1
|
Geng X, Tang Y, Gu C, Zeng J, Zhao Y, Zhou Q, Jia L, Zhou S, Chen X. Integrin αVβ3 antagonist-c(RGDyk) peptide attenuates the progression of ossification of the posterior longitudinal ligament by inhibiting osteogenesis and angiogenesis. Mol Med 2024; 30:57. [PMID: 38698308 PMCID: PMC11067224 DOI: 10.1186/s10020-024-00822-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 04/22/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Ossification of the posterior longitudinal ligament (OPLL), an emerging heterotopic ossification disease, causes spinal cord compression, resulting in motor and sensory dysfunction. The etiology of OPLL remains unclear but may involve integrin αVβ3 regulating the process of osteogenesis and angiogenesis. In this study, we focused on the role of integrin αVβ3 in OPLL and explored the underlying mechanism by which the c(RGDyk) peptide acts as a potent and selective integrin αVβ3 inhibitor to inhibit osteogenesis and angiogenesis in OPLL. METHODS OPLL or control ligament samples were collected in surgery. For OPLL samples, RNA-sequencing results revealed activation of the integrin family, particularly integrin αVβ3. Integrin αVβ3 expression was detected by qPCR, Western blotting, and immunohistochemical analysis. Fluorescence microscopy was used to observe the targeted inhibition of integrin αVβ3 by the c(RGDyk) peptide on ligaments fibroblasts (LFs) derived from patients with OPLL and endothelial cells (ECs). The effect of c(RGDyk) peptide on the ossification of pathogenic LFs was detected using qPCR, Western blotting. Alkaline phosphatase staining or alizarin red staining were used to test the osteogenic capability. The effect of the c(RGDyk) peptide on angiogenesis was determined by EC migration and tube formation assays. The effects of the c(RGDyk) peptide on heterotopic bone formation were evaluated by micro-CT, histological, immunohistochemical, and immunofluorescence analysis in vivo. RESULTS The results indicated that after being treated with c(RGDyk), the osteogenic differentiation of LFs was significantly decreased. Moreover, the c(RGDyk) peptide inhibited the migration of ECs and thus prevented the nutritional support required for osteogenesis. Furthermore, the c(RGDyk) peptide inhibited ectopic bone formation in mice. Mechanistic analysis revealed that c(RGDyk) peptide could inhibit osteogenesis and angiogenesis in OPLL by targeting integrin αVβ3 and regulating the FAK/ERK pathway. CONCLUSIONS Therefore, the integrin αVβ3 appears to be an emerging therapeutic target for OPLL, and the c(RGDyk) peptide has dual inhibitory effects that may be valuable for the new therapeutic strategy of OPLL.
Collapse
Affiliation(s)
- Xiangwu Geng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Yifan Tang
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Changjiang Gu
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Junkai Zeng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Yin Zhao
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Quanwei Zhou
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Lianshun Jia
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Shengyuan Zhou
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| | - Xiongsheng Chen
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| |
Collapse
|
2
|
Chen JR, Zhao JT, Xie ZZ. Integrin-mediated cancer progression as a specific target in clinical therapy. Biomed Pharmacother 2022; 155:113745. [DOI: 10.1016/j.biopha.2022.113745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/15/2022] Open
|
3
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
4
|
Zhang J, Zhang P, Zou Q, Li X, Fu J, Luo Y, Liang X, Jin Y. Co-Delivery of Gemcitabine and Paclitaxel in cRGD-Modified Long Circulating Nanoparticles with Asymmetric Lipid Layers for Breast Cancer Treatment. Molecules 2018; 23:molecules23112906. [PMID: 30405089 PMCID: PMC6278289 DOI: 10.3390/molecules23112906] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 12/19/2022] Open
Abstract
Combination chemotherapy is a common clinical practice in cancer treatment. Here, cyclic RGD (arginylglycylaspartic acid) peptide was introduced to the surface of lipid/calcium/phosphate (LCP) asymmetric lipid layer nanoparticles for the co-delivery of paclitaxel (PTX) and gemcitabine monophosphate (GMP) (P/G-NPs). The sphere-like morphology of P/G-NPs displays a well-distributed particle size, and high entrapment efficiency and drug loading for both PTX and GMP, with a positive zeta potential. P/G-NPs were stable for up to 15 days. The cellular uptake of these cyclic RGD-modified nanoparticles was significantly higher than that of unmodified nanoparticles over 2 h incubation. Compared with the combination of free PTX and GMP (P/G-Free), P/G-NPs exhibited a longer circulation lifetime and improved absorption for PTX and GMP. Polyethylene glycol was responsible for a higher plasma concentration and a decreased apparent volume of distribution (Vz). Nanoparticles enhanced the drug accumulation in tumors compared with other major organs after 24 h. P/G-NPs nearly halted tumor growth, with little evidence of general toxicity, whereas P/G-Free had only a modest inhibitory effect at 16 mg/kg of GMP and 2.0 mg/kg of PTX. Increased levels of apoptosis within tumors were detected in P/G-NPs group by approximately 43.6% (TUNEL assay). When compared with GMP NPs, PTX NPs, and P/G-Free, P/G-NPs decreased expression of B-cell lymphoma-2 and B-cell lymphoma-extra large proteins, and increased expression of cleaved poly-ADP-ribose polymerase-1. Calreticulin expression in tumors also increased upon the co-delivery of PTX and GMP. The antitumor effect of P/G-NPs is more powerful than P/G-Free, GMP NP, or PTX NP alone, without obvious toxicity.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Peng Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Qian Zou
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Xiang Li
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China.
| | - Jianjiang Fu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Ying Luo
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Xinli Liang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China.
| |
Collapse
|
5
|
Iagaru A, Mosci C, Mittra E, Zaharchuk G, Fischbein N, Harsh G, Li G, Nagpal S, Recht L, Gambhir SS. Glioblastoma Multiforme Recurrence: An Exploratory Study of (18)F FPPRGD2 PET/CT. Radiology 2015; 277:497-506. [PMID: 25965900 DOI: 10.1148/radiol.2015141550] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE To prospectively evaluate fluorine 18 ((18)F) 2-fluoropropionyl-labeled PEGylated dimeric arginine-glycine-aspartic acid (RGD) peptide (PEG3-E[c{RGDyk}]2) (FPPRGD2) positron emission tomography (PET) in patients with glioblastoma multiforme (GBM). MATERIALS AND METHODS The institutional review board approved this HIPAA-compliant protocol. Written informed consent was obtained from each patient. (18)F FPPRGD2 uptake was measured semiquantitatively in the form of maximum standardized uptake values (SUV(max)) and uptake volumes before and after treatment with bevacizumab. Vital signs and laboratory results were collected before, during, and after the examinations. A nonparametric version of multivariate analysis of variance was used to assess safety outcome measures simultaneously across time points. A paired two-sample t test was performed to compare SUV(max). RESULTS A total of 17 participants (eight men, nine women; age range, 25-65 years) were enrolled prospectively. (18)F FPPRGD2 PET/computed tomography (CT), (18)F fluorodeoxyglucose (FDG) PET/CT, and brain magnetic resonance (MR) imaging were performed within 3 weeks, prior to the start of bevacizumab therapy. In eight of the 17 patients (47%), (18)F FPPRGD2 PET/CT was repeated 1 week after the start of bevacizumab therapy; six patients (35%) underwent (18)F FPPRGD2 PET/CT a third time 6 weeks after starting bevacizumab therapy. There were no changes in vital signs, electrocardiographic findings, or laboratory values that qualified as adverse events. One patient (6%) had recurrent GBM identified only on (18)F FPPRGD2 PET images, and subsequent MR images enabled confirmation of recurrence. Of the 17 patients, 14 (82%) had recurrent GBM identified on (18)F FPPRGD2 PET and brain MR images, while (18)F FDG PET enabled identification of recurrence in 13 (76%) patients. Two patients (12%) had no recurrent GBM. CONCLUSION (18)F FPPRGD2 is a safe PET radiopharmaceutical that has increased uptake in GBM lesions. Larger cohorts are required to confirm these preliminary findings.
Collapse
Affiliation(s)
- Andrei Iagaru
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Camila Mosci
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Erik Mittra
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Greg Zaharchuk
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Nancy Fischbein
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Griffith Harsh
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Gordon Li
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Seema Nagpal
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Lawrence Recht
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| | - Sanjiv Sam Gambhir
- From the Division of Nuclear Medicine and Molecular Imaging (A.I., C.M., E.M.), Department of Radiology, Neuroradiology Section (G.Z., N.F.), Division of Neurosurgery (G.H., G.L.), and Division of Neuro Oncology (S.N., L.R.), Stanford University Medical Center, 300 Pasteur Dr, Room H-2200, Stanford, CA 94305; and Departments of Radiology, Bioengineering, Materials Science, and Engineering, Stanford University School of Medicine, Stanford, Calif (S.S.G.)
| |
Collapse
|
6
|
Iagaru A, Mosci C, Shen B, Chin FT, Mittra E, Telli ML, Gambhir SS. 18F-FPPRGD2 PET/CT: Pilot Phase Evaluation of Breast Cancer Patients. Radiology 2014; 273:549-59. [DOI: 10.1148/radiol.14140028] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
7
|
Mamuya FA, Wang Y, Roop VH, Scheiblin DA, Zajac JC, Duncan MK. The roles of αV integrins in lens EMT and posterior capsular opacification. J Cell Mol Med 2014; 18:656-70. [PMID: 24495224 PMCID: PMC4000117 DOI: 10.1111/jcmm.12213] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/18/2013] [Indexed: 12/23/2022] Open
Abstract
Posterior capsular opacification (PCO) is the major complication arising after cataract treatment. PCO occurs when the lens epithelial cells remaining following surgery (LCs) undergo a wound healing response producing a mixture of α-smooth muscle actin (α-SMA)-expressing myofibroblasts and lens fibre cells, which impair vision. Prior investigations have proposed that integrins play a central role in PCO and we found that, in a mouse fibre cell removal model of cataract surgery, expression of αV integrin and its interacting β-subunits β1, β5, β6, β8 are up-regulated concomitant with α-SMA in LCs following surgery. To test the hypothesis that αV integrins are functionally important in PCO pathogenesis, we created mice lacking the αV integrin subunit in all lens cells. Adult lenses lacking αV integrins are transparent and show no apparent morphological abnormalities when compared with control lenses. However, following surgical fibre cell removal, the LCs in control eyes increased cell proliferation, and up-regulated the expression of α-SMA, β1-integrin, fibronectin, tenascin-C and transforming growth factor beta (TGF-β)-induced protein within 48 hrs, while LCs lacking αV integrins exhibited much less cell proliferation and little to no up-regulation of any of the fibrotic markers tested. This effect appears to result from the known roles of αV integrins in latent TGF-β activation as αV integrin null lenses do not exhibit detectable SMAD-3 phosphorylation after surgery, while this occurs robustly in control lenses, consistent with the known roles for TGF-β in fibrotic PCO. These data suggest that therapeutics antagonizing αV integrin function could be used to prevent fibrotic PCO following cataract surgery.
Collapse
Affiliation(s)
- Fahmy A Mamuya
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | | | | | | | | | | |
Collapse
|
8
|
Zhu JH, Yuan Y, Li D, Liao SJ, Zhou YH, Wang Q, Shu Y, Yan B, Wei JJ, Sun R, Zhang GM, Feng ZH. Targeting nuclear factor-κB suppresses the negative effect of toll-like receptor 4 signaling on antimetastasis therapy based on targeting αvβ3. Cancer Sci 2012; 103:1319-26. [PMID: 22494046 DOI: 10.1111/j.1349-7006.2012.02299.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 04/02/2012] [Accepted: 04/02/2012] [Indexed: 12/28/2022] Open
Abstract
The targeting of αvβ3 is a promising therapeutic strategy for suppressing tumor metastasis. However, it is unclear whether the therapeutic efficacy could be influenced by metastasis-promoting factor(s) in vivo. Here we report that Toll-like receptor 4 (TLR4) ligand released from damaged tumor cells or bacteria had a negative effect on the therapeutic effect of a recombinant CBD-HepII polypeptide of fibronectin (CH50) that suppresses tumor metastasis by targeting αvβ3. The TLR4 ligand could antagonize the inhibitory effect of CH50 on tumor cell adhesion and invasion by promoting the expression and activity of αvβ3 in tumor cells. The TLR4 ligand also reduced the antimetastasis effect of CH50 by promoting tumor cell survival in circulation. Moreover, TLR4 ligands released by tumor cells in circulation could increase the survival and proliferation capacity of tumor cells after extravasation, resulting in the formation of more metastatic nodules. The effect of TLR4 signaling was mainly mediated by nuclear factor-κB (NF-κB). Inhibiting NF-κB could abrogate the negative effect of TLR4 ligand, and augment the inhibitory effect of CH50 on tumor metastasis. Consistently, the combination of NF-κB inhibitor and CH50 significantly inhibited metastasis of tumor cells in vivo and prolonged the survival of mice. The findings in this study suggest that the combination of NF-κB inhibitor and αvβ3 antagonist would be a novel therapeutic option for the prevention of tumor metastasis.
Collapse
Affiliation(s)
- Jian-Hua Zhu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Yoncheva K, Momekov G. Antiangiogenic anticancer strategy based on nanoparticulate systems. Expert Opin Drug Deliv 2011; 8:1041-56. [DOI: 10.1517/17425247.2011.585155] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
10
|
Mas-Moruno C, Rechenmacher F, Kessler H. Cilengitide: the first anti-angiogenic small molecule drug candidate design, synthesis and clinical evaluation. Anticancer Agents Med Chem 2011; 10:753-68. [PMID: 21269250 PMCID: PMC3267166 DOI: 10.2174/187152010794728639] [Citation(s) in RCA: 480] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 02/02/2011] [Indexed: 12/29/2022]
Abstract
Cilengitide, a cyclic RGD pentapeptide, is currently in clinical phase III for treatment of glioblastomas and in phase II for several other tumors. This drug is the first anti-angiogenic small molecule targeting the integrins αvβ3, αvβ5 and αvβ1. It was developed by us in the early 90s by a novel procedure, the spatial screening. This strategy resulted in c(RGDfV), the first superactive αvβ3 inhibitor (100 to 1000 times increased activity over the linear reference peptides), which in addition exhibited high selectivity against the platelet receptor αIIbβ3. This cyclic peptide was later modified by N-methylation of one peptide bond to yield an even greater antagonistic activity in c(RGDf(NMe)V). This peptide was then dubbed Cilengitide and is currently developed as drug by the company Merck-Serono (Germany). This article describes the chemical development of Cilengitide, the biochemical background of its activity and a short review about the present clinical trials. The positive anti-angiogenic effects in cancer treatment can be further increased by combination with "classical" anti-cancer therapies. Several clinical trials in this direction are under investigation.
Collapse
Affiliation(s)
- Carlos Mas-Moruno
- Institute for Advance Study, Department Chemie, Technische Universität München, Garching, Germany
| | | | | |
Collapse
|
11
|
Mittra ES, Goris ML, Iagaru AH, Kardan A, Burton L, Berganos R, Chang E, Liu S, Shen B, Chin FT, Chen X, Gambhir SS. Pilot pharmacokinetic and dosimetric studies of (18)F-FPPRGD2: a PET radiopharmaceutical agent for imaging α(v)β(3) integrin levels. Radiology 2011; 260:182-91. [PMID: 21502381 DOI: 10.1148/radiol.11101139] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE To assess the safety, biodistribution, and dosimetric properties of the positron emission tomography (PET) radiopharmaceutical agent fluorine 18 ((18)F) FPPRGD2 (2-fluoropropionyl labeled PEGylated dimeric RGD peptide [PEG3-E{c(RGDyk)}2]), which is based on the dimeric arginine-glycine-aspartic acid (RGD) peptide sequence and targets α(v)β(3) integrin, in the first volunteers imaged with this tracer. MATERIALS AND METHODS The protocol was approved by the institutional review board, and written informed consent was obtained from all participants. Five healthy volunteers underwent whole-body combined PET-computed tomography 0.5, 1.0, 2.0, and 3.0 hours after tracer injection (mean dose, 9.5 mCi ± 3.4 [standard deviation] [351.5 MBq ± 125.8]; mean specific radioactivity, 1200 mCi/mmol ± 714 [44.4 GBq/mmol ± 26.4]). During this time, standard vital signs, electrocardiographic (ECG) readings, and blood sample values (for chemistry, hematologic, and liver function tests) were checked at regular intervals and 1 and 7 days after the injection. These data were used to evaluate tracer biodistribution and dosimetric properties, time-activity curves, and the stability of laboratory values. Significant changes in vital signs and laboratory values were evaluated by using a combination of population-averaged generalized estimating equation regression and exact paired Wilcoxon tests. RESULTS The administration of (18)F-FPPRGD2 was well tolerated, with no marked effects on vital signs, ECG readings, or laboratory values. The tracer showed the same pattern of biodistribution in all volunteers: primary clearance through the kidneys (0.360 rem/mCi ± 0.185 [0.098 mSv/MBq ± 0.050]) and bladder (0.862 rem/mCi ± 0.436 [0.233 mSv/MBq ± 0.118], voiding model) and uptake in the spleen (0.250 rem/mCi ± 0.168 [0.068 mSv/MBq ± 0.046]) and large intestine (0.529 rem/mCi ± 0.236 [0.143 mSv/MBq ± 0.064]). The mean effective dose of (18)F-FPPRGD2 was 0.1462 rem/mCi ± 0.0669 (0.0396 mSv/MBq ± 0.0181). With an injected dose of 10 mCi (370 MBq) and a 1-hour voiding interval, a patient would be exposed to an effective radiation dose of 1.5 rem (15 mSv). Above the diaphragm, there was minimal uptake in the brain ventricles, salivary glands, and thyroid gland. Time-activity curves showed rapid clearance from the vasculature, with a mean 26% ± 17 of the tracer remaining in the circulation at 30 minutes and most of the activity occurring in the plasma relative to cells (mean whole blood-plasma ratio, 0.799 ± 0.096). CONCLUSION (18)F-FPPRGD2 has desirable pharmacokinetic and biodistribution properties. The primary application is likely to be PET evaluation of oncologic patients-especially those with brain, breast, or lung cancer. Specific indications may include tumor staging, identifying patients who would benefit from antiangiogenesis therapy, and separating treatment responders from nonresponders early.
Collapse
Affiliation(s)
- Erik S Mittra
- Molecular Imaging Program, Department of Radiology, Division of Nuclear Medicine, Stanford Hospital and Clinics, 300 Pasteur Dr, Room H2200, Stanford, CA 94305-5281, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Bridoux A, Khan RA, Chen C, Chevé G, Cui H, Dyskin E, Yasri A, Mousa SA. Design, synthesis, and biological evaluation of bifunctional thyrointegrin inhibitors: new anti-angiogenesis analogs. J Enzyme Inhib Med Chem 2011; 26:871-82. [DOI: 10.3109/14756366.2011.557023] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alexandre Bridoux
- Pharmaceutical Research Institute, Rensselaer, NY, USA
- Vascular Vision Pharmaceuticals, Rensselaer, NY, USA
| | - Riaz A. Khan
- Pharmaceutical Research Institute, Rensselaer, NY, USA
- Department of Chemistry, Manav Rachna International University (MRIU), Faridabad, Haryana, India
| | - Celei Chen
- Pharmaceutical Research Institute, Rensselaer, NY, USA
| | - Gwenaël Chevé
- NOVADECISION, Rond point Benjamin Franklin–C539521, 34950 Montpellier Cedex 2, France
| | - Huadong Cui
- Pharmaceutical Research Institute, Rensselaer, NY, USA
| | - Evgeny Dyskin
- Pharmaceutical Research Institute, Rensselaer, NY, USA
| | - Aziz Yasri
- NOVADECISION, Rond point Benjamin Franklin–C539521, 34950 Montpellier Cedex 2, France
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Rensselaer, NY, USA
- Vascular Vision Pharmaceuticals, Rensselaer, NY, USA
- King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
13
|
Lu D, Scully M, Kakkar V, Lu X. ADAM-15 disintegrin-like domain structure and function. Toxins (Basel) 2010; 2:2411-27. [PMID: 22069559 PMCID: PMC3153164 DOI: 10.3390/toxins2102411] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 10/13/2010] [Accepted: 10/18/2010] [Indexed: 12/23/2022] Open
Abstract
The ADAM (a disintegrin-like and metalloproteinase) proteins are a family of transmembrane cell-surface proteins with important functions in adhesion and proteolytic processing in all animals. Human ADAM-15 is the only member of the ADAM family with the integrin binding motif Arg-Gly-Asp (RGD) in its disintegrin-like domain. This motif is also found in most snake venom disintegrins and other disintegrin-like proteins. This unique RGD motif within ADAM-15 serves as an integrin ligand binding site, through which it plays a pivotal role in interacting with integrin receptors, a large family of heterodimeric transmembrane glycoproteins. This manuscript will present a review of the RGD-containing disintegrin-like domain structures and the structural features responsible for their activity as antagonists of integrin function in relation to the canonical RGD template.
Collapse
Affiliation(s)
- Dong Lu
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR, UK; (D.L.); (M.S.); (V.K.)
- Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Mike Scully
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR, UK; (D.L.); (M.S.); (V.K.)
| | - Vijay Kakkar
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR, UK; (D.L.); (M.S.); (V.K.)
| | - Xinjie Lu
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR, UK; (D.L.); (M.S.); (V.K.)
- Author to whom correspondence should be addressed; ; Tel.: +44-0207-351-8312; Fax: +44-0207-351-8324
| |
Collapse
|
14
|
Janik ME, Lityńska A, Vereecken P. Cell migration-the role of integrin glycosylation. Biochim Biophys Acta Gen Subj 2010; 1800:545-55. [PMID: 20332015 DOI: 10.1016/j.bbagen.2010.03.013] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 03/11/2010] [Accepted: 03/17/2010] [Indexed: 12/25/2022]
Abstract
BACKGROUND Cell migration is an essential process in organ homeostasis, in inflammation, and also in metastasis, the main cause of death from cancer. The extracellular matrix (ECM) serves as the molecular scaffold for cell adhesion and migration; in the first phase of migration, adhesion of cells to the ECM is critical. Engagement of integrin receptors with ECM ligands gives rise to the formation of complex multiprotein structures which link the ECM to the cytoplasmic actin skeleton. Both ECM proteins and the adhesion receptors are glycoproteins, and it is well accepted that N-glycans modulate their conformation and activity, thereby affecting cell-ECM interactions. Likely targets for glycosylation are the integrins, whose ability to form functional dimers depends upon the presence of N-linked oligosaccharides. Cell migratory behavior may depend on the level of expression of adhesion proteins, and their N-glycosylation that affect receptor-ligand binding. SCOPE OF REVIEW The mechanism underlying the effect of integrin glycosylation on migration is still unknown, but results gained from integrins with artificial or mutated N-glycosylation sites provide evidence that integrin function can be regulated by changes in glycosylation. GENERAL SIGNIFICANCE A better understanding of the molecular mechanism of cell migration processes could lead to novel diagnostic and therapeutic approaches and applications. For this, the proteins and oligosaccharides involved in these events need to be characterized.
Collapse
Affiliation(s)
- Marcelina E Janik
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, Krakow, Poland.
| | | | | |
Collapse
|
15
|
Abstract
The role of cell adhesion molecules (CAMs) and extracellular matrix (ECM) proteins in various pathological processes, including angiogenesis, thrombosis, inflammation, apoptosis, cell migration, and proliferation is well documented. These processes can lead to both acute and chronic disease states such as ocular diseases, metastasis, unstable angina, myocardial infarction, stroke, osteoporosis, a wide range of inflammatory diseases, vascular remodeling, and neurodegenerative disorders. A key success in this field was identification of the role of platelet glycoprotein (GP)IIb/IIIa in the prevention and diagnosis of various thromboembolic disorders. The use of soluble adhesion molecules as potential diagnostic markers for acute and chronic leukocyte, platelet, and endothelial cell insult is becoming increasingly common. The development of various therapeutic and diagnostic candidates based on the key role of CAMs, with special emphasis on integrins in various diseases, as well as the structure-function aspects of cell adhesion and signaling of the different CAMs and ECM are highlighted.
Collapse
Affiliation(s)
- Shaker A Mousa
- Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| |
Collapse
|
16
|
Rerat V, Dive G, Cordi AA, Tucker GC, Bareille R, Amédée J, Bordenave L, Marchand-Brynaert J. αvβ3 Integrin-Targeting Arg-Gly-Asp (RGD) Peptidomimetics Containing Oligoethylene Glycol (OEG) Spacers. J Med Chem 2009; 52:7029-43. [DOI: 10.1021/jm901133z] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Vincent Rerat
- Unité de Chimie Organique et Médicinale, Université Catholique de Louvain, Bâtiment Lavoisier, Place L. Pasteur 1, 1348 Louvain-la-Neuve, Belgium
| | - Georges Dive
- Centre d’Ingénierie des Protéines, Université de Liège, Bâtiment B6, Allée de la Chimie, 4000 Sart-Tilman, Belgium
| | - Alex A. Cordi
- Institut de Recherches Servier, Rue des Moulineaux 11, 92150 Suresnes, France
| | - Gordon C. Tucker
- Institut de Recherches Servier, Rue des Moulineaux 11, 92150 Suresnes, France
| | - Reine Bareille
- INSERM, U577, Université Victor Segalen Bordeaux 2, Rue Léo Saignat 146, 33076 Bordeaux Cedex, France
| | - Joëlle Amédée
- INSERM, U577, Université Victor Segalen Bordeaux 2, Rue Léo Saignat 146, 33076 Bordeaux Cedex, France
| | - Laurence Bordenave
- INSERM, U577, Université Victor Segalen Bordeaux 2, Rue Léo Saignat 146, 33076 Bordeaux Cedex, France
- CIC-IT Biomatériaux, INSERM, Pessac, F-33604 France; CHU Bordeaux, Hôpital Xavier Arnozan, Pessac, 33604, France
| | - Jacqueline Marchand-Brynaert
- Unité de Chimie Organique et Médicinale, Université Catholique de Louvain, Bâtiment Lavoisier, Place L. Pasteur 1, 1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
17
|
Burvenich I, Schoonooghe S, Vervoort L, Dumolyn C, Coene E, Vanwalleghem L, Van Huysse J, Praet M, Cuvelier C, Mertens N, De Vos F, Slegers G. Monoclonal antibody 14C5 targets integrin alphavbeta5. Mol Cancer Ther 2009; 7:3771-9. [PMID: 19074852 DOI: 10.1158/1535-7163.mct-08-0600] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study identifies and characterizes the antigen recognized by monoclonal antibody (mAb) 14C5. We compared the expression of antigen 14C5 with the expression of eight integrin subunits (alpha1, alpha2, alpha3, alphav, beta1, beta2, beta3, and beta4) and three integrin heterodimers (alphavbeta3, alphavbeta5, and alpha5beta1) by flow cytometry. Antigen 14C5 showed a similar expression to alphavbeta5 in eight different epithelial cancer cell lines (A549, A2058, C32, Capan-2, Colo16, HT-1080, HT-29, and SKBR-3). Specific binding of P1F6, an anti-alphavbeta5 specific antibody, was blocked by mAb 14C5. After transient expression of alphavbeta5 in 14C5-negative Colo16 cells, mAb 14C5 was able to bind a subpopulation of alphavbeta5-positive cells. We evaluated the tissue distribution of the 14C5 antigen in colon (n = 20) and lung (n = 16) cancer tissues. The colon carcinoma cells stained positive for 14C5 in 50% of tumors analyzed, whereas bronchoalveolar lung carcinoma and typical carcinoid were not positive for the antigen. More common types of non-small cell lung cancer, i.e., squamous (n = 5) and adenocarcinoma (n = 3), stained positive in 2 of 5 squamous carcinomas and in 1 of 3 investigated adenocarcinoma. Colon (95%) and lung (50%) carcinoma tissues showed extensive expression of antigen 14C5 in the stroma surrounding the tumor cells and on the membrane of the adjacent fibroblasts. We show for the first time that mAb 14C5 binds the vascular integrin alphavbeta5, suggesting that mAb 14C5 can be used as a screening agent to select colon and lung cancer patients that are eligible for anti-alphavbeta5-based therapies.
Collapse
Affiliation(s)
- Ingrid Burvenich
- Laboratory of Radiopharmacy, Harelbekestraat 72, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Chung J, Kim TH. Integrin-dependent translational control: Implication in cancer progression. Microsc Res Tech 2008; 71:380-6. [PMID: 18300291 DOI: 10.1002/jemt.20566] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The importance of translational control in cancer progression has been underscored by a number of recent studies. However, little is known how cancer cells maintain their high efficiency of translation. Here, we summarize studies that support the role of integrins in translational control, especially at the initiation step, and discuss the various mechanisms by which integrins regulate the recruitment of translational machinery. This review also examines the hypothesis that integrins contribute to various aspects of cancer progression such as proliferation, survival, angiogenesis, and invasion through translational control.
Collapse
Affiliation(s)
- Jun Chung
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA.
| | | |
Collapse
|
19
|
Lu X, Lu D, Scully M, Kakkar V. The Role of Integrins in Cancer and the Development of Anti-Integrin Therapeutic Agents for Cancer Therapy. PERSPECTIVES IN MEDICINAL CHEMISTRY 2008. [DOI: 10.1177/1177391x0800200003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Integrins have been reported to mediate cell survival, proliferation, differentiation, and migration programs. For this reason, the past few years have seen an increased interest in the implications of integrin receptors in cancer biology and tumor cell aggression. This review considers the potential role of integrins in cancer and also addresses why integrins are present attractive targets for drug design. It discusses of the several properties of the integrin-based chemotherapeutic agents currently under consideration clinically and provides an insight into cancer drug development using integrin as a target.
Collapse
Affiliation(s)
- Xinjie Lu
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR U.K
| | - Dong Lu
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, U.K
| | - Mike Scully
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR U.K
| | - Vijay Kakkar
- Thrombosis Research Institute, Manresa Road, London, SW3 6LR U.K
| |
Collapse
|
20
|
Havaki S, Kouloukoussa M, Amawi K, Drosos Y, Arvanitis LD, Goutas N, Vlachodimitropoulos D, Vassilaros SD, Katsantoni EZ, Voloudakis-Baltatzis I, Aleporou-Marinou V, Kittas C, Marinos E. Altered expression pattern of integrin alphavbeta3 correlates with actin cytoskeleton in primary cultures of human breast cancer. Cancer Cell Int 2007; 7:16. [PMID: 17910753 PMCID: PMC2116995 DOI: 10.1186/1475-2867-7-16] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Accepted: 10/02/2007] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Integrins are transmembrane adhesion receptors that provide the physical link between the actin cytoskeleton and the extracellular matrix. It has been well established that integrins play a major role in various cancer stages, such as tumor growth, progression, invasion and metastasis. In breast cancer, integrin alphavbeta3 has been associated with high malignant potential in cancer cells, signaling the onset of widespread metastasis. Many preclinical breast cancer studies are based on established cell lines, which may not represent the cell behavior and phenotype of the primary tumor of origin, due to undergone genotypic and phenotypic changes. In the present study, short-term primary breast cancer cell cultures were developed. Integrin alphavbeta3 localization was studied in correlation with F-actin cytoskeleton by means of immunofluorescence and immunogold ultrastructural localization. Integrin fluorescence intensities were semi-quantitatively assessed by means of computerized image analysis, while integrin and actin expression was evaluated by Western immunoblotting. RESULTS In the primary breast cancer epithelial cells integrin alphavbeta3 immunofluorescence was observed in the marginal cytoplasmic area, whereas in the primary normal breast epithelial cells it was observed in the main cell body, i.e. in the ventrally located perinuclear area. In the former, F-actin cytoskeleton appeared well-formed, consisting of numerous and thicker stress fibers, compared to normal epithelial cells. Furthermore, electron microscopy showed increased integrin alphavbeta3 immunogold localization in epithelial breast cancer cells over the area of stress fibers at the basal cell surface. These findings were verified with Western immunoblotting by the higher expression of integrin beta3 subunit and actin in primary breast cancer cells, revealing their reciprocal relation, in response to the higher motility requirements, determined by the malignant potential of the breast cancer cells. CONCLUSION A model system of primary breast cancer cell cultures was developed, in an effort to maintain the closest resembling environment to the tumor of origin. Using the above system model as an experimental tool the study of breast tumor cell behavior is possible concerning the adhesion capacity and the migrating potential of these cells, as defined by the integrin alphavbeta3 distribution in correlation with F-actin cytoskeleton.
Collapse
Affiliation(s)
- Sophia Havaki
- Laboratory of Histology and Embryology, Medical School, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Greece
| | - Mirsini Kouloukoussa
- Laboratory of Histology and Embryology, Medical School, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Greece
| | - Kawther Amawi
- Laboratory of Histology and Embryology, Medical School, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Greece
| | - Yiannis Drosos
- Department of Genetics and Biotechnology, School of Biology, University of Athens, Panepistimioupoli, 15701 Ilissia, Greece
| | - Leonidas D Arvanitis
- Department of Anatomy and Pathology, Medical School, University of Thessaly, 22 Papakiriazi Str., 41222, Larissa, Greece
| | - Nikos Goutas
- Laboratory of Forensic Medicine and Toxicology, Medical School, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Greece
| | - Dimitrios Vlachodimitropoulos
- Laboratory of Forensic Medicine and Toxicology, Medical School, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Greece
| | | | - Eleni Z Katsantoni
- Hematology Division, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece
| | - Irene Voloudakis-Baltatzis
- Department of Electron Microscopy and Cell Biology, Research Centre of Oncology "G. Papanikolaou", Saint Savvas Anticancer Hospital, Alexandras Av. 171, Athens, Greece
| | - Vassiliki Aleporou-Marinou
- Department of Genetics and Biotechnology, School of Biology, University of Athens, Panepistimioupoli, 15701 Ilissia, Greece
| | - Christos Kittas
- Laboratory of Histology and Embryology, Medical School, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Greece
| | - Evangelos Marinos
- Laboratory of Histology and Embryology, Medical School, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Greece
| |
Collapse
|
21
|
Nemeth JA, Nakada MT, Trikha M, Lang Z, Gordon MS, Jayson GC, Corringham R, Prabhakar U, Davis HM, Beckman RA. Alpha-v integrins as therapeutic targets in oncology. Cancer Invest 2007; 25:632-46. [PMID: 18027153 DOI: 10.1080/07357900701522638] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Integrins are heterodimeric cell adhesion receptors that mediate intercellular communication through cell-extracellular matrix interactions and cell-cell interactions. Integrins have been demonstrated to play a direct role in cancer progression, specifically in tumor cell survival, tumor angiogenesis, and metastasis. Therefore, agents targeted against integrin function have potential as effective anticancer therapies. Numerous anti-integrin agents, including monoclonal antibodies and small-molecule inhibitors, are in clinical development for the treatment of solid and hematologic tumors. This review focuses on the role of alpha(v) integrins in cancer progression, the current status of integrin-targeted agents in development, and strategies for the clinical development of anti-integrin therapies.
Collapse
|
22
|
Niu G, Xiong Z, Cheng Z, Cai W, Gambhir SS, Xing L, Chen X. In vivo bioluminescence tumor imaging of RGD peptide-modified adenoviral vector encoding firefly luciferase reporter gene. Mol Imaging Biol 2007; 9:126-34. [PMID: 17297551 PMCID: PMC4165526 DOI: 10.1007/s11307-007-0079-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE The goal of this study is to demonstrate the feasibility of chemically modified human adenovirus (Ad) vectors for tumor retargeting. PROCEDURES E1- and E3-deleted Ad vectors carrying firefly luciferase reporter gene under cytomegalovirus promoter (AdLuc) was surface-modified with cyclic arginine-glycine-aspartic acid (RGD) peptides through a bifunctional poly(ethyleneglycol) linker (RGD-PEG-AdLuc) for integrin alpha(v)beta(3) specific delivery. The Coxsackie and adenovirus viral receptor (CAR) and integrin alpha(v)beta(3) expression in various tumor cell lines was determined by reverse transcriptase PCR and fluorescence-activated cell sorting. Bioluminescence imaging was performed in vitro and in vivo to evaluate RGD-modified AdLuc infectivity. RESULTS RGD-PEG-AdLuc abrogated the native CAR tropism and exhibited significantly enhanced transduction efficiency of integrin-positive tumors than AdLuc through intravenous administration. CONCLUSION This approach provides a robust platform for site-specific gene delivery and noninvasive monitoring of the transgene delivery efficacy and homing.
Collapse
Affiliation(s)
- Gang Niu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, P095, Stanford, CA 94305-5484, USA
| | - Zhengming Xiong
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, P095, Stanford, CA 94305-5484, USA
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, P095, Stanford, CA 94305-5484, USA
| | - Weibo Cai
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, P095, Stanford, CA 94305-5484, USA
| | - Sanjiv S. Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, P095, Stanford, CA 94305-5484, USA
| | - Lei Xing
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoyuan Chen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, P095, Stanford, CA 94305-5484, USA
| |
Collapse
|
23
|
Sroka TC, Marik J, Pennington ME, Lam KS, Cress AE. The minimum element of a synthetic peptide required to block prostate tumor cell migration. Cancer Biol Ther 2006; 5:1556-62. [PMID: 17102593 PMCID: PMC2704570 DOI: 10.4161/cbt.5.11.3461] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Human prostate tumor cell invasion and metastasis are dependent in part on cell adhesion to extracellular matrix proteins and cell migration. We previously identified a synthetic D-amino acid tumor cell adhesion peptide called HYD1 (kikmviswkg) that supported adhesion of tumor cells derived from breast, prostate, ovary and pancreas tissue. Alanine substitution analysis and a peptide deletion strategy were used to determine the minimal element of HYD1 necessary for bioactivity in a prostate cancer cell line called PC3N. Bioactivity was measured by assays of cell adhesion, migration and ERK signaling. The most potent element of HYD1 necessary to support cell adhesion was kmvixw, the block to migration required xkmviswxx and activation of ERK signaling required ikmviswxx. The shortest sequence active in all three assays was iswkg. The HYD1 peptide contains overlapping elements required for adhesion, blocking migration and the activation of ERK signaling. These linear peptide sequences provide the starting point for development of novel compounds to target cancer cell adhesion and migration.
Collapse
Affiliation(s)
- Thomas C. Sroka
- The Arizona Cancer Center, University of Arizona, Tucson, Arizona USA
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona USA
| | - Jan Marik
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, California USA
| | | | - Kit S. Lam
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, California USA
| | - Anne E. Cress
- The Arizona Cancer Center, University of Arizona, Tucson, Arizona USA
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona USA
- Correspondence to: Anne E. Cress; Arizona Cancer Center; 1515 N. Campbell Ave.; Tucson, Arizona 85724 USA; Tel.: 520.626.7479; Fax: 520.626.4979;
| |
Collapse
|
24
|
Vukmirica J, Monzo P, Le Marchand-Brustel Y, Cormont M. The Rab4A effector protein Rabip4 is involved in migration of NIH 3T3 fibroblasts. J Biol Chem 2006; 281:36360-8. [PMID: 17001082 DOI: 10.1074/jbc.m602920200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The small GTP-binding protein Rab4 has been involved in the recycling of alphavbeta3 integrins in response to platelet-derived growth factor (PDGF) stimulation suggesting a role for Rab4 in cell adhesion and migration. In this study, we explored the role of Rabip4 and Rabip4', two Rab4 effector proteins, in migration of NIH 3T3 fibroblasts. In these cells, Rabip4 and Rabip4', collectively named Rabip4s, were partially co-localized with the early endosomal marker EEA1. PDGF treatment re-distributed endogenous Rabip4s toward the cell periphery where they colocalized with F-actin. In cells expressing green fluorescent protein (GFP)-Rabip4 or GFP-Rabip4', constitutive appearance of GFP-Rabip4s at the cell periphery was accompanied by local increase in cortical F-actin in membrane ruffles at the leading edge. The expression of GFP-Rabip4 induced an increased migration compared with control cells expressing GFP alone, even in the absence of PDGF stimulation. On the contrary, in cells expressing a mutated form of Rabip4s unable to interact with Rab4, lack of typical leading edge was observed. Furthermore, PDGF treatment did not stimulate the migration of these cells. Furthermore, down-regulation of the expression of Rabip4s inhibited PDGF-stimulated cell migration. Endogenous Rabip4s were localized with alphav integrins at the leading edge following PDGF treatment, whereas in cells expressing GFP-Rabip4s, alphav integrins, together with GFP-Rabip4s, were constitutively localized at the leading edge. In contrast, reduction in Rabip4s expression levels using small interfering RNA was associated with impaired PDGF-induced translocation of alphav integrins toward the leading edge. Taken together, our data provide evidence that Rabip4s, possibly via their interaction with Rab4, regulate integrin trafficking and are involved in the migration of NIH 3T3 fibroblasts.
Collapse
Affiliation(s)
- Jelena Vukmirica
- INSERM U568, UFR Médecine, 06107 Nice Cedex 02 and Université de Nice-Sophia-Antipolis, UFR Sciences, 06002 Nice, France
| | | | | | | |
Collapse
|
25
|
Maubant S, Saint-Dizier D, Boutillon M, Perron-Sierra F, Casara PJ, Hickman JA, Tucker GC, Van Obberghen-Schilling E. Blockade of alpha v beta3 and alpha v beta5 integrins by RGD mimetics induces anoikis and not integrin-mediated death in human endothelial cells. Blood 2006; 108:3035-44. [PMID: 16835373 DOI: 10.1182/blood-2006-05-023580] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alpha v integrins are thought to play an important role in tumor angiogenesis. However, discrepancies between findings with Arg-Gly-Asp (RGD) mimetics, which block angiogenesis in animal models, and knockout mice, in which loss of some alpha v integrins enhances tumor angiogenesis, raise questions concerning the function of these integrins and the precise role of alpha v substrate mimetics in antiangiogenic therapies. We have examined the effects of a novel non-peptide RGD mimetic, S 36578-2, on human endothelial cells to elucidate its antagonist activity and to identify possible agonist functions. S 36578-2 is highly selective for alpha v beta3 and alpha v beta5 integrins and induces detachment, caspase-8 activation, and apoptosis in human umbilical endothelial cells (HUVECs) plated on vitronectin. Importantly, the compound has no effect on the morphology or survival of cells plated on interstitial matrix components such as fibronectin, and it does not potentiate the apoptotic process in suspended cells. Identical results were obtained with a cyclic RGD peptide with similar target specificity. In microvascular endothelial cells, S 36578-2-induced death was also linked to its antiadhesive effect, with established lines markedly more resistant than primary cultures to the antiadhesive and proapoptotic effects. Altogether, these findings have important implications for the development of this class of antiangiogenics.
Collapse
Affiliation(s)
- Sylvie Maubant
- Centre National de Recherche Scientifique, Unité Mixte de Recherche (CNRS UMR) 6543, Centre Antoine Lacassagne, Nice, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Raboisson P, Manthey CL, Chaikin M, Lattanze J, Crysler C, Leonard K, Pan W, Tomczuk BE, Marugán JJ. Novel potent and selective αvβ3/αvβ5 integrin dual antagonists with reduced binding affinity for human serum albumin. Eur J Med Chem 2006; 41:847-61. [PMID: 16697080 DOI: 10.1016/j.ejmech.2006.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/07/2006] [Accepted: 03/09/2006] [Indexed: 11/30/2022]
Abstract
The binding of lead compounds and drugs to human serum albumin (HSA) is a ubiquitous problem in drug discovery since it modulates the availability of the leads and drugs to their intended target, which is linked to biological efficacy. In our continuing efforts to identify small molecule alpha(V)beta(3) and alpha(V)beta(5) dual antagonists, we recently reported indoles 2-4 as potent and selective alpha(V)beta(3)/alpha(V)beta(5) antagonists with good oral bioavailability profile. In spite of subnanomolar binding affinity of these compounds to human alpha(V)beta(3) and alpha(V)beta(5) integrins, high HSA binding (96.5-97.3%) emerged as a limiting feature for these leads. Structure-activity HSA binding data of organic acids reported in the literature have demonstrated that the incorporation of polar groups into a given molecule can dramatically decrease the affinity toward HSA. We sought to apply this strategy by examining the effects of such modifications in both the central core constrain and the substituent beta to the carboxylate. Most of these derivatives were prepared in good yields through a cesium fluoride-catalyzed coupling reaction. This reaction was successful with a variety of nitrogen-containing scaffolds (20, 33, and 43) and selected acetylenic derivatives (16, 19, and 34). Among the compounds synthesized, the 3-[5-[2-(5,6,7,8-tetrahydro [1,8]naphthyridin-2-yl)ethoxy]indol-1-yl]-3-[5-(N,N-dimethylaminomethyl)-3-pyridyl]propionic acid (25) was found to be the most promising derivative within this novel series with a subnanomolar affinity for both alpha(v)beta(3) and alpha(v)beta(5) (IC(50) = 0.29 and 0.16 nM, respectively), similar to our initial lead receptor antagonists 2-4, and exhibiting a low HSA protein binding (40% bound, K(d) = 1.1+/-0.4 x 10(3) microM) and an improved in vitro stability profile toward human and mouse microsomes (99.9% and 98.7% remaining after 10 min). Moreover, the selectivity of 25 toward alpha(5)beta(1) and IIbIIIa integrins was perfectly maintained when compared to the parent leads 2-4. Thus, compound 25 was selected as a new lead with improved drug-like properties for further evaluations in the field of oncology and osteoporosis.
Collapse
Affiliation(s)
- Pierre Raboisson
- Department of Medicinal Chemistry, Johnson & Johnson Pharmaceutical Research and Development, LLC, Exton, PA 19341, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Nagarajan SR, Meyer JM, Miyashiro JM, Engleman VW, Freeman SK, Griggs DW, Klover JA, Nickols GA. Discovery of Diphenylmethanepropionic and Dihydrostilbeneacetic Acids as Antagonists of the Integrin alphavbeta3. Chem Biol Drug Des 2006; 67:177-81. [PMID: 16492166 DOI: 10.1111/j.1747-0285.2006.00344.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
A peptidomimetic inhibitor of the integrin alpha(v)beta(3) has been substantially modified to produce several new nonpeptidic antagonists. These inhibitors are simpler to synthesize and belong to new classes of scaffolds. Some of the compounds served as the initial lead for further optimization, which led to the discovery of potent and selective inhibitors of the integrin alpha(v)beta(3).
Collapse
Affiliation(s)
- Srinivasan R Nagarajan
- Pfizer Global Research and Development, St Louis Laboratories, Pfizer, Inc., Chesterfield, MO 63017, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Reig F, Juvé A, Ortiz A, Sospedra P, Alsina MA. Effect of a laminin amphiphatic sequence on DPPC ordered bilayers. LUMINESCENCE 2005; 20:326-30. [PMID: 16134200 DOI: 10.1002/bio.851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A peptide sequence, stearoyl-GESIKVAVS(NH2), related to a laminin fragment, has been synthesized. Formation of aggregates was controlled by titrating a sodium anilinonaphthalene sulphonate (ANS) solution with peptide and recording fluorescence intensity increases. The results show that this system experiences a sudden increase in fluorescence at peptide concentrations around 2.5 x 10(-4) mol/L. The interaction of this hydrophobic peptide with DPPC vesicles has been studied using fluorescence techniques. Its influence on the microviscosity of bilayers was determined by studying polarization/temperature dependence for ANS and diphenyl hexatriene (DPH) fluorescent probes. With both markers the presence of peptide promotes a clear increase in anisotropy values. This indicates a rigidifying effect. Leakage studies carried out with liposomes loaded with carboxyfluorescein (CF) indicate a stabilizing effect of the peptide on bilayers, in agreement with results obtained with fluorescent probes.
Collapse
Affiliation(s)
- F Reig
- Peptides Department, Institute for Environmental and Biological Chemistry, CSIC, Jordi Girona 18, 08034 Barcelona, Spain
| | | | | | | | | |
Collapse
|
29
|
Sinner EK, Reuning U, Kök FN, Saccà B, Moroder L, Knoll W, Oesterhelt D. Incorporation of integrins into artificial planar lipid membranes: characterization by plasmon-enhanced fluorescence spectroscopy. Anal Biochem 2005; 333:216-24. [PMID: 15450795 DOI: 10.1016/j.ab.2004.05.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Indexed: 11/27/2022]
Abstract
An optimized peptide-tethered artificial lipid membrane system has been developed. Integrins (cell adhesion receptors) were functionally incorporated into this membrane model and integrin-ligand interactions were analyzed by surface plasmon-enhanced fluorescence spectroscopy (SPFS). The transmembrane receptors alpha(v)beta(3) and alpha(1)beta(1) of the integrin superfamily were incorporated into a lipid-functionalized peptide layer by vesicle spreading. Consecutive layer formations were monitored by surface plasmon spectroscopy (SPS). Orientation and accessibility of the membrane receptor alpha(v)beta(3) was reliably assessed by specific and reproducible binding of selective antibodies. Moreover, full retention of the functional properties of this receptor was verified by specific and reversible binding of natural ligands. Functional integrity of incorporated integrins was maintained over a time period of 72 h. The integrin/extracellular matrix ligand complexes, whose formations are known to depend on the presence of divalent cations, were lost upon addition of ethylenediaminetetraacetate. Therefore, regeneration of the surface for further binding experiments with minimized unspecific ligand association was possible. These results demonstrate that integrins can be functionally incorporated into peptide-tethered artificial membranes. In combination with the SPS/SPFS method, this artificial membrane system provides a reliable experimental platform for investigation of isolated membrane proteins under experimental conditions resembling those of their native environment.
Collapse
|
30
|
Biltresse S, Attolini M, Marchand-Brynaert J. Cell adhesive PET membranes by surface grafting of RGD peptidomimetics. Biomaterials 2005; 26:4576-87. [PMID: 15722127 DOI: 10.1016/j.biomaterials.2004.11.042] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2004] [Accepted: 11/30/2004] [Indexed: 11/30/2022]
Abstract
A non-peptide mimic of the Arg-Gly Asp (RGD) active sequence of adhesive proteins (such as vitronectin) has been equipped with two different spacer-arms for surface anchorage. The covalent grafting on poly(ethylene terephthalate) (PET) membrane was realized via the activation of the hydroxyl polymer chain-ends by tosylation followed by nucleophilic substitution. The surface density of peptidomimetics was determined by X-ray photoelectron spectroscopy (XPS), on the basis of F/C atomic ratios since a fluorine tag was incorporated into the RGD-like compounds. The biological activity of soluble peptidomimetics was evaluated versus isolated human integrin alpha(v)beta(3) (vitronectin receptor), and versus CaCo2 cells. Inhibition of cellular adhesion was observed after pre-incubation of CaCo2 cells with soluble peptidomimetics. On the other hand a significant promotion of cellular adhesion resulted from the surface grafting of peptidomimetics on the PET culture substrate. The best performance was obtained with the RGD-like integrin ligand bearing a triethylene glycol spacer-arm.
Collapse
Affiliation(s)
- Stéphane Biltresse
- Unité de chimie Organique et Médicinale, Université catholique de Louvain, Bâtiment Lavoisier, Place Louis Pasteur 1, B-1348 Louvain-la-Neuve, Belgium
| | | | | |
Collapse
|
31
|
Pattillo CB, Sari-Sarraf F, Nallamothu R, Moore BM, Wood GC, Kiani MF. Targeting of the Antivascular Drug Combretastatin to Irradiated Tumors Results in Tumor Growth Delay. Pharm Res 2005; 22:1117-20. [PMID: 16028012 DOI: 10.1007/s11095-005-5646-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Accepted: 04/25/2005] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of the study is to evaluate the effects of targeting the antivascular drug combretastatin to irradiated mouse melanomas. METHODS Combretastatin was incorporated into liposomes with surfaces modified by the addition of cyclo(Arg-Gly-Asp-D-Phe-Cys) (RGD) to create an immunoliposome (IL). This addition of RGD allows the liposome to be preferentially targeted to alphavbeta3, an integrin up-regulated in the vasculature of irradiated tumors. C57BL mice bearing a transplanted B16-F10 melanoma were randomly assigned to one of the following treatment groups: untreated, a single dose of 5-Gy radiation (IR), IL (14.5 mg/kg of combretastatin), 5-Gy radiation plus IL, and a systemic administration of free drug (81.0 mg/kg of combretastatin). RESULTS In this transplanted tumor model, there was no significant increase in the volume of the IL + IR (5 Gy) treated tumors during the initial 6 days posttreatment; all other treatment groups exhibited exponential growth curves after day 3. The IL + IR (5 Gy) treatment resulted in a 5.1-day tumor growth delay compared to untreated controls. CONCLUSIONS These findings indicate that preferential targeting of antivascular drugs to irradiated tumors results in significant tumor growth delay.
Collapse
Affiliation(s)
- Christopher B Pattillo
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
The process of formation of new vessels from pre-existing capillaries is called angiogenesis. Angiogenesis is a complex process which involves distinct cells, soluble components and factors related to the extra-cellular matrix and which is highly important in a large variety of physiological and pathological processes in the body. Angiogenesis regulation takes place through a perfect equilibrium between the production and release of different stimulatory and inhibitory factors which vary in relation to needs and tissue types. A large number of diseases are characterized by alterations in the angiogenic process, either by an insufficiency or by excessive angiogenesis. The requirement of blood vessel proliferation for tumor growth was observed more than a century ago. Angiogenic treatment would have an indirect antitumoral action, inhibiting tumor vascularization and impairing the supply of essential nutrients for tumoral growth and development.
Collapse
Affiliation(s)
- José L Mauriz
- Departamento de Fisiología, Universidad de León, León, Spain
| | | | | | | |
Collapse
|
33
|
Li S, Crothers J, Haqq CM, Blackburn EH. Cellular and Gene Expression Responses Involved in the Rapid Growth Inhibition of Human Cancer Cells by RNA Interference-mediated Depletion of Telomerase RNA. J Biol Chem 2005; 280:23709-17. [PMID: 15831499 DOI: 10.1074/jbc.m502782200] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Inhibition of the up-regulated telomerase activity in cancer cells has previously been shown to slow cell growth but only after prior telomere shortening. Previously, we have reported that, unexpectedly, a hairpin short interfering RNA specifically targeting human telomerase RNA rapidly inhibits the growth of human cancer cells independently of p53 or telomere length and without bulk telomere shortening (Li, S., Rosenberg, J. E., Donjacour, A. A., Botchkina, I. L., Hom, Y. K., Cunha, G. R., and Blackburn, E. H. (2004) Cancer Res. 64, 4833-4840). Here we have demonstrated that such telomerase RNA knockdown in cancer cells does not cause telomere uncapping but rather induces changes in the global gene expression profile indicative of a novel response pathway, which includes suppression of specific genes implicated in angiogenesis and metastasis, and is distinct from the expression profile changes induced by telomere-uncapping mutant template telomerase RNAs. These cellular responses to depleting telomerase in human cancer cells together suggest that cancer cells are "telomerase-addicted" and uncover functions of telomerase in tumor growth and progression in addition to telomere maintenance.
Collapse
Affiliation(s)
- Shang Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
34
|
Gonzalez Guerrico AM, Jaffer ZM, Page RE, Braunewell KH, Chernoff J, Klein-Szanto AJP. Visinin-like protein-1 is a potent inhibitor of cell adhesion and migration in squamous carcinoma cells. Oncogene 2005; 24:2307-16. [PMID: 15735716 DOI: 10.1038/sj.onc.1208476] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tumor cell invasion is a highly integrated and complex process comprising several biologically distinct functions such as cell adhesion, motility, proteolysis, etc. Visinin-like protein-1 (VILIP-1), a member of the neuronal EF-hand calcium-sensor protein family, plays a role in regulating tumor cell invasiveness of mouse squamous cell carcinoma (SCC). VILIP-1 enhances cyclic adenosine monophosphate levels through PKA induction. However, the mechanism by which VILIP-1 reduces cell invasiveness is not well understood. In this study, we show that VILIP-1 decreased cell adhesion and migration/invasiveness of highly invasive mouse SCC cells. Forced expression of VILIP-1 reduced cell adhesion to fibronectin in parallel to downregulating alphav and alpha5 integrin subunit levels. VILIP-1 overexpression also led to decreased migration ability. Conversely, short hairpin RNA-mediated VILIP-1 knock-down of SCC cells' characterized by little or no invasiveness, correlated with increased adhesion to fibronectin and enhanced expression of alphav and alpha5 integrin subunits together with increased cell migration. Function-blocking assays with inhibitory anti-alpha5 and anti-alphav integrin antibodies showed that both subunits contributed to cell adhesion, migration, and invasiveness of highly invasive SCC cell lines. These results point to a critical role of VILIP-1 in regulating cell adhesion and migration by downregulation of fibronectin receptor expression. Decreased or absent VILIP-1 expression in SCC cell subpopulations may lead to a more advanced malignant phenotype characterized by changes in adhesive ability and increased cell motility, suggestive of a tumor suppressor function.
Collapse
|
35
|
Wartchow CA, Alters SE, Garzone PD, Li L, Choi S, DeChene NE, Doede T, Huang L, Pease JS, Shen Z, Knox SJ, Cleland JL. Enhancement of the efficacy of an antagonist of an extracellular receptor by attachment to the surface of a biocompatible carrier. Pharm Res 2004; 21:1880-5. [PMID: 15553236 DOI: 10.1023/b:pham.0000045243.98010.b6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE In order to improve the in vitro and in vivo efficacy of an integrin antagonist (IA) of the extracellular domain of the alphavbeta3 integrin, a receptor upregulated on tumor neovasculature, the IA was attached to the surface of a dextran-coated liposome (DCL). IA-DCLs were characterized in vitro, and the pharmacokinetic and antitumor properties were assessed in vivo. METHODS The in vitro binding properties were measured with purified integrin, endothelial cells, and melanoma cells. The pharmacokinetic parameters were measured in healthy mice with 14C-labeled IA-DCLs and anti-tumor efficacy was assessed with the M21 human melanoma xenograft mouse model. RESULTS In vitro, IC50 values for IA-DCLs and IA are similar, and IA-DCLs inhibit cell proliferation relative to controls. IA-DCLs are stable in serum, and the pharmacokinetic half-life in mice is 23 h. In the M21/mouse model, statistically significant inhibition of tumor growth was observed for mice treated with IA-DCLs, whereas controls including saline, DCLs lacking IA, and cyclo(RGDfV) were ineffective. Increased apoptosis and a reduction in vessel counts relative to controls were present in tumors from animals treated with IA-DCLs. CONCLUSIONS These results demonstrate that IA-DCLs are potent anti-angiogenic therapeutic agents with superior in vivo activity and pharmacology compared to unmodified IA.
Collapse
|
36
|
Biltresse S, Attolini M, Dive G, Cordi A, Tucker GC, Marchand-Brynaert J. Novel RGD-like molecules based on the tyrosine template: design, synthesis, and biological evaluation on isolated integrins αVβ3/αIIbβ3 and in cellular adhesion tests. Bioorg Med Chem 2004; 12:5379-93. [PMID: 15388165 DOI: 10.1016/j.bmc.2004.07.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Accepted: 07/23/2004] [Indexed: 11/25/2022]
Abstract
RGD (Arg-Gly-Asp) peptidomimetics have been designed for covalent anchorage on biomaterials. The tyrosine template was thus equipped with (i) a basic side chain of various flexibility, (ii) an acidic side chain, which incorporated the XPS fluorine tag, and (iii) a spacer-arm terminated by a primary amine for surface grafting. The most active compounds showed IC50 values in the nanomolar range versus isolated human integrins alphaVbeta3 and alphaIIbbeta3. Preincubation of CaCo2 cells with soluble peptidomimetics (2 and 19a) prevented cellular adhesion on culture plates coated with vitronectin. On the other hand, peptidomimetics (19a and 19b) immobilized on a poly(ethylene)terephthalate membrane (PET) promoted CaCo2 cells adhesion. A modeling study at the ab initio level in MINI-1' basis allowed to compare the various synthetic ligands of integrins and to propose novel pharmacophore structures.
Collapse
Affiliation(s)
- Stephane Biltresse
- Unité de Chimie Organique et Médicinale, Université catholique de Louvain, Bâtiment Lavoisier, Place Louis Pasteur, 1, B-1348 Louvain-la-Neuve, Belgium
| | | | | | | | | | | |
Collapse
|
37
|
De Corte BL, Kinney WA, Liu L, Ghosh S, Brunner L, Hoekstra WJ, Santulli RJ, Tuman RW, Baker J, Burns C, Proost JC, Tounge BA, Damiano BP, Maryanoff BE, Johnson DL, Galemmo RA. Piperidine-containing β-arylpropionic acids as potent antagonists of αvβ3/αvβ5 integrins. Bioorg Med Chem Lett 2004; 14:5227-32. [PMID: 15380233 DOI: 10.1016/j.bmcl.2004.06.061] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2004] [Revised: 06/18/2004] [Accepted: 06/18/2004] [Indexed: 11/16/2022]
Abstract
The synthesis and SAR of a new class of piperidine-based alphavbeta3/alphavbeta5 integrin antagonists is described. Replacement of an amide bond in a prototype isonipecotamide by a C-C isostere, and adjustment of the spacer length between the carboxylic acid and basic moieties, led to low nanomolar antagonists of alphavbeta3 and/or alphavbeta5 integrins with excellent selectivity versus alpha(IIb)beta3.
Collapse
Affiliation(s)
- Bart L De Corte
- Drug Discovery, Johnson & Johnson Pharmaceutical Research and Development, Welsh & McKean Roads, Spring House, PA 19477-0776, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The integrin family of cell adhesion proteins promotes the attachment and migration of cells on the surrounding extracellular matrix (ECM). Through signals transduced upon integrin ligation by ECM proteins or immunoglobulin superfamily molecules, this family of proteins plays key roles in regulating tumour growth and metastasis as well as tumour angiogenesis. Several integrins play key roles in promoting tumour angiogenesis and tumour metastasis. Antagonists of several integrins (alpha5beta1, alphavbeta3 and alphavbeta5) are now under evaluation in clinical trials to determine their potential as therapeutics for cancer and other diseases.
Collapse
Affiliation(s)
- H Jin
- John and Rebecca Moores Comprehensive Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA
| | - J Varner
- John and Rebecca Moores Comprehensive Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA
- John and Rebecca Moores Comprehensive Cancer Center, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA. E-mail:
| |
Collapse
|
39
|
Abstract
Although the genetic basis of tumorigenesis may vary greatly between different cancer types, the cellular and molecular steps required for metastasis are similar for all cancer cells. Not surprisingly, the molecular mechanisms that propel invasive growth and metastasis are also found in embryonic development, and to a less perpetual extent, in adult tissue repair processes. It is increasingly apparent that the stromal microenvironment, in which neoplastic cells develop, profoundly influences many steps of cancer progression, including the ability of tumor cells to metastasize. In carcinomas, the influences of the microenvironment are mediated, in large part, by bidirectional interactions (adhesion, survival, proteolysis, migration, immune escape mechanisms lymph-/angiogenesis, and homing on target organs) between epithelial tumor cells and neighboring stromal cells, such as fibroblasts as well as endothelial and immune cells. In this review, we summarize recent advances in understanding the molecular mechanisms that govern this frequently lethal metastatic progression along an axis from primary tumor to regional lymph nodes to distant organ sites. Affected proteins include growth factor signaling molecules, chemokines, cell-cell adhesion molecules (cadherins, integrins) as well as extracellular proteases (matrix metalloproteinases). We then discuss promising new therapeutic approaches targeting the microenvironment. We note, however, that there is still too little knowledge of how the many events are coordinated and integrated by the cancer cell, with conspiratorial help by the stromal component of the host. Before drug development can proceed with a legitimate chance of success, significant gaps in basic knowledge need to be filled.
Collapse
|
40
|
Affiliation(s)
- John D Hood
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
41
|
Hapke S, Kessler H, Luber B, Benge A, Hutzler P, Höfler H, Schmitt M, Reuning U. Ovarian cancer cell proliferation and motility is induced by engagement of integrin alpha(v)beta3/Vitronectin interaction. Biol Chem 2003; 384:1073-83. [PMID: 12956424 DOI: 10.1515/bc.2003.120] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
During tumor metastasis, a fine-tuned balance between the formation and loosening of adhesive cell contacts has to occur, a process based on the regulated expression of integrins. Human ovarian OV-MZ-6 cancer cells express the integrin alpha(v)beta3, which associates with vitronectin (VN) and correlates with ovarian cancer progression. Adhesion and spreading of OV-MZ-6 cells on VN was accompanied by the formation of focal adhesion contacts and the recruitment of activated tyrosine-phosphorylated focal adhesion kinase. Cultivation of OV-MZ-6 cells on VN resulted in a significantly induced cell proliferation. This VN effect could be mimicked by cultivating cells on the immobilized alpha(v)beta3 directed peptide cyclo-Arg-Gly-Asp-D-Phe-Val (cRGDfV). VN-dependent OV-MZ-6 cell adhesion and proliferation was significantly enhanced by overexpression of alpha(v)beta3 and was accompanied by rapid and transient tyrosine-phosphorylation of p44(erk-1)/p42(erk-2) mitogen-activated protein kinase. Moreover, overexpression of alpha(v)beta3 and OV-MZ-6 cell attachment to VN increased cell motility up to 5-fold accompanied by prominent changes in cytoskeletal organization and cell morphology. Upon alpha(v)beta3/VN interaction, by cDNA expression microarray analysis we identified altered mRNA levels of c-myc, epidermal growth factor receptor (EGF-R), transcription factor Fra-1, prothymosin-alpha (PTMA), integrin-linked kinase (ILK), and the cell adhesion molecule SQM-1, candidates which are possibly involved in changes of the adhesive, migratory, and proliferative phenotype of human ovarian cancer cells.
Collapse
Affiliation(s)
- Sandra Hapke
- Department of Obstetrics and Gynecology, Clinical Research Unit, Technical University Munich, D-81675 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|