51
|
Integrin inhibitors as a therapeutic agent for ovarian cancer. JOURNAL OF ONCOLOGY 2011; 2012:915140. [PMID: 22235205 PMCID: PMC3253465 DOI: 10.1155/2012/915140] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 11/11/2011] [Accepted: 11/17/2011] [Indexed: 12/21/2022]
Abstract
Ovarian cancer is a deadly disease, with a cure rate of only 30%. Despite aggressive treatments, relapse remains almost inevitable in patients with advanced-stage disease. In recent years, great progress has been made towards targeting integrins in cancer treatment, and clinical studies with various integrin inhibitors have demonstrated their effectiveness in blocking cancer progression. Given that the initial critical step of ovarian cancer metastasis is the attachment of cancer cells onto the peritoneum or omentum, in addition to the proven positive clinical results of anti-angiogenic therapy, targeting integrins is likely to be one of the most feasible approaches. This paper summarizes the current understanding of the integrin biology in ovarian cancer metastasis and the various therapeutic approaches attempted with integrin inhibitors. Although no integrin inhibitors have shown favorable results so far, integrin-targeted therapies continue to be a promising approach to be explored for further clinical investigation.
Collapse
|
52
|
Bellone M, Cocco E, Varughese J, Bellone S, Todeschini P, El-Sahwi K, Carrara L, Guzzo F, Schwartz PE, Rutherford TJ, Pecorelli S, Marshall DJ, Santin AD. Expression of αV-integrins in uterine serous papillary carcinomas; implications for targeted therapy with intetumumab (CNTO 95), a fully human antagonist anti-αV-integrin antibody. Int J Gynecol Cancer 2011; 21:1084-90. [PMID: 21633302 PMCID: PMC3690508 DOI: 10.1097/igc.0b013e3182187324] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Uterine serous papillary carcinoma (USPC) is an aggressive variant of endometrial cancer characterized by an innate resistance to chemotherapy and poor prognosis. In this study, we evaluated the expression of αV-integrins in primary USPC cell lines and the in vitro ability of intetumumab (CNTO 95), a fully human monoclonal antibody against αV-integrins, to inhibit USPC cell adhesion and migration. MATERIALS AND METHODS The surface expression of integrins belonging to the αV-family, including αVβ3, αVβ5, and αVβ6, was evaluated in 6 primary USPC cell lines using flow cytometry analysis. To test the ability of intetumumab to inhibit USPC cell adhesion and migration, adhesion assays in the presence of vitronectin and migration assays through an 8.0-μm pore polycarbonate membrane also were performed. RESULTS We found high expression of the αV-subunit on the cell surface of all 6 primary USPC cell lines tested (100% positive cells; mean fluorescence intensity range, 13.1-39.5). When the expression of single heterodimeric integrins was evaluated, αVβ3, αVβ5, and αVβ6 were expressed on 37.5%, 32.0%, and 16.3% of cells (mean fluorescence intensity range, 6.5-16.2, 9.2-32.5, and 6.2-11.5, respectively). Importantly, in functional assays, low doses of intetumumab were effective in inhibiting adhesion (0.15 μg/mL, P = 0.003) and migration (1.25 μg/mL P = 0.02) of primary USPC cell lines. CONCLUSIONS The αV-integrins are overexpressed on the cell surface of primary USPC cell lines. Intetumumab may significantly inhibit USPC cell adhesion and migration pathways and may therefore represent a novel treatment option for patients harboring this rare but highly aggressive variant of endometrial cancer.
Collapse
MESH Headings
- Aged
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal, Humanized
- Cell Line, Tumor/metabolism
- Cystadenocarcinoma, Papillary/drug therapy
- Cystadenocarcinoma, Papillary/metabolism
- Cystadenocarcinoma, Papillary/pathology
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Female
- Flow Cytometry
- Humans
- Integrin alphaV/metabolism
- Middle Aged
- Molecular Targeted Therapy
- Uterine Neoplasms/drug therapy
- Uterine Neoplasms/metabolism
- Uterine Neoplasms/pathology
Collapse
Affiliation(s)
- Marta Bellone
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Emiliano Cocco
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Joyce Varughese
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Stefania Bellone
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Paola Todeschini
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Karim El-Sahwi
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Luisa Carrara
- Division of Gynecologic Oncology, University of Brescia, Brescia, Italy
| | - Federica Guzzo
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Peter E. Schwartz
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Thomas J. Rutherford
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Sergio Pecorelli
- Division of Gynecologic Oncology, University of Brescia, Brescia, Italy
| | | | - Alessandro D. Santin
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
53
|
Veiseh O, Kievit F, Ellenbogen RG, Zhang M. Cancer cell invasion: treatment and monitoring opportunities in nanomedicine. Adv Drug Deliv Rev 2011; 63:582-96. [PMID: 21295093 PMCID: PMC3132387 DOI: 10.1016/j.addr.2011.01.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 01/20/2011] [Accepted: 01/25/2011] [Indexed: 12/19/2022]
Abstract
Cell invasion is an intrinsic cellular pathway whereby cells respond to extracellular stimuli to migrate through and modulate the structure of their extracellular matrix (ECM) in order to develop, repair, and protect the body's tissues. In cancer cells this process can become aberrantly regulated and lead to cancer metastasis. This cellular pathway contributes to the vast majority of cancer related fatalities, and therefore has been identified as a critical therapeutic target. Researchers have identified numerous potential molecular therapeutic targets of cancer cell invasion, yet delivery of therapies remains a major hurdle. Nanomedicine is a rapidly emerging technology which may offer a potential solution for tackling cancer metastasis by improving the specificity and potency of therapeutics delivered to invasive cancer cells. In this review we examine the biology of cancer cell invasion, its role in cancer progression and metastasis, molecular targets of cell invasion, and therapeutic inhibitors of cell invasion. We then discuss how the field of nanomedicine can be applied to monitor and treat cancer cell invasion. We aim to provide a perspective on how the advances in cancer biology and the field of nanomedicine can be combined to offer new solutions for treating cancer metastasis.
Collapse
Affiliation(s)
- Omid Veiseh
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Forrest Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Richard G. Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
54
|
Bäuerle T, Komljenovic D, Merz M, Berger MR, Goodman SL, Semmler W. Cilengitide inhibits progression of experimental breast cancer bone metastases as imaged noninvasively using VCT, MRI and DCE-MRI in a longitudinal in vivo study. Int J Cancer 2011; 128:2453-62. [PMID: 20648558 DOI: 10.1002/ijc.25563] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The aim of this study was to investigate the effect of inhibiting αvβ(3)/α(v) β(5) integrins by cilengitide in experimentally induced breast cancer bone metastases using noninvasive imaging techniques. For this purpose, nude rats bearing established breast cancer bone metastases were treated with cilengitide, a small molecule inhibitor of αvβ(3) and αvβ(5) integrins (75 mg/kg, five days per week; n = 12 rats) and compared to vehicle-treated control rats (n = 12). In a longitudinal study, conventional magnetic resonance imaging (MRI) and flat panel volumetric computed tomography were used to assess the volume of the soft tissue tumor and osteolysis, respectively, and dynamic contrast-enhanced (DCE-) MRI was performed to determine functional parameters of the tumor vasculature reflecting blood volume and blood vessel permeability. In rats treated with cilengitide, VCT and MRI showed that osteolytic lesions and the respective bone metastatic soft tissue tumors progressed more slowly than in vehicle-treated controls. DCE-MRI indicated a decrease in blood volume and an increase in vessel permeability and immunohistology revealed increased numbers of immature vessels in cilengitide-treated rats compared to vehicle controls. In conclusion, treatment of experimental breast cancer bone metastases with cilengitide resulted in pronounced antiresorptive and antitumor effects, suggesting that αvβ(3)/αvβ(5) inhibition may be a promising therapeutic approach for bone metastases.
Collapse
Affiliation(s)
- Tobias Bäuerle
- Department of Medical Physics in Radiology, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
55
|
Signaling mechanism of cell adhesion molecules in breast cancer metastasis: potential therapeutic targets. Breast Cancer Res Treat 2011; 128:7-21. [PMID: 21499686 DOI: 10.1007/s10549-011-1499-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 03/31/2011] [Indexed: 01/13/2023]
Abstract
Metastasis is responsible for the majority of breast cancer-related deaths. The metastatic spread of cancer cells is a complicated process that requires considerable flexibility in the adhesive properties of both tumor cells and other interacting cells. Cell adhesion molecules (CAMs) are membrane receptors that mediate cell-cell and cell-matrix interactions, and are essential for transducing intracellular signals responsible for adhesion, migration, invasion, angiogensis, and organ-specific metastasis. This review will discuss the recent advances in our understanding on the biological functions, signaling mechanisms, and therapeutic potentials of important CAMs involved in breast cancer metastasis.
Collapse
|
56
|
Abstract
INTRODUCTION Integrins, which are heterodimeric membrane glycoproteins, consist of a family of cell-surface receptors mediating cell-matrix and cell-cell adhesion. Analysis of tumor-associated integrins has revealed an important relationship between integrins and tumor development, bringing new insights into integrin-based cancer therapies. Hepatocellular carcinoma (HCC) is one of the most malignant tumors worldwide and integrins appeal to be a novel group of potential therapeutic targets for HCC. AREAS COVERED This review summarizes the current knowledge of integrins involved in HCC and the potential of integrin-targeted drugs in HCC therapy. A brief introduction on the structure, biological function and regulatory mechanism of integrins is given. The distinct expression patterns and biological functions of HCC-associated integrins are described. Finally, the current situation of integrin-based therapies in HCC and other tumor types are extensively discussed in the light of their implications in preclinical and clinical trials. EXPERT OPINION To date, increasing numbers of integrin-targeted drugs are undergoing development and they exhibit diverse effects in cancer clinical trials. Tumor heterogeneity should be emphasized in developing effective integrin-targeted drugs specific for HCC. A better understanding of how integrins cooperatively function in HCC will assist in designing more successful integrin-targeted therapeutic drugs and corresponding approaches.
Collapse
Affiliation(s)
- Yanhua Wu
- Fudan University, Institute of Genetics, State Key Laboratory of Genetic Engineering, 220 Handan Road, Shanghai, 200433, P. R. China
| | | | | | | |
Collapse
|
57
|
Clinical and pharmacologic evaluation of two dose levels of intetumumab (CNTO 95) in patients with melanoma or angiosarcoma. Invest New Drugs 2011; 30:1074-81. [PMID: 21331745 DOI: 10.1007/s10637-011-9639-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 01/25/2011] [Indexed: 10/18/2022]
Abstract
PURPOSE In this Phase 1, multicenter, open-label study, intetumumab (CNTO 95), a fully human anti-αv integrin monoclonal antibody was evaluated for safety, pharmacokinetics, and pharmacodynamic activity in patients with melanoma or angiosarcoma. PATIENTS AND METHODS Patients with histologically-confirmed inoperable melanoma or angiosarcoma refractory to standard treatment were allocated to treatment with 10 mg/kg or 20 mg/kg intetumumab, administered once every 3 weeks for up to four cycles unless unacceptable toxicity or disease progression occurred. Extended dosing was available for patients who responded with stable disease or better. RESULTS Eight patients received 10 mg/kg and 11 received 20 mg/kg intetumumab. Baseline patient characteristics were comparable between treatment groups; 18 patients had metastatic malignant melanoma and one had angiosarcoma. No dose-limiting toxicities were observed. Headache was the most common adverse event across both dose groups. Vomiting, nausea and chills were more common, and uveitic reactions lasted longer, in patients treated with 20 mg/kg compared with 10 mg/kg intetumumab. No patient developed antibodies to intetumumab. Intetumumab drug exposure as assessed by area under the curve and maximum serum concentration appeared to increase approximately dose-proportionally from 10 to 20 mg/kg, while volume of distribution remained constant for both doses. Stable disease was observed in two patients with metastatic malignant melanoma (one in each dose group) for at least 6 weeks. CONCLUSIONS In patients with metastatic malignant melanoma and angiosarcoma in this study, intetumumab demonstrated manageable toxicity, was well tolerated, and presented approximately dose-proportional pharmacokinetics for the 10 mg/kg and 20 mg/kg doses.
Collapse
|
58
|
Zhang Y, Yang M, Ji Q, Fan D, Peng H, Yang C, Xiong D, Zhou Y. Anoikis induction and metastasis suppression by a new integrin αvβ3 inhibitor in human melanoma cell line M21. Invest New Drugs 2010; 29:666-73. [PMID: 21170668 DOI: 10.1007/s10637-010-9616-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 12/03/2010] [Indexed: 11/29/2022]
Abstract
Integrin αvβ3 plays a critical role in the survival and metastasis process of cancer cells. It is therefore desirable to develop new types of small molecule inhibitors of integrin αvβ3. IH1062 (3, 5-dichloro-phenylbiguanide) is a novel small molecule inhibitor of integrin αvβ3 that we have recently discovered. In this study, we investigated the induction effects of anoikis in human melanoma cell line M21 by IH1062, by detecting caspase activity, measuring the expression levels of apoptosis-related proteins, and performing the AnnexinV/PI apoptosis assay. Furthermore, we established a melanoma pulmonary metastasis mouse model in order to evaluate the suppression of metastasis by IH1062 in vivo. Our results demonstrate that IH1062 triggered human melanoma M21 cells to undergo anoikis by interrupting the attachment of M21 cells to extracellular matrix, reducing the phosphorylation of focal adhesion kinase, decreasing survivin and the ratio of Bcl-2/Bax proteins, and activating caspase cascades in vitro. Additionally, IH1062 showed markedly anti-metastatic effects in the pulmonary metastasis model in vivo, which makes it a promising lead to develop new drugs for anti-metastasis therapies.
Collapse
Affiliation(s)
- Yongci Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
59
|
Muldoon LL, Gahramanov S, Li X, Marshall DJ, Kraemer DF, Neuwelt EA. Dynamic magnetic resonance imaging assessment of vascular targeting agent effects in rat intracerebral tumor models. Neuro Oncol 2010; 13:51-60. [PMID: 21123368 DOI: 10.1093/neuonc/noq150] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We used dynamic MRI to evaluate the effects of monoclonal antibodies targeting brain tumor vasculature. Female athymic rats with intracerebral human tumor xenografts were untreated or treated with intetumumab, targeting α(V)-integrins, or bevacizumab, targeting vascular endothelial growth factor (n = 4-6 per group). Prior to treatment and at 1, 3, and 7 days after treatment, we performed standard MRI to assess tumor volume, dynamic susceptibility-contrast MRI with the blood-pool iron oxide nanoparticle ferumoxytol to evaluate relative cerebral blood volume (rCBV), and dynamic contrast-enhanced MRI to assess tumor vascular permeability. Tumor rCBV increased by 27 ± 13% over 7 days in untreated rats; intetumumab increased tumor rCBV by 65 ± 10%, whereas bevacizumab reduced tumor rCBV by 31 ± 10% at 7 days (P < .001 for group and day). Similarly, intetumumab increased brain tumor vascular permeability compared with controls at 3 and 7 days after treatment, whereas bevacizumab decreased tumor permeability within 24 hours (P = .0004 for group, P = .0081 for day). All tumors grew over the 7-day assessment period, but bevacizumab slowed the increase in tumor volume on MRI. We conclude that the vascular targeting agents intetumumab and bevacizumab had diametrically opposite effects on dynamic MRI of tumor vasculature in rat brain tumor models. Targeting α(V)-integrins increased tumor vascular permeability and blood volume, whereas bevacizumab decreased both measures. These findings have implications for chemotherapy delivery and antitumor efficacy.
Collapse
Affiliation(s)
- Leslie L Muldoon
- Department of Neurology, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, L603, Portland, OR 97239-3098, USA
| | | | | | | | | | | |
Collapse
|
60
|
Abstract
Angiogenesis is an important component of cancer growth, invasion and metastasis. Therefore, inhibition of angiogenesis is an attractive strategy for treatment of cancer. We describe existing clinical trials of antiangiogenic agents and the challenges facing the clinical development and optimal use of these agents for the treatment of breast cancer. Currently, the most promising approach has been the use of bevacizumab, a humanized monoclonal antibody directed against the most potent pro-angiogenic factor, vascular endothelial growth factor (VEGF). Small molecular inhibitors of VEGF tyrosine kinase activity, such as sorafenib, appear promising. While, the role of sunitinib and inhibitors of mammalian target of rapamycin (mTOR) in breast cancer has to be defined. Several unanswered questions remain, such as choice of drug(s), optimal duration of therapy and patient selection criteria.
Collapse
Affiliation(s)
- Dorte Lisbet Nielsen
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Michael Andersson
- Department of Oncology, Finsen Centre, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Jon Lykkegaard Andersen
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Claus Kamby
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| |
Collapse
|
61
|
Ning S, Tian J, Marshall DJ, Knox SJ. Anti-alphav integrin monoclonal antibody intetumumab enhances the efficacy of radiation therapy and reduces metastasis of human cancer xenografts in nude rats. Cancer Res 2010; 70:7591-9. [PMID: 20841470 DOI: 10.1158/0008-5472.can-10-1639] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We previously reported that intetumumab (CNTO 95), a fully human anti-αv integrin monoclonal antibody, is a radiosensitizer in mice with xenograft tumors. Because intetumumab does not cross-react with mouse integrins, but has cross-reactivity with rat integrins, we next studied the potential combined use of radiation therapy and intetumumab in human cancer xenograft models in nude rats to assess effects on both tumor cells and the tumor microenvironment. Nude rats bearing human head and neck cancer and non-small cell lung cancer (NSCLC) xenografts were treated with intetumumab and fractionated local tumor radiotherapy. Effects on tumor growth and metastasis, blood perfusion, oxygenation, and gastrointestinal toxicity were studied. Intetumumab alone had a moderate effect on tumor growth. When combined with fractionated radiation therapy, intetumumab significantly inhibited tumor growth and produced a tumor response rate that was significantly better than with radiation therapy alone. Treatment with intetumumab also significantly reduced lung metastasis in the A549 NSCLC xenograft model. The oxygenation and blood perfusion in xenograft tumors measured by microbubble-enhanced ultrasound imaging were substantially increased after treatment with intetumumab. The combined use of intetumumab and radiation therapy reduced the microvessel density and increased apoptosis in tumor cells and the tumor microenvironment. Toxicity studies showed that treatment with intetumumab did not cause the histopathologic changes in the lungs and did not sensitize the sensitive gastrointestinal epithelium to the effect of radiation therapy. Intetumumab can potentiate the efficacy of fractionated radiation therapy in human cancer xenograft tumors in nude rats without increased toxicity.
Collapse
Affiliation(s)
- Shoucheng Ning
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, California 94305-5152, USA
| | | | | | | |
Collapse
|
62
|
Tabrizi M, Funelas C, Suria H. Application of quantitative pharmacology in development of therapeutic monoclonal antibodies. AAPS JOURNAL 2010; 12:592-601. [PMID: 20652780 DOI: 10.1208/s12248-010-9220-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 06/25/2010] [Indexed: 11/30/2022]
Abstract
The advancement of therapeutic monoclonal antibodies during various stages of the drug development process can be effectively streamlined when appropriate translational strategies are applied. Design of successful translational strategies for development of monoclonal antibodies should allow for understanding of the dose- and concentration-response relationships with respect to both beneficial and toxic effects from early phases of drug development. Evaluation of relevant biomarkers during early stages of drug development should facilitate the successful design of safe and effective dosing strategies. Moreover, application of quantitative pharmacology is critical for translation of exposure-response relationships early on.
Collapse
|
63
|
Pharmacology and placental transfer of a human alphav integrin monoclonal antibody in rabbits. ACTA ACUST UNITED AC 2010; 89:116-23. [PMID: 20151458 DOI: 10.1002/bdrb.20232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Intetumumab is a human IgG1 anti-alphav-integrin monoclonal antibody that inhibits angiogenesis. Integrin binding and angiogenesis are important in reproduction including fertilization, implantation, and embryofetal development. These studies were designed to determine the pharmacological relevance of the rabbit for the evaluation of potential effects on embryofetal development and to evaluate the placental transfer of intetumumab in rabbits. METHODS In vitro pharmacology studies evaluated the binding of intetumumab to rabbit cells and the inhibition of vessel sprouting from rabbit aorta. For the evaluation of placental transfer, pregnant rabbits (8/group) were injected intravenously with intetumumab 50 or 100 mg/kg every 2 days from Gestation Day (GD)7 to GD19. Maternal sera, fetal homogenates/sera, and amniotic fluid were collected at necropsy on GD19 or GD28 for evaluation of intetumumab concentrations. Clinical condition of the dams was monitored and fetuses were screened for abnormalities. RESULTS Intetumumab (5-40 microg/mL) inhibited aortic cell adhesion to vitronectin and vessel sprouting from rabbit aortic rings. Immunohistochemical staining of rabbit tissues demonstrated binding of intetumumab to placenta. Administration of intetumumab to pregnant rabbits was well tolerated by the dams and the fetuses did not show major abnormalities. Fetal exposure to intetumumab relative to maternal exposure was <0.1% on GD19 and 100-130% on GD29. CONCLUSIONS The rabbit is a pharmacologically relevant species for evaluation of potential developmental effects of intetumumab. Intetumumab crosses the rabbit placenta during the fetal period (GD 19-28).
Collapse
|
64
|
Abstract
IMPORTANCE OF THE FIELD Recently, there has been substantial progress in the development of integrin targeted pharmaceuticals and drug delivery systems. Integrin is an important member in the cell adhesion molecule family, which is involved in regulation of complex biological conditions, from keeping normal physiological activities to causing cellular dysfunction in diseased cells. Hence, it is timely to summarize the recent developments in integrin targeted drug and gene delivery systems to understand better their advantages and limitations. AREAS COVERED IN THIS REVIEW In this review, advances in the discovery and clinical trials of these integrin antagonists against different integrin subunits are summarized and discussed. Besides using integrin inhibitor as a single therapeutic agent, integrin antagonists that were conjugated to cytotoxic drugs by synthetic chemistry or coupled to biomacromolecules by either DNA recombination technology or fusion protein technology for integrin targeted therapy have been explored. Furthermore, nanoparticles with integrin targeting ligands for both drug and gene delivery, typically for antiangiogenesis and anticancer therapy, are highlighted and evaluated. WHAT THE READER WILL GAIN This review sheds light on the future development of integrin targeted drug and/or gene delivery systems. TAKE HOME MESSAGE Although thus far there are still limitations, integrin targeted delivery systems have already shown their potential as important pharmaceuticals in the near future.
Collapse
Affiliation(s)
- Zhe Wang
- National University of Singapore, Department of Pharmacy, 18 Science Drive 4, Singapore 117543, Singapore
| | | | | |
Collapse
|
65
|
Abstract
The integrin family of cell adhesion receptors regulates a diverse array of cellular functions crucial to the initiation, progression and metastasis of solid tumours. The importance of integrins in several cell types that affect tumour progression has made them an appealing target for cancer therapy. Integrin antagonists, including the alphavbeta3 and alphavbeta5 inhibitor cilengitide, have shown encouraging activity in Phase II clinical trials and cilengitide is currently being tested in a Phase III trial in patients with glioblastoma. These exciting clinical developments emphasize the need to identify how integrin antagonists influence the tumour and its microenvironment.
Collapse
Affiliation(s)
- Jay S Desgrosellier
- Department of Pathology, Moores University of California at San Diego Cancer Center, La Jolla, 92093-0803, United States
| | | |
Collapse
|
66
|
Lowin T, Straub RH, Neumann E, Bosserhoff A, Vogel C, Moissl C, Anders S, Müller-Ladner U, Schedel JÃ. Glucocorticoids increase α5 integrin expression and adhesion of synovial fibroblasts but inhibit ERK signaling, migration, and cartilage invasion. ACTA ACUST UNITED AC 2009; 60:3623-32. [DOI: 10.1002/art.24985] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
67
|
Makrilia N, Kollias A, Manolopoulos L, Syrigos K. Cell adhesion molecules: role and clinical significance in cancer. Cancer Invest 2009; 27:1023-37. [PMID: 19909018 DOI: 10.3109/07357900902769749] [Citation(s) in RCA: 242] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
There is a growing body of evidence suggesting that alterations in the adhesion properties of neoplastic cells endow them with an invasive and migratory phenotype. Indeed, changes in the expression or function of cell adhesion molecules have been implicated in all steps of tumor progression, including detachment of tumor cells from the primary site, intravasation into the blood stream, extravasation into distant target organs, and formation of the secondary lesions. This review presents recent data regarding the role of cell adhesion molecules in tumor development and progress with concern to their clinical exploitation as potential biomarkers in neoplastic diseases.
Collapse
Affiliation(s)
- Nektaria Makrilia
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, Athens School of Medicine, Greece
| | | | | | | |
Collapse
|
68
|
Ren DM, Li M, Dong YY, Cheng Y, Yang XR, Zou YD, Zheng HY, Bai H, Chu XG, Wang JB. Realtime Cytotoxicity Characterization with Cell-Microelectronic Sensing of Water Soluble Carbon Nanotubes. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/15533170903328461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Dong-Mei Ren
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Ming Li
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Yi-Yang Dong
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Yan Cheng
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Xiao-Ran Yang
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Yun-Dong Zou
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Hong-Yan Zheng
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Hua Bai
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Xiao-Gang Chu
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| | - Jun-Bing Wang
- a Chinese Academy of Inspection and Quarantine Science No.3A , North Gaobeidian Road, Chaoyang District, Beijing, P. R. China
| |
Collapse
|
69
|
NIP1/DUOXA1 expression in epithelial breast cancer cells: regulation of cell adhesion and actin dynamics. Breast Cancer Res Treat 2009; 119:773-86. [DOI: 10.1007/s10549-009-0372-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Accepted: 03/06/2009] [Indexed: 12/11/2022]
|
70
|
Alshamsan A, Haddadi A, Incani V, Samuel J, Lavasanifar A, Uludağ H. Formulation and Delivery of siRNA by Oleic Acid and Stearic Acid Modified Polyethylenimine. Mol Pharm 2008; 6:121-33. [DOI: 10.1021/mp8000815] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Aws Alshamsan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton T6G 2N8, Canada, Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G 2V2, Canada, and Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton T6G 2G6, Canada
| | - Azita Haddadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton T6G 2N8, Canada, Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G 2V2, Canada, and Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton T6G 2G6, Canada
| | - Vanessa Incani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton T6G 2N8, Canada, Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G 2V2, Canada, and Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton T6G 2G6, Canada
| | - John Samuel
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton T6G 2N8, Canada, Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G 2V2, Canada, and Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton T6G 2G6, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton T6G 2N8, Canada, Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G 2V2, Canada, and Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton T6G 2G6, Canada
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton T6G 2N8, Canada, Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G 2V2, Canada, and Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton T6G 2G6, Canada
| |
Collapse
|
71
|
Abstract
Integrins, which are transmembrane receptors for extracellular matrix proteins, play a key role in cell survival, proliferation, migration, gene expression, and activation of growth factor receptors. Their functions and expression are deregulated in several types of cancer, including prostate cancer. In this article, we review the role of integrins in prostate cancer progression and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology and Cancer Center, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | |
Collapse
|
72
|
Abstract
Anti-angiogenesis is a promising strategy for the treatment of cancer. Integrins, consisting of two noncovalently bound transmembrane alpha and beta subunits, are an important molecular family involved in tumor angiogenesis. The blockade of integrin signaling has been demonstrated to be efficient to inhibit tumor growth, angiogenesis, and metastasis. Among all the integrins, alpha(v)beta(3) seems to be the most important one during tumor angiogenesis. The inhibition of integrin alpha(v)beta(3) signaling with antibodies, peptides, peptidomimetics, and other antagonists has great potential in the treatment of cancer. In addition, integrin alpha(v)beta(3) is highly expressed on activated endothelial cells, new-born vessels as well as some tumor cells, but is not present in resting endothelial cells and most normal organ systems, making it a suitable target for anti-angiogenic therapy. In this article we will review the role of integrin alpha(v)beta(3) in angiogenesis, present recent progress in the use of integrin alpha(v)beta(3) antagonists and integrin-targeted delivery systems as potential cancer therapeutics, and discuss future perspectives.
Collapse
Affiliation(s)
- Zhaofei Liu
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Biophysics, and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305
- Medical Isotopes Research Center (MIRC), Peking University, Beijing, 100083, China
| | - Fan Wang
- Medical Isotopes Research Center (MIRC), Peking University, Beijing, 100083, China
| | - Xiaoyuan Chen
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Biophysics, and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|