651
|
Giavazzi R, Bani MR, Taraboletti G. Tumor’host interaction in the optimization of paclitaxel-based combination therapies with vascular targeting compounds. Cancer Metastasis Rev 2007; 26:481-8. [PMID: 17896168 DOI: 10.1007/s10555-007-9074-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Targeting of the tumor stroma, including the tumor vasculature, represents a new frontier in the treatment of malignancy. Preclinical studies and clinical experiences have established that stroma-directed novel agents must be combined with conventional therapies in order to achieve relevant therapeutic efficacy. Here we review our preclinical experience on combinations of paclitaxel with a tyrosine kinase receptor inhibitor of angiogenesis (SU6668) and a vascular disrupting agent (VDA, ZD6126), and discuss the critical factors that determine the outcome of these treatments. We also analyze the relevance of the intrinsic sensitivity of the tumor to the drugs, as well as the possibility that the two combined agents synergistically affect the vasculature or independently target the host and the tumor compartments. Finally, we discuss the need to carefully optimize scheduling and sequencing, through the use of reliable end points, in order to avoid negative pharmacological interactions and to improve the antineoplastic efficacy of paclitaxel-based combination treatments.
Collapse
Affiliation(s)
- Raffaella Giavazzi
- Laboratory of Biology and Treatment of Metastasis, Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo, Italy.
| | | | | |
Collapse
|
652
|
Varache-Lembège M, Moreau S, Larrouture S, Montaudon D, Robert J, Nuhrich A. Synthesis and antiproliferative activity of aryl- and heteroaryl-hydrazones derived from xanthone carbaldehydes. Eur J Med Chem 2007; 43:1336-43. [PMID: 17949859 DOI: 10.1016/j.ejmech.2007.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 07/27/2007] [Accepted: 09/06/2007] [Indexed: 01/06/2023]
Abstract
In order to explore the antiproliferative effect associated with the xanthone framework, several arylhydrazonomethyl derivatives were synthesized from various isomeric 1,3-dihydroxyxanthone carbaldehydes. Variation in the position of the aldehydic function led to three sets of compounds, bearing the hydrazonomethyl chain at positions 5, 6 or 7 on the xanthone nucleus, respectively. The antiproliferative effect of the compounds was evaluated in vitro using the MTT colorimetric method against two human cancer cell lines (MCF-7, breast adenocarcinoma, and KB 3.1, squamous cell oral carcinoma) for two time periods (24 h and 72 h). Among the series, four compounds exhibited interesting growth inhibitory effects against both the cell lines, with IC(50) values in the micromolar concentration range. When compared with doxorubicin, the xanthone derivatives showed moderate cytotoxic effects. Surprisingly, unlike doxorubicin, these compounds displayed no significant time-dependent change in the concentration causing 50% inhibitory effect in proliferation. This unusual cytotoxicity profile led to the hypothesis that these molecules could be endowed with a mechanism of action distinct to that of doxorubicin.
Collapse
Affiliation(s)
- Martine Varache-Lembège
- Laboratoire de Chimie Thérapeutique, EA2962, Faculté de Pharmacie, Université Victor Segalen Bordeaux 2, 146 Rue Léo-Saignat, 33076 Bordeaux Cedex, France
| | | | | | | | | | | |
Collapse
|
653
|
Shaked Y, Kerbel RS. Antiangiogenic strategies on defense: on the possibility of blocking rebounds by the tumor vasculature after chemotherapy. Cancer Res 2007; 67:7055-8. [PMID: 17671170 DOI: 10.1158/0008-5472.can-07-0905] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rapid or accelerated tumor cell repopulation after significant tumor cell killing induced by various cytotoxic agents often compromises the expected therapeutic benefit of such tumor responses. Here, we discuss the concept that tumor cell repopulation after certain cytotoxic therapies, using vascular disrupting agents as an example, may be aided by a reactive, systemic host response involving the mobilization of bone marrow-derived circulating cells, including endothelial progenitor cells, which subsequently home to the vasculature of treated tumors and promote tumor neovascularization. These vasculogenic "rebounds" can be blocked, at least in some cases, by treatment with an antiangiogenic drug. There is limited preliminary evidence that maximum tolerated dose chemotherapy causes a similar effect. This could constitute one way by which antiangiogenic therapy could increase the efficacy of conventional cytotoxic chemotherapy regimens; it also raises the specter of new molecular targets for systemic cancer therapies which are involved in therapy-induced bone marrow-derived cell mobilization, homing to tumors, and tumor retention.
Collapse
Affiliation(s)
- Yuval Shaked
- Sunnybrook Health Sciences Centre, Molecular and Cellular Biology Research, and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
654
|
Martinelli M, Bonezzi K, Riccardi E, Kuhn E, Frapolli R, Zucchetti M, Ryan AJ, Taraboletti G, Giavazzi R. Sequence dependent antitumour efficacy of the vascular disrupting agent ZD6126 in combination with paclitaxel. Br J Cancer 2007; 97:888-94. [PMID: 17848949 PMCID: PMC2360417 DOI: 10.1038/sj.bjc.6603969] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The clinical success of small-molecule vascular disrupting agents (VDAs) depends on their combination with conventional therapies. Scheduling and sequencing remain key issues in the design of VDA-chemotherapy combination treatments. This study examined the antitumour activity of ZD6126, a microtubule destabilising VDA, in combination with paclitaxel (PTX), a microtubule-stabilising cytotoxic drug, and the influence of schedule and sequence on the efficacy of the combination. Nude mice bearing MDA-MB-435 xenografts received weekly cycles of ZD6126 (200 mg kg(-1) i.p.) administered at different times before or after PTX (10, 20, and 40 mg kg(-1) i.v.). ZD6126 given 2 or 24 h after PTX showed no significant benefit, a result that was attributed to a protective effect of PTX against ZD6126-induced vascular damage and tumour necrosis, a hallmark of VDA activity. Paclitaxel counteracting activity was reduced by distancing drug administrations, and ZD6126 given 72 h after PTX potentiated the VDA's antitumour activity. Schedules with ZD6126 given before PTX improved therapeutic activity, which was paralleled by a VDA-induced increase in cell proliferation in the viable tumour tissue. Paclitaxel given 72 h after ZD6126 yielded the best response (50% tumours regressing). A single treatment with ZD6126 followed by weekly administration of PTX was sufficient to achieve a similar response (57% remissions). These findings show that schedule, sequence and timing are crucial in determining the antitumour efficacy of PTX in combination with ZD6126. Induction of tumour necrosis and increased proliferation in the remaining viable tumour tissue could be exploited as readouts to optimise schedules and maximise therapeutic efficacy.
Collapse
Affiliation(s)
- M Martinelli
- Laboratory of Biology and Treatment of Metastasis, Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo 24125, Italy
| | - K Bonezzi
- Laboratory of Biology and Treatment of Metastasis, Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo 24125, Italy
| | - E Riccardi
- Laboratory of Biology and Treatment of Metastasis, Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo 24125, Italy
| | - E Kuhn
- Laboratory of Cancer Pharmacology, Department of Oncology, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - R Frapolli
- Laboratory of Cancer Pharmacology, Department of Oncology, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - M Zucchetti
- Laboratory of Cancer Pharmacology, Department of Oncology, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - A J Ryan
- AstraZeneca, Alderley Park, Macclesfield SK10 4TG, UK
| | - G Taraboletti
- Laboratory of Biology and Treatment of Metastasis, Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo 24125, Italy
- E-mail:
| | - R Giavazzi
- Laboratory of Biology and Treatment of Metastasis, Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo 24125, Italy
| |
Collapse
|
655
|
Liou JP, Wu CY, Hsieh HP, Chang CY, Chen CM, Kuo CC, Chang JY. 4- and 5-aroylindoles as novel classes of potent antitubulin agents. J Med Chem 2007; 50:4548-52. [PMID: 17685504 DOI: 10.1021/jm070557q] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A novel series of 4- and 5-aroylindole derivatives was prepared and evaluated for antitumor activity. Several compounds showed excellent antiproliferative activity as inhibitors of tubulin polymerization. Compounds 13, 14, 15, and 18, with IC50 values of 1.9, 1.1, 1.2, and 1.8 microM, respectively, exhibited more potent inhibition of tubulin polymerization than colchicine. They also displayed antiproliferative activity, with IC50 values ranging from 10 to 43 nM in a variety of human cell lines from different organs.
Collapse
Affiliation(s)
- Jing-Ping Liou
- College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
656
|
Farace F, Massard C, Borghi E, Bidart JM, Soria JC. Vascular disrupting therapy-induced mobilization of circulating endothelial progenitor cells. Ann Oncol 2007; 18:1421-2. [PMID: 17693656 DOI: 10.1093/annonc/mdm367] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
657
|
Meng F, Cai X, Duan J, Matteucci MG, Hart CP. A novel class of tubulin inhibitors that exhibit potent antiproliferation and in vitro vessel-disrupting activity. Cancer Chemother Pharmacol 2007; 61:953-63. [PMID: 17639393 DOI: 10.1007/s00280-007-0549-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 06/05/2007] [Indexed: 01/03/2023]
Abstract
PURPOSE Since anticancer agents that interfere with microtubule function are in widespread use and have a broad spectrum of activity against both hematological malignancies and solid tumors, there is an urgent need to develop novel tubulin inhibitors with broader activities and avoiding drug resistance. METHODS AND RESULTS In this study, we describe the characterization of select lead compounds from a novel class of indazole-based tubulin inhibitors. Three lead compounds, TH-337, TH-482 and TH-494, exhibit potent antiproliferative activity against cell lines derived from human pancreatic carcinoma, human breast adenocarcinoma and human colorectal adenocarcinoma cells. The three compounds were also tested for cytotoxicity against a panel of clinically relevant drug resistant cancer cell lines that either overexpress the drug resistance pumps MDR-1, MRP-1 and BCRP-1 or have altered Topoisomerase II activity. TH-482 and -494 retained cytotoxic activities against all of the resistant cell lines tested; however, TH-337 exhibited decreased cytotoxicity in the cell line overexpressing BCRP-1, indicating that TH-337 is a substrate of that pump. We show that TH-482's antiproliferative activity is due to cell cycle arrest at the G(2)/M phase. We demonstrate that TH-482 binds specifically to the colchicine site of tubulin and that it inhibits tubulin polymerization in vitro in a concentration-dependent manner. The in vitro anti-vascular activities of TH-482 were assessed using the HUVEC-C cell line. TH-482 inhibits in vitro neovessel formation and disrupts pre-established vessels using HUVEC-C cells. TH-482 also increases permeability of vascular endothelial cells in a concentration- and time-dependent manner. CONCLUSIONS TH-482 demonstrates potent in vitro efficacy as a novel tubulin-targeted anti-proliferative and anti-vascular agent and notably is more potent in antiproliferative assays than the benchmark compound combretastatin A-4. These results identify TH-482 as a potent tubulin inhibitor, and support the investigation of its in vivo efficacy and pharmacokinetic properties as the prototype of a new class of anti-tubulin agents.
Collapse
Affiliation(s)
- Fanying Meng
- Threshold Pharmaceuticals, Inc., 1300 Seaport Blvd, Redwood City, CA 94063, USA.
| | | | | | | | | |
Collapse
|
658
|
Palmer BD, Henare K, Woon ST, Sutherland R, Reddy C, Wang LCS, Kieda C, Ching LM. Synthesis and biological activity of azido analogues of 5,6-dimethylxanthenone-4-acetic acid for use in photoaffinity labeling. J Med Chem 2007; 50:3757-64. [PMID: 17616114 DOI: 10.1021/jm0702175] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
5,6-Dimethylxanthenone-4-acetic acid (1) is scheduled for phase III clinical trials as a vascular disrupting agent. However, its biochemical receptor(s) have yet to be identified. In this report, the synthesis of azido analogues of 1 that could be used for photoaffinity labeling of proteins as an approach toward identifying its molecular targets is described. While 5-azidoxanthenone-4-acetic acid (2) and 5-azido-6-methylxantheone-4-acetic acid (3) were found to have biological activities similar to that of 1, 6-azido-5-methylxanthenone-4-acetic acid (4) was unstable and could not be evaluated. Both azido compounds 2 and 3 activated NF-kappaB, induced the production of tumor necrosis factor in cultured mouse splenocytes, and induced hemorrhagic necrosis of colon 38 tumors in mice. Photoreaction of lysates from spleen cells with tritiated 2 resulted in two radiolabeled protein bands at 50 and 14 kDa that could be competitively inhibited with cold 1 and cold 2. The azido compounds 2 and 3 exhibit all the requirements for use in photoaffinity labeling of potential receptor(s) for 1.
Collapse
Affiliation(s)
- Brian D Palmer
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
659
|
Salmon BA, Salmon HW, Siemann DW. Monitoring the treatment efficacy of the vascular disrupting agent CA4P. Eur J Cancer 2007; 43:1622-9. [PMID: 17451938 PMCID: PMC2962830 DOI: 10.1016/j.ejca.2007.03.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Revised: 03/19/2007] [Accepted: 03/23/2007] [Indexed: 02/08/2023]
Abstract
The purpose of this study was to investigate two non-invasive methods for determining the treatment efficacy of the vascular disrupting agent (VDA) CA4P: gadolinium enhanced dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) for perfusion analysis and enzyme-linked immunosorbent assay (ELISA) of blood samples. Candidate proteins were identified by multi-analyte profile analysis of plasma from KHT sarcoma-bearing C3H/HeJ mice after CA4P administration. Candidate proteins were further analysed by ELISA of plasma from treated C3H/HeJ, BALBc and C57BL6 mice. Changes in selected proteins, tumour perfusion and tumour necrotic fraction after CA4P treatment were then compared in individual animals. The cytokines KC and MCP-1 were observed to increase after CA4P treatment in all tested models. No correlation was found between KC or MCP-1 levels and tumour necrosis. However, tumour perfusion correlated (r=0.89, p<0.00001) with CA4P treatment efficacy as measured by necrotic fraction, suggesting that DCE-MRI may have utility in a clinical setting.
Collapse
Affiliation(s)
- Beth A. Salmon
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | | | - Dietmar W. Siemann
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
- Department of Radiation Oncology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
660
|
Abstract
Clinical trials showing longer survival when chemotherapy is combined with antiangiogenic agents (AAs) have led to growing interest in designing combined modality protocols that exploit abnormalities in tumor vasculature. Approved agents include bevacizumab, a recombinant monoclonal antibody that binds to vascular endothelial growth factor, and two small molecule multitargeted tyrosine kinase inhibitors of angiogenesis (SU11248 and BAY-43-9006) that have been approved for therapy of renal cancer. Targeting tumor vasculature has a strong biological rationale in radiation therapy, and preclinical studies consistently show an increase in radiosensitization with combined treatment. Preclinical studies indicate that excessive damage to tumor vasculature can result in radioresistance in some situations, and early clinical data suggest that treatment sequencing may be important when combining AAs with radiation. Radiation itself appears to antagonize any hypoxia that can be induced by long-term administration of AAs. The optimal biological doses of AAs with radiotherapy are unknown, and surrogate markers of efficacy remain to be validated. Early clinical trials should therefore include studies designed to identify mechanisms of interaction and increases in tumor hypoxia. This review highlights preclinical and early clinical data that are relevant for clinical trial design. Optimal radiation planning and delivery is required to minimize the volume of irradiated normal organs and to establish safe dose-volume parameters for phase II-III clinical trials.
Collapse
Affiliation(s)
- Suresh Senan
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, The Netherlands.
| | | |
Collapse
|
661
|
Toffoli S, Feron O, Raes M, Michiels C. Intermittent hypoxia changes HIF-1alpha phosphorylation pattern in endothelial cells: unravelling of a new PKA-dependent regulation of HIF-1alpha. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1558-71. [PMID: 17662481 DOI: 10.1016/j.bbamcr.2007.06.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 05/22/2007] [Accepted: 06/06/2007] [Indexed: 01/09/2023]
Abstract
Vascularized tumors are exposed to intermittent hypoxia, that is, hypoxia followed by periods of reoxygenation. Abnormal structure and dysfunction of tumor blood vessels are responsible for these conditions. These repeated short periods of hypoxia concern tumor cells as well as endothelial cells. However, the effects of intermittent hypoxia are poorly understood. The aim of this study was to investigate the effects of intermittent hypoxia on endothelial cells and particularly on HIF-1alpha, a central actor in adaptive response to hypoxia. For that, endothelial cells were exposed to four repeated cycles of 1-h hypoxia followed by 30 min of reoxygenation. We showed that repeated cycles of hypoxia/reoxygenation induced a modification in HIF-l alpha phosphorylation pattern: a progressive increase in HIF-1alpha phosphorylated form was observed during the hypoxic periods. Activation of p42/p44, Akt and PKA was observed in parallel. PKA was shown to be involved in the phosphorylation of HIF-lalpha under intermittent hypoxia, while p42/p44 and Akt were not. As HIF-1 activity is often associated with enhanced cell survival, a better knowledge of the effects of intermittent hypoxia on endothelial cells and the highlight of particular mechanisms induced by intermittent hypoxia are essential to understand the behavior of endothelial cells during neo-angiogenesis.
Collapse
Affiliation(s)
- Sébastien Toffoli
- Laboratory of Biochemistry and Cellular Biology, FUNDP-University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium
| | | | | | | |
Collapse
|
662
|
Roberts ZJ, Goutagny N, Perera PY, Kato H, Kumar H, Kawai T, Akira S, Savan R, van Echo D, Fitzgerald KA, Young HA, Ching LM, Vogel SN. The chemotherapeutic agent DMXAA potently and specifically activates the TBK1-IRF-3 signaling axis. ACTA ACUST UNITED AC 2007; 204:1559-69. [PMID: 17562815 PMCID: PMC2118649 DOI: 10.1084/jem.20061845] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vascular disrupting agents (VDAs) represent a novel approach to the treatment of cancer, resulting in the collapse of tumor vasculature and tumor death. 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is a VDA currently in advanced phase II clinical trials, yet its precise mechanism of action is unknown despite extensive preclinical and clinical investigations. Our data demonstrate that DMXAA is a novel and specific activator of the TANK-binding kinase 1 (TBK1)–interferon (IFN) regulatory factor 3 (IRF-3) signaling pathway. DMXAA treatment of primary mouse macrophages resulted in robust IRF-3 activation and ∼750-fold increase in IFN-β mRNA, and in contrast to the potent Toll-like receptor 4 (TLR4) agonist lipopolysaccharide (LPS), signaling was independent of mitogen-activated protein kinase (MAPK) activation and elicited minimal nuclear factor κB–dependent gene expression. DMXAA-induced signaling was critically dependent on the IRF-3 kinase, TBK1, and IRF-3 but was myeloid differentiation factor 88–, Toll–interleukin 1 receptor domain–containing adaptor inducing IFN-β–, IFN promoter-stimulator 1–, and inhibitor of κB kinase–independent, thus excluding all known TLRs and cytosolic helicase receptors. DMXAA pretreatment of mouse macrophages induced a state of tolerance to LPS and vice versa. In contrast to LPS stimulation, DMXAA-induced IRF-3 dimerization and IFN-β expression were inhibited by salicylic acid. These findings detail a novel pathway for TBK1-mediated IRF-3 activation and provide new insights into the mechanism of this new class of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Zachary J Roberts
- Department of Microbiology and Immunology, University of Maryland-Baltimore, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
663
|
Kanthou C, Kranjc S, Sersa G, Tozer G, Zupanic A, Cemazar M. The endothelial cytoskeleton as a target of electroporation-based therapies. Mol Cancer Ther 2007; 5:3145-52. [PMID: 17172418 DOI: 10.1158/1535-7163.mct-06-0410] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Electroporation-based therapies, such as electrochemotherapy and electrogene therapy, result in the disruption of blood vessel networks in vivo and cause changes in blood flow and vascular permeability. The effects of electroporation on the cytoskeleton of cultured primary endothelial cells and on endothelial monolayer permeability were investigated to elucidate possible mechanisms involved. Human umbilical vein endothelial cells (HUVECs) were electroporated in situ and then immunofluorescence staining for filamentous actin, beta-tubulin, vimentin, and VE-cadherin as well as Western blotting analysis of levels of phosphorylated myosin light chain and cytoskeletal proteins were performed. Endothelial permeability was determined by monitoring the passage of FITC-coupled dextran through endothelial monolayers. Exposure of endothelial cells to electric pulses resulted in a profound disruption of microfilament and microtubule cytoskeletal networks, loss of contractility, and loss of vascular endothelial cadherin from cell-to-cell junctions immediately after electroporation. These effects were voltage dependent and reversible because cytoskeletal structures recovered within 60 min of electroporation with up to 40 V, without any significant loss of cell viability. The cytoskeletal effects of electroporation were paralleled by a rapid increase in endothelial monolayer permeability. These results suggest that the remodeling of the endothelial cytoskeleton and changes in endothelial barrier function could contribute to the vascular disrupting actions of electroporation-based therapies and provide an insight into putative mechanisms responsible for the observed increase in permeability and cessation of blood flow in vivo.
Collapse
Affiliation(s)
- Chryso Kanthou
- Cancer Research UK Tumour Microcirculation Group, Academic Unit of Surgical Oncology, Royal Hallamshire Hospital, University of Sheffield, UK
| | | | | | | | | | | |
Collapse
|
664
|
Kador PF, Blessing K, Randazzo J, Makita J, Wyman M. Evaluation of the vascular targeting agent combretastatin a-4 prodrug on retinal neovascularization in the galactose-fed dog. J Ocul Pharmacol Ther 2007; 23:132-42. [PMID: 17444801 DOI: 10.1089/jop.2006.0103] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Combretastatin A-4 (CA-4) is a vascular targeting agent known to rapidly shut off blood flow in new vessels and, as a result, regress neovascularization. In this pilot study, the ability of CA-4 to modify retinal neovascularization, which results in altered retinal vessel blood flow and retinal permeability, was evaluated in aphakic long-term galactose-fed beagles, an animal model that develops diabetes-like retinal neovascularization. METHODS Two (2) groups of aphakic dogs, each group comprised of 4 galactose-fed dogs and 2 age-matched controls dogs, were utilized. Each group initially received the combretastatin A-4-phosphate prodrug (CA-4P) as either sub-Tenon's injections, administered at the corneoscleral junction, or intravitreal injections. Six (6) weeks after this treatment, all dogs also received systemic (intravenous) injections of CA-4P. Retinal vascular changes were monitored at 2-week intervals by fluorescein angiography. RESULTS All galactose-fed dogs demonstrated the presence of retinal neovascular lesions by fluorescein angiograms. Fluorescein leakage or perfusion through neovascular vessels was not altered by either sub-Tenon's, intravitreal, or systemic CA-4P administration. Whereas CA-4P was well tolerated by the healthy eyes of the control animals, its administration to some galactose-fed dogs was associated with corneal edema and increases in intraocular pressure following sub-Tenon's and intraocular injections. CONCLUSIONS Neovascularization in the galactose-fed dog progresses over a period of years, similar to that observed with clinical diabetic retinopathy. The failure of CA-4P to ameliorate neovascularization suggests that chronic, long-term administration may be required to destroy the slowly growing retinal endothelial cells.
Collapse
Affiliation(s)
- Peter F Kador
- Laboratory of Ocular Therapeutics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | |
Collapse
|
665
|
Salmon BA, Siemann DW. Characterizing the tumor response to treatment with combretastatin A4 phosphate. Int J Radiat Oncol Biol Phys 2007; 68:211-7. [PMID: 17448875 PMCID: PMC1868579 DOI: 10.1016/j.ijrobp.2006.12.051] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2006] [Revised: 12/08/2006] [Accepted: 12/22/2006] [Indexed: 12/22/2022]
Abstract
PURPOSE To examine the pathophysiologic impact of treatment with combretastatin A4 phosphate (CA4P) in regions of tumors that ultimately either necrose or survive treatment with this agent. METHODS AND MATERIALS Proliferation, perfusion, vessel density, and expression of vascular endothelial growth factor (VEGF) were analyzed in the KHT tumor model after treatment with CA4P. Analyses were conducted in the whole tumor and the tumor periphery. RESULTS Perfusion in the tumor periphery decreased 4 h after treatment, but returned to baseline 20 h later. Whole-tumor perfusion also decreased 4 h after treatment, but did not return to baseline. Vessel density decreased in the tumor as a whole, but not in the tumor periphery. No significant effect on the expression of VEGF was observed, but a decrease in proliferation in the whole tumor and the periphery was noted. CONCLUSIONS The present study shows that those areas of a tumor that survive treatment with CA4P are affected by CA4P exposure, though only transiently. The decrease in perfusion could negatively affect therapies utilizing the combination of CA4P and conventional anticancer agents by decreasing drug delivery and tissue oxygenation. These findings suggest that the timing of CA4P treatments when used in conjunction with conventional anticancer therapies should be considered carefully.
Collapse
Affiliation(s)
- Beth A Salmon
- Department of Pharmacology and Therapeutics, Shands Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | | |
Collapse
|
666
|
Bouzin C, Feron O. Targeting tumor stroma and exploiting mature tumor vasculature to improve anti-cancer drug delivery. Drug Resist Updat 2007; 10:109-20. [PMID: 17452119 DOI: 10.1016/j.drup.2007.03.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 03/06/2007] [Accepted: 03/06/2007] [Indexed: 02/08/2023]
Abstract
The identification of a critical role of tumour stroma in the regulation of tumour interstitial fluid pressure and the simultaneous discovery of the impact of anti-angiogenic drugs on tumour hemodynamics have provided new potential for improving tumour delivery of anti-cancer drugs. Here, we review the most recent studies investigating how tumour-associated fibroblasts and macrophages as well as the extracellular matrix itself may be targeted to facilitate delivery of both low-molecular weight drugs and macromolecules. In addition, we summarize the current understanding of the use of vasoactive compounds, radiotherapy and vascular-disrupting agents as potential adjuvants to maximize tumour delivery of anti-cancer drugs. The impact of these strategies on the diffusive and convective modes of drug transport is discussed in the light of Fick's and Starling's laws. Finally, we discuss how transcytosis through caveolae may also be exploited to optimize the selective delivery of conventional chemotherapy to the subendothelial tumour cell compartment.
Collapse
Affiliation(s)
- Caroline Bouzin
- UCL Medical School, Unit of Pharmacology and Therapeutics (FATH5349), Angiogenesis and Cancer Research Group, 52 Avenue E. Mounier, B-1200 Brussels, Belgium
| | | |
Collapse
|
667
|
Rajeshkumar NV, Matwyshyn G, Gulati A. IRL-1620, a tumor selective vasodilator, augments the uptake and efficacy of chemotherapeutic agents in prostate tumor rats. Prostate 2007; 67:701-13. [PMID: 17342753 DOI: 10.1002/pros.20556] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND IRL-1620, a potent endothelin B receptor agonist, enhanced the efficacy of paclitaxel in a breast tumor model, but its effect in prostate cancer is not known. The present study was conducted to evaluate the effect of IRL-1620 on tumor perfusion, uptake of [(14)C]-doxorubicin in the tumor and efficacy of doxorubicin (DOX), and 5-flurouracil (5-FU) in a rat prostate tumor model. METHODS JHU-4 (Mat-Lu) cells inoculated prostate tumor model in Copenhagen rats was used for the study. RESULTS Administration of IRL-1620 (3 nmol/kg, i.v) significantly increased (102.8%) prostate tumor perfusion and tumor uptake of [(14)C]-doxorubicin (115%) compared to vehicle treated rats. Results of the efficacy study demonstrate that IRL-1620 administration 15 min prior to DOX (5 mg/kg) or 5-FU (50 mg/kg) on every third day for a total of four doses significantly reduced tumor volume compared to vehicle treated rats. CONCLUSIONS IRL-1620 significantly enhanced the uptake and efficacy of anticancer agents in prostate cancer.
Collapse
Affiliation(s)
- N V Rajeshkumar
- Department of Biopharmaceutical Sciences, University of Illinois, Chicago, Illinois, USA
| | | | | |
Collapse
|
668
|
Wardman P. Chemical radiosensitizers for use in radiotherapy. Clin Oncol (R Coll Radiol) 2007; 19:397-417. [PMID: 17478086 DOI: 10.1016/j.clon.2007.03.010] [Citation(s) in RCA: 318] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Accepted: 03/13/2007] [Indexed: 12/21/2022]
Abstract
Radiosensitizers are intended to enhance tumour cell killing while having much less effect on normal tissues. Some drugs target different physiological characteristics of the tumour, particularly hypoxia associated with radioresistance. Oxygen is the definitive hypoxic cell radiosensitizer, the large differential radiosensitivity of oxic vs hypoxic cells being an attractive factor. The combination of nicotinamide to reduce acute hypoxia with normobaric carbogen breathing is showing clinical promise. 'Electron-affinic' chemicals that react with DNA free radicals have the potential for universal activity to combat hypoxia-associated radioresistance; a nitroimidazole, nimorazole, is clinically effective at tolerable doses. Hypoxia-specific cytotoxins, such as tirapazamine, are valuable adjuncts to radiotherapy. Nitric oxide is a potent hypoxic cell radiosensitizer; variations in endogenous levels might have prognostic significance, and routes to deliver nitric oxide specifically to tumours are being developed. In principle, many drugs can be delivered selectively to hypoxic tumours using either reductase enzymes or radiation-produced free radicals to activate drug release from electron-affinic prodrugs. A redox-active agent based on a gadolinium chelate is being evaluated clinically. Pyrimidines substituted with bromine or iodine are incorporated into DNA and enhance free radical damage; fluoropyrimidines act by different mechanisms. A wide variety of drugs that influence the nature or repair of DNA damage are being evaluated in conjunction with radiation; it is often difficult to define the mechanisms underlying chemoradiation regimens. Drugs being evaluated include topoisomerase inhibitors (e.g. camptothecin, topotecan), and the hypoxia-activated anthraquinone AQ4N; alkylating agents include temozolomide. Drugs involved in DNA repair pathways being investigated include the potent poly(ADP ribose)polymerase inhibitor, AG14,361. Proteins involved in cell signalling, such as the Ras family, are attractive targets linked to radioresistance, as are epidermal growth factor receptors and linked kinases (drugs including vandetanib [ZD6,474], cetuximab and gefitinib), and cyclooxygenase-2 (celecoxib). The suppression of radioprotective thiols seems to offer more potential with alkylating agents than with radiotherapy, although it remains a strategy worthy of exploration.
Collapse
Affiliation(s)
- P Wardman
- University of Oxford, Gray Cancer Institute, PO Box 100, Mount Vernon Hospital, Northwood HA6 2JR, UK.
| |
Collapse
|
669
|
Chung F, Liu J, Ching LM, Baguley BC. Consequences of increased vascular permeability induced by treatment of mice with 5,6-dimethylxanthenone-4-acetic acid (DMXAA) and thalidomide. Cancer Chemother Pharmacol 2007; 61:497-502. [PMID: 17473922 DOI: 10.1007/s00280-007-0495-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Accepted: 04/04/2007] [Indexed: 10/23/2022]
Abstract
PURPOSE 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) (AS1404), a small-molecule vascular disrupting agent currently in clinical trial, increases vascular permeability and decreases blood flow in both murine and human tumours. DMXAA induces tumour necrosis factor (TNF) in mice and the effects on vascular permeability are hypothesised to result from both direct (DMXAA) and indirect (TNF) effects. Skin temperature decreases in mice treated with high doses of DMXAA, raising the question of whether host toxicity is mediated by the induction of increased vascular permeability in normal tissue. Thalidomide is an anti-inflammatory agent that potentiates the anti-tumour activity of DMXAA but decreases induction of TNF in plasma. We wished to determine how it potentiated the effects of DMXAA. METHODS Vascular permeability was measured in Colon 38 tumour and liver tissue by uptake of Evans Blue dye. Blood haematocrit and body temperature were also measured. RESULTS Tumour vascular permeability was increased following administration of DMXAA (25 mg/kg i.p.), minimally affected following thalidomide (100 mg/kg i.p.) but strongly increased following co-administration of both drugs. In contrast, dye uptake into liver tissue was decreased following administration of DMXAA, thalidomide or both drugs. Administration of DMXAA at a potentially toxic dose (35 mg/kg i.p. or 50 mg/kg orally) was found to decrease body temperature and to increase the blood haematocrit, while administration of thalidomide alone (100 mg/kg i.p.) had no effect. Co-administration of thalidomide potentiated the effects of DMXAA on both body temperature and haematocrit but surprisingly did not increase toxicity. CONCLUSIONS The results are consistent with the hypothesis that the host toxicity of high-dose DMXAA is mediated by effects on host vasculature. Co-administration of thalidomide increases the effective dose of DMXAA by reducing clearance but also, by inhibiting production of circulating TNF, reduces the host toxicity of DMXAA.
Collapse
Affiliation(s)
- Francisco Chung
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | |
Collapse
|
670
|
Bellina F, Cauteruccio S, Rossi R. Synthesis and biological activity of vicinal diaryl-substituted 1H-imidazoles. Tetrahedron 2007. [DOI: 10.1016/j.tet.2007.02.075] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
671
|
Romagnoli R, Baraldi PG, Carrion MD, Lopez Cara C, Preti D, Fruttarolo F, Pavani MG, Tabrizi MA, Tolomeo M, Grimaudo S, Di Cristina A, Balzarini J, Hadfield JA, Brancale A, Hamel E. Synthesis and Biological Evaluation of 2- and 3-Aminobenzo[b]thiophene Derivatives as Antimitotic Agents and Inhibitors of Tubulin Polymerization. J Med Chem 2007; 50:2273-7. [PMID: 17419607 DOI: 10.1021/jm070050f] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two new series of inhibitors of tubulin polymerization based on the 2-amino-3-(3,4,5-trimethoxybenzoyl)benzo[b]thiophene molecular skeleton and its 3-amino positional isomer were synthesized and evaluated for antiproliferative activity, inhibition of tubulin polymerization, and cell cycle effects. Although many more 3-amino derivatives have been synthesized so far, the most promising compound in this series was 2-amino-6-methyl-3-(3,4,5-trimethoxybenzoyl)benzo[b]thiophene, which inhibits cancer cell growth at subnanomolar concentrations and interacts strongly with tubulin by binding to the colchicine site.
Collapse
Affiliation(s)
- Romeo Romagnoli
- Dipartimento di Scienze Farmaceutiche, Università di Ferrara, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
672
|
Patterson DM, Rustin GJS. Vascular damaging agents. Clin Oncol (R Coll Radiol) 2007; 19:443-56. [PMID: 17459681 DOI: 10.1016/j.clon.2007.03.014] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 02/21/2007] [Accepted: 03/22/2007] [Indexed: 12/27/2022]
Abstract
To provide a comprehensive overview on vascular targeting agents and the application of radiobiological principles in pre-clinical and clinical studies, we completed a comprehensive review of published medical studies on vascular targeting agents using Pub Med. Vascular targeting agents are now divided into vascular disrupting agents (VDAs), which target the pre-existing tumour vasculature, and angiogenesis inhibitors (AIs), which prevent the formation of new blood vessels. Modest success has been seen when VDAs and AIs are used as single agents and therefore combination therapies that can work in a complimentary and synergistic manner, targeting both the tumour cells and endothelial cells, are needed. Radiobiological principles have been used to increase our understanding of these agents, and can explain the increased efficacy of combination treatments. In particular, the alteration of the tumour microenvironment by AIs and VDAs can lead to enhanced efficacy when combined with chemotherapy or radiotherapy, with phase II/III trials showing encouraging results. The optimal use and scheduling of AIs and VDAs remains to be determined. Further understanding of the mechanisms of action of these potentially very exciting anti-neoplastic agents is urgently required.
Collapse
Affiliation(s)
- D M Patterson
- Department of Medical Oncology, Mount Vernon Cancer Centre, Rickmansworth Road, Northwood, Middlesex HA6 2RN, UK.
| | | |
Collapse
|
673
|
Seshadri M, Spernyak JA, Maiery PG, Cheney RT, Mazurchuk R, Bellnier DA. Visualizing the acute effects of vascular-targeted therapy in vivo using intravital microscopy and magnetic resonance imaging: correlation with endothelial apoptosis, cytokine induction, and treatment outcome. Neoplasia 2007; 9:128-35. [PMID: 17356709 PMCID: PMC1813934 DOI: 10.1593/neo.06748] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 01/05/2007] [Accepted: 01/08/2007] [Indexed: 01/08/2023] Open
Abstract
The acute effects of the vascular-disrupting agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) were investigated in vivo using intravital microscopy (IVM) and magnetic resonance imaging (MRI). Changes in vascular permeability and blood flow of syngeneic CT-26 murine colon adenocarcinomas were assessed at 4 and 24 hours after DMXAA treatment (30 mg/kg, i.p.) and correlated with induction of tumor necrosis factor-alpha (TNF-alpha), endothelial damage [CD31/terminal deoxynucleotidyl transferase (TdT)], and treatment outcome. Intravital imaging revealed a marked increase in vascular permeability 4 hours after treatment, consistent with increases in intratumoral mRNA and protein levels of TNF-alpha. Parallel contrast-enhanced MRI studies showed a approximately 4-fold increase in longitudinal relaxation rates (DeltaR(1)), indicative of increased contrast agent accumulation within the tumor. Dual immunostained tumor sections (CD31/TdT) revealed evidence of endothelial apoptosis at this time point. Twenty-four hours after treatment, extensive hemorrhage and complete disruption of vascular architecture were observed with IVM, along with a significant reduction in DeltaR(1); and virtual absence of CD31 immunostaining. DMXAA-induced tumor vascular damage resulted in significant long-term (60-day) cures compared to untreated controls. Multimodality imaging approaches are useful in visualizing the effects of antivascular therapy in vivo. Such approaches allow cross validation and correlation of findings with underlying molecular changes contributing to treatment outcome.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/drug therapy
- Adenocarcinoma/pathology
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Capillary Permeability/drug effects
- Colonic Neoplasms/blood supply
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/pathology
- Contrast Media
- Drug Delivery Systems
- Drug Screening Assays, Antitumor
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/ultrastructure
- Female
- Hemorrhage/chemically induced
- Magnetic Resonance Imaging/methods
- Mice
- Mice, Inbred BALB C
- Microscopy/methods
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Transplantation/methods
- Pentetic Acid/analogs & derivatives
- Polylysine/analogs & derivatives
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- Skin Window Technique
- Transplantation, Heterotopic
- Transplantation, Isogeneic
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
- Xanthones/pharmacology
- Xanthones/therapeutic use
Collapse
Affiliation(s)
- Mukund Seshadri
- Preclinical Imaging Resources, Department of Cancer Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | |
Collapse
|
674
|
Ley CD, Horsman MR, Kristjansen PEG. Early effects of combretastatin-A4 disodium phosphate on tumor perfusion and interstitial fluid pressure. Neoplasia 2007; 9:108-12. [PMID: 17356706 PMCID: PMC1813937 DOI: 10.1593/neo.06733] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Revised: 01/08/2007] [Accepted: 01/08/2007] [Indexed: 11/18/2022] Open
Abstract
Combretastatin-A4 disodium phosphate (CA4DP) is a vascular-disruptive agent that causes an abrupt decrease in tumor blood flow. The direct actions of CA4DP include increases in vascular permeability and destabilization of the endothelial cytoskeleton, which are thought to contribute to occlusion of the tumor vasculature. It has been proposed that increased permeability causes a transient increase in interstitial fluid pressure (IFP), which in turn could collapse intratumoral blood vessels. We examined the immediate effects of CA4DP on tumor IFP in C3H mammary carcinoma. Mice were treated with 100 mg/kg CA4DP by intraperitoneal injection. Tumor perfusion was recorded by laser Doppler flowmetry at separate time points, and IFP was recorded continuously by the wick-in-needle method. In this study, we found that CA4DP treatment resulted in a rapid reduction in tumor perfusion, followed by a decrease in IFP; no increases in IFP were observed. This suggests that CA4DP-induced reductions in tumor perfusion are not dependent on increases in IFP.
Collapse
Affiliation(s)
- Carsten D Ley
- Laboratory of Experimental Oncology, Institute of Molecular Pathology, University of Copenhagen, Copenhagen, Denmark.
| | | | | |
Collapse
|
675
|
Ng QS, Goh V, Carnell D, Meer K, Padhani AR, Saunders MI, Hoskin PJ. Tumor Antivascular Effects of Radiotherapy Combined with Combretastatin A4 Phosphate in Human Non–Small-Cell Lung Cancer. Int J Radiat Oncol Biol Phys 2007; 67:1375-80. [PMID: 17275203 DOI: 10.1016/j.ijrobp.2006.11.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 11/16/2006] [Accepted: 11/17/2006] [Indexed: 02/08/2023]
Abstract
PURPOSE The tumor vascular effects of radiotherapy and subsequent administration of the vascular disrupting agent combretastatin A4 phosphate (CA4P) were studied in patients with advanced non-small-cell lung cancer using volumetric dynamic contrast-enhanced computed tomography (CT). PATIENTS AND METHODS Following ethical committee approval and informed consent, 8 patients receiving palliative radiotherapy (27 Gy in six fractions, twice weekly) also received CA4P (50 mg/m(2)) after the second fraction of radiotherapy. Changes in dynamic CT parameters of tumor blood volume (BV) and permeability surface area product (PS) were measured for the whole tumor volume, tumor rim, and center after radiotherapy alone and after radiotherapy in combination with CA4P. RESULTS After the second fraction of radiotherapy, 6 of the 8 patients showed increases in tumor PS (23.6%, p = 0.011). Four hours after CA4P, a reduction in tumor BV (22.9%, p < 0.001) was demonstrated in the same 6 patients. Increase in PS after radiotherapy correlated with reduction in BV after CA4P (r = 0.77, p = 0.026). At 72 h after CA4P, there was a sustained reduction in tumor BV of 29.4% (p < 0.001). Both increase in PS after radiotherapy and reduction in BV after CA4P were greater at the rim of the tumor. The BV reduction at the rim was sustained to 72 h (51.4%, p = 0.014). CONCLUSION Radiotherapy enhances the tumor antivascular activity of CA4P in human non-small-cell lung cancer, resulting in sustained tumor vascular shutdown.
Collapse
Affiliation(s)
- Quan-Sing Ng
- Marie Curie Research Wing, Mount Vernon Hospital, Middlesex, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
676
|
Darwiche N, El-Banna S, Gali-Muhtasib H. Cell cycle modulatory and apoptotic effects of plant-derived anticancer drugs in clinical use or development. Expert Opin Drug Discov 2007; 2:361-79. [DOI: 10.1517/17460441.2.3.361] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
677
|
Rofstad EK, Galappathi K, Mathiesen B, Ruud EBM. Fluctuating and Diffusion-Limited Hypoxia in Hypoxia-Induced Metastasis. Clin Cancer Res 2007; 13:1971-8. [PMID: 17360973 DOI: 10.1158/1078-0432.ccr-06-1967] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Most tumors develop regions with hypoxic cells during growth, owing to permanent limitations in oxygen diffusion (chronic or diffusion-limited hypoxia) and/or transient limitations in blood perfusion (acute or fluctuating hypoxia). The aim of this study was to investigate the relative significance of chronic and acute hypoxia in the development of metastatic disease. EXPERIMENTAL DESIGN D-12 and R-18 human melanoma xenografts were used as models of human cancer. D-12 tumors metastasize to the lungs, whereas R-18 tumors develop lymph node metastases. Fraction of radiobiologically hypoxic cells (HF(Rad)) was measured in individual primary tumors by using a radiobiological assay based on the paired survival curve method. Fraction of immunohistochemically hypoxic cells (HF(Imm)) was assessed in the same tumors by using a pimonidazole-based immunohistochemical assay optimized with respect to achieving selective staining of chronically hypoxic cells. HF(Imm) and the difference between HF(Rad) and HF(Imm), HF(Rad) - HF(Imm), were verified to be adequate variables for fraction of chronically hypoxic cells and fraction of acutely hypoxic cells, respectively. RESULTS Chronic as well as acute hypoxia were found to promote spontaneous metastasis of D-12 and R-18 tumors. Acute hypoxia influenced metastasis to a greater extent than chronic hypoxia, partly because the fraction of acutely hypoxic cells was larger than the fraction of chronically hypoxic cells in most tumors and partly because acutely hypoxic cells showed a higher metastatic potential than chronically hypoxic cells. CONCLUSIONS It may be beneficial to focus on fluctuating hypoxia rather than diffusion-limited hypoxia when searching for hypoxia-related prognostic variables and predictive assays.
Collapse
Affiliation(s)
- Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway.
| | | | | | | |
Collapse
|
678
|
Bentzen SM, Harari PM, Bernier J. Exploitable mechanisms for combining drugs with radiation: concepts, achievements and future directions. ACTA ACUST UNITED AC 2007; 4:172-80. [PMID: 17327857 DOI: 10.1038/ncponc0744] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Accepted: 10/06/2006] [Indexed: 12/27/2022]
Abstract
Widening indications for combining radiation therapy with cytotoxic or molecular-targeted drugs have mainly been driven by pragmatic clinical trials. With a flurry of novel drugs in various stages of preclinical and clinical development there is a need to revise the framework that has traditionally been used for discussing possible drug-radiation interactions, especially because many of the new drugs are directed at a specific molecular target. Spatial cooperation, cytotoxic enhancement, biological cooperation, temporal modulation and normal tissue protection are proposed as five primary exploitable mechanisms for the rational combination of drugs with radiation for cancer therapy. These five mechanisms produce different clinical outcomes and, therefore, the optimum clinical end point for assessing therapeutic benefit will depend on the mechanism tested. The dependence of outcome on these mechanisms also affects the selection of preclinical models and the optimum scheduling of the two modalities, i.e. the timing and dosing of the drug relative to the radiation dose fractions. These considerations are discussed in some detail for each mechanism and illustrated with specific clinical examples. Multi-modality therapy for head and neck squamous-cell carcinoma is used to illustrate these concepts. Further clinical progress in this field will require hypothesis-driven trials to ensure efficient identification of treatments with the most favorable risk:benefit ratio.
Collapse
Affiliation(s)
- Søren M Bentzen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, K4/316 Clinical Sciences Center, Madison, WI 53792, USA.
| | | | | |
Collapse
|
679
|
Döme B, Hendrix MJC, Paku S, Tóvári J, Tímár J. Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1-15. [PMID: 17200177 PMCID: PMC1762709 DOI: 10.2353/ajpath.2007.060302] [Citation(s) in RCA: 260] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although cancer cells are not generally controlled by normal regulatory mechanisms, tumor growth is highly dependent on the supply of oxygen, nutrients, and host-derived regulators. It is now established that tumor vasculature is not necessarily derived from endothelial cell sprouting; instead, cancer tissue can acquire its vasculature by co-option of pre-existing vessels, intussusceptive microvascular growth, postnatal vasculogenesis, glomeruloid angiogenesis, or vasculogenic mimicry. The best-known molecular pathway driving tumor vascularization is the hypoxia-adaptation mechanism. However, a broad and diverse spectrum of genetic aberrations is associated with the development of the "angiogenic phenotype." Based on this knowledge, novel forms of antivascular modalities have been developed in the past decade. When applying these targeted therapies, the stage of tumor progression, the type of vascularization of the given cancer tissue, and the molecular machinery behind the vascularization process all need to be considered. A further challenge is finding the most appropriate combinations of antivascular therapies and standard radio- and chemotherapies. This review intends to integrate our recent knowledge in this field into a rational strategy that could be the basis for developing effective clinical modalities using antivascular therapy for cancer.
Collapse
Affiliation(s)
- Balázs Döme
- Department of Tumor Biology and Thoracic Oncology, National Koranyi Institute of Pulmonology, Budapest, Hungary
| | | | | | | | | |
Collapse
|
680
|
Kerr DJ, Hamel E, Jung MK, Flynn BL. The concise synthesis of chalcone, indanone and indenone analogues of combretastatin A4. Bioorg Med Chem 2007; 15:3290-8. [PMID: 17360188 DOI: 10.1016/j.bmc.2007.02.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Revised: 02/07/2007] [Accepted: 02/08/2007] [Indexed: 11/27/2022]
Abstract
A series of aryl- and aroyl-substituted chalcone analogues of the tubulin binding agent combretastatin A4 (1) were prepared, using a recently introduced one-pot palladium-mediated hydrostannylation-coupling reaction sequence. These chalcones were converted to indanones by Nazarov cyclisation, followed by oxidation to give the corresponding indenones. Indenones were also prepared using a palladium-mediated formal [3+2]-cycloaddition process between ortho-halobenzaldehydes and diarylpropynones. All compounds were assessed as inhibitors of tubulin polymerisation, but only E-31 had activity similar to that of 1. However, compound E-31 did not exhibit antiproliferative activity against the MCF-7 cell line.
Collapse
Affiliation(s)
- Daniel J Kerr
- Department of Medicinal Chemistry, Faculty of Pharmacy, Monash University, 381 Royal Pde, Parkville, Australia
| | | | | | | |
Collapse
|
681
|
Hallow DM, Mahajan AD, Prausnitz MR. Ultrasonically targeted delivery into endothelial and smooth muscle cells in ex vivo arteries. J Control Release 2007; 118:285-93. [PMID: 17291619 PMCID: PMC1892790 DOI: 10.1016/j.jconrel.2006.12.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 12/22/2006] [Accepted: 12/28/2006] [Indexed: 11/24/2022]
Abstract
This study tested the hypothesis that ultrasound can target intracellular uptake of drugs into vascular endothelial cells (ECs) at low to intermediate energy and into smooth muscle cells (SMCs) at high energy. Ultrasound-enhanced delivery has been shown to enhance and target intracellular drug and gene delivery in the vasculature to treat cardiovascular disease, but quantitative studies of the delivery process are lacking. Viable ex vivo porcine carotid arteries were placed in a solution containing a model drug, TO-PRO(R)-1, and Optison microbubbles. Arteries were exposed to ultrasound at 1.1 MHz and acoustic energies of 5.0, 66, or 630 J/cm(2). Using confocal microscopy and fluorescent labeling of cells, the artery endothelium and media were imaged to determine the localization and to quantify intracellular uptake and cell death. At low to intermediate ultrasound energy, ultrasound was shown to target intracellular delivery into viable cells that represented 9-24% of exposed ECs. These conditions also typically caused 7-25% EC death. At high energy, intracellular delivery was targeted to SMCs, which was associated with denuding or death of proximal ECs. This work represents the first known in-depth study to evaluate intracellular uptake into cells in tissue. We conclude that significant intracellular uptake of molecules can be targeted into ECs and SMCs by ultrasound-enhanced delivery suggesting possible applications for treatment of cardiovascular diseases and dysfunctions.
Collapse
MESH Headings
- Animals
- Carotid Arteries/cytology
- Carotid Arteries/drug effects
- Carotid Arteries/physiology
- Drug Delivery Systems/methods
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiology
- Female
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Organic Chemicals/administration & dosage
- Swine
- Ultrasonics
Collapse
Affiliation(s)
- Daniel M Hallow
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | | | | |
Collapse
|
682
|
Persano L, Crescenzi M, Indraccolo S. Anti-angiogenic gene therapy of cancer: current status and future prospects. Mol Aspects Med 2007; 28:87-114. [PMID: 17306361 DOI: 10.1016/j.mam.2006.12.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Accepted: 12/18/2006] [Indexed: 12/14/2022]
Abstract
The discovery of endogenous inhibitors of angiogenesis has made it possible to test the hypothesis that blocking the angiogenic switch may keep tumor growth in check, and has added a new investigational arm to the field of cancer gene therapy. Angiogenesis inhibitors are heterogeneous in origin and potency, and their growing list includes proteolysis products of larger molecules with a different function, such as angiostatin, endostatin and vasostatin, modulators of vascular endothelial growth factor activity, such as sFLT-1, and some cytokines/chemokines with marked anti-endothelial activity, such as IL-12, IFN-alpha, and CXCL10. Pre-clinical studies have clearly indicated that these factors are essentially cytostatic and that they need long-term administration in order to obtain prolonged anti-tumor effects, representing a rational basis for their delivery by a gene therapy approach. The experimental approaches attempted to date, reviewed herein, indicate overall that anti-angiogenic gene therapy has efficacy mainly as an early intervention strategy and that a better understanding of the biological mechanisms underlying resistance to angiogenesis inhibition, as well as appropriate combined treatments, are required to generate a conceptual advancement which could drive the field towards successful management of established tumors.
Collapse
Affiliation(s)
- Luca Persano
- Department of Oncology and Surgical Sciences, Oncology Section, University of Padova, Via Gattamelata, 64, 35128 Padova, Italy
| | | | | |
Collapse
|
683
|
Liou JP, Mahindroo N, Chang CW, Guo FM, Lee SWH, Tan UK, Yeh TK, Kuo CC, Chang YW, Lu PH, Tung YS, Lin KT, Chang JY, Hsieh HP. Structure-activity relationship studies of 3-aroylindoles as potent antimitotic agents. ChemMedChem 2007; 1:1106-18. [PMID: 16952120 DOI: 10.1002/cmdc.200600125] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The concise synthesis and structure-activity relationship (SAR) studies of 3-aroylindoles were carried out in an effort to improve the potency and solubility of anticancer drug candidate BPR0L075 (8) by exploring structure modifications through three regimens: substitution of the B ring, at the N1 position, and of the 3-carbonyl linker. The SAR information revealed that the methoxy group of the B ring could be replaced with an electron-donating group such as methyl (in compound 9) or N,N-dimethylamino (in compound 13) while retaining both strong cytotoxic and antitubulin activities. The introduction of amide (compounds 30-33) and carbamate (compounds 34-37) functionalities at the N1 position of 8 gave analogues with potent antiproliferative activities. The cytotoxic potency of 8 was improved by replacing the carbonyl group with sulfide (compound 41) or oxygen (compound 43), indicating that the carbonyl moiety is important but not essential. The N,N-dimethylamino derivative 13 not only displayed potent cytotoxicity and antitubulin activity, but also showed a markedly improved physicochemical profile relative to the parent compound.
Collapse
Affiliation(s)
- Jing-Ping Liou
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 350, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
684
|
Zhao L, Marshall ES, Kelland LR, Baguley BC. Evidence for the involvement of p38 MAP kinase in the action of the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA). Invest New Drugs 2007; 25:271-6. [PMID: 17203401 DOI: 10.1007/s10637-006-9029-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Accepted: 11/27/2006] [Indexed: 10/23/2022]
Abstract
AIMS DMXAA (AS1404), a small-molecule vascular disrupting agent that has now completed Phase II clinical trial, induces endothelial cell apoptosis, increased vascular permeability and decreased tumour blood flow in vivo. Its action is incompletely understood and we wished to develop an in vitro system to study its effects. METHODS Human tumour cell lines developed from aggressive tumours were grown on Matrigel to simulate a tumour microenvironment. Cells were analysed by light microscopy and by gene expression profiling. RESULTS Several cell lines formed networks when grown on Matrigel and the NZM7 melanoma cell line was chosen for further study. Addition of DMXAA at a clinically achievable concentration (30 microg/mL) prevented network formation, but co-addition of SB203580 (10 microM), a selective inhibitor of p38 MAP kinase, reversed the effect of DMXAA and restored network formation. Analysis of expression genes for endothelial and related functions showed that cells growing on Matrigel expressed a pattern similar to that of NZM7 cells growing as xenografts in vivo but different from that of cells grown on standard tissue culture plates. Addition of DMXAA resulted in the inhibition of expression of several genes including the transcriptional activator Ets1 and matrix metalloproteinase-2 (MMP2), but co-addition of SB203580 did not reverse these effects of DMXAA on gene expression. CONCLUSION The results suggest that p38 MAP kinase plays an important role in the action of DMXAA and that growth of tumour cells on Matrigel provides a promising model for further studies on the action of this drug.
Collapse
Affiliation(s)
- Liangli Zhao
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
685
|
Lippert JW. Vascular disrupting agents. Bioorg Med Chem 2007; 15:605-15. [PMID: 17070061 DOI: 10.1016/j.bmc.2006.10.020] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 10/03/2006] [Accepted: 10/11/2006] [Indexed: 11/25/2022]
Abstract
A clear definition for vascular targeting agents (VTAs) and vascular disrupting agents (VDAs) has separated the two as distinct methods of cancer treatment. VDAs differ from VTAs (antiangiogenesis drugs) in their mechanism of action. VTAs attempt to keep new blood vessels from forming and do not act on blood vessels that already feed existing tumors. In contrast, VDAs cause the vascular structure inside a solid tumor to collapse, depriving the tumor of blood and oxygen it needs to survive. Therefore, VDAs are an attractive way to approach the cancer problem by combating developed tumors. The following review discusses six small molecule VDAs, namely DMXAA, ZD6126, TZT1027, CA4P, AVE8062, and Oxi4503, their synthesis, biological mechanism of action, and current clinical status.
Collapse
Affiliation(s)
- John W Lippert
- Medicinal Chemistry Department, Albany Molecular Research, Inc., PO Box 15098, Albany, NY 12212-5098, USA.
| |
Collapse
|
686
|
Abstract
Cancer stem cells (CSCs) are thought to be critical for initiation and propagation of many types of cancer. Because these cells are resistant to conventional therapies, they have been very difficult to eliminate. A study in this issue of Cancer Cell suggests that brain tumor CSCs live in a "vascular niche" that promotes their long-term growth and self-renewal. Disrupting this niche impairs CSC self-renewal and thereby significantly inhibits the growth of tumors. Targeting the unique microenvironment of CSCs may be the key to effective cancer therapy.
Collapse
Affiliation(s)
- Zeng-Jie Yang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
687
|
Abstract
Hypoxia is an integral characteristic of the tumor microenvironment, primarily due to the microvascular defects that accompany the accelerated neoplastic growth. The presence of tumor hypoxic areas correlates with negative outcome after radiotherapy, chemotherapy, and surgery, as hypoxia not only provides an environment directly facilitating chemo- and radio-resistance, but also encourages the evolution of phenotypic changes inducing permanent resistance to treatment and metastatic spread. Therefore, successful treatment of hypoxic cells has the potential to not only improve local control but also impact overall patient survival. Specific and selective targeting of hypoxic tumor areas can be achieved at all three steps of a gene therapy treatment: delivery of the therapeutic gene to the tumor, regulation of gene expression, and therapeutic efficacy. In this review the latest developments and innovations in hypoxia-targeted gene therapy are discussed. In particular, approaches such as hypoxia-conditionally replicating viruses, cellular vehicles, and gene therapy means to disrupt the hypoxia-inducible factor (HIF) signaling are outlined.
Collapse
Affiliation(s)
- Olga Greco
- Tumour Microcirculation Group, University of Sheffield, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | | |
Collapse
|
688
|
Horsman MR, Siemann DW. Pathophysiologic Effects of Vascular-Targeting Agents and the Implications for Combination with Conventional Therapies. Cancer Res 2006; 66:11520-39. [PMID: 17178843 DOI: 10.1158/0008-5472.can-06-2848] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A functional vascular supply is critical for the continued growth and development of solid tumors. It also plays a major role in metastatic spread of tumor cells. This importance has led to the concept of targeting the vasculature of the tumor as a form of cancer therapy. Two major types of vascular-targeting agent (VTA) have now emerged: those that prevent the angiogenic development of the neovasculature of the tumor and those that specifically damage the already established tumor vascular supply. When used alone neither approach readily leads to tumor control, and so, for VTAs to be most successful in the clinic they will need to be combined with more conventional therapies. However, by affecting the tumor vascular supply, these VTAs should induce pathophysiologic changes in variables, such as blood flow, pH, and oxygenation. Such changes could have negative or positive influences on the tumor response to more conventional therapies. This review aims to discuss the pathophysiologic changes induced by VTAs and the implications of these effects on the potential use of VTAs in combined modality therapy.
Collapse
Affiliation(s)
- Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | | |
Collapse
|
689
|
Abstract
A recent publication reveals the biosynthetic building blocks, genetic code, and broad substrate tolerance of the enzymes of the cryptophycin biosynthetic pathway. This work lays the foundation for the production of poorly accessible yet very promising members of this family of anticancer compounds from lichen cyanobacterial symbionts. Chemoenzymatic production or precursor-directed biosynthesis might bring candidates from this family of natural products back to clinical trials.
Collapse
Affiliation(s)
- Jürgen Rohr
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 725 Rose Street, Lexington, Kentucky 40536-0082, USA.
| |
Collapse
|
690
|
Dachs GU, Steele AJ, Coralli C, Kanthou C, Brooks AC, Gunningham SP, Currie MJ, Watson AI, Robinson BA, Tozer GM. Anti-vascular agent Combretastatin A-4-P modulates hypoxia inducible factor-1 and gene expression. BMC Cancer 2006; 6:280. [PMID: 17156434 PMCID: PMC1702548 DOI: 10.1186/1471-2407-6-280] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2006] [Accepted: 12/07/2006] [Indexed: 11/15/2022] Open
Abstract
Background A functional vascular network is essential for the survival, growth and spread of solid tumours, making blood vessels a key target for therapeutic strategies. Combretastatin A-4 phosphate (CA-4-P) is a tubulin-depolymerising agent in Phase II clinical trials as a vascular disrupting agent. Not much is known of the molecular effect of CA-4-P under tumour conditions. The tumour microenvironment differs markedly from that in normal tissue, specifically with respect to oxygenation (hypoxia). Gene regulation under tumour conditions is governed by hypoxia inducible factor 1 (HIF-1), controlling angiogenic and metastatic pathways. Methods We investigated the effect of CA-4-P on factors of the upstream and downstream signalling pathway of HIF-1 in vitro. Results CA-4-P treatment under hypoxia tended to reduce HIF-1 accumulation in a concentration-dependent manner, an effect which was more prominent in endothelial cells than in cancer cell lines. Conversely, CA-4-P increased HIF-1 accumulation under aerobic conditions in vitro. At these concentrations of CA-4-P under aerobic conditions, nuclear factor κB was activated via the small GTPase RhoA, and expression of the HIF-1 downstream angiogenic effector gene, vascular endothelial growth factor (VEGF-A), was increased. Conclusion Our findings advance the understanding of signal transduction pathways involved in the actions of the anti-vascular agent CA-4-P.
Collapse
Affiliation(s)
- Gabi U Dachs
- Angiogenesis Research Group, Department of Pathology, Christchurch School of Medicine and Health Sciences, University of Otago, Christchurch, New Zealand
- Tumour Microcirculation Group, Gray Cancer Institute, Mount Vernon Hospital, Northwood, HA6 2JR, UK
| | - Andrew J Steele
- Tumour Microcirculation Group, Gray Cancer Institute, Mount Vernon Hospital, Northwood, HA6 2JR, UK
| | - Claudia Coralli
- Tumour Microcirculation Group, Gray Cancer Institute, Mount Vernon Hospital, Northwood, HA6 2JR, UK
| | - Chryso Kanthou
- Tumour Microcirculation Group, Gray Cancer Institute, Mount Vernon Hospital, Northwood, HA6 2JR, UK
- Academic Unit of Surgical Oncology, Division of Clinical Sciences, University of Sheffield, Sheffield, S10 2JF, UK
| | - Andrew C Brooks
- Tumour Microcirculation Group, Gray Cancer Institute, Mount Vernon Hospital, Northwood, HA6 2JR, UK
| | - Sarah P Gunningham
- Angiogenesis Research Group, Department of Pathology, Christchurch School of Medicine and Health Sciences, University of Otago, Christchurch, New Zealand
| | - Margaret J Currie
- Angiogenesis Research Group, Department of Pathology, Christchurch School of Medicine and Health Sciences, University of Otago, Christchurch, New Zealand
| | - Ally I Watson
- Angiogenesis Research Group, Department of Pathology, Christchurch School of Medicine and Health Sciences, University of Otago, Christchurch, New Zealand
| | - Bridget A Robinson
- Angiogenesis Research Group, Department of Pathology, Christchurch School of Medicine and Health Sciences, University of Otago, Christchurch, New Zealand
| | - Gillian M Tozer
- Tumour Microcirculation Group, Gray Cancer Institute, Mount Vernon Hospital, Northwood, HA6 2JR, UK
- Academic Unit of Surgical Oncology, Division of Clinical Sciences, University of Sheffield, Sheffield, S10 2JF, UK
| |
Collapse
|
691
|
Hu L, Li ZR, Li Y, Qu J, Ling YH, Jiang JD, Boykin DW. Synthesis and structure-activity relationships of carbazole sulfonamides as a novel class of antimitotic agents against solid tumors. J Med Chem 2006; 49:6273-82. [PMID: 17034133 DOI: 10.1021/jm060546h] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two series of carbazole sulfonamides related to Combretastatin A4 (1) were synthesized and evaluated for antiproliferative activity. Thirteen of the 26 new sulfonamides exhibited IC(50) values of <1 muM against CEM leukemia cells. Five compounds were evaluated against a panel of eight human tumor cell lines. 9-Ethyl-N-(3,4,5-trimethoxyphenyl)-carbazole-3-sulfonamide (11a) showed significant antitumor activity in two human xenograft models (MCF-7 and Bel-7402). Preliminary studies with 11a showed that the mode of action involves arrest of M-phase cell cycle and induction of apoptosis by increasing expression of p53 and promoting bcl-2 phosphorylation. Unexpectedly, 11a only weakly inhibits tubulin polymerization, which suggests that the mode of action of 11a differs from 1 and involves an unidentified target(s). Also, the SAR information gleaned from ring A-substituted analogues varies significantly from that of 1. Carbazole sulfonamides are a novel promising class of antimitotic agents with clinical development potential.
Collapse
Affiliation(s)
- Laixing Hu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303-3083, USA
| | | | | | | | | | | | | |
Collapse
|
692
|
Gilhuis HJ, van der Laak JAWM, Pomp J, Kappelle AC, Gijtenbeek JMM, Wesseling P. Three-dimensional (3D) reconstruction and quantitative analysis of the microvasculature in medulloblastoma and ependymoma subtypes. Angiogenesis 2006; 9:201-8. [PMID: 17109194 DOI: 10.1007/s10456-006-9054-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 09/05/2006] [Indexed: 11/29/2022]
Abstract
In the World Health Organisation (WHO) classification of tumours of the nervous system, four main histopathological subtypes of medulloblastomas (classic medulloblastoma, desmoplastic medulloblastoma, medulloblastoma with extensive nodularity and advanced neuronal differentiation and large cell/anaplastic medulloblastoma) as well as of ependymal tumours (low-grade ependymoma, anaplastic ependymoma, myxopapillary ependymoma and subependymoma) are recognised. Under the hypothesis that the microvascular architecture of tumours is a reflection of the histopathological subtype, we performed three-dimensional reconstructions of the microvasculature in these subtypes of medulloblastomas and ependymal tumours using computerised image analysis. In addition, we quantitatively assessed three microvascular parameters (number, area, perimeter) in these neoplasms. Three-dimensional reconstructions showed a dense pattern of irregular vessels in classic and large cell medulloblastoma. In desmoplastic medulloblastoma and medulloblastoma with extensive nodularity, the vessels were more unevenly distributed and organised around the nodular areas. Classic medulloblastoma and large cell medulloblastoma had on average the largest vessel area and perimeter. The highest number of vessels was seen in classic medulloblastoma and medulloblastoma with extensive nodularity. Three-dimensional analysis of ependymal tumours showed that low-grade ependymoma had larger but fewer vessels compared to anaplastic ependymoma, while myxopapillary ependymoma had a complex, heterogeneous pattern of vessels and subependymoma few but regular vessels. In ependymal tumours, the highest values for vessel number, vessel area and vessel perimeter were found in anaplastic ependymoma and the lowest values in subependymoma. We conclude that our three-dimensional reconstructions shed unprecedented light on the tumour vasculature in medulloblastomas and ependymal tumours and expect that such reconstructions are helpful tools for further studies on tumour angiogenesis.
Collapse
Affiliation(s)
- H Jacobus Gilhuis
- Department of Neurology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
693
|
Bix G, Castello R, Burrows M, Zoeller JJ, Weech M, Iozzo RA, Cardi C, Thakur ML, Barker CA, Camphausen K, Iozzo RV. Endorepellin In Vivo: Targeting the Tumor Vasculature and Retarding Cancer Growth and Metabolism. ACTA ACUST UNITED AC 2006; 98:1634-46. [PMID: 17105986 DOI: 10.1093/jnci/djj441] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The antiangiogenic approach to controlling cancer requires a better understanding of angiogenesis and the discovery of new compounds that modulate this key biological process. Here we investigated the role of endorepellin, an angiostatic protein fragment that is derived from the C-terminus of perlecan, a heparan sulfate proteoglycan, in controlling tumor angiogenesis in vivo. METHODS We administered human recombinant endorepellin systemically to mice bearing orthotopic squamous carcinoma xenografts or syngeneic Lewis lung carcinoma tumors. We monitored tumor growth, angiogenesis, metabolism, hypoxia, and mitotic index by using quantitative immunohistochemistry and positron emission tomography scan imaging. In addition, we determined the localization of injected endorepellin using near-infrared labeling and immunohistochemistry of frozen tumor sections. Finally, we isolated tumor-derived endothelial cells and tested whether endorepellin could interact with these cells and disrupt in vitro capillary morphogenesis. All statistical tests were two-sided. RESULTS Endorepellin specifically targeted the tumor vasculature as determined by immunohistochemical analysis and accumulated in the tumor perivascular zones where it persisted for several days as discrete deposits. This led to inhibition of tumor angiogenesis (as measured by decreased CD31-positive cells, mean control = 1902 CD31-positive pixels, mean endorepellin treated = 343.9, difference between means = 1558, 95% confidence interval [CI] = 1296 to 1820, P<.001), enhanced tumor hypoxia, and a statistically significant decrease in tumor metabolism and mitotic index (as measured by decreased Ki67-positive cells, mean control Ki67 pixels = 5970, mean endorepellin-treated Ki67 pixels = 3644, difference between means = 2326, 95% CI = 1904 to 2749, P<.001) compared to untreated controls. Endorepellin was actively internalized by tumor-derived endothelial cells causing a redistribution of alpha2beta1 integrin such that both proteins colocalized to punctate deposits in the perivascular region. Endorepellin treatment inhibited in vitro capillary morphogenesis of both normal and tumor-derived endothelia. CONCLUSIONS Our results provide support for the hypothesis that endorepellin is an effective antitumor vasculature agent that could be used as a therapeutic modality to combat cancer.
Collapse
MESH Headings
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Apoptosis
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/metabolism
- Cell Hypoxia
- Cell Line, Tumor
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Female
- Heparan Sulfate Proteoglycans/administration & dosage
- Heparan Sulfate Proteoglycans/metabolism
- Heparan Sulfate Proteoglycans/therapeutic use
- Humans
- Immunohistochemistry
- Infusions, Parenteral
- Integrin alpha2beta1/metabolism
- Male
- Mice
- Mice, Nude
- Mitotic Index
- Neoplasms/blood supply
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neovascularization, Pathologic/drug therapy
- Peptide Fragments/administration & dosage
- Peptide Fragments/metabolism
- Peptide Fragments/therapeutic use
- Positron-Emission Tomography
- Random Allocation
- Recombinant Proteins/therapeutic use
- Tomography, X-Ray Computed
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Gregory Bix
- Department of Pathology, Anatomy and Cell Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
694
|
Breidahl T, Nielsen FU, Stødkilde-Jørgensen H, Maxwell RJ, Horsman MR. The effects of the vascular disrupting agents combretastatin A-4 disodium phosphate, 5,6-dimethylxanthenone-4-acetic acid and ZD6126 in a murine tumour: a comparative assessment using MRI and MRS. Acta Oncol 2006; 45:306-16. [PMID: 16644574 DOI: 10.1080/02841860600570465] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The aim of this study was to use magnetic resonance (MR) techniques to non-invasively compare the effects of the three leading vascular disrupting agents, namely combretastatin A-4 disodium phosphate (CA4DP), 5,6-dimethylxanthenone-4-acetic acid (DMXAA) and ZD6126. A C3H mouse mammary carcinoma grown in the right rear foot of female CDF1 mice was used and treatments performed when tumours had reached 200 mm3 in volume. Drugs were prepared fresh before each experiment and intraperitoneally injected into restrained non-anaesthetised mice. Tumour response was evaluated using 31P-MR spectroscopy and T1- and T2- weighted imaging with a 7-Tesla, horizontal bore magnet, before and up to 24 hours after treatment. All three drugs significantly decreased bioenergetic status and pH, and did so in a time and dose dependent fashion, but there were differences; the decrease by CA4DP occurred more rapidly than for DMXAA or ZD6126, while DMXAA had a narrow window of activity compared to CA4DP and ZD6126. Changes in T1 weighted images for all three agents suggested a dose dependent increase in tumour oedema within three hours after treatment, consistent with an increase in vessel permeability. Using T2 weighted images there was some evidence of haemorrhagic necrosis by DMXAA, but such necrosis was limited following treatment with CA4DP or ZD6126.
Collapse
Affiliation(s)
- Tomas Breidahl
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | |
Collapse
|
695
|
Bellina F, Cauteruccio S, Monti S, Rossi R. Novel imidazole-based combretastatin A-4 analogues: Evaluation of their in vitro antitumor activity and molecular modeling study of their binding to the colchicine site of tubulin. Bioorg Med Chem Lett 2006; 16:5757-62. [PMID: 16950621 DOI: 10.1016/j.bmcl.2006.08.087] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Revised: 08/18/2006] [Accepted: 08/19/2006] [Indexed: 10/24/2022]
Abstract
The in vitro antitumor activity of novel combretastatin-like 1,5- and 1,2-diaryl-1H-imidazoles was evaluated against the NCI 60 human tumor cell lines panel. Compounds 2d and 2g proved to be more cytotoxic than CA-4 in tests involving their evaluation over a 10(-4)-10(-8) range. Docking experiments showed a good correlation between the MG_MID Log GI(50) values of all these compounds and their calculated interaction energies with the colchicine binding site of alphabeta-tubulin.
Collapse
Affiliation(s)
- Fabio Bellina
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via Risorgimento 35, I-56126 Pisa, Italy.
| | | | | | | |
Collapse
|
696
|
Bertolini F, Shaked Y, Mancuso P, Kerbel RS. The multifaceted circulating endothelial cell in cancer: towards marker and target identification. Nat Rev Cancer 2006; 6:835-45. [PMID: 17036040 DOI: 10.1038/nrc1971] [Citation(s) in RCA: 440] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Increases in the number of circulating endothelial cells (CECs) and progenitors (CEPs) have been reported in various pathological conditions including cancer. Preclinical studies have shown that CEC and CEP kinetics correlate well with several standard laboratory angiogenesis assays, which cannot be used in humans. At the clinical level, evidence is emerging that CEC kinetics and viability might correlate with clinical outcomes in cancer patients who undergo anti-angiogenic treatment. Therefore, CEC and CEP measurement has potential as a surrogate marker for monitoring anti-angiogenic treatment and drug activity, and could help to determine the optimal biological dose of anti-angiogenic drugs, which are being used with increasing frequency in medical oncology.
Collapse
Affiliation(s)
- Francesco Bertolini
- Division of Hematology-Oncology, Department of Medicine, European Institute of Oncology, 20141 Milan, Italy.
| | | | | | | |
Collapse
|
697
|
Miglarese MR, Carlson RO. Development of new cancer therapeutic agents targeting mitosis. Expert Opin Investig Drugs 2006; 15:1411-25. [PMID: 17040200 DOI: 10.1517/13543784.15.11.1411] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Targeting cellular proliferation persists as a mainstay of cancer therapeutic strategy. Although microtubule-targeting drugs (such as taxanes and vinca alkaloids) have been used successfully in the clinic to treat a variety of cancers, they carry substantial liabilities that have spurred drug companies to aggressively pursue new tubulin-targeting drug candidates with improved efficacy and toxicity profiles. The recent discoveries of new mitotic targets for cancer therapy (such as kinesin spindle protein, Aurora kinases and Polo-like kinase-1) have also stimulated intense work focused on identifying novel antimitotic drugs directed at these new targets. A number of novel antimitotic drugs have demonstrated encouraging activity in preclinical models and have progressed into clinical development. This review focuses on selected new antimitotic drugs under evaluation in clinical trials.
Collapse
Affiliation(s)
- Mark R Miglarese
- Array BioPharma, Inc., 3200 Walnut Street, Boulder, CO 80301, USA.
| | | |
Collapse
|
698
|
Abstract
It has been recognized for more than a century that most tumors tend to become more aggressive in clinical behavior over time, although this time course may be variable. This phenomenon has been termed "cancer progression," a process that appears to develop in a stepwise fashion through qualitatively different stages. Cancer progression relies on the ability of neoplastic cells to abandon their primary site of accretion, trespass tissue boundaries, and penetrate into the vasculature to colonize and repopulate distant sites. Among the various properties associated with cancer progression, the acquisition by neoplastic cells of the capacity to invade locally and to metastasize is of great clinical significance, and is still the fundamental definition of malignancy. This process represents the aberrant counterpart of a physiological morphogenetic program, known as invasive growth, occurring during embryo development and, in some instances, in adulthood for the generation and maintenance of normal organ complexity and architecture. Here we summarize some of the strategies adopted to inhibit cancer cell growth and spreading. We also review the current findings about cancer and metastasis inhibitors. As we suggest possible directions for drug development, we propose the receptor for the hepatocyte growth factor, Met, as an ideal target for tackling cancer progression.
Collapse
Affiliation(s)
- Massimiliano Mazzone
- Division of Molecular Oncology, Institute for Cancer Research and Treatment (IRCC), University of Torino Medical School, Strada Provinciale 142, km 3.95, I-10060 Candiolo (Torino), Italy.
| | | |
Collapse
|
699
|
Shaked Y, Ciarrocchi A, Franco M, Lee CR, Man S, Cheung AM, Hicklin DJ, Chaplin D, Foster FS, Benezra R, Kerbel RS. Therapy-induced acute recruitment of circulating endothelial progenitor cells to tumors. Science 2006; 313:1785-7. [PMID: 16990548 DOI: 10.1126/science.1127592] [Citation(s) in RCA: 476] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The contribution of bone marrow-derived circulating endothelial progenitor cells (CEPs) to tumor angiogenesis has been controversial, primarily because of their low numbers in blood vessels of untreated tumors. We show that treatment of tumor-bearing mice with vascular disrupting agents (VDAs) leads to an acute mobilization of CEPs, which home to the viable tumor rim that characteristically remains after such therapy. Disruption of this CEP spike by antiangiogenic drugs or by genetic manipulation resulted in marked reductions in tumor rim size and blood flow as well as enhanced VDA antitumor activity. These findings also provide a mechanistic rationale for the enhanced efficacy of VDAs when combined with antiangiogenic drugs.
Collapse
Affiliation(s)
- Yuval Shaked
- Department of Molecular and Cellular Biology Research, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
700
|
Chang JY, Yang MF, Chang CY, Chen CM, Kuo CC, Liou JP. 2-Amino and 2‘-Aminocombretastatin Derivatives as Potent Antimitotic Agents. J Med Chem 2006; 49:6412-5. [PMID: 17034147 DOI: 10.1021/jm060616k] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A novel series of 2-amino and 2'-aminocombretastatin derivatives were synthesized and evaluated for antitumor activity. Several compounds had excellent antiproliferative activity as inhibitors of tubulin polymerization. Compounds 11, 20, and 21 with IC(50) values of 1.6, 1.7, and 1.8 microM, respectively, exhibited more potent inhibition of tubulin polymerization than colchicine and approximately as active as combretastatin A-4. They also displayed antiproliferative activity with an IC(50) values ranging from 11 to 44 nM in a variety of human cell lines from different organs. Structure activity relationship information suggests that the NH(2) substituent at the 2-position of either ring A or ring B in combretastatin molecular skeleton may play an important role in the bioactivity of this series of compounds.
Collapse
Affiliation(s)
- Jang-Yang Chang
- Institute of Cancer Research, National Health Research Institutes, Taipei 114, Taiwan
| | | | | | | | | | | |
Collapse
|