both kill proliferating endothelial cells and inhibit VEGF production Recombinant H-1PV vectors can also transduce tumor cells with chemokines endowed with anti-angiogenesis properties, and warrant further validation for the treatment of highly vascularized tumors.
Collapse
Affiliation(s)
- Muriel Lavie
- Tumor Virology Division, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
99
|
Cao Z, Yang P, Zhou Q. Multiple biological functions and pharmacological effects of lycorine. Sci China Chem 2013; 56:1382-1391. [PMID: 32215001 PMCID: PMC7088923 DOI: 10.1007/s11426-013-4967-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 06/28/2013] [Indexed: 12/20/2022]
Abstract
Lycorine is the major active component from the amaryllidaceae family plant Lycoris radiate, a represent traditional Chinese medicinal herb, and is one of the typical alkaloids with pyrrolophenanthridine nucleus core. Lycorine has drawn great interest in medicinal field due to its divergent chemical structures and multiple biological functions, as well as pharmacological effects on various diseases. Accumulated evidence shows that lycorine not only possesses strong pharmacological effects on many diseases, including anti-leukemia, anti-tumor, anti-angiogenesis, anti-virus, anti-bacteria, anti-inflammation, and antimalaria, but also exerts many other biological functions, such as inhibition of acetylcholinesterase and topoisomerase, suppression of ascorbic acid biosynthesis, and control of circadian period length. Notably, lycorine exhibits its numerous pharmacological effects on various diseases with very low toxicity and mild side effects. The divergent chemical structures, multiple biological functions, and very low toxicity of lycorine imply that the agent is a potential drug candidate that warrants for further preclinical and clinic investigation.
Collapse
Affiliation(s)
- ZhiFei Cao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University; Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, 215006 China
| | - Ping Yang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University; Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, 215006 China
| | - QuanSheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University; Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, 215006 China
| |
Collapse
|
100
|
Cao Z, Shang B, Zhang G, Miele L, Sarkar FH, Wang Z, Zhou Q. Tumor cell-mediated neovascularization and lymphangiogenesis contrive tumor progression and cancer metastasis. Biochim Biophys Acta Rev Cancer 2013; 1836:273-86. [PMID: 23933263 DOI: 10.1016/j.bbcan.2013.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/22/2013] [Accepted: 08/01/2013] [Indexed: 12/12/2022]
Abstract
Robust neovascularization and lymphangiogenesis have been found in a variety of aggressive and metastatic tumors. Endothelial sprouting angiogenesis is generally considered to be the major mechanism by which new vasculature forms in tumors. However, increasing evidence shows that tumor vasculature is not solely composed of endothelial cells (ECs). Some tumor cells acquire processes similar to embryonic vasculogenesis and produce new vasculature through vasculogenic mimicry, trans-differentiation of tumor cells into tumor ECs, and tumor cell-EC vascular co-option. In addition, tumor cells secrete various vasculogenic factors that induce sprouting angiogenesis and lymphangiogenesis. Vasculogenic tumor cells actively participate in the formation of vascular cancer stem cell niche and a premetastatic niche. Therefore, tumor cell-mediated neovascularization and lymphangiogenesis are closely associated with tumor progression, cancer metastasis, and poor prognosis. Vasculogenic tumor cells have emerged as key players in tumor neovascularization and lymphangiogenesis and play pivotal roles in tumor progression and cancer metastasis. However, the mechanisms underlying tumor cell-mediated vascularity as they relate to tumor progression and cancer metastasis remain unclear. Increasing data have shown that various intrinsic and extrinsic factors activate oncogenes and vasculogenic genes, enhance vasculogenic signaling pathways, and trigger tumor neovascularization and lymphangiogenesis. Collectively, tumor cells are the instigators of neovascularization. Therefore, targeting vasculogenic tumor cells, genes, and signaling pathways will open new avenues for anti-tumor vasculogenic and metastatic drug discovery. Dual targeting of endothelial sprouting angiogenesis and tumor cell-mediated neovascularization and lymphangiogenesis may overcome current clinical problems with anti-angiogenic therapy, resulting in significantly improved anti-angiogenesis and anti-cancer therapies.
Collapse
Affiliation(s)
- Zhifei Cao
- Cyrus Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu 215006, China
| | | | | | | | | | | | | |
Collapse
|