201
|
Kenny LM, Tomasi G, Turkheimer F, Larkin J, Gore M, Brock CS, Mangar S, Aboagye EO. Preliminary clinical assessment of the relationship between tumor alphavbeta3 integrin and perfusion in patients studied with [(18)F]fluciclatide kinetics and [ (15)O]H 2O PET. EJNMMI Res 2014; 4:30. [PMID: 26055935 PMCID: PMC4884000 DOI: 10.1186/s13550-014-0030-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 04/10/2014] [Accepted: 05/20/2014] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND [(18)F]fluciclatide, a peptide ligand with high affinity for αvβ3/αvβ5 integrins, is a proposed biomarker of tumor angiogenesis. The study rationale was to perform a preliminary evaluation of the relationship between tumor [(18)F]fluciclatide uptake and perfusion by [(15)O]H2O PET. METHODS Patients with non-small cell lung cancer and melanoma underwent dynamic imaging with arterial sampling following injection of [(15)O]H2O and [(18)F]fluciclatide. Quantification was performed using a one-tissue compartmental model for [(15)O]H2O and a two-tissue model for [(18)F]fluciclatide at volume-of-interest level, and SUV at voxel level. RESULTS Tumor binding potential (k 3/k 4 ratio) of [(18)F]fluciclatide tumor was 5.39 ± 1.46, consistent with previous studies in breast cancer metastases. Voxel-by-voxel maps of [(18)F]fluciclatide delivery strongly correlated with [(15)O]H2O-based perfusion (p < 10(-4) tumor, 1,794 ± 1,331 voxels). Interestingly, this correlation was lost when retention of [(18)F]fluciclatide at late time-points was compared with perfusion (p > 0.15). CONCLUSIONS Our study suggests tumor [(18)F]fluciclatide retention is unrelated to tumor perfusion, supporting use of late (60-min) imaging protocols in patients.
Collapse
Affiliation(s)
- Laura M Kenny
- />Department of Surgery and Cancer, Hammersmith Campus, Imperial College London, London, UK
- />Department of Medical Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Giampaolo Tomasi
- />Department of Surgery and Cancer, Hammersmith Campus, Imperial College London, London, UK
| | - Federico Turkheimer
- />Centre for Neuroimaging, Institute of Psychiatry, King’s College London, London UK
| | | | | | - Cathryn S Brock
- />Department of Medical Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Stephen Mangar
- />Department of Medical Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Eric O Aboagye
- />Department of Surgery and Cancer, Hammersmith Campus, Imperial College London, London, UK
| |
Collapse
|
202
|
Tsiapa I, Loudos G, Fragogeorgi EA, Bouziotis P, Psimadas D, Xanthopoulos S, Paravatou-Petsotas M, Palamaris L, Varvarigou AD, Karnabatidis D, Kagadis GC. Evaluation of ανβ3-mediated tumor expression with a 99mTc-labeled ornithine-modified RGD derivative during glioblastoma growth in vivo. Cancer Biother Radiopharm 2014; 29:444-50. [PMID: 25405951 DOI: 10.1089/cbr.2014.1672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this study, a novel way of distinguishing the intrinsic relationship between ανβ3 integrin targeting and detection of tumor growth by using a radiolabeled tracer based on a cyclic Arg-Gly-Asp (RGD) peptide was provided. The potential of the in vivo scintigraphic imaging of the developing vasculature from the early stage of tumor growth was evaluated. Alongside with the scintigraphic images, biodistribution studies were performed at distinct time points to validate this noninvasive imaging approach. The ability to noninvasively assess the tumor growth of ανβ3 integrin-positive glioblastoma tumors provides a method to better understand tumor angiogenesis in vivo and allows for a direct assessment of anti-integrin treatment efficacy.
Collapse
Affiliation(s)
- Irene Tsiapa
- 1 Department of Medical Physics, School of Medicine, University of Patras , Rion, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Davis PJ, Hercbergs A, Luidens MK, Lin HY. Recurrence of differentiated thyroid carcinoma during full TSH suppression: is the tumor now thyroid hormone dependent? Discov Oncol 2014; 6:7-12. [PMID: 25292307 PMCID: PMC4309911 DOI: 10.1007/s12672-014-0204-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 09/29/2014] [Indexed: 01/09/2023] Open
Abstract
Well-standardized primary treatment and long-term management of differentiated thyroid carcinoma (DTC) include lowering or suppression of host thyrotropin (TSH) with exogenous L-thyroxine (T4). This treatment recognizes the trophic action of TSH on DTC cells. Suppression of endogenous TSH with T4 is continued in recurrent disease. However, T4 can induce proliferation of follicular and papillary thyroid carcinoma cell lines and of other human carcinoma cells. The proliferative mechanism is initiated at a cell surface receptor for T4 on integrin αvβ3, a receptor by which the hormone also inhibits p53-dependent apoptosis in tumor cells. In recurrent DTC with satisfactory suppression of endogenous TSH, we discuss here the possibility that the tumor is no longer TSH dependent and that T4 has become a critical growth factor for the cancer.
Collapse
Affiliation(s)
- Paul J Davis
- Department of Medicine, Albany Medical College, Albany, NY, USA,
| | | | | | | |
Collapse
|
204
|
Thomaidis T, Maderer A, Formentini A, Bauer S, Trautmann M, Schwarz M, Neumann W, Kittner JM, Schad A, Link KH, Rey JW, Weinmann A, Hoffman A, Galle PR, Kornmann M, Moehler M. Proteins of the VEGFR and EGFR pathway as predictive markers for adjuvant treatment in patients with stage II/III colorectal cancer: results of the FOGT-4 trial. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:83. [PMID: 25272957 PMCID: PMC4192339 DOI: 10.1186/s13046-014-0083-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/24/2014] [Indexed: 12/12/2022]
Abstract
Background Unlike metastatic colorectal cancer (CRC) there are to date few reports concerning the predictive value of molecular biomarkers on the clinical outcome in stage II/III CRC patients receiving adjuvant chemotherapy. Aim of this study was to assess the predictive value of proteins related with the EGFR- and VEGFR- signalling cascades in these patients. Methods The patients' data examined in this study were from the collective of the 5-FU/FA versus 5-FU/FA/irinotecan phase III FOGT-4 trial. Tumor tissues were stained by immunohistochemistry for VEGF-C, VEGF-D, VEGFR-3, Hif-1 α, PTEN, AREG and EREG expression and evaluated by two independent, blinded investigators. Survival analyses were calculated for all patients receiving adjuvant chemotherapy in relation to expression of all makers above. Results Patients with negative AREG and EREG expression on their tumor had a significant longer DFS in comparison to AREG/EREG positive ones (p< 0.05). The benefit on DFS in AREG-/EREG- patients was even stronger in the group that received 5-FU/FA/irinotecan as adjuvant treatment (p=0.002). Patients with strong expression of PTEN profited more in terms of OS under adjuvant treatment containing irinotecan (p< 0.05). Regarding markers of the VEGFR- pathway we found no correlation of VEGF-C- and VEGFR-3 expression with clinical outcome. Patients with negative VEGF-D expression had a trend to live longer when treated with 5-FU/FA (p=0.106). Patients who were negative for Hif-1 α, were disease-free in more than 50% at the end of the study and showed significant longer DFS-rates than those positive for Hif-1 α (p=0.007). This benefit was even stronger at the group treated with 5-FU/FA/irinotecan (p=0.026). Finally, AREG-/EREG-/PTEN+ patients showed a trend to live longer under combined treatment combination. Conclusions The addition of irinotecan to adjuvant treatment with 5-FU/FA does not provide OS or DFS benefit in patients with stage II/III CRC. Nevertheless, AREG/EREG negative, PTEN positive and Hif-1 α negative patients might profit significantly in terms of DFS from a treatment containing fluoropyrimidines and irinotecan. Our results suggest a predictive value of these biomarkers concerning adjuvant chemotherapy with 5-FU/FA +/− irinotecan in stage II/III colorectal cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13046-014-0083-8) contains supplementary material, which is available to authorized users.
Collapse
|
205
|
|
206
|
Kim BR, Dong SM, Seo SH, Lee JH, Lee JM, Lee SH, Rho SB. Lysyl oxidase-like 2 (LOXL2) controls tumor-associated cell proliferation through the interaction with MARCKSL1. Cell Signal 2014; 26:1765-73. [DOI: 10.1016/j.cellsig.2014.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 05/15/2014] [Indexed: 01/25/2023]
|
207
|
Coller BS. The platelet: life on the razor's edge between hemorrhage and thrombosis. Transfusion 2014; 54:2137-46. [PMID: 25092268 DOI: 10.1111/trf.12806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 06/24/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Barry S Coller
- Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, New York
| |
Collapse
|
208
|
Tsiapa I, Efthimiadou EK, Fragogeorgi E, Loudos G, Varvarigou AD, Bouziotis P, Kordas GC, Mihailidis D, Nikiforidis GC, Xanthopoulos S, Psimadas D, Paravatou-Petsotas M, Palamaris L, Hazle JD, Kagadis GC. (99m)Tc-labeled aminosilane-coated iron oxide nanoparticles for molecular imaging of ανβ3-mediated tumor expression and feasibility for hyperthermia treatment. J Colloid Interface Sci 2014; 433:163-175. [PMID: 25128864 DOI: 10.1016/j.jcis.2014.07.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/22/2014] [Accepted: 07/23/2014] [Indexed: 12/18/2022]
Abstract
HYPOTHESIS Dual-modality imaging agents, such as radiolabeled iron oxide nanoparticles (IO-NPs), are promising candidates for cancer diagnosis and therapy. We developed and evaluated aminosilane coated Fe3O4 (10±2nm) as a tumor imaging agent in nuclear medicine through 3-aminopropyltriethoxysilane (APTES) functionalization. We evaluated this multimeric system of targeted (99m)Tc-labeled nanoparticles (NPs) conjugated with a new RGD derivate (cRGDfK-Orn3-CGG), characterized as NPs-RGD as a potential thermal therapy delivery vehicle. EXPERIMENTS Transmission Electron Microscopy (TEM) and spectroscopy techniques were used to characterize the IO-NPs indicating their functionalization with peptides. Radiolabeled IO-NPs (targeted, non-targeted) were evaluated with regard to their radiochemical, radiobiological and imaging characteristics. In vivo studies were performed in normal and ανβ3-positive tumor (U87MG glioblastoma) bearing mice. We also demonstrated that this system could reach ablative temperatures in vivo. FINDINGS Both radiolabeled IO-NPs were obtained in high radiochemical yield (>98%) and proved stable in vitro. The in vivo studies for both IO-NPs have shown significant liver and spleen uptake at all examined time points in normal and U87MG glioblastoma tumor-bearing mice, due to their colloidal nature. We have confirmed through in vivo biodistribution studies that the non-targeted (99m)Tc-NPs poorly internalized in the tumor, while the targeted (99m)Tc-NPs-RGD, present 9-fold higher tumor accumulation at 1h p.i. Accumulation of both IO-NPs in other organs was negligible. Blocking experiments indicated target specificity for integrin receptors in U87MG glioblastoma cells. The preliminary in vivo study of applied alternating magnetic field showed that the induced hyperthermia is feasible due to the aid of IO-NPs.
Collapse
Affiliation(s)
- Irene Tsiapa
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece; Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Eleni K Efthimiadou
- Sol-Gel Laboratory, Institute for Advanced Materials, Physicochemical Processes, Nanotechnology & Microsystems, NCSR "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Eirini Fragogeorgi
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece; Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - George Loudos
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece
| | - Alexandra D Varvarigou
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Penelope Bouziotis
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - George C Kordas
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece
| | | | - George C Nikiforidis
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece
| | - Stavros Xanthopoulos
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Dimitrios Psimadas
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece; Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Maria Paravatou-Petsotas
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Lazaros Palamaris
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece
| | - John D Hazle
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George C Kagadis
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece; Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
209
|
Liu H, Wu Y, Wang F, Liu Z. Molecular imaging of integrin αvβ6 expression in living subjects. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2014; 4:333-345. [PMID: 24982819 PMCID: PMC4074499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/15/2014] [Indexed: 06/03/2023]
Abstract
Integrins, a family of cell adhesion molecules composed of α and β heterodimeric subunits, are involved in a wide range of cell-extracellular matrix and cell-cell interactions. The study of integrin family members as targets for molecular imaging and therapy has been generally limited with the exception of integrin αvβ3. vβ6, a member of the integrin family, is expressed at low or undetectable levels in normal tissues, but is widely upregulated during many pathological and physiological processes, especially cancer and fibrosis, making it a promising target for molecular imaging. Noninvasive and quantitative imaging of integrin vβ6 expression would be very useful for disease diagnosis, treatment monitoring, and prognosis assessment. Although various molecular probes have been developed for positron emission tomography and single-photon emission computed tomography imaging of integrin vβ6 expression in preclinical animal models, further research efforts are required to optimize integrin vβ6-targeting probes for future potential clinical applications in the fields of oncology and beyond.
Collapse
Affiliation(s)
- Hao Liu
- Medical Isotopes Research Center, Peking UniversityBeijing 100191, China
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking UniversityBeijing 100191, China
| | - Yue Wu
- Medical Isotopes Research Center, Peking UniversityBeijing 100191, China
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking UniversityBeijing 100191, China
| | - Fan Wang
- Medical Isotopes Research Center, Peking UniversityBeijing 100191, China
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking UniversityBeijing 100191, China
| | - Zhaofei Liu
- Medical Isotopes Research Center, Peking UniversityBeijing 100191, China
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking UniversityBeijing 100191, China
| |
Collapse
|
210
|
Nanoparticle-formulated siRNA targeting integrins inhibits hepatocellular carcinoma progression in mice. Nat Commun 2014; 5:3869. [PMID: 24844798 PMCID: PMC4107318 DOI: 10.1038/ncomms4869] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/11/2014] [Indexed: 02/07/2023] Open
Abstract
Integrins play an important role during development, regulating cell differentiation, proliferation and survival. Here we show that knockdown of integrin subunits slows down the progression of hepatocellular carcinoma (HCC). Using nanoparticulate delivery of short interfering RNAs targeting β1 and αv integrin subunits we downregulate all integrin receptors in hepatocytes. Short-term integrin knockdown (two weeks) does not cause apparent structural or functional perturbations of normal liver tissue. Alterations in liver morphology accumulate upon sustained integrin downregulation (seven weeks). The integrin knockdown leads to significant retardation of HCC progression, reducing proliferation and increasing tumour cell death. This tumour retardation is accompanied by reduced activation of MET oncogene as well as expression of its mature form on the cell surface. Our data suggest that transformed proliferating cells from HCC are more sensitive to knockdown of integrins than normal quiescent hepatocytes, highlighting the potential of siRNA-mediated inhibition of integrins as an anti-cancer therapeutic approach.
Collapse
|
211
|
Abstract
Unlike other blood cells, platelets are small anucleate structures derived from marrow megakaryocytes. Thought for almost a century to possess solely hemostatic potentials, platelets, however, play a much wider role in tissue regeneration and repair and interact intimately with tumor cells. On one hand, tumor cells induce platelet aggregation (TCIPA), known to act as the trigger of cancer-associated thrombosis. On the other hand, platelets recruited to the tumor microenvironment interact, directly, with tumor cells, favoring their proliferation, and, indirectly, through the release of a wide palette of growth factors, including angiogenic and mitogenic proteins. In addition, the role of platelets is not solely confined to the primary tumor site. Indeed, they escort tumor cells, helping their intravasation, vascular migration, arrest, and extravasation to the tissues to form distant metastasis. As expected, nonspecific or specific inhibition of platelets and their content represents an attractive novel approach in the fight against cancer. This review illustrates the role played by platelets at primary tumor sites and in the various stages of the metastatic process.
Collapse
Affiliation(s)
- Hadi A Goubran
- Saskatoon Cancer Centre and Division of Oncology, Department of Medicine, College of Medicine, University of Saskatchewan, SK, Canada.
| | - Julie Stakiw
- Saskatoon Cancer Centre and Division of Oncology, Department of Medicine, College of Medicine, University of Saskatchewan, SK, Canada
| | | | - Thierry Burnouf
- Institute of Biomedical Materials and Tissue Engineering, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
212
|
Landowski TH, Gard J, Pond E, Pond GD, Nagle RB, Geffre CP, Cress AE. Targeting integrin α6 stimulates curative-type bone metastasis lesions in a xenograft model. Mol Cancer Ther 2014; 13:1558-66. [PMID: 24739392 DOI: 10.1158/1535-7163.mct-13-0962] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Laminin-binding integrin receptors are key mediators of epithelial cell migration and tumor metastasis. Recent studies have demonstrated a role for the α6 integrin (ITGA6/CD49f) in maintaining stem cell compartments within normal bone marrow and in residency of tumors metastatic to bone. In this study, we tested a function-blocking antibody specific for ITGA6, called J8H, to determine if preexisting cancer lesions in bone could be slowed and/or animal survival improved. Human prostate tumors were established by intracardiac injection into male SCID mice and treatment with J8H antibody was initiated after 1 week. Tumor progression was monitored by micro-computed tomography (CT) imaging of skeletal lesions. Animals that received weekly injections of the anti-ITGA6 antibody showed radiographic progression in only 40% of osseous tumors (femur or tibia), compared with control animals, where 80% of the lesions (femur or tibia) showed progression at 5 weeks. Kaplan-Meier survival analysis demonstrated a significant survival advantage for J8H-treated animals. Unexpectedly, CT image analysis revealed an increased proportion of bone lesions displaying a sclerotic rim of new bone formation, encapsulating the arrested lytic lesions in animals that received the anti-ITGA6 antibody treatment. Histopathology of the sclerotic lesions demonstrated well-circumscribed tumor within bone, surrounded by fibrosis. These data suggest that systemic targeting of the ITGA6-dependent function of established tumors in bone may offer a noncytotoxic approach to arrest the osteolytic progression of metastatic prostate cancer, thereby providing a new therapeutic strategy for advanced disease.
Collapse
Affiliation(s)
- Terry H Landowski
- Authors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, ArizonaAuthors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona
| | - Jaime Gard
- Authors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona
| | - Erika Pond
- Authors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona
| | - Gerald D Pond
- Authors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona
| | - Raymond B Nagle
- Authors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, ArizonaAuthors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona
| | - Christopher P Geffre
- Authors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona
| | - Anne E Cress
- Authors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, ArizonaAuthors' Affiliations: University of Arizona Cancer Center; Departments of Medicine, Medical Imaging, Pathology, and Cellular and Molecular Medicine, University of Arizona Cancer Center, Tucson, Arizona
| |
Collapse
|
213
|
Ha SY, Shin J, Kim JH, Kang MS, Yoo HY, Kim HH, Um SH, Kim SH. Overexpression of integrin αv correlates with poor prognosis in colorectal cancer. J Clin Pathol 2014; 67:576-81. [PMID: 24695839 DOI: 10.1136/jclinpath-2013-202163] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIMS Integrin αv subunits are involved in tumour angiogenesis and tumour progression in various types of cancers. Clinical trials evaluating agents targeting integrin αv are ongoing. Integrin αv expression has been reported in several cancers in association with tumour progression or poor survival. However, no study has addressed the prognostic influence of integrin αv expression on survival of patients with colorectal cancer (CRC). METHODS Immunohistochemical staining of integrin αv was performed in 198 CRC samples to evaluate its prognostic significance. RESULTS High expression of integrin αv was observed in 58.1% (115/189) of colorectal adenocarcinoma samples, while only in 11.5% (3/26) of tubular adenoma samples and in none of normal mucosa or hyperplastic polyp samples. It was more frequently found in female patients and less frequently observed in well differentiated tumours. The proportion of cases with high expression of integrin αv showed an increasing trend with increased T stage (p=0.032), N stage (p=0.006) and TNM stage (p=0.001). Patients displaying exuberant expression of integrin αv showed shorter overall survival (p=0.001) and disease-free survival (p=0.004). Elevated integrin αv expression was an independent prognostic factor for overall survival (HR: 2.04, 95% CI 1.16 to 3.56; p=0.013) and disease-free survival (HR: 2.19, 95% CI 1.16 to 4.13; p=0.015). CONCLUSIONS Overexpression of integrin αv is associated with advanced T and N stage and as an independent prognostic factor in CRC.
Collapse
Affiliation(s)
- Sang Yun Ha
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Juyoun Shin
- Division of Cancer Biology, Comparative Biomedicine Research Branch, Research Institute of National Cancer Center, Goyang, Korea
| | - Jeong Hoon Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung Soo Kang
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hae-Yong Yoo
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyeon-Ho Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung-Hee Um
- Department of Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Seok-Hyung Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea Department of Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
214
|
Mahalingam B, Van Agthoven JF, Xiong JP, Alonso JL, Adair BD, Rui X, Anand S, Mehrbod M, Mofrad MRK, Burger C, Goodman SL, Arnaout MA. Atomic basis for the species-specific inhibition of αV integrins by monoclonal antibody 17E6 is revealed by the crystal structure of αVβ3 ectodomain-17E6 Fab complex. J Biol Chem 2014; 289:13801-9. [PMID: 24692540 DOI: 10.1074/jbc.m113.546929] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The function-blocking, non-RGD-containing, and primate-specific mouse monoclonal antibody 17E6 binds the αV subfamily of integrins. 17E6 is currently in phase II clinical trials for treating cancer. To elucidate the structural basis of recognition and the molecular mechanism of inhibition, we crystallized αVβ3 ectodomain in complex with the Fab fragment of 17E6. Protein crystals grew in presence of the activating cation Mn(2+). The integrin in the complex and in solution assumed the genuflected conformation. 17E6 Fab bound exclusively to the Propeller domain of the αV subunit. At the core of αV-Fab interface were interactions involving Propeller residues Lys-203 and Gln-145, with the latter accounting for primate specificity. The Propeller residue Asp-150, which normally coordinates Arg of the ligand Arg-Gly-Asp motif, formed contacts with Arg-54 of the Fab that were expected to reduce soluble FN10 binding to cellular αVβ3 complexed with 17E6. This was confirmed in direct binding studies, suggesting that 17E6 is an allosteric inhibitor of αV integrins.
Collapse
Affiliation(s)
| | | | | | - José Luis Alonso
- the Leukocyte Biology and Inflammation Program, Departments of Medicine and Developmental & Regenerative Biology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | | | - Xianliang Rui
- the Leukocyte Biology and Inflammation Program, Departments of Medicine and Developmental & Regenerative Biology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - Saurabh Anand
- the Leukocyte Biology and Inflammation Program, Departments of Medicine and Developmental & Regenerative Biology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - Mehrdad Mehrbod
- the Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California 94720
| | - Mohammad R K Mofrad
- the Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California 94720
| | - Christa Burger
- Merck KGaA and Discovery Technologies, Molecular Pharmacology, and
| | - Simon L Goodman
- Merck KGaA and Therapeutic Innovation Platform, Oncology, Darmstadt 64271, Germany
| | - M Amin Arnaout
- From the Structural Biology Program and the Leukocyte Biology and Inflammation Program, Departments of Medicine and Developmental & Regenerative Biology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129,
| |
Collapse
|
215
|
Correlation of breast cancer subtypes, based on estrogen receptor, progesterone receptor, and HER2, with functional imaging parameters from ⁶⁸Ga-RGD PET/CT and ¹⁸F-FDG PET/CT. Eur J Nucl Med Mol Imaging 2014; 41:1534-43. [PMID: 24652232 DOI: 10.1007/s00259-014-2744-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/21/2014] [Indexed: 12/19/2022]
Abstract
PURPOSE Imaging biomarkers from functional imaging modalities were assessed as potential surrogate markers of disease status. Specifically, in this prospective study, we investigated the relationships between functional imaging parameters and histological prognostic factors and breast cancer subtypes. METHODS In total, 43 patients with large or locally advanced invasive ductal carcinoma (IDC) were analyzed (47.6 ± 7.5 years old). (68)Ga-Labeled arginine-glycine-aspartic acid (RGD) and (18)F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) were performed. The maximum and average standardized uptake values (SUVmax and SUVavg) from RGD PET/CT and SUVmax and SUVavg from FDG PET/CT were the imaging parameters used. For histological prognostic factors, estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression was identified using immunohistochemistry (IHC) or fluorescent in situ hybridization (FISH). Four breast cancer subtypes, based on ER/PR and HER2 expression (ER/PR+,Her2-, ER/PR+,Her2+, ER/PR-,Her2+, and ER/PR-,Her2-), were considered. RESULTS Quantitative FDG PET parameters were significantly higher in the ER-negative group (15.88 ± 8.73 vs 10.48 ± 6.01, p = 0.02 for SUVmax; 9.40 ± 5.19 vs 5.92 ± 4.09, p = 0.02 for SUVavg) and the PR-negative group (8.37 ± 4.94 vs 4.79 ± 3.93, p = 0.03 for SUVavg). Quantitative RGD PET parameters were significantly higher in the HER2-positive group (2.42 ± 0.59 vs 2.90 ± 0.75, p = 0.04 for SUVmax; 1.60 ± 0.38 vs 1.95 ± 0.53, p = 0.04 for SUVavg) and showed a significant positive correlation with the HER2/CEP17 ratio (r = 0.38, p = 0.03 for SUVmax and r = 0.46, p < 0.01 for SUVavg). FDG PET parameters showed significantly higher values in the ER/PR-,Her2- subgroup versus the ER/PR+,Her2- or ER/PR+,Her2+ subgroups, while RGD PET parameters showed significantly lower values in the ER/PR-,Her2- subgroup versus the other subgroups. There was no correlation between FDG and RGD PET parameters in the overall group. Only the ER/PR-,Her2- subgroup showed a significant positive correlation between FDG and RGD PET parameters (r = 0.59, p = 0.03 for SUVmax). CONCLUSION (68)Ga-RGD and (18)F-FDG PET/CT are promising functional imaging modalities for predicting biomarkers and molecular phenotypes in breast cancer patients.
Collapse
|
216
|
Fitzpatrick P, Shattil SJ, Ablooglu AJ. C-terminal COOH of integrin β1 is necessary for β1 association with the kindlin-2 adapter protein. J Biol Chem 2014; 289:11183-11193. [PMID: 24599960 DOI: 10.1074/jbc.m113.535369] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein-protein interactions are driving forces in cellular processes. As a prime example, transmembrane integrins link extracellular matrix and intracellular proteins, resulting in bidirectional signaling that regulates cell migration, proliferation, differentiation, and survival. Here we provide the first evidence that interaction between the integrin β1 cytoplasmic tail and kindlin-2, a member of a family of adapters implicated in human disease pathogenesis, is mainly governed by the β1 C-terminal carboxylate moiety and is required for laterality organ development in zebrafish. Affinity measurements indicate that this unusual protein-protein interaction mode is coordinated by a putative carboxylate-binding motif in the kindlin-2 FERM subdomain F3. Contrary to the C terminus of proteins that engage PDZ domains, the C-terminal three residues of β1, per se, do not contribute to kindlin-2 binding or to laterality organ development. Thus, by employing zebrafish as an in situ physiological tool to correlate protein structure and function, we have discovered an unexpected association chemistry between an integrin and a key adapter involved in integrin signaling.
Collapse
Affiliation(s)
- Paul Fitzpatrick
- Department of Medicine, University of California San Diego, La Jolla, California 92093-0726
| | - Sanford J Shattil
- Department of Medicine, University of California San Diego, La Jolla, California 92093-0726
| | - Ararat J Ablooglu
- Department of Medicine, University of California San Diego, La Jolla, California 92093-0726.
| |
Collapse
|
217
|
Suppression of tumour growth by orally administered osteopontin is accompanied by alterations in tumour blood vessels. Br J Cancer 2014; 110:1269-77. [PMID: 24473400 PMCID: PMC3950862 DOI: 10.1038/bjc.2014.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/11/2013] [Accepted: 12/13/2013] [Indexed: 12/16/2022] Open
Abstract
Background: The integrin-binding protein osteopontin is strongly associated with tumour development, yet is an abundant dietary component as a constituent of human and bovine milk. Therefore, we tested the effect of orally administered osteopontin (o-OPN) on the development of subcutaneous tumours in mice. Methods: Bovine milk osteopontin was administered in drinking water to tumour-bearing immune-competent mice. Tumour growth, proliferation, necrosis, apoptosis and blood vessel size and number were measured. Expression of the α9 integrin was determined. Results: o-OPN suppressed tumour growth, increased the extent of necrosis, and induced formation of abnormally large blood vessels. Anti-OPN reactivity detected in the plasma of OPN-null mice fed OPN suggested that tumour-blocking peptides were absorbed during digestion, but the o-OPN effect was likely distinct from that of an RGD peptide. Expression of the α9 integrin was detected on both tumour cells and blood vessels. Potential active peptides from the α9 binding site of OPN were identified by mass spectrometry following in vitro digestion, and injection of these peptides suppressed tumour growth. Conclusions: These results suggest that peptides derived from o-OPN are absorbed and interfere with tumour growth and normal vessel development. o-OPN-derived peptides that target the α9 integrin are likely involved.
Collapse
|
218
|
Terry SYA, Abiraj K, Frielink C, van Dijk LK, Bussink J, Oyen WJ, Boerman OC. Imaging integrin αvβ3 on blood vessels with 111In-RGD2 in head and neck tumor xenografts. J Nucl Med 2014; 55:281-6. [PMID: 24408894 DOI: 10.2967/jnumed.113.129668] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Arginine-glycine-aspartic acid (RGD)-based imaging tracers allow specific imaging of integrin αvβ3, a protein overexpressed during angiogenesis, leading to the possibility that it might serve as a tool to stratify patients for antiangiogenic treatment. However, these tracers have generally been characterized in xenograft models in which integrin αvβ3 was constitutively expressed by the tumor cells themselves. In the studies presented here, the use of (111)In-RGD2 as a tracer to image only integrin αvβ3 expression on blood vessels in the tumor was determined using tumor xenografts in which tumor cells were integrin αvβ3-negative. METHODS DOTA-E-[c(RGDfK)]2 was radiolabeled with (111)In ((111)In-RGD2), and biodistribution studies were performed in squamous cell carcinoma of the head and neck (HNSCC) xenograft mouse models to determine the optimal peptide dose to image angiogenesis. Next, biodistribution and imaging studies were performed at the optimal peptide dose in 3 HNSCC mouse models, FaDu, SCCNij3, and SCCNij202. Immunohistochemical analysis of tumor vascular and cell surface expression of integrin αvβ3 and correlation analysis of vascular integrin αvβ3 and autoradiography were completed. RESULTS All 3 HNSCC xenografts expressed integrin αvβ3 on the vessels only. The optimal peptide dose of (111)In-RGD2 was 1 μg or less for specific integrin αvβ3-mediated uptake of the tracer. SPECT/CT imaging showed clear uptake of the tracer in the periphery of the tumors, corresponding with well-vascularized areas of the tumor. Within the tumor, (111)In-RGD2 autoradiography coincided with vascular integrin αvβ3 expression, as determined immunohistochemically. Integrin αvβ3-mediated uptake was also detected in nontumor tissues, which, through immunohistochemical analysis, proved positive for integrin αvβ3. CONCLUSION (111)In-RGD2 allows the visualization of integrin αvβ3 in xenograft models in which integrin αvβ3 is expressed only on the neovasculature, such as in the HNSCC tumors. Thus, (111)In-RGD2 allows specific visualization of angiogenesis in tumor models lacking constitutive tumoral integrin αvβ3 expression but may be less useful for this purpose in many tumors in which tumor cells express integrin αvβ3.
Collapse
|
219
|
Steri V, Ellison TS, Gontarczyk AM, Weilbaecher K, Schneider JG, Edwards D, Fruttiger M, Hodivala-Dilke KM, Robinson SD. Acute depletion of endothelial β3-integrin transiently inhibits tumor growth and angiogenesis in mice. Circ Res 2014; 114:79-91. [PMID: 24103390 DOI: 10.1161/circresaha.114.301591] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 10/08/2013] [Indexed: 11/16/2022]
Abstract
RATIONALE The dramatic upregulation of αvβ3-integrin that occurs in the vasculature during tumor growth has long suggested that the endothelial expression of this molecule is an ideal target for antiangiogenic therapy to treat cancer. This discovery led to the development of small-molecule inhibitors directed against αvβ3-integrin that are currently in clinical trials. In 2002, we reported that β3-integrin-knockout mice exhibit enhanced tumor growth and angiogenesis. However, as β3-integrin is expressed by a wide variety of cells, endothelial cell-specific contributions to tumor angiogenesis are muddied by the use of a global knockout of β3-integrin function. OBJECTIVE Our aim was to examine the endothelial-specific contribution β3-integrin makes to tumor growth and angiogenesis. METHODS AND RESULTS We have crossed β3-integrin-floxed (β3-floxed) mice to 2 endothelial-specific Cre models and examined angiogenic responses in vivo, ex vivo, and in vitro. We show that acute depletion of endothelial β3-integrin inhibits tumor growth and angiogenesis preventatively, but not in already established tumors. However, the effects are transient, and long-term depletion of the molecule is ineffective. Furthermore, long-term depletion of the molecule correlates with many molecular changes, such as reduced levels of focal adhesion kinase expression and a misbalance in focal adhesion kinase phosphorylation, which may lead to a release from the inhibitory effects of decreased endothelial β3-integrin expression. CONCLUSIONS Our findings imply that timing and length of inhibition are critical factors that need to be considered when targeting the endothelial expression of β3-integrin to inhibit tumor growth and angiogenesis.
Collapse
Affiliation(s)
- Veronica Steri
- From the School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (V.S., T.S.E., A.M.G., D.E., S.D.R.); Department of Internal Medicine, Division of Molecular Oncology, Washington University in St Louis, MO (K.W.); Luxembourg Center for Systems Biomedicine, University of Luxembourg, Luxembourg and Saarland University Medical Center, Internal Medicine II, Homburg, Germany (J.G.S.); UCL Institute of Ophthalmology, University College London, London, United Kingdom (M.F.); Barts Cancer Institute - a Cancer Research UK Centre of Excellence, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London, United Kingdom (K.M.H.-D.)
| | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Copper-free azide–alkyne cycloaddition of targeting peptides to porous silicon nanoparticles for intracellular drug uptake. Biomaterials 2014; 35:1257-66. [DOI: 10.1016/j.biomaterials.2013.10.065] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/22/2013] [Indexed: 01/07/2023]
|
221
|
Berezin V, Walmod PS, Filippov M, Dityatev A. Targeting of ECM molecules and their metabolizing enzymes and receptors for the treatment of CNS diseases. PROGRESS IN BRAIN RESEARCH 2014; 214:353-88. [DOI: 10.1016/b978-0-444-63486-3.00015-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
222
|
Transcriptional targeting of primary and metastatic tumor neovasculature by an adenoviral type 5 roundabout4 vector in mice. PLoS One 2013; 8:e83933. [PMID: 24376772 PMCID: PMC3871592 DOI: 10.1371/journal.pone.0083933] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 11/10/2013] [Indexed: 11/25/2022] Open
Abstract
New approaches targeting metastatic neovasculature are needed. Payload capacity, cellular transduction efficiency, and first-pass cellular uptake following systemic vector administration, motivates persistent interest in tumor vascular endothelial cell (EC) adenoviral (Ad) vector targeting. While EC transductional and transcriptional targeting has been accomplished, vector administration approaches of limited clinical utility, lack of tumor-wide EC expression quantification, and failure to address avid liver sequestration, challenged prior work. Here, we intravenously injected an Ad vector containing 3 kb of the human roundabout4 (ROBO4) enhancer/promoter transcriptionally regulating an enhanced green fluorescent protein (EGFP) reporter into immunodeficient mice bearing 786-O renal cell carcinoma subcutaneous (SC) xenografts and kidney orthotopic (KO) tumors. Initial experiments performed in human coxsackie virus and adenovirus receptor (hCAR) transgenic:Rag2 knockout mice revealed multiple ECs with high-level Ad5ROBO4-EGFP expression throughout KO and SC tumors. In contrast, Ad5CMV-EGFP was sporadically expressed in a few tumor vascular ECs and stromal cells. As the hCAR transgene also facilitated Ad5ROBO4 and control Ad5CMV vector EC expression in multiple host organs, follow-on experiments engaged warfarin-mediated liver vector detargeting in hCAR non-transgenic mice. Ad5ROBO4-mediated EC expression was undetectable in most host organs, while the frequencies of vector expressing intratumoral vessels and whole tumor EGFP protein levels remained elevated. In contrast, AdCMV vector expression was only detectable in one or two stromal cells throughout the whole tumor. The Ad5ROBO4 vector, in conjunction with liver detargeting, provides tractable genetic access for in-vivo EC genetic engineering in malignancies.
Collapse
|
223
|
Abstract
During metastasis, cancer cells disseminate to other parts of the body by entering the bloodstream in a process that is called intravasation. They then extravasate at metastatic sites by attaching to endothelial cells that line blood vessels and crossing the vessel walls of tissues or organs. This Review describes how cancer cells cross the endothelial barrier during extravasation and how different receptors, signalling pathways and circulating cells such as leukocytes and platelets contribute to this process. Identification of the mechanisms that underlie cancer cell extravasation could lead to the development of new therapies to reduce metastasis.
Collapse
Affiliation(s)
- Nicolas Reymond
- 1] Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK. [2] Centre de Recherche de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique (CNRS) - UMR5237, 1919 Route de Mende, 34293 Montpellier, Cedex 5, France. [3]
| | | | | |
Collapse
|
224
|
Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev 2013; 65:1866-79. [PMID: 24120656 PMCID: PMC5812459 DOI: 10.1016/j.addr.2013.09.019] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 09/29/2013] [Accepted: 09/30/2013] [Indexed: 12/27/2022]
Abstract
Nanomedicine is an emerging form of therapy that focuses on alternative drug delivery and improvement of the treatment efficacy while reducing detrimental side effects to normal tissues. Cancer drug resistance is a complicated process that involves multiple mechanisms. Here we discuss the major forms of drug resistance and the new possibilities that nanomedicines offer to overcome these treatment obstacles. Novel nanomedicines that have a high ability for flexible, fast drug design and production based on tumor genetic profiles can be created making drug selection for personal patient treatment much more intensive and effective. This review aims to demonstrate the advantage of the young medical science field, nanomedicine, for overcoming cancer drug resistance. With the advanced design and alternative mechanisms of drug delivery known for different nanodrugs including liposomes, polymer conjugates, micelles, dendrimers, carbon-based, and metallic nanoparticles, overcoming various forms of multi-drug resistance looks promising and opens new horizons for cancer treatment.
Collapse
Affiliation(s)
- Janet L Markman
- Nanomedicine Research Center, Department of Neurosurgery at Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | | | | | |
Collapse
|
225
|
El-Kenawi AE, El-Remessy AB. Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales. Br J Pharmacol 2013; 170:712-29. [PMID: 23962094 PMCID: PMC3799588 DOI: 10.1111/bph.12344] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/25/2013] [Accepted: 07/30/2013] [Indexed: 12/17/2022] Open
Abstract
Angiogenesis, a process of new blood vessel formation, is a prerequisite for tumour growth to supply the proliferating tumour with oxygen and nutrients. The angiogenic process may contribute to tumour progression, invasion and metastasis, and is generally accepted as an indicator of tumour prognosis. Therefore, targeting tumour angiogenesis has become of high clinical relevance. The current review aimed to highlight mechanistic details of anti-angiogenic therapies and how they relate to classification and treatment rationales. Angiogenesis inhibitors are classified into either direct inhibitors that target endothelial cells in the growing vasculature or indirect inhibitors that prevent the expression or block the activity of angiogenesis inducers. The latter class extends to include targeted therapy against oncogenes, conventional chemotherapeutic agents and drugs targeting other cells of the tumour micro-environment. Angiogenesis inhibitors may be used as either monotherapy or in combination with other anticancer drugs. In this context, many preclinical and clinical studies revealed higher therapeutic effectiveness of the combined treatments compared with individual treatments. The proper understanding of synergistic treatment modalities of angiogenesis inhibitors as well as their wide range of cellular targets could provide effective tools for future therapies of many types of cancer.
Collapse
Affiliation(s)
- Asmaa E El-Kenawi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura UniversityMansoura, Egypt
| | - Azza B El-Remessy
- Center for Pharmacy and Experimental Therapeutics, University of GeorgiaAugusta, GA, USA
- Department of Pharmacology and Toxicology, Georgia Regents UniversityAugusta, GA, USA
- Charlie Norwood VA Medical CenterAugusta, GA, USA
| |
Collapse
|
226
|
Mackow ER, Gorbunova EE, Dalrymple NA, Gavrilovskaya IN. Role of vascular and lymphatic endothelial cells in hantavirus pulmonary syndrome suggests targeted therapeutic approaches. Lymphat Res Biol 2013; 11:128-35. [PMID: 24024573 DOI: 10.1089/lrb.2013.0006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Hantaviruses in the Americas cause a highly lethal acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). Hantaviruses nonlytically infect microvascular and lymphatic endothelial cells and cause dramatic changes in barrier functions without disrupting the endothelium. Hantaviruses cause changes in the function of infected endothelial cells that normally regulate fluid barrier functions. The endothelium of arteries, veins, and lymphatic vessels are unique and central to the function of vast pulmonary capillary beds that regulate pulmonary fluid accumulation. RESULTS We have found that HPS-causing hantaviruses alter vascular barrier functions of microvascular and lymphatic endothelial cells by altering receptor and signaling pathway responses that serve to permit fluid tissue influx and clear tissue edema. Infection of the endothelium provides several mechanisms for hantaviruses to cause acute pulmonary edema, as well as potential therapeutic targets for reducing the severity of HPS disease. CONCLUSIONS Here we discuss interactions of HPS-causing hantaviruses with the endothelium, roles for unique lymphatic endothelial responses in HPS, and therapeutic targeting of the endothelium as a means of reducing the severity of HPS disease.
Collapse
Affiliation(s)
- Erich R Mackow
- Department of Molecular Genetics and Microbiology, Stony Brook University , Stony Brook, New York
| | | | | | | |
Collapse
|
227
|
Marelli UK, Rechenmacher F, Sobahi TRA, Mas-Moruno C, Kessler H. Tumor Targeting via Integrin Ligands. Front Oncol 2013; 3:222. [PMID: 24010121 PMCID: PMC3757457 DOI: 10.3389/fonc.2013.00222] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/13/2013] [Indexed: 01/02/2023] Open
Abstract
Selective and targeted delivery of drugs to tumors is a major challenge for an effective cancer therapy and also to overcome the side-effects associated with current treatments. Overexpression of various receptors on tumor cells is a characteristic structural and biochemical aspect of tumors and distinguishes them from physiologically normal cells. This abnormal feature is therefore suitable for selectively directing anticancer molecules to tumors by using ligands that can preferentially recognize such receptors. Several subtypes of integrin receptors that are crucial for cell adhesion, cell signaling, cell viability, and motility have been shown to have an upregulated expression on cancer cells. Thus, ligands that recognize specific integrin subtypes represent excellent candidates to be conjugated to drugs or drug carrier systems and be targeted to tumors. In this regard, integrins recognizing the RGD cell adhesive sequence have been extensively targeted for tumor-specific drug delivery. Here we review key recent examples on the presentation of RGD-based integrin ligands by means of distinct drug-delivery systems, and discuss the prospects of such therapies to specifically target tumor cells.
Collapse
Affiliation(s)
- Udaya Kiran Marelli
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , Garching , Germany
| | | | | | | | | |
Collapse
|
228
|
Berberine Reduces the Metastasis of Chondrosarcoma by Modulating the α v β 3 Integrin and the PKC δ , c-Src, and AP-1 Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:423164. [PMID: 24027594 PMCID: PMC3763569 DOI: 10.1155/2013/423164] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 07/16/2013] [Indexed: 02/06/2023]
Abstract
Chondrosarcoma is a primary malignant bone cancer, with a potent capacity to invade locally and cause distant metastasis, especially to the lungs. Patients diagnosed with chondrosarcoma have poor prognosis. Berberine, an active component of the Ranunculaceae and Papaveraceae families of plant, has been proven to induce tumor apoptosis and to prevent the metastasis of cancer cells. However, the effects of berberine in human chondrosarcoma are largely unknown. In this study, we found that berberine did not induce cell apoptosis in human primary chondrocytes and chondrosarcoma cells. However, at noncytotoxic concentrations, berberine reduced the migration and invasion of chondrosarcoma cancer cells. Integrins are the major adhesive molecules in mammalian cells and have been associated with the metastasis of cancer cells. We also found that incubation of chondrosarcoma cells with berberine reduced mRNA transcription for, and cell surface expression of, the αvβ3 integrin, with additional inhibitory effects on PKCδ, c-Src, and NF-κB activation. Thus, berberine may be a novel antimetastasis agent for the treatment of metastatic chondrosarcoma.
Collapse
|
229
|
Mullins CS, Schubert J, Schneider B, Linnebacher M, Classen CF. Cilengitide response in ultra-low passage glioblastoma cell lines: relation to molecular markers. J Cancer Res Clin Oncol 2013; 139:1425-31. [PMID: 23749036 DOI: 10.1007/s00432-013-1457-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 05/25/2013] [Indexed: 01/30/2023]
Abstract
PURPOSE In glioblastoma multiforme (GBM), a tumor still characterized by dismal prognosis, recent research focuses on novel-targeted compounds, in addition to standard temozolomide (TMZ) chemotherapy. One of these emerging compounds is cilengitide (CGT), which by binding to integrins (i.e., αvβ3 and αvβ5) may inhibit angiogenesis and also is directly cytotoxic to tumor cells by interfering with intracellular signaling pathways. METHODS A total of ten patient-derived ultra-low passage GBM cell lines were treated with increasing doses of CGT, TMZ, and a combination of both substances. Inhibitory concentrations of 50% (IC₅₀) were determined for the single agents and as a combination. Cell lines were stratified according to MGMT promoter methylation. The expression of relevant integrins was assessed by flow cytometry. RESULTS In monotherapy, all GBM cell lines showed higher sensitivity to CGT than to TMZ, as determined by IC₅₀ values in relation to clinically relevant patient plasma levels. MGMT promoter methylation correlated with a significantly higher TMZ response, but tended to be associated with a lower CGT response. Response to CGT was not correlated with cell surface integrin expression as measured by flow cytometry. Finally, addition of CGT to TMZ enhanced growth inhibition, but only in those cell lines with a methylated MGMT promoter. CONCLUSIONS As suggested by this analysis, patients with MGMT promoter-methylated GBM may benefit from addition of CGT to the standard TMZ treatment, while patients with MGMT promoter-unmethylated GBM may better respond to CGT monotherapy.
Collapse
Affiliation(s)
- Christina S Mullins
- University Children's Hospital, University Medicine, Ernst-Heydemann-Straße 8, 18057 Rostock, Germany.
| | | | | | | | | |
Collapse
|
230
|
The promotion of endothelial cell attachment and spreading using FNIII10 fused to VEGF-A165. Biomaterials 2013; 34:5958-68. [DOI: 10.1016/j.biomaterials.2013.04.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/24/2013] [Indexed: 11/20/2022]
|
231
|
Lin CY, Chen HJ, Li TM, Fong YC, Liu SC, Chen PC, Tang CH. β5 integrin up-regulation in brain-derived neurotrophic factor promotes cell motility in human chondrosarcoma. PLoS One 2013; 8:e67990. [PMID: 23874483 PMCID: PMC3706611 DOI: 10.1371/journal.pone.0067990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/24/2013] [Indexed: 01/22/2023] Open
Abstract
Chondrosarcoma is a primary malignant bone cancer, with a potent capacity to invade locally and cause distant metastasis; it has a poor prognosis and shows a predilection for metastasis to the lungs. Brain derived neurotrophic factor (BDNF) is a small-molecule protein from the neurotrophin family of growth factors that is associated with the disease status and outcomes of cancers. However, the effect of BDNF on migration activity in human chondrosarcoma cells is mostly unknown. Here, we found that human chondrosarcoma tissues showed significant expression of BDNF, which was higher than that in normal cartilage and primary chondrocytes. We also found that BDNF increased the migration and expression of β5 integrin in human chondrosarcoma cells. In addition, knockdown of BDNF expression markedly inhibited migratory activity. BDNF-mediated migration and β5 integrin up-regulation were attenuated by antibody, inhibitor, or siRNA against the TrkB receptor. Pretreatment of chondrosarcoma cells with PI3K, Akt, and NF-κB inhibitors or mutants also abolished BDNF-promoted migration and integrin expression. The PI3K, Akt, and NF-κB signaling pathway was activated after BDNF treatment. Taken together, our results indicate that BDNF enhances the migration of chondrosarcoma by increasing β5 integrin expression through a signal transduction pathway that involves the TrkB receptor, PI3K, Akt, and NF-κB. BDNF thus represents a promising new target for treating chondrosarcoma metastasis.
Collapse
Affiliation(s)
- Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Hui-Jye Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Molecular Systems Biomedicine, China Medical University, Taichung, Taiwan
| | - Te-Mao Li
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Chin Fong
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopaedics, China Medical University Hospital, Taichung, Taiwan
| | - Shan-Chi Liu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Po-Chun Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
232
|
[68Ga]NS3-RGD and [68Ga] Oxo-DO3A-RGD for imaging αvβ3 integrin expression: synthesis, evaluation, and comparison. Nucl Med Biol 2013; 40:65-72. [DOI: 10.1016/j.nucmedbio.2012.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 09/15/2012] [Accepted: 09/20/2012] [Indexed: 11/24/2022]
|
233
|
Sun W. Angiogenesis in metastatic colorectal cancer and the benefits of targeted therapy. J Hematol Oncol 2012; 5:63. [PMID: 23057939 PMCID: PMC3537532 DOI: 10.1186/1756-8722-5-63] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 09/26/2012] [Indexed: 02/08/2023] Open
Abstract
The diverse pathways and molecules involved in angiogenesis, the formation of new blood vessels, have been targeted for the treatment of colorectal and other cancers. Vascular endothelial growth factor (VEGF)-A binding to VEGF receptor (VEGFR)-2 is believed to be the key signaling pathway mediating angiogenesis. Other VEGF pathways involved in angiogenesis include VEGF-A, VEGF-B, and placental growth factor binding to VEGFR-1, and VEGF-C and VEGF-D binding to VEGFR-2 and VEGFR-3. VEGF signaling also intersects with other pathways, including angiopoietin/Tie, Notch, hypoxia-inducible factor, and integrin pathways. The roles of these pathways in tumor angiogenesis and in various human cancers will be explored in this article. In addition, preclinical and clinical data on bevacizumab, aflibercept (known as ziv-aflibercept in the US), and investigational antiangiogenic agents in development for the treatment of colorectal and other cancers will be reviewed.
Collapse
Affiliation(s)
- Weijing Sun
- University of Pittsburgh School of Medicine, UPMC Cancer Pavilion, 5150 Centre Avenue, Fifth Floor, Pittsburgh, PA 15232, USA.
| |
Collapse
|
234
|
Anti-angiogenic effects of thioridazine involving the FAK-mTOR pathway. Microvasc Res 2012; 84:227-34. [PMID: 23022044 DOI: 10.1016/j.mvr.2012.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/27/2012] [Accepted: 09/19/2012] [Indexed: 11/24/2022]
Abstract
Thioridazine is a type of anti-psychotic drug that also includes anti-tumor activity. In this study, we assessed the effects of thioridazine, as a novel anti-angiogenic agent, on the suppression of angiogenesis-mediated cell proliferation. Thioridazine was found to inhibit growth in ovarian cancer cells (OVCAR-3 and 2774), but did not possess any inhibitory effects on normal cell types such as HOSE-E6E7, MCF-10A, MRC-5, and BEAS-2B. Thioridazine also suppressed vascular endothelial growth factor (VEGF)-stimulated HUVEC migration in a dose-time-dependent manner. We also showed that being treated with thioridazine inhibited VEGF-stimulated proliferation, invasion, and capillary-like structure tube formation in vitro. Thioridazine suppressed phosphorylation of the signaling regulators downstream of the focal adhesion kinase (FAK) through αvβ3 integrin, which also include Akt, phosphoinositide-dependent protein kinase 1 (PDK-1), mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase (p70S6K), but had no effect on VEGF-stimulated extracellular signal-regulated kinase (ERK) phosphorylation. We found the molecular mechanism of thioridazine to be a novel anti-angiogenic protein. These results provide evidence for the regulation of endothelial cell functions that are relevant to angiogenesis through the suppression of the αvβ3/FAK/mTOR signaling pathway.
Collapse
|
235
|
The Role of the Endothelium in HPS Pathogenesis and Potential Therapeutic Approaches. Adv Virol 2012; 2012:467059. [PMID: 22811711 PMCID: PMC3395186 DOI: 10.1155/2012/467059] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 05/16/2012] [Accepted: 05/18/2012] [Indexed: 02/07/2023] Open
Abstract
American hantaviruses cause a highly lethal acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). Hantaviruses nonlytically infect endothelial cells and cause dramatic changes in barrier functions of the endothelium without disrupting the endothelium. Instead hantaviruses cause changes in the function of infected endothelial cells that normally regulate fluid barrier functions of capillaries. The endothelium of arteries, veins, and lymphatic vessels is unique and central to the function of vast pulmonary capillary beds, which regulate pulmonary fluid accumulation. The endothelium maintains vascular barrier functions through a complex series of redundant receptors and signaling pathways that serve to both permit fluid and immune cell efflux into tissues and restrict tissue edema. Infection of the endothelium provides several mechanisms for hantaviruses to alter capillary permeability but also defines potential therapeutic targets for regulating acute pulmonary edema and HPS disease. Here we discuss interactions of HPS causing hantaviruses with the endothelium, potential endothelial cell-directed permeability mechanisms, and therapeutic targeting of the endothelium as a means of reducing the severity of HPS disease.
Collapse
|
236
|
Andes virus infection of lymphatic endothelial cells causes giant cell and enhanced permeability responses that are rapamycin and vascular endothelial growth factor C sensitive. J Virol 2012; 86:8765-72. [PMID: 22696643 DOI: 10.1128/jvi.00817-12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Hantaviruses primarily infect endothelial cells (ECs) and nonlytically cause vascular changes that result in hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). Acute pulmonary edema during HPS may be caused by capillary leakage and failure of lymphatic vessels to clear fluids. Uniquely regulated lymphatic ECs (LECs) control fluid clearance, although roles for lymphatics in hantavirus disease remain undetermined. Here we report that hantaviruses productively infect LECs and that LEC infection by HPS causing Andes virus (ANDV) and HFRS causing Hantaan virus (HTNV) are inhibited by α(v)β(3) integrin antibodies. Although α(v)β(3) integrins regulate permeabilizing responses directed by vascular endothelial growth factor receptor 2 (VEGFR2), we found that only ANDV-infected LECs were hyperpermeabilized by the addition of VEGF-A. However, VEGF-C activation of LEC-specific VEGFR3 receptors blocked ANDV- and VEGF-A-induced LEC permeability. In addition, ∼75% of ANDV-infected LECs became viable mononuclear giant cells, >4 times larger than normal, in response to VEGF-A. Giant cells are associated with constitutive mammalian target of rapamycin (mTOR) activation, and we found that both giant LECs and LEC permeability were sensitive to rapamycin, an mTOR inhibitor, and VEGF-C addition. These findings indicate that ANDV uniquely alters VEGFR2-mTOR signaling responses of LECs, resulting in giant cell and LEC permeability responses. This suggests that ANDV infection alters normal LEC and lymphatic vessel functions which may contribute to edematous fluid accumulation during HPS. Moreover, the ability of VEGF-C and rapamycin to normalize LEC responses suggests a potential therapeutic approach for reducing pulmonary edema and the severity of HPS following ANDV infection.
Collapse
|
237
|
Vascular density and endothelial cell expression of integrin alpha v beta 3 and E-selectin in murine tumours. Tumour Biol 2012; 33:1709-17. [PMID: 22669616 DOI: 10.1007/s13277-012-0428-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 05/23/2012] [Indexed: 01/17/2023] Open
Abstract
The endothelial cell adhesion molecules, including the integrin alpha v beta 3 (αvβ3) and E-selectin, are involved in the process of angiogenesis required for tumour growth, cell migration and metastasis. The purpose of this study was to assess and compare widely used tumour models to select the ones most suitable for angiogenesis research. Fifteen murine tumours were selected including melanoma (B16), colon (C26, C38, C51), mammary (MA13, MA16, MA16/Adr, MA17, MA17/Adr, MA25, MA44), pancreatic (PO2, PO3), Glasgow osteogenic sarcoma (GOS) and Lewis lung carcinoma (LLC). The tumour vascular density, assessed using the platelet endothelial cell adhesion molecule 1 (PECAM-1; CD31) immunostaining, revealed that B16 melanoma was poorly vascularized (<5 %), whereas the colon and mammary tumours were well vascularized (5-15 %). The most vascularized tumours (>15 %) were the pancreatic tumours (PO2 and PO3), the sarcoma (GOS) and the lung tumour (LLC). The integrin αvβ3 and E-selectin, evaluated by immunohistology, showed that 7/15 tumours expressed the αvβ3 integrin which was homogeneously distributed on all tumour sections (B16, C26, MA17/Adr, MA25, MA44, PO2, LLC). E-selectin was expressed in 4/15 tumours and its expression was restricted to the tumour periphery. Only 2/15 tumours (B16 and C26) were shown to express both integrin αvβ3 and E-selectin. In conclusion, these data not only contribute to a better understanding of the tumour biology of murine tumours but can also guide the choice of appropriate models for antiangiogenic therapy, for selective drug delivery to tumours and the validation of tumour imaging modalities targeting these endothelial cell adhesion molecules.
Collapse
|