101
|
Uncovering Metabolic Effects of Anti-angiogenic Therapy in Tumors by Induced Metabolic Bioluminescence Imaging. Methods Mol Biol 2016. [PMID: 27858366 DOI: 10.1007/978-1-4939-3999-2_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Induced metabolic bioluminescence imaging (imBI) is an imaging technique which enables detection of various metabolites associated with glycolysis in tumor sections. Signals captured by imBI can be used to chart the topographic distribution of lactate, glucose, pyruvate, and ATP and quantify their absolute amount. ImBi can enable us to perform metabolic classification of tumors as well as to detect metabolic changes in the glycolytic pathway associated with certain therapies, such as anti-angiogenic drugs.
Collapse
|
102
|
Carbohydrate-based peptidomimetics targeting neuropilin-1: Synthesis, molecular docking study and in vitro biological activities. Bioorg Med Chem 2016; 24:5315-5325. [DOI: 10.1016/j.bmc.2016.08.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/24/2016] [Accepted: 08/27/2016] [Indexed: 12/31/2022]
|
103
|
Collins DC, Velázquez-Kennedy K, Deady S, Brady AP, Sweeney P, Power DG. National Incidence, Management and Survival of Urachal Carcinoma. Rare Tumors 2016; 8:6257. [PMID: 27746878 PMCID: PMC5064294 DOI: 10.4081/rt.2016.6257] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 02/10/2016] [Accepted: 02/15/2016] [Indexed: 12/31/2022] Open
Abstract
Urachal carcinoma is an uncommon cancer whose rarity has precluded its study and evidence-based management strategies are lacking. This study assessed all urachal carcinomas in Ireland and clinical parameters in order to improve understanding. Urachal carcinomas diagnosed from 1994 to 2011 were identified from the National Cancer Registry in Ireland. Data obtained included patient age, gender, diagnostic year, pathology, tumor stage, patient treatment strategies and survival. Twenty-six urachal carcinomas were identified, the majority being adenocarcinoma. This comprised 0.3% of all invasive bladder tumors. Patients were predominantly male (62%) and over 50 years of age (58%). Twenty-two patients (85%) underwent surgery, with only six (23%) undergoing chemotherapy. On average, median overall survival was 2.6 years (range 0-15.2 yrs). Survival was longer in women (5 vs. 1.9 yrs), patients under 50 years of age (3.6 vs. 1.9 yrs), those without confirmed metastasis (4.1 vs. 0.7 yrs) and those who received chemotherapy (3.6 vs. 2.6 yrs). The overall survival of urachal carcinoma in Ireland is less than expected from published literature. This study highlights the need for centralization of rare tumors with international collaboration to identify the optimal treatment strategy and improve outcome.
Collapse
Affiliation(s)
| | | | - Sandra Deady
- National Cancer Registry Ireland , Cork, Ireland
| | - Adrian P Brady
- Department of Radiology, Mercy University Hospital , Cork, Ireland
| | - Paul Sweeney
- Department of Uro-Oncological Surgery, Mercy University Hospital , Cork, Ireland
| | - Derek G Power
- Department of Medical Oncology, Cork University Hospital , Cork, Ireland
| |
Collapse
|
104
|
Kim S, Cayre A, Jary M, Jacquin M, Arbez-Gindre F, Fein F, Lakkis Z, Nguyen T, Borg C. Complete Response to Aflibercept-FOLFIRI in One Patient With Colorectal Cancer Refractory to Bevacizumab-FOLFOX: A Possible Autocrine Vascular Endothelial Growth Factor Receptor 2-Related Mechanism. Clin Colorectal Cancer 2016; 15:e229-e234. [PMID: 27600470 DOI: 10.1016/j.clcc.2016.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/15/2016] [Accepted: 07/28/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Stefano Kim
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France; Clinical Investigational Center, CIC-1431, University Hospital of Besançon, Besançon, France; INSERM, Unit 1098, University of Bourgogne Franche-Comté, Besançon, France.
| | - Anne Cayre
- Department of Pathology, Jean Perrin Center, Clermont-Ferrand, France
| | - Marine Jary
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France; Clinical Investigational Center, CIC-1431, University Hospital of Besançon, Besançon, France; INSERM, Unit 1098, University of Bourgogne Franche-Comté, Besançon, France
| | - Marion Jacquin
- Clinical Investigational Center, CIC-1431, University Hospital of Besançon, Besançon, France
| | | | - Francine Fein
- Department of Gastroenterology, University Hospital of Besançon, Besançon, France
| | - Zaher Lakkis
- Department of Digestive Surgery and Liver Transplantation, University Hospital of Besançon, Besançon, France
| | - Thierry Nguyen
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France; Clinical Investigational Center, CIC-1431, University Hospital of Besançon, Besançon, France; INSERM, Unit 1098, University of Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
105
|
Al-Hujaily EM, Oldham RAA, Hari P, Medin JA. Development of Novel Immunotherapies for Multiple Myeloma. Int J Mol Sci 2016; 17:E1506. [PMID: 27618026 PMCID: PMC5037783 DOI: 10.3390/ijms17091506] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a disorder of terminally differentiated plasma cells characterized by clonal expansion in the bone marrow (BM). It is the second-most common hematologic malignancy. Despite significant advances in therapeutic strategies, MM remains a predominantly incurable disease emphasizing the need for the development of new treatment regimens. Immunotherapy is a promising treatment modality to circumvent challenges in the management of MM. Many novel immunotherapy strategies, such as adoptive cell therapy and monoclonal antibodies, are currently under investigation in clinical trials, with some already demonstrating a positive impact on patient survival. In this review, we will summarize the current standards of care and discuss major new approaches in immunotherapy for MM.
Collapse
Affiliation(s)
- Ensaf M Al-Hujaily
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Robyn A A Oldham
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Parameswaran Hari
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Jeffrey A Medin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
106
|
Fanale D, Castiglia M, Bazan V, Russo A. Involvement of Non-coding RNAs in Chemo- and Radioresistance of Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 937:207-28. [DOI: 10.1007/978-3-319-42059-2_11] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
107
|
Blood-based biomarkers for monitoring antiangiogenic therapy in non-small cell lung cancer. Med Oncol 2016; 33:105. [PMID: 27568331 DOI: 10.1007/s12032-016-0824-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/20/2016] [Indexed: 12/30/2022]
Abstract
Tumor angiogenesis pathways have been identified as important therapeutic targets in non-small cell lung cancer. However, no biomarkers have been described as predictors of response to antiangiogenic therapy in these patients. In this study, plasma levels of VEGF, bFGF, E-selectin, and S-ICAM and gene expression profiles of peripheral blood mononuclear cells from non-small cell lung cancer patients treated with chemotherapy plus bevacizumab were analyzed before and after treatment. Values were correlated with clinicopathological characteristics and treatment response. Plasma factor levels were measured using commercially available ELISA kits. The TaqMan(®) human angiogenesis array was used to investigate the effect of treatment on gene expression profiles. Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analysis was performed for differentially expressed genes using WEB-based GEne SeT AnaLysis Toolkit. Our results suggest a benefit for patients with increased plasma levels of VEGF, E-selectin, and S-ICAM in the course of bevacizumab treatment. Also, we identified differentially expressed genes between paired blood samples from patients before and after treatment, and significantly perturbed pathways were predicted. These changes in gene expression and levels of plasma factors could be used to assess the effectiveness of antiangiogenic therapy, in addition to standard clinical and radiological evaluations.
Collapse
|
108
|
Kim BR, Seo SH, Park MS, Lee SH, Kwon Y, Rho SB. sMEK1 inhibits endothelial cell proliferation by attenuating VEGFR-2-dependent-Akt/eNOS/HIF-1α signaling pathways. Oncotarget 2016; 6:31830-43. [PMID: 26378810 PMCID: PMC4741643 DOI: 10.18632/oncotarget.5570] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 08/15/2015] [Indexed: 12/31/2022] Open
Abstract
The suppressor of MEK null (sMEK1) protein possesses pro-apoptotic activities. In the current study, we reveal that sMEK1 functions as a novel anti-angiogenic factor by suppressing vascular endothelial growth factor (VEGF)-induced cell proliferation, migration, and capillary-like tubular structure in vitro. In addition, sMEK1 inhibited the phosphorylation of the signaling components up- and downstream of Akt, including phospholipase Cγ1 (PLC-γ1), 3-phosphoinositide-dependent protein kinase 1 (PDK1), endothelial nitric oxide synthetase (eNOS), and hypoxia-inducible factor 1 (HIF-1α) during ovarian tumor progression via binding with vascular endothelial growth factor receptor 2 (VEGFR-2). Furthermore, sMEK1 decreased tumor vascularity and inhibited tumor growth in a xenograft human ovarian tumor model. These results supply convincing evidence that sMEK1 controls endothelial cell function and subsequent angiogenesis by suppressing VEGFR-2-mediated PI3K/Akt/eNOS signaling pathway. Taken together, our results clearly suggest that sMEK1 might be a novel anti-angiogenic and anti-tumor agent for use in ovarian tumor.
Collapse
Affiliation(s)
- Boh-Ram Kim
- Research Institute, National Cancer Center, Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do, Republic of Korea.,College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Global Top 5 Program, Ewha Womans University, Seoul, Republic of Korea
| | - Seung Hee Seo
- Research Institute, National Cancer Center, Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do, Republic of Korea
| | - Mi Sun Park
- Research Institute, National Cancer Center, Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do, Republic of Korea
| | - Seung-Hoon Lee
- Department of Life Science, Yong In University, Samga-dong, Cheoin-gu, Yongin-si Gyeonggi-do, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Global Top 5 Program, Ewha Womans University, Seoul, Republic of Korea
| | - Seung Bae Rho
- Research Institute, National Cancer Center, Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do, Republic of Korea
| |
Collapse
|
109
|
Jerez S, Araya H, Thaler R, Charlesworth MC, López-Solís R, Kalergis AM, Céspedes PF, Dudakovic A, Stein GS, van Wijnen AJ, Galindo M. Proteomic Analysis of Exosomes and Exosome-Free Conditioned Media From Human Osteosarcoma Cell Lines Reveals Secretion of Proteins Related to Tumor Progression. J Cell Biochem 2016; 118:351-360. [PMID: 27356893 DOI: 10.1002/jcb.25642] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022]
Abstract
Osteosarcomas are the most prevalent bone tumors in pediatric patients, but can also occur later in life. Bone tumors have the potential to metastasize to lung and occasionally other vital organs. To understand how osteosarcoma cells interact with their micro-environment to support bone tumor progression and metastasis, we analyzed secreted proteins and exosomes from three human osteosarcoma cell lines. Exosome isolation was validated by transmission electron microscopy (TEM) and immuno-blotting for characteristic biomarkers (CD63, CD9, and CD81). Exosomal and soluble proteins (less than 100 kDa) were identified by mass spectrometry analysis using nanoLC-MS/MS and classified by functional gene ontology clustering. We identified a secretome set of >3,000 proteins for both fractions, and detected proteins that are either common or unique among the three osteosarcoma cell lines. Protein ontology comparison of proteomes from exosomes and exosome-free fractions revealed differences in the enrichment of functional categories associated with different biological processes, including those related to tumor progression (i.e., angiogenesis, cell adhesion, and cell migration). The secretome characteristics of osteosarcoma cells are consistent with the pathological properties of tumor cells with metastatic potential. J. Cell. Biochem. 118: 351-360, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sofía Jerez
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Héctor Araya
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roman Thaler
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street S.W., MSB 3-69, Rochester, Minnesota 55905
| | | | - Remigio López-Solís
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Céspedes
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Amel Dudakovic
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street S.W., MSB 3-69, Rochester, Minnesota 55905
| | - Gary S Stein
- Department of Biochemistry, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, Vermont 05405
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street S.W., MSB 3-69, Rochester, Minnesota 55905
| | - Mario Galindo
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
110
|
Kalmuk J, Folaron M, Buchinger J, Pili R, Seshadri M. Multimodal imaging guided preclinical trials of vascular targeting in prostate cancer. Oncotarget 2016. [PMID: 26203773 PMCID: PMC4695192 DOI: 10.18632/oncotarget.4463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The high mortality rate associated with castration-resistant prostate cancer (CRPC) underscores the need for improving therapeutic options for this patient population. The purpose of this study was to examine the potential of vascular targeting in prostate cancer. Experimental studies were carried out in subcutaneous and orthotopic Myc-CaP prostate tumors implanted into male FVB mice to examine the efficacy of a novel microtubule targeted vascular disrupting agent (VDA), EPC2407 (Crolibulin™). A non-invasive multimodality imaging approach based on magnetic resonance imaging (MRI), bioluminescence imaging (BLI), and ultrasound (US) was utilized to guide preclinical trial design and monitor tumor response to therapy. Imaging results were correlated with histopathologic assessment, tumor growth and survival analysis. Contrast-enhanced MRI revealed potent antivascular activity of EPC2407 against subcutaneous and orthotopic Myc-CaP tumors. Longitudinal BLI of Myc-CaP tumors expressing luciferase under the androgen response element (Myc-CaP/ARE-luc) revealed changes in AR signaling and reduction in intratumoral delivery of luciferin substrate following castration suggestive of reduced blood flow. This reduction in blood flow was validated by US and MRI. Combination treatment resulted in sustained vascular suppression, inhibition of tumor regrowth and conferred a survival benefit in both models. These results demonstrate the therapeutic potential of vascular targeting in combination with androgen deprivation against prostate cancer.
Collapse
Affiliation(s)
- James Kalmuk
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Current address: SUNY Upstate Medical University, Syracuse, NY, USA
| | - Margaret Folaron
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Julian Buchinger
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Current address: University at Buffalo - School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Roberto Pili
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Mukund Seshadri
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
111
|
Treps L, Conradi LC, Harjes U, Carmeliet P. Manipulating Angiogenesis by Targeting Endothelial Metabolism: Hitting the Engine Rather than the Drivers-A New Perspective? Pharmacol Rev 2016; 68:872-87. [PMID: 27363442 DOI: 10.1124/pr.116.012492] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Excessive angiogenesis (i.e., the formation of new blood vessels) contributes to different pathologies, among them cancer and ocular disorders. Conversely, dysfunction of endothelial cells (ECs) contributes to cardiovascular complications, as is the case in diabetes. Inhibition of pathologic angiogenesis in blinding eye disease and cancer by targeting growth factors such as vascular endothelial growth factor has become an accepted therapeutic strategy. However, recent studies also unveiled the emerging importance of EC metabolism in controlling angiogenesis. In this overview, we will discuss recent insights in the metabolic regulation of angiogenesis, focusing on the best-characterized metabolic pathways, and highlight deregulation of EC metabolism in cancer and diabetes. We will give an outlook on how targeting EC metabolism can be used for blocking pathologic angiogenesis and for normalizing EC dysfunction.
Collapse
Affiliation(s)
- Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, and Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| | - Lena-Christin Conradi
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, and Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| | - Ulrike Harjes
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, and Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, and Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| |
Collapse
|
112
|
Chang Y, Niu W, Lian PL, Wang XQ, Meng ZX, Liu Y, Zhao R. Endocan-expressing microvessel density as a prognostic factor for survival in human gastric cancer. World J Gastroenterol 2016; 22:5422-5429. [PMID: 27340359 PMCID: PMC4910663 DOI: 10.3748/wjg.v22.i23.5422] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 04/12/2016] [Accepted: 05/04/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of endocan in tumour vessels and the relationships between endocan and the expression of vascular endothelial growth factor (VEGF) and prognosis in gastric cancer.
METHODS: This study included 142 patients with confirmed gastric cancer in a single cancer centre between 2008 and 2009. Clinicopathologic features were determined, and an immunohistochemical analysis of endocan-expressing microvessel density (MVD) (endocan-MVD), VEGF and vascular endothelial growth factor receptor 2 (VEGFR2) was performed. Potential relationships between endocan-MVD and clinicopathological variables were assessed using a Student’s t-test or an analysis of variance test. Spearman’s rank correlation was applied to evaluate the relationship between endocan-MVD and the expression of VEGF/VEGFR2. Long-term survival of these patients was analysed using univariate and multivariate analyses.
RESULTS: Positive staining of endocan was observed in most of the gastric cancer tissues (108/142) and in fewer of the normal gastric tissues. Endocan-MVD was not associated with gender or histological type (P > 0.05), while endocan-MVD was associated with tumour size, Borrmann type, tumour differentiation, tumour invasion, lymph node metastasis and TNM stage (P < 0.05). According to the Spearman’s rank correlation analysis, endocan-MVD had a positive correlation with VEGF (r = 0.167, P = 0.047) and VEGFR2 (r = 0.410, P = 0.000). The univariate analysis with a log-rank test indicated that the patients with a high level of endocan-MVD had a significantly poorer overall survival rate than those with a low level of endocan-MVD (17.9% vs 64.0%, P = 0.000). The multivariate analysis showed that a high level of endocan-MVD was a valuable prognostic factor.
CONCLUSION: Endocan-MVD significantly correlates with the expression of VEGF and VEGFR2 and is a valuable prognostic factor for survival in human gastric cancer.
Collapse
|
113
|
Development of a robust reporter-based assay for the bioactivity determination of anti-VEGF therapeutic antibodies. J Pharm Biomed Anal 2016; 125:212-8. [DOI: 10.1016/j.jpba.2016.03.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/18/2016] [Accepted: 03/21/2016] [Indexed: 12/21/2022]
|
114
|
MacDonald DA, Martin J, Muthusamy KK, Luo JK, Pyles E, Rafique A, Huang T, Potocky T, Liu Y, Cao J, Bono F, Delesque N, Savi P, Francis J, Amirkhosravi A, Meyer T, Romano C, Glinka M, Yancopoulos GD, Stahl N, Wiegand SJ, Papadopoulos N. Aflibercept exhibits VEGF binding stoichiometry distinct from bevacizumab and does not support formation of immune-like complexes. Angiogenesis 2016; 19:389-406. [PMID: 27234973 PMCID: PMC4930479 DOI: 10.1007/s10456-016-9515-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/13/2016] [Indexed: 12/13/2022]
Abstract
Anti-vascular endothelial growth factor (VEGF) therapies have improved clinical outcomes for patients with cancers and retinal vascular diseases. Three anti-VEGF agents, pegaptanib, ranibizumab, and aflibercept, are approved for ophthalmic indications, while bevacizumab is approved to treat colorectal, lung, and renal cancers, but is also used off-label to treat ocular vascular diseases. The efficacy of bevacizumab relative to ranibizumab in treating neovascular age-related macular degeneration has been assessed in several trials. However, questions persist regarding its safety, as bevacizumab can form large complexes with dimeric VEGF165, resulting in multimerization of the Fc domain and platelet activation. Here, we compare binding stoichiometry, Fcγ receptor affinity, platelet activation, and binding to epithelial and endothelial cells in vitro for bevacizumab and aflibercept, in the absence or presence of VEGF. In contrast to bevacizumab, aflibercept forms a homogenous 1:1 complex with each VEGF dimer. Unlike multimeric bevacizumab:VEGF complexes, the monomeric aflibercept:VEGF complex does not exhibit increased affinity for low-affinity Fcγ receptors, does not activate platelets, nor does it bind to the surface of epithelial or endothelial cells to a greater degree than unbound aflibercept or control Fc. The latter finding reflects the fact that aflibercept binds VEGF in a unique manner, distinct from antibodies not only blocking the amino acids necessary for VEGFR1/R2 binding but also occluding the heparin-binding site on VEGF165.
Collapse
Affiliation(s)
- Douglas A MacDonald
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Joel Martin
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Kathir K Muthusamy
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Jiann-Kae Luo
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Erica Pyles
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Ashique Rafique
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Tammy Huang
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Terra Potocky
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Yang Liu
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Jingtai Cao
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | | | | | | | - John Francis
- Center for Thrombosis Research, Florida Hospital, Orlando, FL, USA
| | - Ali Amirkhosravi
- Center for Thrombosis Research, Florida Hospital, Orlando, FL, USA
| | - Todd Meyer
- Center for Thrombosis Research, Florida Hospital, Orlando, FL, USA
| | - Carmelo Romano
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Meredith Glinka
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - George D Yancopoulos
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Neil Stahl
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Stanley J Wiegand
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Nicholas Papadopoulos
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA.
| |
Collapse
|
115
|
Puddu A, Sanguineti R, Traverso CE, Viviani GL, Nicolò M. Response to anti-VEGF-A treatment of endothelial cells in vitro. Exp Eye Res 2016; 146:128-136. [DOI: 10.1016/j.exer.2015.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 11/23/2015] [Accepted: 12/31/2015] [Indexed: 10/22/2022]
|
116
|
Zhang Y, Liu J, Wang S, Luo X, Li Y, Lv Z, Zhu J, Lin J, Ding L, Ye Q. The DEK oncogene activates VEGF expression and promotes tumor angiogenesis and growth in HIF-1α-dependent and -independent manners. Oncotarget 2016; 7:23740-56. [PMID: 26988756 PMCID: PMC5029660 DOI: 10.18632/oncotarget.8060] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/29/2016] [Indexed: 11/25/2022] Open
Abstract
The DEK oncogene is overexpressed in various cancers and overexpression of DEK correlates with poor clinical outcome. Vascular endothelial growth factor (VEGF) is the most important regulator of tumor angiogenesis, a process essential for tumor growth and metastasis. However, whether DEK enhances tumor angiogenesis remains unclear. Here, we show that DEK is a key regulator of VEGF expression and tumor angiogenesis. Using chromatin immunoprecipitation assay, we found that DEK promoted VEGF transcription in breast cancer cells (MCF7, ZR75-1 and MDA-MB-231) by directly binding to putative DEK-responsive element (DRE) of the VEGF promoter and indirectly binding to hypoxia response element (HRE) upstream of the DRE through its interaction with the transcription factor hypoxia-inducible factor 1α (HIF-1α), a master regulator of tumor angiogenesis and growth. DEK is responsible for recruitment of HIF-1α and the histone acetyltransferase p300 to the VEGF promoter. DEK-enhanced VEGF increases vascular endothelial cell proliferation, migration and tube formation as well as angiogenesis in the chick chorioallantoic membrane. DEK promotes tumor angiogenesis and growth in nude mice in HIF-1α-dependent and -independent manners. Immunohistochemical staining showed that DEK expression positively correlates with the expression of VEGF and microvessel number in 58 breast cancer patients. Our data establish DEK as a sequence-specific binding transcription factor, a novel coactivator for HIF-1α in regulation of VEGF transcription and a novel promoter of angiogenesis.
Collapse
MESH Headings
- Animals
- Apoptosis
- Biomarkers, Tumor
- Breast Neoplasms/blood supply
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Proliferation
- Chick Embryo
- Chorioallantoic Membrane/metabolism
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Mice
- Mice, Nude
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Poly-ADP-Ribose Binding Proteins/genetics
- Poly-ADP-Ribose Binding Proteins/metabolism
- Response Elements
- Signal Transduction
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, People's Republic of China
| | - Jie Liu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Shibin Wang
- First Affiliated Hospital, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiaoli Luo
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Yang Li
- First Affiliated Hospital, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhaohui Lv
- Department of Endocrinology, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, People's Republic of China
| | - Jie Zhu
- Department of Endocrinology, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, People's Republic of China
| | - Jing Lin
- First Affiliated Hospital, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lihua Ding
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, People's Republic of China
| |
Collapse
|
117
|
Hwang DE, Ryou JH, Oh JR, Han JW, Park TK, Kim HS. Anti-Human VEGF Repebody Effectively Suppresses Choroidal Neovascularization and Vascular Leakage. PLoS One 2016; 11:e0152522. [PMID: 27015541 PMCID: PMC4807815 DOI: 10.1371/journal.pone.0152522] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/15/2016] [Indexed: 12/27/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss and blindness among people over the age of 60. Vascular endothelial growth factor (VEGF) plays a major role in pathological angiogenesis in AMD. Herein, we present the development of an anti- human VEGF repebody, which is a small-sized protein binder consisting of leucine-rich repeat (LRR) modules. The anti-VEGF repebody selected through a phage-display was shown to have a high affinity and specificity for human VEGF. We demonstrate that this repebody effectively inhibits in vitro angiogenic cellular processes, such as proliferation and migration, by blocking the VEGF-mediated signaling pathway. The repebody was also shown to have a strong suppression effect on choroidal neovascularization (CNV) and vascular leakage in vivo. Our results indicate that the anti-VEGF repebody has a therapeutic potential for treating neovascular AMD as well as other VEGF-involved diseases including diabetic retinopathy and metastatic cancers.
Collapse
Affiliation(s)
- Da-Eun Hwang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305–701, Korea
| | - Jeong-Hyun Ryou
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305–701, Korea
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305–701, Korea
| | - Jong Rok Oh
- Department of Ophthalmology, Soonchunhyang University, College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University, College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University, College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305–701, Korea
- * E-mail:
| |
Collapse
|
118
|
Ben-Batalla I, Cubas-Cordova M, Udonta F, Wroblewski M, Waizenegger JS, Janning M, Sawall S, Gensch V, Zhao L, Martinez-Zubiaurre I, Riecken K, Fehse B, Pantel K, Bokemeyer C, Loges S. Cyclooxygenase-2 blockade can improve efficacy of VEGF-targeting drugs. Oncotarget 2016; 6:6341-58. [PMID: 25849942 PMCID: PMC4467441 DOI: 10.18632/oncotarget.3437] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/21/2015] [Indexed: 01/05/2023] Open
Abstract
Anti-angiogenic therapies were approved for different cancers. However, significant primary and secondary resistance hampers efficacy in several tumor types including breast cancer. Thus, we need to develop clinically applicable strategies to enhance efficacy of anti-angiogenic drugs. We report that anti-angiogenic therapies can induce upregulation of cyclooxygenase-2 (Cox-2) and of its product prostaglandin E2 (PGE2) in breast cancer models. Upon Cox-2 inhibition PGE2 levels were normalized and efficacy of anti-vascular endothelial growth factor receptor 2 (anti-VEGFR-2) antibodies and sunitinib was enhanced. Interestingly, both treatments exerted additive anti-angiogenic effects. Following Cox-2 inhibition, we observed reduced infiltration of tumors with cancer-associated fibroblasts (CAFs) and lower levels of pro-angiogenic factors active besides the VEGF axis including hepatocyte growth factor (HGF) and basic fibroblast growth factor (FGF2). Mechanistic studies indicated that Cox-2 inhibition reduced PGE2-induced migration and proliferation of CAFs via inhibiting phosphorylation of Akt. Hence, Cox-2 inhibition can increase efficacy of anti-angiogenic treatments and our findings might pave the road for clinical investigations of concomitant blockade of Cox-2 and VEGF-signaling.
Collapse
Affiliation(s)
- Isabel Ben-Batalla
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Miguel Cubas-Cordova
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Udonta
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mark Wroblewski
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas S Waizenegger
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Janning
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefanie Sawall
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Victoria Gensch
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lin Zhao
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Clinic for Stem Cell Transplantation, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Clinic for Stem Cell Transplantation, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Loges
- Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
119
|
Montero AJ, Kwon D, Flores A, Kovacs K, Trent JC, Benedetto P, Rocha-Lima C, Merchan JR. A Phase I Clinical, Pharmacokinetic, and Pharmacodynamic Study of Weekly or Every Three Week Ixabepilone and Daily Sunitinib in Patients with Advanced Solid Tumors. Clin Cancer Res 2016; 22:3209-17. [PMID: 26864210 DOI: 10.1158/1078-0432.ccr-15-2184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE To evaluate the safety, MTD, pharmacokinetics/pharmacodynamics, and early clinical activity of ixabepilone given either weekly or every 3 weeks in combination with daily sunitinib in patients with advanced solid tumors. EXPERIMENTAL DESIGN Eligible patients received either weekly (schedule A) or every 3 weeks (schedule B) ixabepilone at escalating doses (schedule A: 7.5, 15, or 20 mg/m(2); schedule B: 20, 30, or 40 mg/m(2)), and oral sunitinib (37.5 mg daily), starting on day 8 of cycle 1. Dose-limiting toxicities (DLT) were assessed during cycle 1. RESULTS The ixabepilone and sunitinib combination was fairly well tolerated. DLTs were observed in 3 subjects (1 in schedule 3A and 2 in schedule 3B). The most common grade 3-4 hematologic and nonhematologic adverse events were leukopenia and fatigue, respectively. Four patients (3 in schedule A) achieved a partial response, while 13 patients had stable disease. Nine of 17 heavily pretreated colorectal cancer patients had clinical benefit. Coadministration of sunitinib with ixabepilone on a weekly (but not every 3 week) schedule was associated with a significant increase in the half-life and a significant decrease in the clearance of ixabepilone. Correlative studies demonstrated a significant association between higher baseline plasma angiogenic activity (PAA) and clinical benefit in schedule A patients. Weekly, but not every 3 weeks, ixabepilone led to a significant decrease in PAA postbaseline. CONCLUSIONS Coadministration of ixabepilone with sunitinib has acceptable toxicity and encouraging clinical activity in heavily pretreated patients, particularly in patients with metastatic colorectal cancer. Clin Cancer Res; 22(13); 3209-17. ©2016 AACR.
Collapse
Affiliation(s)
- Alberto J Montero
- Department of Medicine, Division of Hematology-Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida.
| | - Deukwoo Kwon
- Biostatistics and Bioinformatics Core, Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Aurea Flores
- Department of Medicine, Division of Hematology-Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Krisztina Kovacs
- Department of Medicine, Division of Hematology-Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Jonathan C Trent
- Department of Medicine, Division of Hematology-Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Pasquale Benedetto
- Department of Medicine, Division of Hematology-Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Caio Rocha-Lima
- Department of Medicine, Division of Hematology-Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Jaime R Merchan
- Department of Medicine, Division of Hematology-Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida.
| |
Collapse
|
120
|
Avallone A, Piccirillo MC, Aloj L, Nasti G, Delrio P, Izzo F, Di Gennaro E, Tatangelo F, Granata V, Cavalcanti E, Maiolino P, Bianco F, Aprea P, De Bellis M, Pecori B, Rosati G, Carlomagno C, Bertolini A, Gallo C, Romano C, Leone A, Caracò C, de Lutio di Castelguidone E, Daniele G, Catalano O, Botti G, Petrillo A, Romano GM, Iaffaioli VR, Lastoria S, Perrone F, Budillon A. A randomized phase 3 study on the optimization of the combination of bevacizumab with FOLFOX/OXXEL in the treatment of patients with metastatic colorectal cancer-OBELICS (Optimization of BEvacizumab scheduLIng within Chemotherapy Scheme). BMC Cancer 2016; 16:69. [PMID: 26857924 PMCID: PMC4746902 DOI: 10.1186/s12885-016-2102-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite the improvements in diagnosis and treatment, colorectal cancer (CRC) is the second cause of cancer deaths in both sexes. Therefore, research in this field remains of great interest. The approval of bevacizumab, a humanized anti-vascular endothelial growth factor (VEGF) monoclonal antibody, in combination with a fluoropyrimidine-based chemotherapy in the treatment of metastatic CRC has changed the oncology practice in this disease. However, the efficacy of bevacizumab-based treatment, has thus far been rather modest. Efforts are ongoing to understand the better way to combine bevacizumab and chemotherapy, and to identify valid predictive biomarkers of benefit to avoid unnecessary and costly therapy to nonresponder patients. The BRANCH study in high-risk locally advanced rectal cancer patients showed that varying bevacizumab schedule may impact on the feasibility and efficacy of chemo-radiotherapy. METHODS/DESIGN OBELICS is a multicentre, open-label, randomised phase 3 trial comparing in mCRC patients two treatment arms (1:1): standard concomitant administration of bevacizumab with chemotherapy (mFOLFOX/OXXEL regimen) vs experimental sequential bevacizumab given 4 days before chemotherapy, as first or second treatment line. Primary end point is the objective response rate (ORR) measured according to RECIST criteria. A sample size of 230 patients was calculated allowing reliable assessment in all plausible first-second line case-mix conditions, with a 80% statistical power and 2-sided alpha error of 0.05. Secondary endpoints are progression free-survival (PFS), overall survival (OS), toxicity and quality of life. The evaluation of the potential predictive role of several circulating biomarkers (circulating endothelial cells and progenitors, VEGF and VEGF-R SNPs, cytokines, microRNAs, free circulating DNA) as well as the value of the early [(18)F]-Fluorodeoxyglucose positron emission tomography (FDG-PET) response, are the objectives of the traslational project. DISCUSSION Overall this study could optimize bevacizumab scheduling in combination with chemotherapy in mCRC patients. Moreover, correlative studies could improve the knowledge of the mechanisms by which bevacizumab enhance chemotherapy effect and could identify early predictors of response. EudraCT Number: 2011-004997-27 TRIAL REGISTRATION: ClinicalTrials.gove number, NCT01718873.
Collapse
Affiliation(s)
- Antonio Avallone
- Multidisciplinary Treatment Unit, Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy. .,Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | | | - Luigi Aloj
- Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Guglielmo Nasti
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Paolo Delrio
- Colorectal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Vincenza Granata
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Ernesta Cavalcanti
- Laboratory Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Francesco Bianco
- Gastrointestinal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Pasquale Aprea
- Vascular Access Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Mario De Bellis
- Endoscopy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Biagio Pecori
- Radiotherapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Gerardo Rosati
- Medical Oncology Unit, San Carlo Hospital, Potenza, Italy.
| | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy.
| | | | - Ciro Gallo
- Medical Statistics Unit, Second University, Naples, Italy.
| | - Carmela Romano
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Corradina Caracò
- Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | | | - Gennaro Daniele
- Clinical Trials Unit, Istituto Nazionale Tumori "Fondazione G. Pascale" - IRCCS, Napoli, Italy.
| | - Orlando Catalano
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Giovanni M Romano
- Gastrointestinal Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Vincenzo R Iaffaioli
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori "Fondazione G. Pascale" - IRCCS, Napoli, Italy.
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS, Napoli, Italy.
| |
Collapse
|
121
|
Indraccolo S, Mueller-Klieser W. Potential of Induced Metabolic Bioluminescence Imaging to Uncover Metabolic Effects of Antiangiogenic Therapy in Tumors. Front Oncol 2016; 6:15. [PMID: 26870694 PMCID: PMC4733917 DOI: 10.3389/fonc.2016.00015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/14/2016] [Indexed: 11/13/2022] Open
Abstract
Tumor heterogeneity at the genetic level has been illustrated by a multitude of studies on the genomics of cancer, but whether tumors can be heterogeneous at the metabolic level is an issue that has been less systematically investigated so far. A burning-related question is whether the metabolic features of tumors can change either following natural tumor progression (i.e., in primary tumors versus metastasis) or therapeutic interventions. In this regard, recent findings by independent teams indicate that antiangiogenic drugs cause metabolic perturbations in tumors as well as metabolic adaptations associated with increased malignancy. Induced metabolic bioluminescence imaging (imBI) is an imaging technique that enables detection of key metabolites associated with glycolysis, including lactate, glucose, pyruvate, and ATP in tumor sections. Signals captured by imBI can be used to visualize the topographic distribution of these metabolites and quantify their absolute amount. imBI can be very useful for metabolic classification of tumors as well as to track metabolic changes in the glycolytic pathway associated with certain therapies. Imaging of the metabolic changes induced by antiangiogenic drugs in tumors by imBI or other emerging technologies is a valuable tool to uncover molecular sensors engaged by metabolic stress and offers an opportunity to understand how metabolism-based approaches could improve cancer therapy.
Collapse
Affiliation(s)
- Stefano Indraccolo
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto - IRCCS , Padova , Italy
| | - Wolfgang Mueller-Klieser
- University Medical Center of the Johannes Gutenberg University, Institute of Pathophysiology , Mainz , Germany
| |
Collapse
|
122
|
Abstract
Glioblastoma is the most common and aggressive primary brain tumor in adults. Defining histopathologic features are necrosis and endothelial proliferation, resulting in the assignment of grade IV, the highest grade in the World Health Organization (WHO) classification of brain tumors. The classic clinical term "secondary glioblastoma" refers to a minority of glioblastomas that evolve from previously diagnosed WHO grade II or grade III gliomas. Specific point mutations of the genes encoding isocitrate dehydrogenase (IDH) 1 or 2 appear to define molecularly these tumors that are associated with younger age and more favorable outcome; the vast majority of glioblastomas are IDH wild-type. Typical molecular changes in glioblastoma include mutations in genes regulating receptor tyrosine kinase (RTK)/rat sarcoma (RAS)/phosphoinositide 3-kinase (PI3K), p53, and retinoblastoma protein (RB) signaling. Standard treatment of glioblastoma includes surgery, radiotherapy, and alkylating chemotherapy. Promoter methylation of the gene encoding the DNA repair protein, O(6)-methylguanyl DNA methyltransferase (MGMT), predicts benefit from alkylating chemotherapy with temozolomide and guides choice of first-line treatment in elderly patients. Current developments focus on targeting the molecular characteristics that drive the malignant phenotype, including altered signal transduction and angiogenesis, and more recently, various approaches of immunotherapy.
Collapse
|
123
|
Ayed AO, Chang LJ, Moreb JS. Immunotherapy for multiple myeloma: Current status and future directions. Crit Rev Oncol Hematol 2015; 96:399-412. [DOI: 10.1016/j.critrevonc.2015.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 04/26/2015] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
|
124
|
Coelho AL, Araújo AM, Gomes MP, Catarino RJ, Andrade EB, Lopes AM, Medeiros RM. Combined Ang-2 and VEGF serum levels: holding hands as a new integral biomarker in non-small-cell lung cancers. Future Oncol 2015; 11:3233-42. [PMID: 26562248 DOI: 10.2217/fon.15.207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM Evaluate if serum levels of VEGF and Ang-2 are correlated in non-small-cell lung cancers (NSCLCs) and its implications in the diagnostic and prognostic of the disease. PATIENTS & METHODS Unselected cohort of 145 NSCLC patients and 30 control individuals. The serum levels of Ang-2 and VEGF of each patient were measured by ELISA prior to treatment. RESULTS & CONCLUSIONS Serum levels of Ang-2 and VEGF are correlated (p < 0.0001). High serum levels of Ang-2 and VEGF isolated and both combined (high(Ang-2/VEGF)) correlate with likelihood of presenting NSCLC (p = 0.016; p = 0.003; p < 0.0001, respectively). Serum levels of Ang-2 and high(Ang-2/VEGF) but not VEGF alone are independent prognostic factors (p = 0.001; p = 0.619; p = 0.005). High(Ang-2/VEGF) serum levels could be exploited as a new valuable integral biomarker in NSCLC.
Collapse
Affiliation(s)
- Ana Luísa Coelho
- Instituto Português de Oncologia - Porto, Molecular Oncology Group, Portugal.,Faculdade de Medicina - University of Porto, Porto, Portugal
| | - António Manuel Araújo
- Centro Hospitalar do Porto - Medical Oncology Department, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar - University of Porto, Porto, Portugal
| | - Mónica Patrícia Gomes
- Instituto Português de Oncologia - Porto, Molecular Oncology Group, Portugal.,Instituto de Ciências Biomédicas Abel Salazar - University of Porto, Porto, Portugal
| | - Raquel Jorge Catarino
- Instituto Português de Oncologia - Porto, Molecular Oncology Group, Portugal.,Instituto de Ciências Biomédicas Abel Salazar - University of Porto, Porto, Portugal
| | - Elva Bonifácio Andrade
- Instituto de Biologia Molecular e Celular - Immunobiology Research Group, Porto, Portugal
| | - Agostinho Marques Lopes
- Faculdade de Medicina - University of Porto, Porto, Portugal.,Centro Hospitalar de S. João - Pulmonology Department, Porto, Portugal
| | - Rui Manuel Medeiros
- Instituto Português de Oncologia - Porto, Molecular Oncology Group, Portugal.,Instituto de Ciências Biomédicas Abel Salazar - University of Porto, Porto, Portugal.,Liga Portuguesa Contra o Cancro (NRNorte) - Research Department, Porto, Portugal
| |
Collapse
|
125
|
Woo J, Cohen SA, Grim JE. Targeted therapy in gastroesophageal cancers: past, present and future. Gastroenterol Rep (Oxf) 2015; 3:316-29. [PMID: 26510453 PMCID: PMC4650980 DOI: 10.1093/gastro/gov052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/09/2015] [Indexed: 12/12/2022] Open
Abstract
Gastroesophageal cancer is a significant global problem that frequently presents at an incurable stage and has very poor survival with standard chemotherapy approaches. This review will examine the epidemiology and molecular biology of gastroesophageal cancer and will focus on the key deregulated signaling pathways that have been targeted in the clinic. A comprehensive overview of clinical data highlighting successes and failures with targeted agents will be presented. Most notably, HER2-targeted therapy with the monoclonal antibody trastuzumab has proven beneficial in first-line therapy and has been incorporated into standard practice. Targeting the VEGF pathway has also proven beneficial, and the VEGFR-targeted monoclonal antibody ramucirumab is now approved for second-line therapy. In contrast to these positive results, agents targeting the EGFR and MET pathways have been evaluated extensively in gastroesophageal cancer but have repeatedly failed to show benefit. An increased understanding of the molecular predictors of response to targeted therapies is sorely needed. In the future, improved molecular pathology approaches should subdivide this heterogeneous disease entity to allow individualization of cancer therapy based on integrated and global identification of deregulated signaling pathways. Better patient selection, rational combinations of targeted therapies and incorporation of emerging immunotherapeutic approaches should further improve the treatment of this deadly disease.
Collapse
Affiliation(s)
- Janghee Woo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Division of Medical Oncology, University of Washington, Seattle, WA, USA and
| | - Stacey A Cohen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Division of Medical Oncology, University of Washington, Seattle, WA, USA and
| | - Jonathan E Grim
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Division of Medical Oncology, University of Washington, Seattle, WA, USA and Hospital and Specialty Medicine, VA Puget Sound Health Care System, Seattle, WA, USA
| |
Collapse
|
126
|
Rodriguez-Freixinos V, Mackay HJ. Breaking down the evidence for bevacizumab in advanced cervical cancer: past, present and future. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2015; 2:8. [PMID: 27231568 PMCID: PMC4881045 DOI: 10.1186/s40661-015-0015-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/16/2015] [Indexed: 12/15/2022]
Abstract
Despite the introduction of screening and, latterly, vaccination programs in the developed world, globally cervical cancer remains a significant health problem. For those diagnosed with advanced or recurrent disease even within resource rich communities, prognosis remains poor with an overall survival (OS) of just over 12 months. New therapeutic interventions are urgently required. Advances in our understanding of the mechanisms underlying tumor growth and the downstream effects of human papilloma virus (HPV) infection identified angiogenesis as a rational target for therapeutic intervention in cervical cancer. Anti-angiogenic agents showed promising activity in early phase clinical trials culminating in a randomized phase III study of the humanized monoclonal antibody to vascular endothelial growth factor (VEGF), bevacizumab, in combination with chemotherapy. This pivotal study, the Gynecologic Oncology Group protocol 240, met its primary endpoint demonstrating a significant improvement in OS. Bevacizumab became the first targeted agent to be granted regulatory approval by the United States Food and Drug Administration for use alongside chemotherapy in adults with persistent, recurrent or metastatic carcinoma of the cervix. This review outlines the rationale for targeting angiogenesis in cervical cancer focusing on the current indications for the use of bevacizumab in this disease and future directions.
Collapse
Affiliation(s)
- Victor Rodriguez-Freixinos
- Division of Medical Oncology and Hematology, Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, Ontario M5G 2 M9 Canada
| | - Helen J. Mackay
- Division of Medical Oncology and Hematology, Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, Ontario M5G 2 M9 Canada
| |
Collapse
|
127
|
Teuwen LA, Van den Mooter T, Dirix L. Management of pulmonary toxicity associated with targeted anticancer therapies. Expert Opin Drug Metab Toxicol 2015; 11:1695-707. [PMID: 26293379 DOI: 10.1517/17425255.2015.1080687] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Targeted anticancer therapies act by interfering with defined molecular entities and/or biologic pathways. Because of their more specific mechanism of action, adverse events (AEs) on healthy tissues are intended to be minimal, resulting in a different toxicity profile from that observed with conventional cytotoxic chemotherapy. Pulmonary AEs are rare but potentially life-threatening and it is, therefore, critical to recognize early on and manage appropriately. AREAS COVERED In this review, we aim to offer an overview of both more frequent and rare pulmonary AEs caused by targeted anticancer therapies and discuss possible treatment algorithms. Anti-vascular endothelial growth factor, anti-human epidermal growth factor receptor and anti-CD20 therapy will be reviewed, as well as immune checkpoint inhibitors, anaplastic lymphoma kinase inhibitors and mammalian target of rapamycin inhibitors. EXPERT OPINION Novel agents used in the treatment of cancer have specific side-effects, the result of allergic reactions, on-target and off-target effects. Clinical syndromes associated with pulmonary toxicity vary from bronchospasms, hypersensitivity reactions, pneumonitis, acute respiratory distress, lung bleeding, pleural effusion to pneumothorax. Knowledge of risk factors, a high index of suspicion and a complete diagnostic work-up are essential for limiting the risk of these events becoming life threatening. The development of treatment algorithms is extremely helpful in managing these events. It is probable that these toxicities will be even more frequent with the introduction of combination therapies with the obvious challenge of discerning the responsible agent.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- a 1 Sint-Augustinus, Resident in Internal Medicine , Oosterveldlaan 24, 2610 Wilrijk-Antwerp, Belgium
| | - Tom Van den Mooter
- b 2 Sint-Augustinus, Resident in Medical Oncology , Oosterveldlaan 24, 2610 Wilrijk-Antwerp, Belgium
| | - Luc Dirix
- c 3 Sint-Augustinus, Medical Oncology , Oosterveldlaan 24, 2610 Wilrijk-Antwerp, Belgium +32 34 433 737 ; +32 34 430 09 ;
| |
Collapse
|
128
|
Abstract
Angiogenesis is the process through which new blood vessels are formed from pre-existing vessels and is essential for the growth of all solid tumors. Vascular endothelial growth factor (VEGF) is a regulator of angiogenesis, which is crucial for tumor growth and metastasis. Its inhibition with antiangiogenic drugs is thought to improve delivery of chemotherapy through vascular normalization and disruption of tumor vasculature. Aflibercept is a recombinant fusion protein of the VEGF receptor (VEGFR)1 and VEGFR2 extracellular domains that binds to VEGF-A, VEGF-B, placental growth factor (PlGF) 1 and 2. Aflibercept has demonstrated preclinical efficacy in different tumor types and exerts its antiangiogenic effects through regression of tumor vasculature, remolding of vasculature, and inhibition of new tumor vessel growth. This review examines the effects of aflibercept on tumor vasculature and on different types of solid tumors, and explores the preclinical and clinical benefits of inclusion aflibercept into anticancer treatment strategies.
Collapse
Affiliation(s)
- Vincenzo Ricci
- Oncology Department, San Raffaele Institute, 60, Olgettina St., 20132 Milan, Italy
| | - Monica Ronzoni
- Oncology Department, San Raffaele Institute, 60, Olgettina St., 20132 Milan, Italy
| | - Teresa Fabozzi
- San Raffaele Institute, 60, Olgettina St., 20132 Milan, Italy
| |
Collapse
|
129
|
Flaherty KT, Hamilton BK, Rosen MA, Amaravadi RK, Schuchter LM, Gallagher M, Chen H, Sehgal C, O'Dwyer PJ. Phase I/II Trial of Imatinib and Bevacizumab in Patients With Advanced Melanoma and Other Advanced Cancers. Oncologist 2015; 20:952-9. [PMID: 26084808 DOI: 10.1634/theoncologist.2015-0108] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/21/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor and platelet-derived growth factor signaling in the tumor microenvironment appear to cooperate in promoting tumor angiogenesis. PATIENTS AND METHODS We conducted a phase I trial combining bevacizumab (i.v. every 2 weeks) and imatinib (oral daily). Once a recommended phase II dose combination was established, a phase II trial was initiated in patients with metastatic melanoma. A Simon 2-stage design was used with 23 patients required in the first stage and 41 patients in total should the criteria to proceed be met. We required that 50% of the patients be progression-free at 16 weeks. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and power Doppler ultrasonography were performed in patients with metastatic tumors amenable to imaging with these methods at baseline and after 4 weeks. RESULTS A total of 17 patients were accrued to 4 dose and combination levels. Bevacizumab 10 mg/kg every 2 weeks could be safely combined with imatinib 800 mg daily. Common toxicities included fatigue, nausea, vomiting, edema, proteinuria, and anemia, but were not commonly severe. A total of 23 patients with metastatic melanoma (48% with American Joint Commission on Cancer stage M1c; median age, 63 years) were enrolled in the first stage of phase II. The 16-week progression-free survival rate was 35%, leading to termination of phase II after the first stage. In the small subset of patients who remained on study with lesions evaluable by DCE-MRI, significant decreases in tumor vascular permeability were noted, despite early disease progression using the Response Evaluation Criteria In Solid Tumors. CONCLUSION Bevacizumab and imatinib can be safely combined at the maximum doses used for each agent. We did not observe significant clinical activity with this regimen in melanoma patients.
Collapse
Affiliation(s)
- Keith T Flaherty
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Betty K Hamilton
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark A Rosen
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Ravi K Amaravadi
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Lynn M Schuchter
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Maryann Gallagher
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Helen Chen
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Chandra Sehgal
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Peter J O'Dwyer
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
130
|
Carneiro RM, Carneiro BA, Agulnik M, Kopp PA, Giles FJ. Targeted therapies in advanced differentiated thyroid cancer. Cancer Treat Rev 2015; 41:690-8. [PMID: 26105190 DOI: 10.1016/j.ctrv.2015.06.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 12/12/2022]
Abstract
Differentiated thyroid cancer is the most common endocrine malignancy, and its incidence has been rising rapidly over the past 10 years. Although most patients with this disease have an excellent prognosis, a subset develops a more aggressive disease phenotype refractory to conventional therapies. Until recently, there was no effective therapy for these patients. With increasing knowledge of the molecular pathogenesis of thyroid cancer, novel targeted therapies are being developed for this group of patients. Sorafenib and lenvatinib, small-molecule multikinase inhibitors, were approved for the treatment of progressive, symptomatic, radioactive iodine refractory, advanced differentiated thyroid cancer in 2013 and 2015, respectively. This represents a major innovation in the therapy of patients with advanced thyroid cancer. However, these therapies still have many limitations and further research needs to be pursued with the ultimate goal of providing safe and effective personalized therapy for patients with advanced thyroid cancer.
Collapse
Affiliation(s)
- Raquel M Carneiro
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, United States; Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, United States; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, United States.
| | - Benedito A Carneiro
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, United States; Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, United States; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, United States.
| | - Mark Agulnik
- Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, United States; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, United States.
| | - Peter A Kopp
- Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, United States; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, United States.
| | - Francis J Giles
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, United States; Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, United States; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, United States.
| |
Collapse
|
131
|
Lee SH, Jeong D, Han YS, Baek MJ. Pivotal role of vascular endothelial growth factor pathway in tumor angiogenesis. Ann Surg Treat Res 2015; 89:1-8. [PMID: 26131438 PMCID: PMC4481026 DOI: 10.4174/astr.2015.89.1.1] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/06/2015] [Accepted: 02/27/2015] [Indexed: 12/18/2022] Open
Abstract
The shaping of new blood vessels is a significant event in cancer growth and metastasis. Therefore, the molecular system of cancer angiogenesis has garnered considerable interest in cancer research. The vascular endothelial growth factor (VEGF) and VEGF receptor pathway are recognized as the key regulators of the angiogenic process. Activation of the VEGF/VEGF-receptor pathway initiates signaling cascades that promote endothelial cell growth, migration, and differentiation. Recently, VEGF was shown to play a role in the recruitment of bone marrow-derived endothelial progenitor cells to neovascularization sites. The role of VEGF in promoting tumor angiogenesis and the occurrence of human cancers has led to the rational design and development of agents that selectively target this pathway. Moreover, these anti-VEGF/VEGF receptor agents show therapeutic potential by inhibition of angiogenesis and tumor growth in preclinical models. In this review, we summarize the role of the VEGF pathway during tumor angiogenesis.
Collapse
Affiliation(s)
- Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea. ; Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Dongjun Jeong
- Department of Pathology, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Moo Jun Baek
- Department of Surgery, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
132
|
Zhang SD, McCrudden CM, Meng C, Lin Y, Kwok HF. The significance of combining VEGFA, FLT1, and KDR expressions in colon cancer patient prognosis and predicting response to bevacizumab. Onco Targets Ther 2015; 8:835-43. [PMID: 25926745 PMCID: PMC4403689 DOI: 10.2147/ott.s80518] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Targeting angiogenesis through inhibition of the vascular endothelial growth factor (VEGF) pathway has been successful in the treatment of late stage colorectal cancer. However, not all patients benefit from inhibition of VEGF. Ras status is a powerful biomarker for response to anti-epidermal growth factor receptor therapy; however, an appropriate biomarker for response to anti-VEGF therapy is yet to be identified. VEGF and its receptors, FLT1 and KDR, play a crucial role in colon cancer progression; individually, these factors have been shown to be prognostic in colon cancer; however, expression of none of these factors alone was predictive of tumor response to anti-VEGF therapy. In the present study, we analyzed the expression levels of VEGFA, FLT1, and KDR in two independent colon cancer datasets and found that high expression levels of all three factors afforded a very poor prognosis. The observation was further confirmed in another independent colon cancer dataset, wherein high levels of expression of this three-gene signature was predictive of poor prognosis in patients with proficient mismatch repair a wild-type KRas status, or mutant p53 status. Most importantly, this signature also predicted tumor response to bevacizumab, an antibody targeting VEGFA, in a cohort of bevacizumab-treated patients. Since bevacizumab has been proven to be an important drug in the treatment of advanced stage colon cancer, our results suggest that the three-gene signature approach is valuable in terms of its prognostic value, and that it should be further evaluated in a prospective clinical trial to investigate its predictive value to anti-VEGF treatment.
Collapse
Affiliation(s)
- Shu-Dong Zhang
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau, Special Administrative Region of the People’s Republic of China
- Center for Cancer Research and Cell Biology, Queen’s University Belfast, United Kingdom
| | | | - Chen Meng
- College of Life Sciences, Fujian Normal University, Fujian, People’s Republic of China
| | - Yao Lin
- College of Life Sciences, Fujian Normal University, Fujian, People’s Republic of China
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau, Special Administrative Region of the People’s Republic of China
- School of Pharmacy, Queen’s University Belfast, United Kingdom
| |
Collapse
|
133
|
Hu Q, Wu M, Fang C, Cheng C, Zhao M, Fang W, Chu PK, Ping Y, Tang G. Engineering nanoparticle-coated bacteria as oral DNA vaccines for cancer immunotherapy. NANO LETTERS 2015; 15:2732-9. [PMID: 25806599 DOI: 10.1021/acs.nanolett.5b00570] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Live attenuated bacteria are of increasing importance in biotechnology and medicine in the emerging field of cancer immunotherapy. Oral DNA vaccination mediated by live attenuated bacteria often suffers from low infection efficiency due to various biological barriers during the infection process. To this end, we herein report, for the first time, a new strategy to engineer cationic nanoparticle-coated bacterial vectors that can efficiently deliver oral DNA vaccine for efficacious cancer immunotherapy. By coating live attenuated bacteria with synthetic nanoparticles self-assembled from cationic polymers and plasmid DNA, the protective nanoparticle coating layer is able to facilitate bacteria to effectively escape phagosomes, significantly enhance the acid tolerance of bacteria in stomach and intestines, and greatly promote dissemination of bacteria into blood circulation after oral administration. Most importantly, oral delivery of DNA vaccines encoding autologous vascular endothelial growth factor receptor 2 (VEGFR2) by this hybrid vector showed remarkable T cell activation and cytokine production. Successful inhibition of tumor growth was also achieved by efficient oral delivery of VEGFR2 with nanoparticle-coated bacterial vectors due to angiogenesis suppression in the tumor vasculature and tumor necrosis. This proof-of-concept work demonstrates that coating live bacterial cells with synthetic nanoparticles represents a promising strategy to engineer efficient and versatile DNA vaccines for the era of immunotherapy.
Collapse
MESH Headings
- Administration, Oral
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/chemistry
- Cell Line, Tumor
- Coated Materials, Biocompatible/chemical synthesis
- Humans
- Immunotherapy, Active/methods
- Nanocapsules/administration & dosage
- Nanocapsules/chemistry
- Nanocapsules/ultrastructure
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/microbiology
- Neoplasms, Experimental/pathology
- Salmonella/physiology
- Transformation, Bacterial
- Treatment Outcome
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/chemistry
Collapse
Affiliation(s)
- Qinglian Hu
- †Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, China
| | - Min Wu
- †Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, China
| | - Chun Fang
- ‡College of Animal Science, Zhejiang University, Hangzhou 310028, China
| | - Changyong Cheng
- ‡College of Animal Science, Zhejiang University, Hangzhou 310028, China
| | - Mengmeng Zhao
- †Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, China
| | - Weihuan Fang
- ‡College of Animal Science, Zhejiang University, Hangzhou 310028, China
| | - Paul K Chu
- §Department of Physics and Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Yuan Ping
- ∥School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Guping Tang
- †Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, China
| |
Collapse
|
134
|
Targeting diverse protein-protein interaction interfaces with α/β-peptides derived from the Z-domain scaffold. Proc Natl Acad Sci U S A 2015; 112:4552-7. [PMID: 25825775 DOI: 10.1073/pnas.1420380112] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Peptide-based agents derived from well-defined scaffolds offer an alternative to antibodies for selective and high-affinity recognition of large and topologically complex protein surfaces. Here, we describe a strategy for designing oligomers containing both α- and β-amino acid residues ("α/β-peptides") that mimic several peptides derived from the three-helix bundle "Z-domain" scaffold. We show that α/β-peptides derived from a Z-domain peptide targeting vascular endothelial growth factor (VEGF) can structurally and functionally mimic the binding surface of the parent peptide while exhibiting significantly decreased susceptibility to proteolysis. The tightest VEGF-binding α/β-peptide inhibits the VEGF165-induced proliferation of human umbilical vein endothelial cells. We demonstrate the versatility of this strategy by showing how principles underlying VEGF signaling inhibitors can be rapidly extended to produce Z-domain-mimetic α/β-peptides that bind to two other protein partners, IgG and tumor necrosis factor-α. Because well-established selection techniques can identify high-affinity Z-domain derivatives from large DNA-encoded libraries, our findings should enable the design of biostable α/β-peptides that bind tightly and specifically to diverse targets of biomedical interest. Such reagents would be useful for diagnostic and therapeutic applications.
Collapse
|
135
|
Smith J, Liu F, Beyer B, Morales K, Reilly A, Cole R, Herron BJ. Angiogenesis QTL on Mouse Chromosome 8 Colocalizes with Differential β-Defensin Expression. J Biomol Tech 2015; 26:45-53. [PMID: 25802489 DOI: 10.7171/jbt.15-2602-002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Identification of genetic factors that modify complex traits is often complicated by gene-environment interactions that contribute to the observed phenotype. In model systems, the phenotypic outcomes quantified are typically traits that maximize observed variance, which in turn, should maximize the detection of quantitative trait loci (QTL) in subsequent mapping studies. However, when the observed trait is dependent on multiple interacting factors, it can complicate genetic analysis, reducing the likelihood that the modifying mutation will ultimately be found. Alternatively, by focusing on intermediate phenotypes of a larger condition, we can reduce a model's complexity, which will, in turn, limit the number of QTL that contribute to variance. We used a novel method to follow angiogenesis in mice that reduces environmental variance by measuring endothelial cell growth from culture of isolated skin biopsies that varies depending on the genetic source of the tissue. This method, in combination with a backcross breeding strategy, is intended to reduce genetic complexity and limit the phenotypic effects to fewer modifier loci. We determined that our approach was an efficient means to generate recombinant progeny and used this cohort to map a novel s.c. angiogenesis QTL to proximal mouse chromosome (Chr.) 8 with suggestive QTL on Chr. 2 and 7. Global mRNA expression analysis of samples from parental reference strains revealed β-defensins as potential candidate genes for future study.
Collapse
Affiliation(s)
- Jason Smith
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Fang Liu
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Barbara Beyer
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Krista Morales
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Andrew Reilly
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Richard Cole
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Bruce J Herron
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| |
Collapse
|
136
|
Zeng Y, Liu Y, Shang J, Ma J, Wang R, Deng L, Guo Y, Zhong F, Bai M, Zhang S, Wu D. Phosphorescence monitoring of hypoxic microenvironment in solid-tumors to evaluate chemotherapeutic effects using the hypoxia-sensitive iridium (III) coordination compound. PLoS One 2015; 10:e0121293. [PMID: 25786221 PMCID: PMC4365010 DOI: 10.1371/journal.pone.0121293] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/29/2015] [Indexed: 02/06/2023] Open
Abstract
Objectives To utilize phosphorescence to monitor hypoxic microenvironment in solid-tumors and investigate cancer chemotherapeutic effects in vivo. Methods A hypoxia-sensitive probe named BTP was used to monitor hypoxic microenvironment in solid-tumors. The low-dose metronomic treatment with cisplatin was used in anti-angiogenetic chemotherapeutic programs. The phosphorescence properties of BTP were detected by a spectrofluorometer. BTP cytotoxicity utilized cell necrosis and apoptosis, which were evaluated by trypan blue dye exclusion and Hoechst33342 plus propidium iodide assays. Tumor-bearing mouse models of colon adenocarcinoma were used for tumor imaging in vivo. Monitoring of the hypoxic microenvironment in tumors was performed with a Maestro 2 fluorescence imaging system. Tumor tissues in each group were harvested regularly and treated with pathological hematoxylin and eosin and immunohistochemical staining to confirm imaging results. Results BTP did not feature obvious cytotoxicity for cells, and tumor growth in low-dose metronomic cisplatin treated mice was significantly inhibited by chemotherapy. Hypoxic levels significantly increased due to cisplatin, as proven by the expression level of related proteins. Phosphorescence intensity in the tumors of mice in the cisplatin group was stronger and showed higher contrast than that in tumors of saline treated mice.
Collapse
Affiliation(s)
- Yun Zeng
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Yang Liu
- School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Jin Shang
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Jingwen Ma
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Rong Wang
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Lei Deng
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Youmin Guo
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Fan Zhong
- School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Mingfeng Bai
- Molecular Imaging Laboratory, Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Shaojuan Zhang
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
- Molecular Imaging Laboratory, Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (SZ); (DW)
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
- * E-mail: (SZ); (DW)
| |
Collapse
|
137
|
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: 1.4] [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
|
138
|
Ozturk D, Yonucu S, Yilmaz D, Unlu MB. Influence of vascular normalization on interstitial flow and delivery of liposomes in tumors. Phys Med Biol 2015; 60:1477-96. [PMID: 25611340 DOI: 10.1088/0031-9155/60/4/1477] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Elevated interstitial fluid pressure is one of the barriers of drug delivery in solid tumors. Recent studies have shown that normalization of tumor vasculature by anti-angiogenic factors may improve the delivery of conventional cytotoxic drugs, possibly by increasing blood flow, decreasing interstitial fluid pressure, and enhancing the convective transvascular transport of drug molecules. Delivery of large therapeutic agents such as nanoparticles and liposomes might also benefit from normalization therapy since their transport depends primarily on convection. In this study, a mathematical model is presented to provide supporting evidence that normalization therapy may improve the delivery of 100 nm liposomes into solid tumors, by both increasing the total drug extravasation and providing a more homogeneous drug distribution within the tumor. However these beneficial effects largely depend on tumor size and are stronger for tumors within a certain size range. It is shown that this size effect may persist under different microenvironmental conditions and for tumors with irregular margins or heterogeneous blood supply.
Collapse
Affiliation(s)
- Deniz Ozturk
- Department of Physics, Bogazici University, 34342 Bebek, Istanbul, Turkey. Center for Life Sciences and Technologies, Bogazici University, 34342 Bebek, Istanbul, Turkey
| | | | | | | |
Collapse
|
139
|
Guan YY, Liu HJ, Luan X, Xu JR, Lu Q, Liu YR, Gao YG, Zhao M, Chen HZ, Fang C. Raddeanin A, a triterpenoid saponin isolated from Anemone raddeana, suppresses the angiogenesis and growth of human colorectal tumor by inhibiting VEGFR2 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:103-110. [PMID: 25636878 DOI: 10.1016/j.phymed.2014.11.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/27/2014] [Accepted: 11/15/2014] [Indexed: 06/04/2023]
Abstract
Raddeanin A (RA) is an active triterpenoid saponin from a traditional Chinese medicinal herb, Anemone raddeana Regel. It was previously reported that RA possessed attractive antitumor activity through inhibiting proliferation and inducing apoptosis of multiple cancer cells. However, whether RA can inhibit angiogenesis, an essential step in cancer development, remains unknown. In this study, we found that RA could significantly inhibit human umbilical vein endothelial cell (HUVEC) proliferation, motility, migration, and tube formation. RA also dramatically reduced angiogenesis in chick embryo chorioallantoic membrane (CAM), restrained the trunk angiogenesis in zebrafish, and suppressed angiogenesis and growth of human HCT-15 colorectal cancer xenograft in mice. Western blot assay showed that RA suppressed VEGF-induced phosphorylation of VEGFR2 and its downstream protein kinases including PLCγ1, JAK2, FAK, Src, and Akt. Molecular docking simulation indicated that RA formed hydrogen bonds and hydrophobic interactions within the ATP binding pocket of VEGFR2 kinase domain. Our study firstly provides the evidence that RA has high antiangiogenic potency and explores its molecular basis, demonstrating that RA is a potential agent or lead candidate for antiangiogenic cancer therapy.
Collapse
Affiliation(s)
- Ying-Yun Guan
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Hai-Jun Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Xin Luan
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Jian-Rong Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Ya-Rong Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Yun-Ge Gao
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Mei Zhao
- Department of Pharmacy, Shanghai Institute of Health Sciences and Health School Attached to SJTU-SM, 279 Zhouzhu Road, Shanghai 201318, China.
| | - Hong-Zhuan Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China.
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China.
| |
Collapse
|
140
|
Michael IP, Westenskow PD, Hacibekiroglu S, Greenwald AC, Ballios BG, Kurihara T, Li Z, Warren CM, Zhang P, Aguilar E, Donaldson L, Marchetti V, Baba T, Hussein SM, Sung HK, Iruela-Arispe ML, Rini JM, van der Kooy D, Friedlander M, Nagy A. Local acting Sticky-trap inhibits vascular endothelial growth factor dependent pathological angiogenesis in the eye. EMBO Mol Med 2014; 6:604-23. [PMID: 24705878 PMCID: PMC4023884 DOI: 10.1002/emmm.201303708] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Current therapeutic antiangiogenic biologics used for the treatment of pathological ocular angiogenesis could have serious side effects due to their interference with normal blood vessel physiology. Here, we report the generation of novel antivascular endothelial growth factor-A (VEGF) biologics, termed VEGF “Sticky-traps,” with unique properties that allow for local inhibition of angiogenesis without detectable systemic side effects. Using genetic and pharmacological approaches, we demonstrated that Sticky-traps could locally inhibit angiogenesis to at least the same extent as the original VEGF-trap that also gains whole-body access. Sticky-traps did not cause systemic effects, as shown by uncompromised wound healing and normal tracheal vessel density. Moreover, if injected intravitreally, recombinant Sticky-trap remained localized to various regions of the eye, such as the inner-limiting membrane and ciliary body, for prolonged time periods, without gaining access either to the photoreceptors/choriocapillaris area or the circulation. These unique pharmacological characteristics of Sticky-trap could allow for safe treatment of pathological angiogenesis in patients with diabetic retinopathy and retinopathy of pre-maturity.
Collapse
Affiliation(s)
- Iacovos P Michael
- Lunenfeld-Tanenbaum Research Institute Mount Sinai Hospital, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Amin M, Lockhart AC. The potential role of immunotherapy to treat colorectal cancer. Expert Opin Investig Drugs 2014; 24:329-44. [PMID: 25519074 DOI: 10.1517/13543784.2015.985376] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the fourth most common cancer and the second leading cause of cancer-related death worldwide. Surgery, chemotherapy, radiation therapy and anti-angiogenic therapies form the backbone of treatment for CRC in various stages. Immunotherapy is frequently used either alone or in combination with chemotherapy for the treatment of various cancers such as melanoma, prostate cancer and renal cell cancer. Current CRC research is moving forward to discover ways to incorporate immunotherapies into the treatment of CRC. AREAS COVERED The aim of this review is to summarize the potential role of immunotherapy in CRC. Herein, the authors provide a brief overview of immune modulatory cells, immune surveillance and escape in CRC. They also review vaccine trials in addition to cytokines and monoclonal antibodies. This coverage includes ongoing trials and checkpoint inhibitors such as cytotoxic T lymphocyte antigen-1, programmed cell death-1, and PDL1. EXPERT OPINION Checkpoint inhibitors in combination with either chemotherapy or chemo-antiangiogenic-therapy may represent a future therapeutic approach for CRC incorporating immune system targeting. Given the success of immune-based therapy in other tumor types, the authors anticipate that a similar breakthrough in CRC will be forthcoming.
Collapse
Affiliation(s)
- Manik Amin
- Washington University, Siteman Cancer Center , 660 S. Euclid Ave, Box 8056, St. Louis, MO 63110 , USA
| | | |
Collapse
|
142
|
Tomao F, Papa A, Rossi L, Zaccarelli E, Caruso D, Zoratto F, Benedetti Panici P, Tomao S. Angiogenesis and antiangiogenic agents in cervical cancer. Onco Targets Ther 2014; 7:2237-48. [PMID: 25506227 PMCID: PMC4259513 DOI: 10.2147/ott.s68286] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Standard treatment of cervical cancer (CC) consists of surgery in the early stages and of chemoradiation in locally advanced disease. Metastatic CC has a poor prognosis and is usually treated with palliative platinum-based chemotherapy. Current chemotherapeutic regimens are associated with significant adverse effects and only limited activity, making identification of active and tolerable novel targeted agents a high priority. Angiogenesis is a complex process that plays a crucial role in the development of many types of cancer. The dominant role of angiogenesis in CC seems to be directly related to human papillomavirus-related inhibition of p53 and stabilization of hypoxia-inducible factor-1α. Both of these mechanisms are able to increase expression of vascular endothelial growth factor (VEGF). Activation of VEGF promotes endothelial cell proliferation and migration, favoring formation of new blood vessels and increasing permeability of existing blood vessels. Since bevacizumab, a recombinant humanized monoclonal antibody binding to all isoforms of VEGF, has been demonstrated to significantly improve survival in gynecologic cancer, some recent clinical research has explored the possibility of using novel therapies directed toward inhibition of angiogenesis in CC too. Here we review the main results from studies concerning the use of antiangiogenic drugs that are being investigated for the treatment of CC.
Collapse
Affiliation(s)
- Federica Tomao
- Department of Gynecology and Obstetrics, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Anselmo Papa
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Oncology Unit, ICOT, Latina, Italy
| | - Luigi Rossi
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Oncology Unit, ICOT, Latina, Italy
| | - Eleonora Zaccarelli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Oncology Unit, ICOT, Latina, Italy
| | - Davide Caruso
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Oncology Unit, ICOT, Latina, Italy
| | - Federica Zoratto
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Oncology Unit, ICOT, Latina, Italy
| | - Pierluigi Benedetti Panici
- Department of Gynecology and Obstetrics, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Silverio Tomao
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Oncology Unit, ICOT, Latina, Italy
| |
Collapse
|
143
|
Abstract
Gastric cancer is the fourth most common cancer globally and represents the second most common cause of cancer-related mortality. Early detection, aggressive surgical resection, and postoperative adjuvant therapy have led to survival improvement for early-stage gastric cancer, particularly in Asian countries. Unfortunately, advanced gastric cancer continues to pose a formidable challenge with few gains being reported recently. Trastuzumab was the first targeted agent to be approved for the treatment of advanced gastric cancer in 2010. The failure of the AVAGAST trial was a setback for antiangiogenic therapy for this disease. Ramucirumab is a monoclonal antibody that binds to VEGF-R2 and prevents its activation. The recent REGARD trial was a randomized phase III trial of ramucirumab vs. placebo for patients with advanced, pretreated gastric cancer that met its primary endpoint of increased overall survival. The toxicity of ramucirumab was modest in this setting, with an increased risk of grade 3 or higher hypertension (8% vs. 3%, with ramucirumab and placebo, respectively). The subsequent RAINBOW trial of paclitaxel plus ramucirumab vs. paclitaxel plus placebo for advanced pretreated gastric cancer confirmed the survival advantage of this antiangiogenic agent in gastric cancer. Ramucirumab is the first FDA-approved therapy for advanced gastric cancer after prior chemotherapy.
Collapse
Affiliation(s)
- Milind Javle
- Department of Gastrointestinal Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Elizabeth C Smyth
- Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| |
Collapse
|
144
|
“Programmed packaging” for gene delivery. J Control Release 2014; 193:316-23. [DOI: 10.1016/j.jconrel.2014.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/26/2014] [Accepted: 04/10/2014] [Indexed: 11/21/2022]
|
145
|
Podhorecka M, Markowicz J, Szymczyk A, Pawlowski J. Target Therapy in Hematological Malignances: New Monoclonal Antibodies. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:701493. [PMID: 27433507 PMCID: PMC4897146 DOI: 10.1155/2014/701493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 11/17/2022]
Abstract
Apart from radio- and chemotherapy, monoclonal antibodies (MoAbs) represent a new, more selective tool in the treatment of hematological malignancies. MoAbs bind with the specific antigens of the tumors. This interaction is a basis for targeted therapies which exhibit few side effects and significant antitumor activity. This review provides an overview of the functional characteristics of MoAbs, with some examples of their clinical application. The promising results in the treatment of hematological malignancies have led to the more frequent usage of MoAbs in the therapy. Development of MoAbs is a subject of extensive research. They are a promising method of cancer treatment in the future.
Collapse
Affiliation(s)
- Monika Podhorecka
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Staszica 11, 20-081 Lublin, Poland
| | - Justyna Markowicz
- Students Scientific Association at the Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Szymczyk
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Staszica 11, 20-081 Lublin, Poland
| | - Johannes Pawlowski
- Students Scientific Association at the Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
146
|
Guerrouahen BS, Pasquier J, Kaoud NA, Maleki M, Beauchamp MC, Yasmeen A, Ghiabi P, Lis R, Vidal F, Saleh A, Gotlieb WH, Rafii S, Rafii A. Akt-activated endothelium constitutes the niche for residual disease and resistance to bevacizumab in ovarian cancer. Mol Cancer Ther 2014; 13:3123-36. [PMID: 25319392 DOI: 10.1158/1535-7163.mct-13-1053] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ovarian cancer is the second leading cause of cancer-related death in women worldwide. Despite optimal cytoreduction and adequate adjuvant therapies, initial tumor response is often followed by relapse suggesting the existence of a tumor niche. Targeted therapies have been evaluated in ovarian cancer to overcome resistant disease. Among them, antiangiogenic therapies inhibit new blood vessel growth, induce endothelial cell apoptosis, and block the incorporation of hematopoietic and endothelial progenitor cells into new blood vessels. Despite in vitro and in vivo successes, antivascular therapy with bevacizumab targeting VEGF-A has limited efficacy in ovarian cancer. The precise molecular mechanisms underlying clinical resistance to anti-VEGF therapies are not yet well understood. Among them, tumor and stromal heterogeneity might determine the treatment outcomes. The present study investigates whether abnormalities in the tumor endothelium may contribute to treatment resistance to bevacizumab and promote a residual microscopic disease. Here, we showed that ovarian cancer cells activate Akt phosphorylation in endothelial cells inducing resistance to bevacizumab leading to an autocrine loop based on FGF2 secretion. Altogether, our results point out the role of an activated endothelium in the resistance to bevacizumab and in the constitution of a niche for a residual disease.
Collapse
Affiliation(s)
- Bella S Guerrouahen
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar. Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Jennifer Pasquier
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar. Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Nadine Abu Kaoud
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Mahtab Maleki
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Marie-Claude Beauchamp
- Department of Gynecologic Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Amber Yasmeen
- Department of Gynecologic Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Pegah Ghiabi
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Raphael Lis
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Fabien Vidal
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Ahmed Saleh
- National Center for Cancer Care and Research, Doha, Qatar
| | - Walter H Gotlieb
- Department of Gynecologic Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Shahin Rafii
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Arash Rafii
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar. Department of Genetic Medicine, Weill Cornell Medical College, New York, New York. Department of Gynecologic oncology, University Montpellier, Montpellier, France.
| |
Collapse
|
147
|
Kadomatsu K, Bencsik P, Görbe A, Csonka C, Sakamoto K, Kishida S, Ferdinandy P. Therapeutic potential of midkine in cardiovascular disease. Br J Pharmacol 2014; 171:936-44. [PMID: 24286213 DOI: 10.1111/bph.12537] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/12/2013] [Accepted: 11/20/2013] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED Ischaemic heart disease, stroke and their pathological consequences are life-threatening conditions that account for about half of deaths in developed countries. Pathology of these diseases includes cell death due to ischaemia/reperfusion injury, vascular stenosis and cardiac remodelling. The growth factor midkine plays a pivotal role in these events. Midkine shows an acute cytoprotective effect in ischaemia/reperfusion injury at least in part via its anti-apoptotic effect. Moreover, while midkine promotes endothelial cell proliferation, it also recruits inflammatory cells to lesions. These activities eventually enhance angiogenesis, thereby preventing cardiac tissue remodelling. However, midkine's activity in recruiting inflammatory cells into the vascular wall also triggers neointima formation, and consequently, vascular stenosis. Moreover, midkine is induced in cancer tissues where it enhances angiogenesis. Therefore, midkine may promote tumour formation through its angiogenic and anti-apoptotic activity. This review focuses on the roles of midkine in ischaemic cardiovascular disease and their pathological consequences, that is angiogenesis, vascular stenosis, and cardiac remodelling, and discusses the possible therapeutic potential of modulation of midkine in these diseases. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
148
|
Soria JC, DeBraud F, Bahleda R, Adamo B, Andre F, Dientsmann R, Delmonte A, Cereda R, Isaacson J, Litten J, Allen A, Dubois F, Saba C, Robert R, D'Incalci M, Zucchetti M, Camboni MG, Tabernero J. Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Ann Oncol 2014; 25:2244-2251. [PMID: 25193991 DOI: 10.1093/annonc/mdu390] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Lucitanib is a potent, oral inhibitor fibroblast growth factor receptor types 1 and 2 (FGFR), vascular endothelial growth factor receptor types 1, 2, and 3 (VEGFR), platelet-derived growth factor receptor types α and β (PGFRα/β), which are essential kinases for tumor growth, survival, migration, and angiogenesis. Several tumor types, including breast carcinoma, demonstrate amplification of fibroblast growth factor (FGF)-related genes. There are no approved drugs for molecularly defined FGF-aberrant (FGFR1- or FGF3/4/19-amplified) tumors. METHODS This open-label phase I/IIa study involved a dose-escalation phase to determine maximum tolerated dose (MTD), recommended dose (RD), and pharmacokinetics of lucitanib in patients with advanced solid tumors, followed by a dose-expansion phase to obtain preliminary evidence of efficacy in patients who could potentially benefit from treatment (i.e. with tumors harboring FGF-aberrant pathway or considered angiogenesis-sensitive). RESULTS Doses from 5 to 30 mg were evaluated with dose-limiting toxic effects dominated by vascular endothelial growth factor (VEGF) inhibition-related toxic effects at the 30 mg dose level (one case of grade 4 depressed level of consciousness and two cases of grade 3 thrombotic microangiopathy). The most common adverse events (all grades, all cohorts) were hypertension (91%), asthenia (42%), and proteinuria (57%). Exposure increased with dose and t½ was 31-40 h, suitable for once daily administration. Seventy-six patients were included. All but one had stage IV; 42% had >3 lines of previous chemotherapy. Sixty-four patients were assessable for response; 58 had measurable disease. Clinical activity was observed at all doses tested with durable Response Evaluation Criteria In Solid Tumors (RECIST) partial responses in a variety of tumor types. In the angiogenesis-sensitive group, objective RECIST response rate (complete response + partial response) was 26% (7 of 27) and progression-free survival (PFS) was 25 weeks. In assessable FGF-aberrant breast cancer patients, 50% (6 of 12) achieved RECIST partial response with a median PFS of 40.4 weeks for all treated patients. CONCLUSION Lucitanib has promising efficacy and a manageable side-effect profile. The spectrum of activity observed demonstrates clinical benefit in both FGF-aberrant and angiogenesis-sensitive populations. A comprehensive phase II program is planned.
Collapse
Affiliation(s)
- J-C Soria
- Department of Drug Development, Gustave-Roussy Cancer Campus, Villejuif, France.
| | - F DeBraud
- European Institute of Oncology, Milan, Italy
| | - R Bahleda
- Department of Drug Development, Gustave-Roussy Cancer Campus, Villejuif, France
| | - B Adamo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - F Andre
- Department of Drug Development, Gustave-Roussy Cancer Campus, Villejuif, France
| | - R Dientsmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain; Sage Bionetworks, Fred Hutchinson Cancer Research Center, Seattle
| | - A Delmonte
- European Institute of Oncology, Milan, Italy
| | - R Cereda
- Clovis Oncology, Inc., San Francisco; Clovis Oncology, Inc., Boulder,USA; Clovis Oncology, Inc., Milan, Italy
| | - J Isaacson
- Clovis Oncology, Inc., San Francisco; Clovis Oncology, Inc., Boulder,USA; Clovis Oncology, Inc., Milan, Italy
| | - J Litten
- Clovis Oncology, Inc., San Francisco; Clovis Oncology, Inc., Boulder,USA; Clovis Oncology, Inc., Milan, Italy
| | - A Allen
- Clovis Oncology, Inc., San Francisco; Clovis Oncology, Inc., Boulder,USA; Clovis Oncology, Inc., Milan, Italy
| | - F Dubois
- Institut de Recherche International Servier, Suresnes, France
| | - C Saba
- Institut de Recherche International Servier, Suresnes, France
| | - R Robert
- Institut de Recherche International Servier, Suresnes, France
| | - M D'Incalci
- Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, Milan, Italy
| | - M Zucchetti
- Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, Milan, Italy
| | - M G Camboni
- Clovis Oncology, Inc., San Francisco; Clovis Oncology, Inc., Boulder,USA; Clovis Oncology, Inc., Milan, Italy
| | - J Tabernero
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
149
|
Macarulla T, Sauri T, Tabernero J. Evaluation of aflibercept in the treatment of metastatic colorectal cancer. Expert Opin Biol Ther 2014; 14:1493-505. [DOI: 10.1517/14712598.2014.947956] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
150
|
Yazama H, Kitatani K, Fujiwara K, Kato M, Hashimoto-Nishimura M, Kawamoto K, Hasegawa K, Kitano H, Bielawska A, Bielawski J, Okazaki T. Dietary glucosylceramides suppress tumor growth in a mouse xenograft model of head and neck squamous cell carcinoma by the inhibition of angiogenesis through an increase in ceramide. Int J Clin Oncol 2014; 20:438-46. [PMID: 25080062 DOI: 10.1007/s10147-014-0734-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/14/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND We previously reported that dietary glucosylceramides show cancer-prevention activity in a mouse xenograft model of human head and neck cancer cells (SCCKN). However, the mechanism was unclear. Ceramides, metabolites of glucosylceramides, induce apoptotic cell death in various malignancies. Here, we investigated the inhibitory effects of dietary glucosylceramides on tumor growth in vivo and in vitro. METHODS SCCKN were subcutaneously inoculated into the right flanks of NOD/SCID mice. Mice were treated with or without dietary glucosylceramides (300 mg/kg) daily for 14 consecutive days after confirmation of tumor progression. Microvessel areas around the tumor were assessed by hematoxylin-eosin staining and immunohistochemistry of CD31, and, as markers for angiogenesis, protein levels of VEGF, VEGF receptor-2, and HIF-1α were assessed by Western blotting. Mass spectrometry was performed to measure the levels of sphingolipids in mouse serum after treatment with dietary glucosylceramides. RESULTS Oral administration of glucosylceramides significantly decreased SCCKN growth in the xenograft model with inhibition of angioinvasion. In tumor-invasive areas, VEGF and HIF-1α in the tumor cells, and VEGF receptor-2 in endothelial cells decreased after treatment with dietary glucosylceramides. Dietary glucosylceramides increased serum levels of sphingosine-based ceramides as compared to the control. In SCCKN and UV♀2 cells, C6-ceramide suppressed the expressions of VEGF, VEGF receptor-2, and HIF-1α in vitro. CONCLUSION These results suggest that dietary glucosylceramides trigger the de novo pathway of ceramide synthesis, indicating that sphingosine-based ceramide suppresses the growth of head and neck tumors through the inhibition of pro-angiogenic signals such as VEGF, VEGF receptor-2, and HIF-1α.
Collapse
Affiliation(s)
- Hiroaki Yazama
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, Tottori University, Nishimachi 86, Yonago, 683-8503, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|