1
|
Liu F, Wu Q, Dong Z, Liu K. Integrins in cancer: Emerging mechanisms and therapeutic opportunities. Pharmacol Ther 2023:108458. [PMID: 37245545 DOI: 10.1016/j.pharmthera.2023.108458] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Integrins are vital surface adhesion receptors that mediate the interactions between the extracellular matrix (ECM) and cells and are essential for cell migration and the maintenance of tissue homeostasis. Aberrant integrin activation promotes initial tumor formation, growth, and metastasis. Recently, many lines of evidence have indicated that integrins are highly expressed in numerous cancer types and have documented many functions of integrins in tumorigenesis. Thus, integrins have emerged as attractive targets for the development of cancer therapeutics. In this review, we discuss the underlying molecular mechanisms by which integrins contribute to most of the hallmarks of cancer. We focus on recent progress on integrin regulators, binding proteins, and downstream effectors. We highlight the role of integrins in the regulation of tumor metastasis, immune evasion, metabolic reprogramming, and other hallmarks of cancer. In addition, integrin-targeted immunotherapy and other integrin inhibitors that have been used in preclinical and clinical studies are summarized.
Collapse
Affiliation(s)
- Fangfang Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Qiong Wu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zigang Dong
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Kangdong Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China.
| |
Collapse
|
2
|
Li D, Finley SD. Exploring the Extracellular Regulation of the Tumor Angiogenic Interaction Network Using a Systems Biology Model. Front Physiol 2019; 10:823. [PMID: 31379588 PMCID: PMC6656929 DOI: 10.3389/fphys.2019.00823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/12/2019] [Indexed: 12/31/2022] Open
Abstract
Tumor angiogenesis is regulated by pro- and anti-angiogenic factors. Anti-angiogenic agents target the interconnected network of angiogenic factors to inhibit neovascularization, which subsequently impedes tumor growth. Due to the complexity of this network, optimizing anti-angiogenic cancer treatments requires detailed knowledge at a systems level. In this study, we constructed a tumor tissue-based model to better understand how the angiogenic network is regulated by opposing mediators at the extracellular level. We consider the network comprised of two pro-angiogenic factors: vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (FGF2), and two anti-angiogenic factors: thrombospondin-1 (TSP1) and platelet factor 4 (PF4). The model's prediction of angiogenic factors' distribution in tumor tissue reveals the localization of different factors and indicates the angiogenic state of the tumor. We explored how the distributions are affected by the secretion of the pro- and anti-angiogenic factors, illustrating how the angiogenic network is regulated in the extracellular space. Interestingly, we identified a counterintuitive result that the secretion of the anti-angiogenic factor PF4 can enhance pro-angiogenic signaling by elevating the levels of the interstitial and surface-level pro-angiogenic species. This counterintuitive situation is pertinent to the clinical setting, such as the release of anti-angiogenic factors in platelet activation or the administration of exogenous PF4 for anti-angiogenic therapy. Our study provides mechanistic insights into this counterintuitive result and highlights the role of heparan sulfate proteoglycans in regulating the interactions between angiogenic factors. This work complements previous studies aimed at understanding the formation of angiogenic complexes in tumor tissue and helps in the development of anti-cancer strategies targeting angiogenesis.
Collapse
Affiliation(s)
- Ding Li
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Stacey D Finley
- Department of Biomedical Engineering, Mork Family Department of Chemical Engineering and Materials Science, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
3
|
Validation of Bevacizumab Therapy Effect on Colon Cancer Subtypes by Using Whole Body Imaging in Mice. Mol Imaging Biol 2018; 19:847-856. [PMID: 28315202 DOI: 10.1007/s11307-017-1048-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Preclinical imaging offers a useful tool for monitoring cancer biological behavior and therapy in vivo without the necessity of animal surgery. The following paper describes our examination of tumor progress and anti-angiogenic therapy with Bevacizumab on colon cancer subtypes (SW480 and SW620) by using different non-invasive real-time in vivo imaging techniques. PROCEDURES Color Doppler ultrasound imaging (CDUI) was used to observe the formation of new blood vessels; a homemade fluorescence reflectance imaging (FRI) apparatus was mainly used to test the difference in VEGFR2 expression between the tumor subtypes. Briefly, 15 Balb/c nude mice bearing subcutaneous SW480 and SW620 xenografts were randomly divided into Control and Drug groups. Bevacizumab treatment lasted for 3 weeks. All images were captured pre- and post-treatment. At the end of experiment, all mice were euthanized, and tumor tissue was collected and analyzed by immunohistochemical staining. RESULTS Expression of VEGFR2 was found to be slightly (10 %) but significantly higher for the SW620 cells than for SW480 cells. In addition, SW620 has shown to be more vascularized than SW480 subtype. After 3-week Bevacizumab therapy, no blood vessels were found within 83 % of SW620, while it was 67 % in SW480; the increase of SW620 tumor volume post-treatment was only 3.17-fold compared with the tumor volume pre-treatment, and 4.51-fold higher in SW480. CONCLUSION Our data suggest that SW480 and SW620 cell lines respond differently to Bevacizumab therapy in vivo. Because of higher vascularization, and subsequently higher reduction by drug of new blood vessels and tumor growth rate, xenografts derived from the metastatic SW620 cell line have a better chance of being successfully treated with Bevacizumab compared with those derived from the primary tumor SW480 cell line.
Collapse
|
4
|
Koyanagi T, Suzuki Y, Komori K, Saga Y, Matsubara S, Fujiwara H, Sato Y. Targeting human vasohibin-2 by a neutralizing monoclonal antibody for anti-cancer treatment. Cancer Sci 2017; 108:512-519. [PMID: 28032401 PMCID: PMC5378258 DOI: 10.1111/cas.13149] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/14/2016] [Accepted: 12/18/2016] [Indexed: 01/22/2023] Open
Abstract
There are two members of the vasohibin (VASH) family, VASH1 and VASH2. VASH1 is expressed mainly in endothelial cells to inhibit angiogenesis, whereas VASH2 is expressed mainly in cancer cells to stimulate tumor growth. The aim of the present study was to establish neutralizing monoclonal antibody (mAb) against human VASH2 and apply it as an anti‐cancer treatment. We previously raised mAb against several synthetic peptides of hVASH1, and found that one of them exhibited neutralizing activity against hVASH1. Because of the similarity in the amino acid sequences between VASH1 and VASH2, we hypothesized that they shared the bioactive center. When we mutated four amino acids within the region, the mutant VASH2 lost its pro‐angiogenic activity. Therefore, we raised mAb against a synthetic peptide overlapping the mutated amino acids of hVASH2, and isolated one clone (1760) that almost completely inhibited the stimulatory effect of hVASH2 on the migration of and tube formation by endothelial cells. When we used this clone 1760 antibody for cancer treatment, the peritoneal injection of it inhibited both tumor growth and angiogenesis in a mouse xenograft model of human cancer cells. In terms of anti‐tumor activity, 25 mg/kg of clone 1760 was equivalent to 5 mg/kg of bevacizmab. From these results, we propose the targeting of human VASH2 with neutralizing mAb as a new strategy for cancer treatment.
Collapse
Affiliation(s)
- Takahiro Koyanagi
- Department of Vascular Biology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan.,Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yasuhiro Suzuki
- Department of Vascular Biology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Kazuki Komori
- Department of Vascular Biology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Yasushi Saga
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shigeki Matsubara
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yasufumi Sato
- Department of Vascular Biology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
5
|
Sartori A, Portioli E, Battistini L, Calorini L, Pupi A, Vacondio F, Arosio D, Bianchini F, Zanardi F. Synthesis of Novel c(AmpRGD)-Sunitinib Dual Conjugates as Molecular Tools Targeting the α vβ 3 Integrin/VEGFR2 Couple and Impairing Tumor-Associated Angiogenesis. J Med Chem 2016; 60:248-262. [PMID: 27997164 DOI: 10.1021/acs.jmedchem.6b01266] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
On the basis of a previously discovered anti-αVβ3 integrin peptidomimetic (c(AmpRGD)) and the clinically approved antiangiogenic kinase inhibitor sunitinib, three novel dual conjugates were synthesized (compounds 1-3), featuring the covalent and robust linkage between these two active modules. In all conjugates, the ligand binding competence toward αVβ3 (using both isolated receptors and αVβ3-overexpressing endothelial progenitor EP cells) and the kinase inhibitory activity (toward both isolated kinases and EPCs) remained almost untouched and comparable to the activity of the single active units. Compounds 1-3 showed interesting antiangiogenesis properties in an in vitro tubulogenic assay; furthermore, dimeric-RGD conjugate 3 strongly inhibited in vivo angiogenesis in Matrigel plug assays in FVB mice. These results offer proof-of-concept of how the covalent conjugation of two angiogenesis-related small modules may result in novel and stable molecules, which impair tumor-related angiogenesis with equal or even superior ability as compared to the single modules or their simple combinations.
Collapse
Affiliation(s)
- Andrea Sartori
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Elisabetta Portioli
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Lucia Battistini
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Lido Calorini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Alberto Pupi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy.,Centro Interdipartimentale per lo Sviluppo Preclinico dell'Imaging Molecolare (CISPIM), Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Federica Vacondio
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari, Consiglio Nazionale delle Ricerche , Via Golgi 19, 20133 Milano, Italy
| | - Francesca Bianchini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy.,Centro Interdipartimentale per lo Sviluppo Preclinico dell'Imaging Molecolare (CISPIM), Università degli Studi di Firenze , Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | - Franca Zanardi
- Dipartimento di Farmacia, Università degli Studi di Parma , Parco Area delle Scienze 27A, 43124 Parma, Italy
| |
Collapse
|
6
|
Huang Y, Lichtenberger LM, Taylor M, Bottsford-Miller JN, Haemmerle M, Wagner MJ, Lyons Y, Pradeep S, Hu W, Previs RA, Hansen JM, Fang D, Dorniak PL, Filant J, Dial EJ, Shen F, Hatakeyama H, Sood AK. Antitumor and Antiangiogenic Effects of Aspirin-PC in Ovarian Cancer. Mol Cancer Ther 2016; 15:2894-2904. [PMID: 27638860 PMCID: PMC5136300 DOI: 10.1158/1535-7163.mct-16-0074] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/08/2016] [Accepted: 08/21/2016] [Indexed: 02/07/2023]
Abstract
To determine the efficacy of a novel and safer (for gastrointestinal tract) aspirin (aspirin-PC) in preclinical models of ovarian cancer, in vitro dose-response studies were performed to compare the growth-inhibitory effect of aspirin-PC versus aspirin on three human (A2780, SKOV3ip1, and HeyA8) and a mouse (ID8) ovarian cancer cell line over an 8-day culture period. In the in vivo studies, the aspirin test drugs were studied alone and in the presence of a VEGF-A inhibitor (bevacizumab or B20), due to an emerging role for platelets in tumor growth following antiangiogenic therapy, and we examined their underlying mechanisms. Aspirin-PC was more potent (vs. aspirin) in blocking the growth of both human and mouse ovarian cancer cells in monolayer culture. Using in vivo model systems of ovarian cancer, we found that aspirin-PC significantly reduced ovarian cancer growth by 50% to 90% (depending on the ovarian cell line). The efficacy was further enhanced in combination with Bevacizumab or B20. The growth-inhibitory effect on ovarian tumor mass and number of tumor nodules was evident, but less pronounced for aspirin and the VEGF inhibitors alone. There was no detectable gastrointestinal toxicity. Both aspirin and aspirin-PC also inhibited cell proliferation, angiogenesis, and increased apoptosis of ovarian cancer cells. In conclusion, PC-associated aspirin markedly inhibits the growth of ovarian cancer cells, which exceeds that of the parent drug, in both cell culture and in mouse model systems. We also found that both aspirin-PC and aspirin have robust antineoplastic action in the presence of VEGF-blocking drugs. Mol Cancer Ther; 15(12); 2894-904. ©2016 AACR.
Collapse
Affiliation(s)
- Yan Huang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Lenard M Lichtenberger
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| | - Morgan Taylor
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Justin N Bottsford-Miller
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monika Haemmerle
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Wagner
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yasmin Lyons
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rebecca A Previs
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jean M Hansen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dexing Fang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| | - Piotr L Dorniak
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Justyna Filant
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Dial
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas
| | - Fangrong Shen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hiroto Hatakeyama
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
7
|
Roggiani F, Mezzanzanica D, Rea K, Tomassetti A. Guidance of Signaling Activations by Cadherins and Integrins in Epithelial Ovarian Cancer Cells. Int J Mol Sci 2016; 17:ijms17091387. [PMID: 27563880 PMCID: PMC5037667 DOI: 10.3390/ijms17091387] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/11/2016] [Accepted: 08/13/2016] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the deadliest tumor among gynecological cancer in the industrialized countries. The EOC incidence and mortality have remained unchanged over the last 30 years, despite the progress in diagnosis and treatment. In order to develop novel and more effective therapeutic approaches, the molecular mechanisms involved in EOC progression have been thoroughly investigated in the last few decades. At the late stage, peritoneal metastases originate from the attachment of small clusters of cancer cells that shed from the primary site and carried by the ascites adhere to the abdominal peritoneum or omentum. This behavior suggests that cell–cell or cell–matrix adhesion mechanisms regulate EOC growth and dissemination. Complex downstream signalings, which might be influenced by functional cross-talk between adhesion molecules and co-expressed and activated signaling proteins, can affect the proliferation/survival and the migration/invasion of EOC cells. This review aimed to define the impact of the mechanisms of cell–cell, through cadherins, and cell–extracellular matrix adhesion, through integrins, on the signaling cascades induced by membrane receptors and cytoplasmic proteins known to have a role in the proliferation, migration and invasion of EOC cells. Finally, some novel approaches using peptidomimetic ligands to cadherin and integrins are summarized.
Collapse
Affiliation(s)
- Francesca Roggiani
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| | - Delia Mezzanzanica
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| | - Katia Rea
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| | - Antonella Tomassetti
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy.
| |
Collapse
|
8
|
Ravoori MK, Nishimura M, Singh SP, Lu C, Han L, Hobbs BP, Pradeep S, Choi HJ, Bankson JA, Sood AK, Kundra V. Tumor T1 Relaxation Time for Assessing Response to Bevacizumab Anti-Angiogenic Therapy in a Mouse Ovarian Cancer Model. PLoS One 2015; 10:e0131095. [PMID: 26098849 PMCID: PMC4476738 DOI: 10.1371/journal.pone.0131095] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 05/28/2015] [Indexed: 12/19/2022] Open
Abstract
Purpose To assess whether T1 relaxation time of tumors may be used to assess response to bevacizumab anti-angiogenic therapy. Procedures: 12 female nude mice bearing subcutaneous SKOV3ip1-LC ovarian tumors were administered bevacizumab (6.25ug/g, n=6) or PBS (control, n=6) therapy twice a week for two weeks. T1 maps of tumors were generated before, two days, and 2 weeks after initiating therapy. Tumor weight was assessed by MR and at necropsy. Histology for microvessel density, proliferation, and apoptosis was performed. Results Bevacizumab treatment resulted in tumor growth inhibition (p<0.04, n=6), confirming therapeutic efficacy. Tumor T1 relaxation times increased in bevacizumab treated mice 2 days and 2 weeks after initiating therapy (p<.05, n=6). Microvessel density decreased 59% and cell proliferation (Ki67+) decreased 50% in the bevacizumab treatment group (p<.001, n=6), but not apoptosis. Conclusions Findings suggest that increased tumor T1 relaxation time is associated with response to bevacizumab therapy in ovarian cancer model and might serve as an early indicator of response.
Collapse
Affiliation(s)
- Murali K. Ravoori
- Department of Cancer Systems Imaging, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Masato Nishimura
- Department of Obstetrics and Gynecology, The University of Tokushima Graduate School, Tokushima, Japan
| | - Sheela P. Singh
- Department of Cancer Systems Imaging, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Chunhua Lu
- Department of Gynecologic Oncology, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Lin Han
- Department of Cancer Systems Imaging, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Brian P. Hobbs
- Department of Biostatistics, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Sunila Pradeep
- Department of Gynecologic Oncology, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Hyun J. Choi
- Department of Gynecologic Oncology, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - James A. Bankson
- Department of Imaging Physics, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Anil K. Sood
- Department of Gynecologic Oncology, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Cancer Biology, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Vikas Kundra
- Department of Cancer Systems Imaging, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Radiology, U.T.- M.D. Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
9
|
Monaco A, Michelin O, Prior J, Rüegg C, Scapozza L, Seimbille Y. Synthesis of a non-peptidic PET tracer designed forα5β1integrin receptor. J Labelled Comp Radiopharm 2014; 57:365-70. [DOI: 10.1002/jlcr.3190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 01/16/2014] [Accepted: 01/23/2014] [Indexed: 01/21/2023]
Affiliation(s)
- Alessandra Monaco
- Cyclotron Unit; University Hospital of Geneva; Rue Gabrielle Perret-Gentil 4 1211 Geneva Switzerland
- School of Pharmaceutical Sciences; University of Geneva and University of Lausanne; Quai Ernest Ansermet 30 1211 Geneva Switzerland
| | - Olivier Michelin
- Swiss Institute of Bioinformatics; University of Lausanne; Quartier Sorge 1015 Lausanne Switzerland
| | - John Prior
- CHUV; Department of Nuclear Medicine; Rue du Bugnon 46 1011 Lausanne Switzerland
| | - Curzio Rüegg
- Faculty of Science, Department of Medicine; University of Fribourg; Ch. du Musée 8 1700 Fribourg Switzerland
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences; University of Geneva and University of Lausanne; Quai Ernest Ansermet 30 1211 Geneva Switzerland
| | - Yann Seimbille
- Cyclotron Unit; University Hospital of Geneva; Rue Gabrielle Perret-Gentil 4 1211 Geneva Switzerland
- School of Pharmaceutical Sciences; University of Geneva and University of Lausanne; Quai Ernest Ansermet 30 1211 Geneva Switzerland
| |
Collapse
|
10
|
Hong S, Haibing H, Jialiang H, Xiaojuan Z, Jingjing W, Wenjing W, Caihui N, Hanmei X. PEGylated HM-3 presents anti-rheumatic bioactivity by inhibiting angiogenesis and inflammation. J Mater Chem B 2014; 2:800-813. [DOI: 10.1039/c3tb21100b] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
11
|
Gangadhara S, Barrett-Lee P, Nicholson RI, Hiscox S. Pro-metastatic tumor–stroma interactions in breast cancer. Future Oncol 2012; 8:1427-42. [DOI: 10.2217/fon.12.134] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The vast majority of breast cancer-related deaths are due to metastatic disease. Reciprocal and complex interactions between epithelial tumor cells and the various components of the tumor microenvironment influence tumor progression and metastases although the molecular mechanisms underlying these metastasis-promoting effects are not fully characterized. Identifying and understanding pathways of tumor–stroma cross-talk are likely to lead to the development of novel prognostic biomarkers for metastasis and strategies to prevent metastasis at its earliest stages, resulting in improved patient outcomes.
Collapse
Affiliation(s)
- Sharath Gangadhara
- School of Pharmacy & Pharmaceutical Sciences, Redwood Building, Cardiff University, CF10 3NB, UK
- Velindre Cancer Centre, Velindre Road, Whitchurch, Cardiff, UK
| | | | - Robert I Nicholson
- School of Pharmacy & Pharmaceutical Sciences, Redwood Building, Cardiff University, CF10 3NB, UK
| | - Stephen Hiscox
- School of Pharmacy & Pharmaceutical Sciences, Redwood Building, Cardiff University, CF10 3NB, UK
| |
Collapse
|
12
|
Infusino GA, Jacobson JR. Endothelial FAK as a therapeutic target in disease. Microvasc Res 2011; 83:89-96. [PMID: 22008516 DOI: 10.1016/j.mvr.2011.09.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 09/28/2011] [Accepted: 09/29/2011] [Indexed: 01/14/2023]
Abstract
Focal adhesions (FA) are important mediators of endothelial cytoskeletal interactions with the extracellular matrix (ECM) via transmembrane receptors, integrins and integrin-associated intracellular proteins. This communication is essential for a variety of cell processes including EC barrier regulation and is mediated by the non-receptor protein tyrosine kinase, focal adhesion kinase (FAK). As FA mediate the basic response of EC to a variety of stimuli and FAK is essential to these responses, the idea of targeting EC FAK as a therapeutic strategy for an assortment of diseases is highly promising. In particular, inhibition of FAK could prove beneficial in a variety of cancers via effects on EC proliferation and angiogenesis, in acute lung injury (ALI) via the attenuation of lung vascular permeability, and in rheumatoid arthritis via reductions in synovial angiogenesis. In addition, there are potential therapeutic benefits of FAK inhibition in cardiovascular disease and diabetic nephropathy as well. Several drugs that target EC FAK are now in existence and include agents currently under investigation in preclinical models as well as drugs that are readily available such as the sphingolipid analog FTY720 and statins. As the role of EC FAK in the pathogenesis of a variety of diseases continues to be explored and new insights are revealed, drug targeting of FAK will continue to be an important area of investigation and may ultimately lead to highly novel and effective strategies to treat these diseases.
Collapse
Affiliation(s)
- Giovanni A Infusino
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | |
Collapse
|
13
|
Antagonistic VEGF variants engineered to simultaneously bind to and inhibit VEGFR2 and alphavbeta3 integrin. Proc Natl Acad Sci U S A 2011; 108:14067-72. [PMID: 21825147 DOI: 10.1073/pnas.1016635108] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Significant cross-talk exists between receptors that mediate angiogenesis, such as VEGF receptor-2 (VEGFR2) and α(v)β(3) integrin. Thus, agents that inhibit both receptors would have important therapeutic potential. Here, we used an antagonistic VEGF ligand as a molecular scaffold to engineer dual-specific proteins that bound to VEGFR2 and α(v)β(3) integrin with antibody-like affinities and inhibited angiogenic processes in vitro and in vivo. Mutations were introduced into a single-chain VEGF (scVEGF) ligand that retained VEGFR2 binding, but prevented receptor dimerization and activation. Yeast-displayed scVEGF mutant libraries were created and screened by high-throughput flow cytometric sorting to identify several variants that bound with high affinity to both VEGFR2 and α(v)β(3) integrin. These engineered scVEGF mutants were specific for α(v)β(3) integrin and did not bind to the related integrins α(v)β(5), α(iib)β(3), or α(5)β(1). In addition, surface plasmon resonance and cell binding assays showed that dual-specific scVEGF proteins can simultaneously engage both receptors. Compared to monospecific scVEGF mutants that bind VEGFR2 or α(v)β(3) integrin, dual-specific scVEGF proteins more strongly inhibited VEGF-mediated receptor phosphorylation, capillary tube formation, and proliferation of endothelial cells cultured on Matrigel or vitronectin-coated surfaces. Moreover, dual specificity conferred strong inhibition of VEGF-mediated blood vessel formation in Matrigel plugs in vivo, whereas monospecific scVEGF mutants that bind VEGFR2 or α(v)β(3) integrin were only marginally effective. Instead of relying on antibody associating domains or physical linkage, this work highlights an approach to creating dual-specific proteins where additional functionality is introduced into a protein ligand to complement its existing biological properties.
Collapse
|
14
|
Abstract
The use of monoclonal antibodies (mAbs) has become a general approach for specifically targeting and treating human disease. In oncology, the therapeutic utility of mAbs is usually evaluated in the context of treatment with standard of care, as well as other small molecule targeted therapies. Many anti-cancer antibody modalities have achieved validation, including the targeting of growth factor and angiogenesis pathways, the induction of tumor cell killing or apoptosis, and the blocking of immune inhibitory mechanisms to stimulate anti-tumor responses. But, as with other targeted therapies, few antibodies are curative because of biological complexities that underlie tumor formation and redundancies in molecular pathways that enable tumors to adapt and show resistance to treatment. This review discusses the combinations of antibody therapeutics that are emerging to improve efficacy and durability within a specific biological mechanism (e.g., immunomodulation or the inhibition of angiogenesis) and across multiple biological pathways (e.g., inhibition of tumor growth and induction of tumor cell apoptosis).
Collapse
|
15
|
Abdollahi A, Folkman J. Evading tumor evasion: current concepts and perspectives of anti-angiogenic cancer therapy. Drug Resist Updat 2010; 13:16-28. [PMID: 20061178 DOI: 10.1016/j.drup.2009.12.001] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 12/20/2009] [Accepted: 12/22/2009] [Indexed: 12/20/2022]
Abstract
Within three decades, anti-angiogenic therapy has rapidly evolved into an integral component of current standard anti-cancer treatment. Anti-angiogenic therapy has fulfilled a number of its earlier proposed promises. The universality of this approach is demonstrated by the broad spectrum of malignant and benign tumor entities, as well as non-neoplastic diseases, that are currently treated with anti-angiogenic agents. In contrast to tumor cell targeting therapies, the development of acquired drug resistance (e.g., via mutations in growth factor receptor signaling genes) has not been described yet for the principal target of anti-angiogenic therapy--the tumor endothelium. Moreover, the tumor endothelium has emerged as a critical target of conventional cancer therapies, such as chemotherapy and radiotherapy. The presumption that tumor growth and metastasis are angiogenesis-dependent implies that the number of potential targets of an anti-cancer therapy could be reduced to those that stimulate the angiogenesis process. Therefore, the set of endogenous angiogenesis stimulants might constitute an "Achilles heel" of cancer. Direct targeting of tumor endothelium via, e.g., endogenous angiogenesis inhibitors poses another promising but clinically less explored therapeutic strategy. Indeed, the majority of current anti-angiogenic approaches block the activity of a single or at most a few pro-angiogenic proteins secreted by tumor cells or the tumor stroma. Based on our systems biology work on the angiogenic switch, we predicted that the redundancy of angiogenic signals might limit the efficacy of anti-angiogenic monotherapies. In support of this hypothesis, emerging experimental evidence suggests that tumors may become refractory or even evade the inhibition of a single pro-angiogenic pathway via compensatory upregulation of alternative angiogenic factors. Here, we discuss current concepts and propose novel strategies to overcome tumor evasion of anti-angiogenic therapy. We believe that early detection of tumors, prediction of tumor evasive mechanisms and rational design of anti-angiogenic combinations will direct anti-angiogenic therapy towards its ultimate goal--the conversion of cancer to a dormant, chronic, manageable disease.
Collapse
Affiliation(s)
- Amir Abdollahi
- Center of Cancer Systems Biology, Dept. of Medicine, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135, USA.
| | | |
Collapse
|