51
|
Laschke MW, Elitzsch A, Vollmar B, Vajkoczy P, Menger MD. Combined inhibition of vascular endothelial growth factor (VEGF), fibroblast growth factor and platelet-derived growth factor, but not inhibition of VEGF alone, effectively suppresses angiogenesis and vessel maturation in endometriotic lesions. Hum Reprod 2005; 21:262-8. [PMID: 16172144 DOI: 10.1093/humrep/dei308] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Angiogenesis represents the crucial step in the pathogenesis of endometriosis, because endometriotic lesions require neovascularization to establish, proliferate and invade inside the peritoneal cavity. To elucidate the role of angiogenic factors, we investigated in vivo whether blockade of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), and platelet-derived growth factor (PDGF) affects angiogenesis of ectopic endometrium. METHODS Mechanically isolated endometrial fragments were transplanted into the dorsal skinfold chamber of hormonally synchronized hamsters. Subsequently, we analysed the effect of the VEGF inhibitor SU5416 and the combined VEGF, FGF and PDGF inhibitor SU6668 on angiogenesis of the ectopic endometrium over a time-period of 14 days using intravital fluorescence microscopy. RESULTS Selective blockade of VEGF resulted in a slight reduction of microvessel density when compared to control animals. In contrast, combined inhibition of all three growth factors significantly suppressed angiogenesis of endometrial grafts, as indicated by a reduced size of the microvascular network and a decreased microvessel density. This was caused by an inhibition of blood vessel maturation. CONCLUSIONS Vascularization of endometriotic lesions is not solely driven by VEGF, but depends on the cross-talk between VEGF, FGF and PDGF. Thus, the combined inhibition of these growth factors may represent a novel therapeutic strategy in the treatment of endometriosis.
Collapse
Affiliation(s)
- M W Laschke
- Institute for Clinical & Experimental Surgery, University of Saarland, 66421 Homburg/Saar, Germany.
| | | | | | | | | |
Collapse
|
52
|
Zips D, Eicheler W, Geyer P, Hessel F, Dörfler A, Thames HD, Haberey M, Baumann M. Enhanced susceptibility of irradiated tumor vessels to vascular endothelial growth factor receptor tyrosine kinase inhibition. Cancer Res 2005; 65:5374-9. [PMID: 15958586 DOI: 10.1158/0008-5472.can-04-3379] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Previous experiments with PTK787/ZK222584, a specific inhibitor of vascular endothelial growth factor receptor (VEGFR) tyrosine kinases, using irradiated human FaDu squamous cell carcinoma in nude mice, suggested that radiation-damaged tumor vessels are more sensitive to VEGFR inhibition. To test this hypothesis, the tumor transplantation site (i.e., the right hind leg of nude mice) was irradiated 10 days before transplantation of FaDu to induce radiation damage in the host tissue. FaDu tumors vascularized by radiation-damaged blood vessels appeared later, grew at a slower rate, and showed more necrosis and a smaller vessel area per central tumor section than controls. PTK787/ZK222584 at a daily dose of 50 mg/kg body weight had no impact on growth of control tumors. In contrast, tumors vascularized by radiation-damaged vessels responded to PTK787/ZK222584 with longer latency and slower growth rate than controls, and a trend toward further increase in necrosis, indicating that irradiated tumor vessels are more susceptible to VEGFR inhibition than unirradiated vessels. Although not proving causality, expression analysis of VEGF and VEGFR2 shows that enhanced sensitivity of irradiated vessels to a specific inhibitor of VEGFR tyrosine kinases correlates with increased expression of the molecular target.
Collapse
MESH Headings
- Animals
- Blood Vessels/radiation effects
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/radiotherapy
- Cell Growth Processes/drug effects
- Cell Growth Processes/radiation effects
- Cell Line, Tumor
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Hypopharyngeal Neoplasms/blood supply
- Hypopharyngeal Neoplasms/drug therapy
- Hypopharyngeal Neoplasms/pathology
- Hypopharyngeal Neoplasms/radiotherapy
- Male
- Mice
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/radiotherapy
- Phthalazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyridines/pharmacology
- Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-2/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Daniel Zips
- Department of Radiation Oncology and Experimental Center, Medical Faculty Carl Gustav Carus, University of Technology, Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Hoeben A, Landuyt B, Highley MS, Wildiers H, Van Oosterom AT, De Bruijn EA. Vascular endothelial growth factor and angiogenesis. Pharmacol Rev 2005; 56:549-80. [PMID: 15602010 DOI: 10.1124/pr.56.4.3] [Citation(s) in RCA: 1338] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is a hallmark of wound healing, the menstrual cycle, cancer, and various ischemic and inflammatory diseases. A rich variety of pro- and antiangiogenic molecules have already been discovered. Vascular endothelial growth factor (VEGF) is an interesting inducer of angiogenesis and lymphangiogenesis, because it is a highly specific mitogen for endothelial cells. Signal transduction involves binding to tyrosine kinase receptors and results in endothelial cell proliferation, migration, and new vessel formation. In this article, the role of VEGF in physiological and pathological processes is reviewed. We also discuss how modulation of VEGF expression creates new therapeutic possibilities and describe recent developments in this field.
Collapse
Affiliation(s)
- Ann Hoeben
- Laboratory of Experimental Oncology, University Hospital Gasthuisberg, Catholic University of Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
54
|
Zhou Q, Olivo M, Lye KYK, Moore S, Sharma A, Chowbay B. Enhancing the therapeutic responsiveness of photodynamic therapy with the antiangiogenic agents SU5416 and SU6668 in murine nasopharyngeal carcinoma models. Cancer Chemother Pharmacol 2005; 56:569-77. [PMID: 16001166 DOI: 10.1007/s00280-005-1017-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Accepted: 12/08/2004] [Indexed: 12/27/2022]
Abstract
BACKGROUND Photodynamic therapy (PDT) is a promising therapeutic modality using a tumor localizing photosensitizer and light to destroy tumor cells. A major limitation of PDT is tumor recurrence, which is partly due to neovascularization. PURPOSE The objective of the present study was to determine whether combination therapy with PDT and antiangiogenic agents (i.e. SU5416 and SU6668) would be more effective in controlling tumor recurrence in a mouse model of human CNE2 poorly differentiated nasopharyngeal carcinoma compared with PDT or antiangiogenic agents administered alone. METHODS Athymic mice bearing CNE2 tumor xenografts received daily i.p. injections of 20 mg/kg SU5416 or 100 mg/kg SU6668 for 28 consecutive days either alone or following a single hypericin-PDT treatment. RESULTS Significant inhibition of CNE2 tumor growth was observed in all treatment groups. Differences in 4x tumor growth time, the number of mice with 4x tumor growth, tumor growth inhibition as well as the percent of mice surviving were not statistically significant among individual treatment groups. However, the number of mice with 4x tumor growth observed in SU6668 monotherapy and combined PDT and SU6668 treatment groups was significantly less than that in the control group (P<0.05 and 0.01, respectively). Moreover, compared with the control group, only the combined PDT and SU6668 treatment significantly extended survival of tumor-bearing host mice (P<0.05). The semiquantitative RT-PCR results showed that the expression of HIF-1alpha, VEGF, COX-2 and bFGF were increased in PDT-treated tumor samples collected 24 h post-PDT, suggesting that PDT-induced damage to tumor microvasculature and the resultant hypoxia upregulate the expression of certain proangiogenic factors. CONCLUSIONS The effectiveness of PDT can be enhanced by antiangiogenic treatment with the synthetic RTK inhibitors. Of the two synthetic RTK inhibitors tested, SU6668 was more effective than SU5416 in enhancing tumor responsiveness to PDT.
Collapse
Affiliation(s)
- Qingyu Zhou
- Laboratory of Clinical Pharmacology Division of Clinical Trials & Epidemiological Sciences, National Cancer Centre, 11, Hospital Drive, 169610, Singapore
| | | | | | | | | | | |
Collapse
|
55
|
Brieger J, Schroeder P, Gosepath J, Mann WJ. The cyclooxygenase inhibitor flurbiprofen reduces radiation-induced angiogenic growth factor secretion of squamous cell carcinoma cell lines. Ann N Y Acad Sci 2005; 1030:37-42. [PMID: 15659778 DOI: 10.1196/annals.1329.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Surgical intervention and radiotherapy still represent the gold standard in the therapy of head and neck squamous cell carcinoma (SCC), although often with unsatisfactory results. Radiation might induce the expression and secretion of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) by unknown mechanisms. These two highly active proangiogenic and cytoprotective factors might contribute to a limited therapeutic success by promoting revascularization and cytoprotection of the radiated tumor. The aim of the present study was to analyze the potential of the cyclooxygenase inhibitor flurbiprofen to reduce radiation-induced increase of VEGF and bFGF secretion of tumor cells. We analyzed the expression of VEGF and bFGF at 72 h after radiation with 30 Gy in four SCC cell lines (De-pt, Hun, Lau, and A549) in cell culture with or without added flurbiprofen. Controls were not exposed to radiation and were analyzed at the same time after culture in the same media. We observed increased VEGF levels in all and increased bFGF levels in three of four lines after radiation. In irradiated cultures with flurbiprofen, VEGF was reduced between 13% and 26% and bFGF was reduced between 84% and 93% compared with radiated cultures without flurbiprofen. We found no reduction of VEGF and bFGF secretion in the unirradiated cultures despite added flurbiprofen. We conclude that flurbiprofen is able to alter the radiation-induced secretion of these two growth factors and might be useful in decreasing the resistance of SCC to radiation.
Collapse
Affiliation(s)
- Jürgen Brieger
- Department of Otorhinolaryngology, Mainz Medical School, Germany.
| | | | | | | |
Collapse
|
56
|
Kim TE, Murren JR. Angiogenesis in non-small cell lung cancer. A new target for therapy. ACTA ACUST UNITED AC 2005; 1:325-38. [PMID: 14720035 DOI: 10.1007/bf03256626] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-small cell lung cancer (NSCLC) is cured with surgery in a minority of affected persons. Chemotherapy and radiation can palliate and extend survival of patients with disease not amenable to surgery. Consequently, new treatment options are urgently needed. In the era of molecularly targeted therapeutics, the recent direction in cancer research has been to identify and modulate specific events in tumorigenesis. Angiogenesis, or new vessel formation, is one such event elucidated to be fundamental to the development, growth, and metastasis of cancers and is one of the characteristics that differentiates tumor from host. Thus, targeting of tumor neovasculature continues to generate tremendous enthusiasm and effort in drug development. Extensive research into the role of angiogenesis in NSCLC has produced a host of novel targets; their potential inhibitors, now numbering over 40, are in various phases of clinical testing around the world. The current lead compounds include inhibitors of matrix metalloproteinases, angiogenic growth factors and their receptor tyrosine kinases. Despite their impressive activity in animal models, definitive evidence of their antitumor activity in humans is yet to be established. We face several challenges as we look to advance the field of antiangiogenesis for the treatment of cancer, namely, the need for a better understanding of the optimal timing and dosing of antiangiogenic agents, the validation of imaging and quantification methods of tumor angiogenesis, and a new clinical trials design for a more expedient evaluation of novel cytostatic target modulators.
Collapse
Affiliation(s)
- Tracy E Kim
- Department of Internal Medicine, Section of Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | |
Collapse
|
57
|
Jin GH, Jin SZ, Liu Y, Xu RM, Yang JZ, Pan XN, Liu SZ. Therapeutic effect of gene-therapy in combination with local X-irradiation in a mouse malignant melanoma model. Biochem Biophys Res Commun 2005; 330:975-81. [PMID: 15809091 DOI: 10.1016/j.bbrc.2005.03.070] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2005] [Indexed: 11/20/2022]
Abstract
Plasmid containing mIL-18 and B7.1 genes downstream of Egr-1 promoter was constructed and used in gene-radiotherapy on malignant melanoma in C57BL/6J mice implanted with B16 cells followed by exploration of the immunologic mechanism of the therapeutic effect. The treatment with plasmid pEgr-IL-18-B7.1 plus local X-irradiation showed more effective suppression of tumor growth than the treatment with radiation alone, pEgr-IL-18-B7.1 alone, or single gene pEgr-IL-18 (or pEgr-B7.1) combined with local X-irradiation. Anticancer immunity was found to be significantly upregulated in tumor-bearing mice treated with pEgr-IL-18-B7.1 plus local X-irradiation. IL-18 showed no direct killing effect on malignant melanoma cells in vitro, and the mechanism of the combined therapy with pEgr-IL-18-B7.1 and local X-irradiation was apparently related with the stimulation of host anticancer immunity by increased secretion of IL-18 and upregulated immunogenicity of the tumor cells by increased expression of B7.1 on their surface in addition to the direct effect of local X-irradiation on the tumor cells.
Collapse
Affiliation(s)
- Guang-Hui Jin
- Immunobiology Laboratory, MH Radiobiology Research Unit, Jilin University Norman, Bethune Health Sciences Center, 828 Xinmin Street, Changchun 130021, PR China
| | | | | | | | | | | | | |
Collapse
|
58
|
Gasparini G, Longo R, Fanelli M, Teicher BA. Combination of antiangiogenic therapy with other anticancer therapies: results, challenges, and open questions. J Clin Oncol 2005; 23:1295-311. [PMID: 15718328 DOI: 10.1200/jco.2005.10.022] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Angiogenesis is necessary for tumor growth. Drug discovery efforts have identified several potential therapeutic targets on endothelial cells and selective inhibitors capable of slowing tumor growth or producing tumor regression by blocking angiogenesis in in vivo tumor models. Certain antiangiogenic therapeutics increase the activity of cytotoxic anticancer treatments in preclinical models. More than 75 antiangiogenic compounds have entered clinical trials. Most of the early clinical testing was conducted in patients with advanced disease resistant to standard therapies. Several phase III trials have been undertaken to compare the efficacy of standard chemotherapy versus the same in combination with an experimental angiogenesis inhibitor. Preliminary results of the clinical studies suggest that single-agent antiangiogenic therapy is poorly active in advanced tumors. Although some of the results of combination trials are controversial, recent positive outcomes with an antivascular endothelial growth factor antibody combined with chemotherapy as front-line therapy of metastatic colorectal cancer have renewed enthusiasm for this therapeutic strategy. This article presents an overview of experimental and clinical studies of combined therapy with antiangiogenic agents and highlights the challenges related to the appropriate strategies for selection of the patients, study design, and choice of proper end points for preclinical and clinical studies using these agents.
Collapse
|
59
|
Schuuring J, Bussink J, Bernsen HJJA, Peeters W, van Der Kogel AJ. Irradiation combined with SU5416: microvascular changes and growth delay in a human xenograft glioblastoma tumor line. Int J Radiat Oncol Biol Phys 2005; 61:529-34. [PMID: 15667976 DOI: 10.1016/j.ijrobp.2004.09.063] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2004] [Revised: 09/26/2004] [Accepted: 09/30/2004] [Indexed: 11/29/2022]
Abstract
PURPOSE The combination of irradiation and the antiangiogenic compound SU5416 was tested and compared with irradiation alone in a human glioblastoma tumor line xenografted in nude mice. The aim of this study was to monitor microenvironmental changes and growth delay. METHODS AND MATERIALS A human glioblastoma xenograft tumor line was implanted in nude mice. Irradiations consisted of 10 Gy or 20 Gy with and without SU5416. Several microenvironmental parameters (tumor cell hypoxia, tumor blood perfusion, vascular volume, and microvascular density) were analyzed after imunohistochemical staining. Tumor growth delay was monitored for up to 200 days after treatment. RESULTS SU5416, when combined with irradiation, has an additive effect over treatment with irradiation alone. Analysis of the tumor microenvironment showed a decreased vascular density during treatment with SU5416. In tumors regrowing after reaching only a partial remission, vascular characteristics normalized shortly after cessation of SU5416. However, in tumors regrowing after reaching a complete remission, permanent microenvironmental changes and an increase of tumor necrosis with a subsequent slower tumor regrowth was found. CONCLUSIONS Permanent vascular changes were seen after combined treatment resulting in complete remission. Antiangiogenic treatment with SU5416 when combined with irradiation has an additive effect over treatment with irradiation or antiangiogenic treatment alone.
Collapse
Affiliation(s)
- Janneke Schuuring
- Department of Neurology, University Medical Center, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
60
|
Zips D, Hessel F, Krause M, Schiefer Y, Hoinkis C, Thames HD, Haberey M, Baumann M. Impact of adjuvant inhibition of vascular endothelial growth factor receptor tyrosine kinases on tumor growth delay and local tumor control after fractionated irradiation in human squamous cell carcinomas in nude mice. Int J Radiat Oncol Biol Phys 2005; 61:908-14. [PMID: 15708274 DOI: 10.1016/j.ijrobp.2004.11.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2004] [Revised: 10/28/2004] [Accepted: 11/01/2004] [Indexed: 11/28/2022]
Abstract
PURPOSE Previous experiments have shown that adjuvant inhibition of the vascular endothelial growth factor receptor after fractionated irradiation prolonged tumor growth delay and may also improve local tumor control. To test the latter hypothesis, local tumor control experiments were performed. METHODS AND MATERIALS Human FaDu and UT-SCC-14 squamous cell carcinomas were studied in nude mice. The vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584 (50 mg/kg body weight b.i.d.) was administered for 75 days after irradiation with 30 fractions within 6 weeks. Tumor growth time and tumor control dose 50% (TCD(50)) were determined and compared to controls (carrier without PTK787/ZK222584). RESULTS Adjuvant administration of PTK787/ZK222584 significantly prolonged tumor growth time to reach 5 times the volume at start of drug treatment by an average of 11 days (95% confidence interval 0.06;22) in FaDu tumors and 29 days (0.6;58) in UT-SCC-14 tumors. In both tumor models, TCD(50) values were not statistically significantly different between the groups treated with PTK787/ZK222584 compared to controls. CONCLUSIONS Long-term inhibition of angiogenesis after radiotherapy significantly reduced the growth rate of local recurrences but did not improve local tumor control. This indicates that recurrences after irradiation depend on vascular endothelial growth factor-driven angiogenesis, but surviving tumor cells retain their clonogenic potential during adjuvant antiangiogenic treatment with PTK787/ZK222584.
Collapse
Affiliation(s)
- Daniel Zips
- Department of Radiation Oncology, Medical Faculty Carl Gustav Carus, University of Technology-Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Raben D, Helfrich B. Angiogenesis inhibitors: a rational strategy for radiosensitization in the treatment of non-small-cell lung cancer? Clin Lung Cancer 2005; 6:48-57. [PMID: 15310417 DOI: 10.3816/clc.2004.n.021] [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: 01/28/2023]
Abstract
Angiogenesis is a precondition to invasion and metastasis for all solid tumors. Vascular endothelial growth factor (VEGF) and its family of receptors (VEGFR) play a critical role in cancer progression by promoting new blood vessel formation. Overexpression of VEGF and VEGFR has been correlated with poor prognosis in a variety of malignancies. In this era of targeted therapies for cancer, inhibiting angiogenesis through antiangiogenic and/or vascular targeting agents seems logical. Disturbing the angiogenesis process is an alternative or complementary strategy to inhibition of growth factor signaling. Blocking angiogenesis may enhance conventional anticancer treatments such as radiation therapy in situations where tumors are unresponsive to current antigrowth factor efforts. Compounds currently under investigation in cancer therapy include anti-VEGF/VEGFR antibodies, small molecule VEGFR tyrosine kinase inhibitors, antisense suppression of VEGF, immunotherapy, viral-directed targeting of VEGFR signaling, ribozymes, and various toxin conjugates. Preclinical investigations are exploring the benefits of combining angiogenic inhibitors with radiation. This article will provide an overview of these preclinical studies and the rationale for this therapeutic strategy in the treatment of non-small-cell lung cancer.
Collapse
Affiliation(s)
- David Raben
- Tobacco Related Malignancy Program, University of Colorado Cancer Center and Department of Radiation Oncology, Aurora 80010-0510, USA.
| | | |
Collapse
|
62
|
Ellis LM. Preclinical data targeting vascular endothelial growth factor in colorectal cancer. Clin Colorectal Cancer 2005; 4 Suppl 2:S55-61. [PMID: 15479480 DOI: 10.3816/ccc.2004.s.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Vascular endothelial growth factor (VEGF) is the driving force behind angiogenesis in most solid malignancies. This also holds true for colorectal cancer (CRC), where increased levels of VEGF in primary cancers are associated with increased microvessel density and poor prognosis. These findings have led to preclinical studies evaluating the efficacy of anti-VEGF therapy in inhibiting the growth of CRC in ectopic and orthotopic locations. In preclinical models, numerous approaches to inhibit VEGF activity led to decreased tumor growth and angiogenesis. These studies led to clinical trials in which, unfortunately, single-agent anti-VEGF therapy was relatively ineffective for patients with metastatic CRC. However, combinations of anti-VEGF therapies with chemotherapy have clearly demonstrated clinical benefit. Understanding the mechanisms of the role of VEGF in CRC angiogenesis and the effect of anti-VEGF therapy on the tumor vasculature will allow oncologists to optimize therapeutic regimens targeting VEGF and its receptors.
Collapse
Affiliation(s)
- Lee M Ellis
- Department of Surgical Oncology, Box 444, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030-4009, USA.
| |
Collapse
|
63
|
Nishikawa T, Ramesh R, Munshi A, Chada S, Meyn RE. Adenovirus-mediated mda-7 (IL24) gene therapy suppresses angiogenesis and sensitizes NSCLC xenograft tumors to radiation. Mol Ther 2005; 9:818-28. [PMID: 15194048 DOI: 10.1016/j.ymthe.2004.03.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Accepted: 03/15/2004] [Indexed: 12/11/2022] Open
Abstract
Melanoma differentiation-associated gene-7 (mda-7), recently classified as interleukin-24 (approved gene symbol IL24), is thought to be a tumor suppressor gene based on the loss of its expression in many different types of cancer. Gene therapy by adenovirus-mediated mda-7 (Ad-mda7) gene transfer has been shown to inhibit the growth of several different tumor cell lines, in vitro and in vivo. We previously demonstrated that Ad-mda7 radiosensitized non-small-cell lung cancer (NSCLC) cell lines by enhancing an apoptosis pathway through the activation of JNK and c-Jun. In the present study, we investigated the efficacy of intratumoral administration of Ad-mda7 combined with ionizing radiation for treating A549 xenograft tumors in nude mice. Substantial and long-lasting inhibition of tumor growth was evident following the combined treatment. Histological examination revealed marked reduction of angiogenic factors (bFGF, VEGF) and microvessel density and enhanced apoptosis in the tumors treated with the combination therapy compared to those treated with Ad-mda7 alone or radiation alone. To confirm the radiosensitizing effect of secreted MDA-7 protein, we performed clonogenic survival assays using human umbilical vein endothelial cells (HUVECs), A549 cells, and normal human lung fibroblasts, CCD16 cells, pretreated with the conditioned medium from 293 cells that had been stably transfected with mda-7 or a control vector. The results showed that MDA-7 protein sensitized HUVECs to ionizing radiation but not A549 cells or CCD16 cells. Our results suggest that Ad-mda7 in combination with radiation enhances apoptosis in the tumors and that secreted MDA-7 protein inhibits angiogenesis by sensitizing endothelial cells to ionizing radiation without affecting other normal cells. We conclude that the combination of mda-7 gene therapy and radiotherapy may be a feasible and effective strategy for treatment of NSCLC.
Collapse
Affiliation(s)
- Takashi Nishikawa
- Department of Experimental Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
64
|
Peterson AC, Swiger S, Stadler WM, Medved M, Karczmar G, Gajewski TF. Phase II study of the Flk-1 tyrosine kinase inhibitor SU5416 in advanced melanoma. Clin Cancer Res 2005; 10:4048-54. [PMID: 15217937 DOI: 10.1158/1078-0432.ccr-03-0766] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Vascular endothelial growth factor (VEGF) expression is prognostic in melanoma, and the activity of VEGF is mediated in part through the receptor tyrosine kinase Flk-1. A Phase II study of SU5416, a preferential inhibitor of Flk-1, was carried out in patients with metastatic melanoma to determine clinical response, tolerability, and changes in tumor vascular perfusion. EXPERIMENTAL DESIGN Patients with documented progressive disease and </=1 prior therapy were eligible. Central nervous system metastases were allowed if stable off medication. SU5416 (145 mg/m(2)) was administered via a central catheter twice weekly for 8 weeks. Premedication with dexamethasone, diphenhydramine, and a H(2) blocker was required because of the Cremophor vehicle. Tumor vascular perfusion was assessed before treatment and during week 8 by dynamic contrast magnetic resonance imaging, and plasma was analyzed for VEGF. RESULTS Thirty-one patients were enrolled. Two-thirds had received prior therapy, 21 had visceral metastasis, and 14 had an elevated lactate dehydrogenase. Mean absolute lymphocyte counts were decreased (P = 0.002), and glucose levels were increased (P = 0.001) posttherapy, presumably because of steroid premedication. Four vascular adverse events were observed. Of 26 evaluable patients, 1 experienced a partial response, 1 had stable disease, and 5 had a mixed response. Dynamic contrast magnetic resonance imaging in 5 evaluable patients showed decreased tumor perfusion at week 8 (P = 0.024), and plasma VEGF levels were elevated compared with pretherapy (P = 0.008). CONCLUSIONS SU5146 appears to be relatively well tolerated in this population. Although the modest clinical activity and potential effects on tumor vascularity may support additional exploration of VEGF as a target in melanoma, effects from steroid premedication limit further investigation of this agent.
Collapse
Affiliation(s)
- Amy C Peterson
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Cancer Research Center, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
65
|
Williams KJ, Telfer BA, Brave S, Kendrew J, Whittaker L, Stratford IJ, Wedge SR. ZD6474, a Potent Inhibitor of Vascular Endothelial Growth Factor Signaling, Combined With Radiotherapy. Clin Cancer Res 2004; 10:8587-93. [PMID: 15623642 DOI: 10.1158/1078-0432.ccr-04-1147] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Vascular endothelial growth factor (VEGF) plays a key role in tumor angiogenesis and acts as a radiation survival factor for endothelial cells. ZD6474 (N-(4-bromo-2-fluorophenyl)-6-methoxy-7-[(1-methylpiperidin-4-yl)methoxy]quinazolin-4-amine) is a potent VEGF receptor 2 (KDR) tyrosine kinase inhibitor (TKI) that has additional activity versus the epidermal growth factor receptor. This study was designed to determine the efficacy of combining ZD6474 and radiotherapy in vivo. EXPERIMENTAL DESIGN The Calu-6 (non-small-cell lung cancer) tumor model was selected because it was found to be unresponsive to treatment with a selective epidermal growth factor receptor TKI but responds significantly to treatment with selective VEGF receptor TKIs. Tumor-bearing mice received either vehicle or ZD6474 (50 mg/kg, by mouth, once daily) for the duration of the experiment, with or without radiotherapy (3 x 2 Gy, days 1-3). Two combination schedules were examined: (a) ZD6474 given before each dose of radiation (concurrent schedule); and (b) ZD6474 given 30 minutes after the last dose of radiotherapy (sequential schedule). RESULTS The growth delay induced using the concurrent schedule was greater than that induced by ZD6474 or radiation treatment alone (22 +/- 1 versus 9 +/- 1 and 17 +/- 2 days, respectively; P = 0.03 versus radiation alone). When administered sequentially, the growth delay was markedly enhanced (36 +/- 1 days; P < 0.001 versus radiation alone or the concurrent schedule). Intravenous administration of Hoechst 33342 showed a trend toward reduced tumor perfusion after ZD6474 treatment, and a pairwise comparison (versus control) was significant after three doses of ZD6474 (P = 0.05 by one-tailed t test). Thus, impaired reoxygenation between fractions in the concurrent protocol may be the causal basis for the schedule dependency of the radiopotentiation observed. CONCLUSIONS ZD6474 may be a successful adjuvant to clinical radiotherapy, and scheduling of the treatments could be important to ensure optimal efficacy.
Collapse
Affiliation(s)
- Kaye J Williams
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Manchester, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
66
|
Abstract
The treatment of squamous cell carcinoma of the head and neck (SCCHN) has recently witnessed the introduction of molecularly targeted agents based on disease biology, target discovery, and validation. One class of agents, the epidermal growth factor receptor (EGFR) inhibitors, is currently in phase III trials. There are multiple processes, however, that appear to be suitable for targeted therapy beyond EGFR. These include signal transduction, cell cycle control, prostaglandin synthesis, protein degradation, hypoxia, and angiogenesis. These systems and specific protein targets will be reviewed in detail with emphasis on promising preclinical and early clinical evidence of activity.
Collapse
Affiliation(s)
- Ezra E W Cohen
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637-1470, USA.
| |
Collapse
|
67
|
Fenton BM, Paoni SF, Ding I. Pathophysiological Effects of Vascular Endothelial Growth Factor Receptor-2-Blocking Antibody plus Fractionated Radiotherapy on Murine Mammary Tumors. Cancer Res 2004; 64:5712-9. [PMID: 15313911 DOI: 10.1158/0008-5472.can-04-0434] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although clinical trials of antiangiogenic strategies have been disappointing when administered as single agents, such approaches can play an important role in cancer treatment when combined with conventional therapies. Previous studies have shown that DC101, an antiangiogenic monoclonal antibody against vascular endothelial growth factor receptor-2, can produce significant growth inhibition in spontaneous and transplanted tumors but can also induce substantial hypoxia. Because DC101 appears to potentiate radiotherapy in some tumors, the present studies were undertaken to characterize pathophysiological changes following combined therapy and to determine whether radioresponse is enhanced despite the induction of hypoxia. MCa-4 and MCa-35 mammary carcinomas were treated with: (a) DC101; (b) 5 x 6 Gy radiation fractions; or (c) the combination. Image analysis of frozen tumor sections was used to quantitate: (a) hypoxia; (b) spacing of total and perfused blood vessels; and (c) endothelial and tumor cell apoptosis. For MCa-4, combination treatment schedules produced significant and prolonged delays in tumor growth, whereas single-modality treatments had minor effects. For MCa-35, radiation or the combination led to equivalent growth inhibition. In all tumors, hypoxia increased markedly after either radiation or DC101 alone. Although combination therapy produced no immediate pathophysiological changes, hypoxia ultimately increased after cessation of therapy. Preferential increases in endothelial apoptosis following combination treatment suggest that in addition to blocking tumor angiogenesis, DC101 enhances radiotherapy by specifically sensitizing endothelial cells, leading to degeneration of newly formed blood vessels.
Collapse
Affiliation(s)
- Bruce M Fenton
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York 14642, USA.
| | | | | |
Collapse
|
68
|
Fenton BM, Paoni SF, Ding I. Effect of VEGF receptor-2 antibody on vascular function and oxygenation in spontaneous and transplanted tumors. Radiother Oncol 2004; 72:221-30. [PMID: 15297140 DOI: 10.1016/j.radonc.2004.05.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2003] [Revised: 04/02/2004] [Accepted: 05/10/2004] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND PURPOSE The primary objectives of this study were to address two major questions. (1) Does VEGF receptor-2 antibody (DC101) produce detrimental effects on tumor vascular function and oxygenation that could compromise adjuvant therapies? (2) Is pathophysiological response to such antiangiogenic strategies different in transplanted versus primary spontaneous tumors? MATERIALS AND METHODS The effects of early and late initiation DC101 treatment were evaluated using spontaneous murine mammary carcinomas and two markedly different transplanted mammary tumors, MCa-35 and MCa-4. Mice were administered DC101 or saline, tumors were frozen, and immunohistochemical staining was quantified using image analysis of multiply-stained frozen sections. Total blood vessels were identified using antibodies to CD31 or panendothelial antigen, perfused vessels via i.v. injection of fluorescent DiOC7, and tumor hypoxia by hypoxia marker (EF5) uptake. RESULTS Tumor growth was significantly inhibited following DC101 administration in all tumor models. In general, early initiation DC101 treatment reduced perfused vessel counts and increased tumor hypoxia, while late initiation treatment had no significant impact on either. Results indicate that DC101 slows tumor growth through a decrease in vascular function, leading to increased tumor cell apoptosis and necrosis at sites distant from perfused blood vessels, and suggest that DC101 accelerates the rate at which tumor cells outgrow their functional vascular supply. CONCLUSIONS Although highly variable among individual spontaneous tumors, the overall effects of DC101 on tumor hypoxia were quite similar between spontaneous and transplanted tumors. Since reductions in tumor oxygenation due to antiangiogenic treatment were transient, initial pathophysiological deficiencies that could compromise conventional therapies over the short-term may be of less relevance when administered over more extended treatment schedules.
Collapse
Affiliation(s)
- Bruce M Fenton
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | |
Collapse
|
69
|
Abstract
A functional tumor vasculature is essential for tumor growth and metastasis and makes an attractive target for therapy. Both antiangiogenic and antivascular approaches are being developed for this purpose. In this article, the current antiangiogenic and antivascular approaches to cancer therapy, potential for their combination with radiotherapy, methods for identifying new targets on the tumor vasculature, and methods for evaluating new vascular-targeted strategies in in vivo model systems are reviewed.
Collapse
Affiliation(s)
- Gillian M Tozer
- Tumour Microcirculation Group, Gray Cancer Institute, Mount Vernon Hospital Middlesex, United Kingdom
| | | |
Collapse
|
70
|
Moeller BJ, Cao Y, Li CY, Dewhirst MW. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell 2004; 5:429-41. [PMID: 15144951 DOI: 10.1016/s1535-6108(04)00115-1] [Citation(s) in RCA: 793] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2003] [Revised: 02/25/2004] [Accepted: 03/15/2004] [Indexed: 12/20/2022]
Abstract
Through a poorly understood mechanism, tumors respond to radiation by secreting cytokines capable of inhibiting apoptosis in endothelial cells, thereby diminishing treatment response by minimizing vascular damage. We reveal here that this pathway is governed by a major angiogenesis regulator, HIF-1. Following radiotherapy, tumor reoxygenation leads to: (1) nuclear accumulation of HIF-1 in response to reactive oxygen, and (2) enhanced translation of HIF-1-regulated transcripts secondary to stress granule depolymerization. The resulting increase in HIF-1-regulated cytokines enhances endothelial cell radioresistance. Inhibiting postradiation HIF-1 activation significantly increases tumor radiosensitivity as a result of enhanced vascular destruction. These data describe novel pathways contributing significantly to our understanding of HIF-1 regulation which may be major determinants of tumor radiosensitivity, potentially having high clinical relevance.
Collapse
Affiliation(s)
- Benjamin J Moeller
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
71
|
Lu B, Geng L, Musiek A, Tan J, Cao C, Donnelly E, McMahon G, Choy H, Hallahan DE. Broad spectrum receptor tyrosine kinase inhibitor, SU6668, sensitizes radiation via targeting survival pathway of vascular endothelium. Int J Radiat Oncol Biol Phys 2004; 58:844-50. [PMID: 14967441 DOI: 10.1016/j.ijrobp.2003.10.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2003] [Revised: 10/16/2003] [Accepted: 10/31/2003] [Indexed: 01/10/2023]
Abstract
PURPOSE Recent studies have demonstrated radiosensitization by inhibiting receptor tyrosine kinases (RTKs). Irradiation activates RTKs and their downstream prosurvival molecule, Akt. In this study, we investigated the mechanism by which SU6668, an inhibitor of RTKs involved in angiogenic pathways, enhances effects of irradiation. METHODS AND MATERIALS Western blots were used to determine Akt phosphorylation. Clonogenic assays were performed to determine endothelial survival after combination of SU6668 and irradiation. This combination therapy was also tested in mouse models with Lewis lung carcinoma or glioblastoma multiforme (GL261) for inhibition of tumor growth and tumor vasculature by examining tumor volume, tumor vascular window, and blood flow. RESULTS We found that SU6668 inhibited the Akt activation inducible by irradiation. Clonogenic survival of endothelial cells was decreased after the combined therapy compared with radiotherapy alone. In vivo studies demonstrated reduction of tumor vasculature and blood flow. In addition, 21 Gy in 7 fractions given concurrently with SU6668 resulted in tumor growth delay compared to either treatment alone. CONCLUSION These data suggest that the combination therapy was more effective in destroying tumor vasculature than either treatment alone. SU6668 augments tumor-suppressive effects of radiotherapy in Lewis lung carcinoma and GL261 xenographs, possibly through reducing the survival of tumor endothelium.
Collapse
Affiliation(s)
- Bo Lu
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, Tennessee USA
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Li L, Wartchow CA, Danthi SN, Shen Z, Dechene N, Pease J, Choi HS, Doede T, Chu P, Ning S, Lee DY, Bednarski MD, Knox SJ. A novel antiangiogenesis therapy using an integrin antagonist or anti–Flk-1 antibody coated 90Y-labeled nanoparticles. Int J Radiat Oncol Biol Phys 2004; 58:1215-27. [PMID: 15001266 DOI: 10.1016/j.ijrobp.2003.10.057] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2003] [Revised: 09/29/2003] [Accepted: 10/15/2003] [Indexed: 11/25/2022]
Abstract
PURPOSE Integrin alpha(v)beta(3) and vascular endothelial growth factor receptor 2 (Flk-1) have been shown to be involved in tumor-induced angiogenesis. Selective targeting of upregulated alpha(v)beta(3) and Flk-1 on the neovasculature of tumors is a novel antiangiogenesis strategy for treating a wide variety of solid tumors. In the studies described here, we investigated the potential therapeutic efficacy of two three-component treatment regimens using two murine tumor models. METHODS AND MATERIALS The treatment regimens used nanoparticle (NP) based targeting agents radiolabeled with (90)Y. The small molecule integrin antagonist (IA) 4-[2-(3,4,5,6-tetrahydropyrimidin-2-ylamino)ethoxy]-benzoyl-2-(5)-aminoethylsulfonylamino-beta-alanine, which binds to the integrin alpha(v)beta(3), and a monoclonal antibody against murine Flk-1 (anti-Flk-1 MAb) were used to target the NPs. Murine tumor models K1735-M2 (melanoma) and CT-26 (colon adenocarcinoma) were used to evaluate the treatment efficacy. RESULTS In K1735-M2 and CT-26 tumors, a single treatment with IA-NP-(90)Y (14.2 microg/g IA, 5 or 6 microCi/g (90)Y) caused a significant tumor growth delay compared to untreated control tumors, as well as tumors treated with IA, IA-NP, and NP-(90)Y, respectively (p < 0.025, Wilcoxon test). In K1735-M2 tumors, a single treatment with anti-Flk-1 MAb-NP-(90)Y (0.36 microg/g anti-Flk-1 MAb, 5 microCi/g (90)Y) also caused a significant tumor growth delay (p < 0.05, Wilcoxon test) compared to untreated tumors, as well as tumors treated with anti-Flk-1 MAb, anti-Flk-1 MAb-NP, and conventional radioimmunotherapy with (90)Y-labeled anti-Flk mAb. Anti-CD31 staining showed a marked decrease in vessel density in tumors treated with anti-Flk-1 MAb-NP-(90)Y, which was associated with a high level of apoptotic death in these tumors, as shown by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining. CONCLUSIONS The present studies provide proof of principle that targeted radiotherapy works using different targeting agents on a nanoparticle, to target both the integrin alpha(v)beta(3) and the vascular endothelial growth factor receptor. These encouraging results demonstrate the potential therapeutic efficacy of the IA-NP-(90)Y and anti-Flk-1 MAb-NP-(90)Y complexes as novel therapeutic agents for the treatment of a variety of tumor types.
Collapse
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antibodies, Monoclonal/therapeutic use
- Apoptosis
- Cell Line, Tumor
- Drug Screening Assays, Antitumor
- Female
- In Situ Nick-End Labeling
- Integrins/antagonists & inhibitors
- Liposomes
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/drug therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Nude
- Models, Animal
- Nanotechnology
- Neoplasms/blood supply
- Neoplasms/drug therapy
- Neovascularization, Pathologic/drug therapy
- Pentetic Acid/therapeutic use
- Receptors, Vitronectin/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
Collapse
Affiliation(s)
- Lingyun Li
- Department of Radiation Oncology, Stanford University, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
|
74
|
Harrigan MR. Angiogenic factors in the central nervous system. Neurosurgery 2003; 53:639-60; discussion 660-1. [PMID: 12943581 DOI: 10.1227/01.neu.0000079575.09923.59] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2002] [Accepted: 04/24/2003] [Indexed: 01/08/2023] Open
Abstract
The past decade has seen considerable advances in the understanding of angiogenesis. Blood vessel development and growth in the central nervous system are tightly controlled processes that are regulated by angiogenic factors. Angiogenic factors have been implicated in the pathogenesis of a wide variety of disorders, including primary and metastatic brain tumors, aneurysms, arteriovenous malformations, and cavernous malformations. The potential clinical applications of angiogenesis research include inhibition of angiogenesis to control brain tumors and therapeutic angiogenesis to promote collateral blood vessel formation among patients at risk of ischemia. This article summarizes the processes of blood vessel formation in the brain, examines the angiogenic factors that are prominent in the central nervous system, reviews the clinical use of angiogenesis inhibitors, and identifies areas for future investigation.
Collapse
Affiliation(s)
- Mark R Harrigan
- Department of Neurosurgery and Toshiba Stroke Research Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14209, USA.
| |
Collapse
|
75
|
Ma BBY, Bristow RG, Kim J, Siu LL. Combined-modality treatment of solid tumors using radiotherapy and molecular targeted agents. J Clin Oncol 2003; 21:2760-76. [PMID: 12860956 DOI: 10.1200/jco.2003.10.044] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Molecular targeted agents have been combined with radiotherapy (RT) in recent clinical trials in an effort to optimize the therapeutic index of RT. The appeal of this strategy lies in their potential target specificity and clinically acceptable toxicity. DESIGN This article integrates the salient, published research findings into the underlying molecular mechanisms, preclinical efficacy, and clinical applicability of combining RT with molecular targeted agents. These agents include inhibitors of intracellular signal transduction molecules, modulators of apoptosis, inhibitors of cell cycle checkpoints control, antiangiogenic agents, and cyclo-oxygenase-2 inhibitors. RESULTS Molecular targeted agents can have direct effects on the cytoprotective and cytotoxic pathways implicated in the cellular response to ionizing radiation (IR). These pathways involve cellular proliferation, DNA repair, cell cycle progression, nuclear transcription, tumor angiogenesis, and prostanoid-associated inflammation. These pathways can also converge to alter RT-induced apoptosis, terminal growth arrest, and reproductive cell death. Pharmacologic modulation of these pathways may potentially enhance tumor response to RT though inhibition of tumor repopulation, improvement of tumor oxygenation, redistribution during the cell cycle, and alteration of intrinsic tumor radiosensitivity. CONCLUSION Combining RT and molecular targeted agents is a rational approach in the treatment of solid tumors. Translation of this approach from promising preclinical data to clinical trials is actively underway.
Collapse
Affiliation(s)
- Brigette B Y Ma
- Department of Medical Oncology and Hematology, Precess Margaret Hospital, University Health Network, Suite 5-210, 610 University Ave, Toronto, Ontario, Canada M5G 2M9
| | | | | | | |
Collapse
|
76
|
Garcia-Barros M, Paris F, Cordon-Cardo C, Lyden D, Rafii S, Haimovitz-Friedman A, Fuks Z, Kolesnick R. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science 2003; 300:1155-9. [PMID: 12750523 DOI: 10.1126/science.1082504] [Citation(s) in RCA: 1147] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
About 50% of cancer patients receive radiation therapy. Here we investigated the hypothesis that tumor response to radiation is determined not only by tumor cell phenotype but also by microvascular sensitivity. MCA/129 fibrosarcomas and B16F1 melanomas grown in apoptosis-resistant acid sphingomyelinase (asmase)-deficient or Bax-deficient mice displayed markedly reduced baseline microvascular endothelial apoptosis and grew 200 to 400% faster than tumors on wild-type microvasculature. Thus, endothelial apoptosis is a homeostatic factor regulating angiogenesis-dependent tumor growth. Moreover, these tumors exhibited reduced endothelial apoptosis upon irradiation and, unlike tumors in wild-type mice, they were resistant to single-dose radiation up to 20 grays (Gy). These studies indicate that microvascular damage regulates tumor cell response to radiation at the clinically relevant dose range.
Collapse
Affiliation(s)
- Monica Garcia-Barros
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Solomon B, Hagekyriakou J, Trivett MK, Stacker SA, McArthur GA, Cullinane C. EGFR blockade with ZD1839 ("Iressa") potentiates the antitumor effects of single and multiple fractions of ionizing radiation in human A431 squamous cell carcinoma. Epidermal growth factor receptor. Int J Radiat Oncol Biol Phys 2003; 55:713-23. [PMID: 12573759 DOI: 10.1016/s0360-3016(02)04357-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE Signaling pathways initiated by the epidermal growth factor receptor (EGFR) play important roles in the response to ionizing radiation. In this study the consequences of inhibiting the EGFR on the response of A431 cells (human vulvar squamous cell carcinoma cells that overexpress EGFR) to radiation, were investigated in vitro and in vivo, using the selective EGFR-tyrosine kinase inhibitor, ZD1839 ("Iressa"). METHODS AND MATERIALS The effect of ZD1839 on proliferation, apoptosis, and clonogenic survival after radiation was determined in vitro. For in vivo studies, athymic nude mice with established subcutaneous A431 xenografts (approximately 100 mm(3)) were treated with either a single 10 Gy fraction or 4 daily 2.5 Gy fractions of radiation with or without ZD1839 (75 mg/kg/day intraperitoneally for 10 days) to determine effects on tumor growth delay. RESULTS Treatment of A431 cells with ZD1839 in vitro reduced proliferation, increased apoptosis, and reduced clonogenic survival after radiation. Strikingly greater than additive effects of ZD1839 in combination with radiation on tumor growth delay were observed in vivo after either a single 10 Gy fraction (enhancement ratio: 1.5) or multiple 4 x 2.5 Gy fractions (enhancement ratio: 4). ZD1839 reduced tumor vascularity, as well as levels of vascular endothelial growth factor (VEGF) protein and mRNA induced by stimulation with epidermal growth factor (EGF), suggesting a possible role of inhibition of angiogenesis in the effect. CONCLUSIONS Inhibiting EGFR-mediated signal transduction cascades with ZD1839 potentiates the antitumor effect of single and multiple fractions of radiation. These data provide preclinical rationale for clinical trials of EGFR inhibitors including ZD1839 in combination with radiation.
Collapse
Affiliation(s)
- Benjamin Solomon
- Research Division, Department of Haematology and Medical Oncology, Peter MacCallum Cancer Institute, Melbourne, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
78
|
Siemann DW, Shi W. Targeting the tumor blood vessel network to enhance the efficacy of radiation therapy. Semin Radiat Oncol 2003; 13:53-61. [PMID: 12520464 DOI: 10.1053/srao.2003.50005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
It has been well established that the vascularization of solid tumors is a prerequisite if a clinically relevant size is to be reached. For progressive tumor growth, the vessel network must continuously expand to satisfy the neoplastic cells' nutritional needs and waste product removal requirements. This utter reliance of the tumor on its vasculature provides a logical target for new approaches to cancer therapy. Indeed, there currently exists a great deal of enthusiasm for the development of interventions that compromise the growth and/or function of the tumor neovasculature. Two primary directions are being pursued. Inhibitors of angiogenesis seek to interrupt the angiogenic process to prevent new vessel formation. Antivascular approaches aim to cause direct damage to the tumor endothelium and thus lead to extensive secondary neoplastic cell death. The application of such strategies as adjuvants to conventional radiation treatments offers unique opportunities to develop more effective cancer therapies.
Collapse
Affiliation(s)
- Dietmar W Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL 32610, USA
| | | |
Collapse
|
79
|
|
80
|
Abstract
Antiangiogenic drugs are unique for having highly specific targets while carrying the potential to be effective against a wide variety of tumors. Moreover, some of the major limitations of cytotoxic therapies likely will be avoided by this entirely new class of anticancer weapons. After the realization of the potential advantages of antiangiogenic therapy, the field of angiogenesis research is growing exponentially. Still, there is much to learn about the machinery that tumors use to recruit new blood vessels, and the results of the clinical trials will show the best way to apply that knowledge for cancer therapy.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Angiostatins
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Anticarcinogenic Agents/pharmacology
- Anticarcinogenic Agents/therapeutic use
- Cell Hypoxia/physiology
- Child
- Clinical Trials as Topic
- Collagen/physiology
- Cyclooxygenase Inhibitors/pharmacology
- Cyclooxygenase Inhibitors/therapeutic use
- Drug Design
- Endostatins
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Ephrins/physiology
- Growth Substances/physiology
- Humans
- Immunotherapy
- Integrin alphaVbeta3/antagonists & inhibitors
- Integrin alphaVbeta3/physiology
- Ligases/physiology
- Matrix Metalloproteinase Inhibitors
- Matrix Metalloproteinases/physiology
- Mice
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/physiology
- Neoplasms/blood supply
- Neoplasms/drug therapy
- Neovascularization, Pathologic/drug therapy
- Outcome Assessment, Health Care
- Peptide Fragments/physiology
- Plasminogen/physiology
- Protease Inhibitors/pharmacology
- Protease Inhibitors/therapeutic use
- Receptors, Eph Family/antagonists & inhibitors
- Receptors, Eph Family/physiology
- Receptors, Growth Factor/antagonists & inhibitors
- Receptors, Growth Factor/physiology
- Thrombospondins/physiology
- Tumor Suppressor Proteins
- Ubiquitin-Protein Ligases
- Von Hippel-Lindau Tumor Suppressor Protein
Collapse
Affiliation(s)
- Kerim Kaban
- Department of Thoracic Head and Neck Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|