551
|
Advani A, Kelly DJ, Advani SL, Cox AJ, Thai K, Zhang Y, White KE, Gow RM, Marshall SM, Steer BM, Marsden PA, Rakoczy PE, Gilbert RE. Role of VEGF in maintaining renal structure and function under normotensive and hypertensive conditions. Proc Natl Acad Sci U S A 2007; 104:14448-53. [PMID: 17726104 PMCID: PMC1964850 DOI: 10.1073/pnas.0703577104] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Inhibiting the actions of VEGF is a new therapeutic paradigm in cancer management with antiangiogenic therapy also under intensive investigation in a range of nonmalignant diseases characterized by pathological angiogenesis. However, the effects of VEGF inhibition on organs that constitutively express it in adulthood, such as the kidney, are mostly unknown. Accordingly, we examined the effect of VEGF inhibition on renal structure and function under physiological conditions and in the setting of the common renal stressors: hypertension and activation of the renin-angiotensin system. When compared with normotensive Sprague-Dawley (SD) rats, glomerular VEGF mRNA was increased 2-fold in transgenic (mRen-2)27 rats that overexpress renin with spontaneously hypertensive rat (SHR) kidneys showing VEGF expression levels that were intermediate between them. Administration of either an orally active inhibitor of the type 2 VEGF receptor (VEGFR-2) tyrosine kinase or a VEGF neutralizing antibody to TGR(mRen-2)27 rats resulted in loss of glomerular endothelial cells and transformation to a malignant hypertensive phenotype with severe glomerulosclerosis. VEGFR-2 kinase inhibition treatment was well tolerated in SDs and SHRs; although even in these animals there was detectable endothelial cell loss and rise in albuminuria. Mild mesangial expansion was also noted in hypertensive SHR, but not in SD rats. These studies illustrate: (i) VEGF has a role in the maintenance of glomerular endothelial integrity under physiological circumstances, (ii) glomerular VEGF is increased in response to hypertension and activation of the renin-angiotensin system, and (iii) VEGF signaling plays a protective role in the setting of these renal stressors.
Collapse
Affiliation(s)
- Andrew Advani
- *Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, ON, Canada M5C 2T2
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Victoria 3065, Australia
- School of Clinical Medical Sciences, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom; and
| | - Darren J. Kelly
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Victoria 3065, Australia
| | - Suzanne L. Advani
- *Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, ON, Canada M5C 2T2
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Victoria 3065, Australia
- School of Clinical Medical Sciences, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom; and
| | - Alison J. Cox
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Victoria 3065, Australia
| | - Kerri Thai
- *Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, ON, Canada M5C 2T2
| | - Yuan Zhang
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Victoria 3065, Australia
| | - Kathryn E. White
- School of Clinical Medical Sciences, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom; and
| | - Renae M. Gow
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Victoria 3065, Australia
| | - Sally M. Marshall
- School of Clinical Medical Sciences, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom; and
| | - Brent M. Steer
- *Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, ON, Canada M5C 2T2
| | - Philip A. Marsden
- *Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, ON, Canada M5C 2T2
| | - P. Elizabeth Rakoczy
- Centre for Ophthalmology and Visual Science, University of Western Australia, Nedlands, WA 6009, Australia
| | - Richard E. Gilbert
- *Department of Medicine, University of Toronto, St. Michael's Hospital, Toronto, ON, Canada M5C 2T2
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Victoria 3065, Australia
- To whom correspondence should be addressed at:
Department of Medicine, University of Toronto, Division of Endocrinology, St. Michael's Hospital, 61 Queen Street East, Toronto, ON, Canada M5C 2T2. E-mail:
| |
Collapse
|
552
|
ten Dam GB, van de Westerlo EMA, Purushothaman A, Stan RV, Bulten J, Sweep FCGJ, Massuger LF, Sugahara K, van Kuppevelt TH. Antibody GD3G7 selected against embryonic glycosaminoglycans defines chondroitin sulfate-E domains highly up-regulated in ovarian cancer and involved in vascular endothelial growth factor binding. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1324-33. [PMID: 17717144 PMCID: PMC1988881 DOI: 10.2353/ajpath.2007.070111] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Chondroitin sulfate (CS) is abundantly present in the tumor stroma, and tumor-specific CS modifications might be potential targets to influence tumor development. We applied the phage display technology to select antibodies that identify these tumor-specific CS modifications. Antibody GD3G7 was selected against embryonic glycosaminoglycans, and it reacted strongly with CS-E (rich in GlcA-GalNAc4S6S units). In ovarian adenocarcinomas, strong expression of this CS-E epitope was found in the extracellular matrix, and occasionally on tumor cells. No expression was found in normal ovary and cystadenomas. Differential expression was found in ovarian carcinoma cell lines, which correlated with the gene expression of the GalNAc4S-6st enzyme, involved in biosynthesis of CS-E. Vascular endothelial growth factor (VEGF)-sensitive fenestrated (in normal tissues) and tumor blood vessels were both identified by antibody GD3G7, which might implicate a role for CS-E in VEGF biology. VEGF bound to CS-E and antibody GD3G7 could compete for binding of VEGF to CS-E. In conclusion, antibody GD3G7 identified rare CS-E-like structures that were strongly expressed in ovarian adenocarcinomas. This antibody might therefore be instrumental for identifying tumor-related CS alterations.
Collapse
Affiliation(s)
- Gerdy B ten Dam
- Department of Biochemistry 280, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen Medical Center, PO. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
553
|
Ladewig MS, Karl SE, Hamelmann V, Helb HM, Scholl HPN, Holz FG, Eter N. Combined intravitreal bevacizumab and photodynamic therapy for neovascular age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol 2007; 246:17-25. [PMID: 17701197 DOI: 10.1007/s00417-007-0654-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Revised: 05/04/2007] [Accepted: 07/01/2007] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Our aim was to evaluate the short-term safety and efficacy of combined photodynamic therapy (PDT) with verteporfin and intravitreal bevacizumab in neovascular age-related macular degeneration (AMD). METHODS A prospective non-randomized interventional case series of 30 eyes of 30 patients with choroidal neovascularization (CNV) caused by AMD was studied. All patients were treated with PDT followed by an intravitreal injection of bevacizumab (1.5 mg) on the same day. Ophthalmic evaluations included determination of best-corrected visual acuity by using ETDRS charts. CNV lesion characteristics were determined by fluorescein angiography, and retinal morphology by optical coherence tomography. Review examinations were performed 1, 4, and 12 weeks following treatment. RESULTS The median ETDRS letter scores increased by 3 letters after 4 weeks and 4.3 letters after 12 weeks. Median central retinal thickness decreased from the baseline by 145 microm (week 1), 205 microm (week 4), and 171 microm (week 12), respectively (P < 0.0001, for all comparisons). One patient experienced a transient moderate vision loss after 4 weeks post treatment. Leakage on fluorescein angiography was resolved in all patients at week 12. No significant ocular or systemic side-effects were observed. CONCLUSIONS Short-term results suggest that a single PDT in combination with intravitreal bevacizumab is safe and associated with stabilization of visual acuity and decrease of intraretinal and subretinal fluid accumulation in the macula. Further evaluation of this treatment strategy for neovascular AMD appears warranted.
Collapse
Affiliation(s)
- Markus S Ladewig
- Department of Ophthalmology, University of Bonn, Ernst-Abbe-Strasse 2, 53127, Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|
554
|
D'Amore PA. Vascular endothelial cell growth factor-a: not just for endothelial cells anymore. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:14-8. [PMID: 17591949 PMCID: PMC1941611 DOI: 10.2353/ajpath.2007.070385] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Patricia A D'Amore
- Schepens Eye Institute, Department of Ophthalmology & Pathology, Boston, MA 02114, USA.
| |
Collapse
|
555
|
Nishijima K, Ng YS, Zhong L, Bradley J, Schubert W, Jo N, Akita J, Samuelsson SJ, Robinson GS, Adamis AP, Shima DT. Vascular endothelial growth factor-A is a survival factor for retinal neurons and a critical neuroprotectant during the adaptive response to ischemic injury. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:53-67. [PMID: 17591953 PMCID: PMC1941589 DOI: 10.2353/ajpath.2007.061237] [Citation(s) in RCA: 529] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor-A (VEGF-A) has recently been recognized as an important neuroprotectant in the central nervous system. Given its position as an anti-angiogenic target in the treatment of human diseases, understanding the extent of VEGF's role in neural cell survival is paramount. Here, we used a model of ischemia-reperfusion injury and found that VEGF-A exposure resulted in a dose-dependent reduction in retinal neuron apoptosis. Although mechanistic studies suggested that VEGF-A-induced volumetric blood flow to the retina may be partially responsible for the neuroprotection, ex vivo retinal culture demonstrated a direct neuroprotective effect for VEGF-A. VEGF receptor-2 (VEGFR2) expression was detected in several neuronal cell layers of the retina, and functional analyses showed that VEGFR2 was involved in retinal neuroprotection. VEGF-A was also shown to be involved in the adaptive response to retinal ischemia. Ischemic preconditioning 24 hours before ischemia-reperfusion injury increased VEGF-A levels and substantially decreased the number of apoptotic retinal cells. The protective effect of ischemic preconditioning was reversed after VEGF-A inhibition. Finally, chronic inhibition of VEGF-A function in normal adult animals led to a significant loss of retinal ganglion cells yet had no observable effect on several vascular parameters. These findings have implications for both neural pathologies and ocular vascular diseases, such as diabetic retinopathy and age-related macular degeneration.
Collapse
|
556
|
Izzedine H, Rixe O, Billemont B, Baumelou A, Deray G. Angiogenesis Inhibitor Therapies: Focus on Kidney Toxicity and Hypertension. Am J Kidney Dis 2007; 50:203-18. [PMID: 17660022 DOI: 10.1053/j.ajkd.2007.04.025] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Accepted: 04/24/2007] [Indexed: 12/29/2022]
Abstract
Angiogenesis inhibitors that target the epidermal growth factor (EGF) receptor (EGFR) and vascular endothelial growth factor (VEGF) constitute an important addition to the therapeutic armamentarium for the treatment of patients with metastatic disease. However, because the same growth factors are expressed in the kidneys, these treatment molecules have renal side effects. EGFR is expressed mainly in tubules (mainly distal and collecting segments) and mesangial and parietal epithelial cells. EGF is involved in maintaining tubular integrity and is a potent mitogen for cultured mesangial cells. Few cases of acute renal failure have been reported related to EGFR inhibitors. VEGF and VEGF receptors are still highly expressed in the kidney. VEGF is expressed in podocytes in the glomerulus, and VEGF receptors are present on endothelial, mesangial, and peritubular capillary cells. Signaling between endothelial cells and podocytes is essential for the proper development and maintenance of the filtration function of the kidney glomerulus. The most common renal class effects of VEGF antagonists are both manageable; hypertension and proteinuria commonly regressive on drug withdrawal. There was a dose-dependent increase in risk of proteinuria and hypertension in patients with cancer who received targeted therapies. Furthermore, few patients with glomerulonephritis or thrombotic microangiopathy secondary to treatment were reported. Hypertension is believed to be nitric oxide dependent, whereas proteinuria seems to be related to downregulation of podocyte tight junction protein. This article reviews data relating to hypertension and proteinuria associated with the use of these drugs.
Collapse
Affiliation(s)
- Hassane Izzedine
- Department of Nephrology, Pitie-Salpetriere Hospital, Paris, France.
| | | | | | | | | |
Collapse
|
557
|
McDonald DM. Imaging of angiogenesis in inflammation and cancer: lessons for novel treatment of allergic rhinitis. ACTA ACUST UNITED AC 2007. [DOI: 10.1111/j.1365-2222.2007.00119.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
558
|
VEGF: a key therapeutic target for the treatment of cancer-insights into its role and pharmacological inhibition. Target Oncol 2007. [DOI: 10.1007/s11523-007-0051-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
559
|
Kamba T, McDonald DM. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br J Cancer 2007; 96:1788-95. [PMID: 17519900 PMCID: PMC2359962 DOI: 10.1038/sj.bjc.6603813] [Citation(s) in RCA: 722] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 04/19/2007] [Accepted: 04/25/2007] [Indexed: 12/17/2022] Open
Abstract
Advances in understanding the role of vascular endothelial growth factor (VEGF) in normal physiology are giving insight into the basis of adverse effects attributed to the use of VEGF inhibitors in clinical oncology. These effects are typically downstream consequences of suppression of cellular signalling pathways important in the regulation and maintenance of the microvasculature. Downregulation of these pathways in normal organs can lead to vascular disturbances and even regression of blood vessels, which could be intensified by concurrent pathological conditions. These changes are generally manageable and pose less risk than the tumours being treated, but they highlight the properties shared by tumour vessels and the vasculature of normal organs.
Collapse
Affiliation(s)
- T Kamba
- Department of Urology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - D M McDonald
- Comprehensive Cancer Center and Cardiovascular Research Institute, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0452, USA
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0452, USA
| |
Collapse
|
560
|
Wang GM, Kovalenko B, Wilson EL, Moscatelli D. Vascular density is highest in the proximal region of the mouse prostate. Prostate 2007; 67:968-75. [PMID: 17440972 PMCID: PMC2430188 DOI: 10.1002/pros.20582] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The proximal region of the prostatic ducts harbor the prostatic epithelial stem cells. As stem cell niches in other organs are highly vascularized, we determined if the proximal region is more highly vascularized than the remaining regions of the prostate. The effect of androgen on vascular density in the different prostatic regions was also examined. METHODS Sections from prostates were immunostained with antibodies to CD31, and the vascular density in proximal, intermediate, and distal regions was calculated by image analysis software. Vascular density was compared in prostates from castrated mice that received daily inoculations of testosterone or vehicle alone for 3 days. To examine the role of angiogenic factors in the response to androgen, some animals were also treated with soluble VEGF receptor-2-Fc or Tie-2--Fc fusion proteins, which inhibit the activities of VEGF and angiopoietins, respectively. The endothelial proliferative response to androgen was determined by double staining sections with antibodies to CD31 and Ki-67. RESULTS In prostates from intact mice, vascular density was highest in the proximal region and lowest in the distal region. Administration of testosterone to castrated mice increased vascular density to the greatest extent in the distal and intermediate regions. The increase in vascular density required VEGF and the angiopoietins. Endothelial cell proliferation was less sensitive to androgen in the proximal region than the remainder of the prostate. CONCLUSIONS Vascular density is highest in the proximal region of the prostate, but the proximal vessels are less responsive to testosterone.
Collapse
|
561
|
Peters S, Heiduschka P, Julien S, Ziemssen F, Fietz H, Bartz-Schmidt KU, Schraermeyer U. Ultrastructural findings in the primate eye after intravitreal injection of bevacizumab. Am J Ophthalmol 2007; 143:995-1002. [PMID: 17449002 DOI: 10.1016/j.ajo.2007.03.007] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 03/04/2007] [Accepted: 03/07/2007] [Indexed: 02/08/2023]
Abstract
PURPOSE To examine the ultrastructural effect of intravitreal bevacizumab on primate eyes with particular focus set on the choriocapillaris and to examine the influence of vascular endothelial growth factor (VEGF) inhibition on endothelial cell fenestration. DESIGN Animal study. METHODS Four Cynomolgus monkeys received an intravitreal injection of 1.25 mg bevacizumab. The eyes were enucleated and prepared for light and electron microscopy on days one, four, seven, and 14. Control eyes remained untreated. Choriocapillaris endothelial cell fenestrations were quantified. RESULTS Choriocapillaris endothelial cell fenestrations were significantly reduced after intravitreal injection of bevacizumab. Fenestration was lowest on day four (15.9 +/- 6.7 per 25 microm) and increased again from days seven to 14, but was still significantly lower than in the control (66.2 +/- 9.5 per 25 microm). Densely packed thrombocytes and leukocytes regionally occluded the choriocapillaris lumen of treated eyes. On day one an increased number of leukocytes filled in the choriocapillaris lumen. Photoreceptors were damaged in two of 40 light microscopic sections. On days one to seven, choroidal melanocytes contained giant melanosomes. None of these described features was found in controls. CONCLUSIONS Intravitreal bevacizumab causes ultrastructural changes in the choriocapillaris of primate eyes. A significant reduction of choriocapillaris endothelial cell fenestrations is seen as early as 24 hours after injection and their number increases again after two weeks. These findings may play a role in the early clinical effect of intravitreal bevacizumab for macular edema. Because an increased risk of circulation disturbances in the choriocapillaris cannot be excluded, patients should be carefully monitored.
Collapse
|
562
|
Verheul HMW, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer 2007; 7:475-85. [PMID: 17522716 DOI: 10.1038/nrc2152] [Citation(s) in RCA: 363] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Contrary to initial expectations, angiogenesis inhibitors can cause toxicities in patients with cancer. The toxicity profiles of these inhibitors reflect the disturbance of growth factor signalling pathways that are important for maintaining homeostasis. Experiences with angiogenesis inhibitors in clinical trials indicate that short-term toxicities are mostly manageable. However, these agents will also be given in prolonged treatment strategies, so we need to anticipate possible long-term toxicities. In addition, understanding the molecular mechanisms involved in the toxicity of angiogenesis inhibition should allow more specific and more potent inhibitors to be developed.
Collapse
Affiliation(s)
- Henk M W Verheul
- University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | | |
Collapse
|
563
|
Raab S, Plate KH. Different networks, common growth factors: shared growth factors and receptors of the vascular and the nervous system. Acta Neuropathol 2007; 113:607-26. [PMID: 17492293 DOI: 10.1007/s00401-007-0228-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 04/18/2007] [Accepted: 04/18/2007] [Indexed: 01/13/2023]
Abstract
Growth factors and their respective receptors are key regulators during development and for homeostasis of the nervous system. In addition, changes in growth factor function, availability or downstream signaling is involved in many neuropathological disorders like Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, stroke and brain tumours. Research of the recent years revealed that some growth factors, initially discovered as neural growth factors are also affecting blood vessels [e.g. nerve growth factor (NGF) or brain-derived neurotrophic factor (BDNF)]. Likewise, vascular growth factors, such as vascular endothelial growth factor (VEGF), which was previously described as an endothelial cell specific mitogen, also affect neural cells. The discovery of shared growth factors affecting the vascular and the nervous system is of relevance for potential therapies of vascular and neurological diseases. This review aims to give an overview about the growing field of common growth factors and receptors within the two different networks.
Collapse
Affiliation(s)
- Sabine Raab
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe-University, Frankfurt/Main, Germany
| | | |
Collapse
|
564
|
Lee S, Mandic J, Van Vliet KJ. Chemomechanical mapping of ligand-receptor binding kinetics on cells. Proc Natl Acad Sci U S A 2007; 104:9609-14. [PMID: 17535923 PMCID: PMC1887608 DOI: 10.1073/pnas.0702668104] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The binding kinetics between cell surface receptors and extracellular biomolecules is critical to all intracellular and intercellular activity. Modeling and prediction of receptor-mediated cell functions are facilitated by measurement of the binding properties on whole cells, ideally indicating the subcellular locations or cytoskeletal associations that may affect the function of bound receptors. This dual need is particularly acute vis à vis ligand engineering and clinical applications of antibodies to neutralize pathological processes. Here, we map individual receptors and determine whole-cell binding kinetics by means of functionalized force imaging, enabled by scanning probe microscopy and molecular force spectroscopy of intact cells with biomolecule-conjugated mechanical probes. We quantify the number, distribution, and association/dissociation rate constants of vascular endothelial growth factor receptor-2 with respect to a monoclonal antibody on both living and fixed human microvascular endothelial cells. This general approach to direct receptor imaging simultaneously quantifies both the binding kinetics and the nonuniform distribution of these receptors with respect to the underlying cytoskeleton, providing spatiotemporal visualization of cell surface dynamics that regulate receptor-mediated behavior.
Collapse
Affiliation(s)
- Sunyoung Lee
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Jelena Mandic
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Krystyn J. Van Vliet
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
- *To whom correspondence should be addressed at:
Massachusetts Institute of Technology, Room 8-237, 77 Massachusetts Avenue, Cambridge, MA 02139. E-mail:
| |
Collapse
|
565
|
Fraser HM, Wilson H, Wulff C, Rudge JS, Wiegand SJ. Administration of vascular endothelial growth factor Trap during the 'post-angiogenic' period of the luteal phase causes rapid functional luteolysis and selective endothelial cell death in the marmoset. Reproduction 2007; 132:589-600. [PMID: 17008470 DOI: 10.1530/rep.1.01064] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The intense angiogenesis characteristic of early corpus luteum development is dependent upon vascular endothelial growth factor (VEGF) as inhibitors of VEGF administered at the peri-ovulatory period suppress endothelial cell proliferation and progesterone secretion. We now report that administration of VEGF Trap, a soluble decoy receptor-based inhibitor, at the mid- or the late luteal phase in the marmoset results in a rapid decline in plasma progesterone. Since vascularisation of the corpus luteum is largely complete by the mid-luteal phase, it suggested that this functional luteolysis involved mechanisms other than inhibition of angiogenesis. A second experiment investigated the role of VEGF in maintaining the integrity of the luteal vasculature and hormone-producing cells. VEGF Trap was administered to marmosets in the mid-luteal phase and ovaries were obtained 1, 2, 4 or 8 days later for localisation of activated caspase-3 staining in the corpus luteum and compared with those obtained 2, 4 and 8 days after administration of control protein. The number of cells with activated caspase-3 staining was significantly increased after administration of VEGF Trap. Dual staining of activated caspase-3 with the endothelial cell marker CD31 showed that at 1 day post-treatment, more than 90% caspase-3-stained cells were vascular endothelium, prior to detection of an increasing incidence in death of hormone-producing cells on days 2 and 4. Staining with CD31 showed that the endothelial cell area was decreased after treatment. By 8 days after treatment, corpora lutea had regressed to varying degrees, while all control corpora lutea remained healthy. These results show that VEGF inhibition in the mid- or the late luteal phase induces functional luteolysis in the marmoset that is associated with premature and selective death of endothelial cells.
Collapse
Affiliation(s)
- Hamish M Fraser
- Medical Research Council Human Reproductive Sciences Unit, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | | | | | | | | |
Collapse
|
566
|
Duda DG, Batchelor TT, Willett CG, Jain RK. VEGF-targeted cancer therapy strategies: current progress, hurdles and future prospects. Trends Mol Med 2007; 13:223-30. [PMID: 17462954 PMCID: PMC2686126 DOI: 10.1016/j.molmed.2007.04.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Revised: 03/21/2007] [Accepted: 04/10/2007] [Indexed: 12/26/2022]
Abstract
Despite setbacks, the clinical development of antiangiogenic agents has accelerated remarkably over the past 3-4 years. Consequently, there are currently three direct inhibitors of the VEGF pathway approved for use in cancer therapy. Other agents that block the VEGF pathway are in advanced stages of clinical development and have shown promising results. With these exciting developments come crucial questions regarding the use of these new molecular-targeted agents, alone or in combination with standard cytotoxic or targeted agents. Importantly, the mechanisms of action of anti-VEGF therapy remain unknown. Here, we discuss several potential mechanisms of action such as tumor vascular normalization, bone marrow-derived cell recruitment blockade and cytostatic effects of anti-VEGF therapy. We review the current progress, the major stumbling blocks and the future directions for anti-cancer therapy using anti-VEGF agents, emphasizing clarification of the underlying molecular mechanisms of action and biomarker identification and validation.
Collapse
Affiliation(s)
- Dan G Duda
- Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital, 100 Blossom Street, Cox-734, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
567
|
Ho QT, Kuo CJ. Vascular endothelial growth factor: biology and therapeutic applications. Int J Biochem Cell Biol 2007; 39:1349-57. [PMID: 17537667 PMCID: PMC2752727 DOI: 10.1016/j.biocel.2007.04.010] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 03/29/2007] [Accepted: 04/10/2007] [Indexed: 11/19/2022]
Abstract
While the development of anti-angiogenic therapy, as it pertains to cancer treatment, may still be in its infancy relative to well-established modalities such as chemotherapy, radiation, and surgery, major strides made in the past several decades have allowed translation of basic science discoveries in this field into clinical reality. The discovery of key molecular modulators of angiogenesis, notably vascular endothelial growth factor (VEGF), has catalyzed the development of numerous neutralizing therapeutic agents. The validity of VEGF inhibition as a therapeutic strategy has been well supported in randomized clinical trials, as well as U.S. Food and Drug Administration approval of the VEGF antagonists bevacizumab, sunitinib malate, sorafenib, pegaptinib and ranibizumab. Accordingly, this review will (1) briefly review the basic molecular biology of VEGF and (2) summarize recent progress in targeting the VEGF molecular pathway as therapy for angiogenic diseases such as cancer and age-related macular degeneration.
Collapse
Affiliation(s)
- Quoc T Ho
- Division of Hematology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, USA
| | | |
Collapse
|
568
|
Maharaj AS, D’Amore PA. Roles for VEGF in the adult. Microvasc Res 2007; 74:100-13. [PMID: 17532010 PMCID: PMC2128714 DOI: 10.1016/j.mvr.2007.03.004] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 03/29/2007] [Accepted: 03/30/2007] [Indexed: 12/17/2022]
Abstract
The role of VEGF during development and in pathology is well known, but its function in normal adult tissues is poorly understood. Adverse effects associated with the use of anti-angiogenic therapies targeting VEGF in human pathologies have begun to reveal potential functions of VEGF in quiescent vasculature. Further clues from expression studies of VEGF and its receptors in the adult, from the disease preeclampsia, and from experimental neutralization studies, have suggested that VEGF is involved in endothelial cell survival and fenestration, as well as in the signaling and maintenance of non-endothelial cells. The various biochemical properties of VEGF, and its interaction with other growth factors, may be an important point in determining whether VEGF functions as a maintenance factor versus an angiogenic factor. A thorough understanding of the function of VEGF in the adult may lead to more efficacious pro- and anti-angiogenic therapies.
Collapse
Affiliation(s)
- Arindel S.R. Maharaj
- Schepens Eye Research Institute
- Program in Biological and Biomedical Sciences
- Harvard Medical School, Boston, MA 02114
| | - Patricia A. D’Amore
- Schepens Eye Research Institute
- Program in Biological and Biomedical Sciences
- Harvard Medical School, Boston, MA 02114
- Departments of Ophthalmology and Pathology
| |
Collapse
|
569
|
Yu SW, Friedman B, Cheng Q, Lyden PD. Stroke-evoked angiogenesis results in a transient population of microvessels. J Cereb Blood Flow Metab 2007; 27:755-63. [PMID: 16883352 DOI: 10.1038/sj.jcbfm.9600378] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The role of angiogenesis after stroke is unclear; if angiogenesis supports long-term recovery of blood flow, then microvessel hyperdensity consequent to angiogenesis should persist in infarcted cortex. Here, we assess the long-term stability of ischemia-induced microvessels after 2-h transient rat middle cerebral artery occlusion (tMCAo) followed by 30, 90, or 165 days of reperfusion. Stereological measures of microvessel density were taken adjacent to and within cortical cysts. Vascular permeability was documented by extravasation of immunoglobulin (IgG) and of fluorescein-dextran. After 30 days reperfusion, a significantly increased microvessel volume density (V(V)) was restricted to the inner margin of cystic infarcts as compared with the region external to the infarct or contralateral control cortex (F=42.675, P<0.001). The hyperdense ischemic vasculature was abnormally leaky to IgG and fluorescein-dextran. Between 30 and 90 days of reperfusion, this vessel hyperdensity regressed significantly and then regressed further but less drastically between 90 and 165 days. Phagocytic macrophages were restricted to the infarct and dynamic changes in their number correlated with microvessel regression. Additional ED-1 labeled inflammatory cells were widely distributed inside and external to the infarct, even after 165 days of reperfusion. These data show that ischemia evoked angiogenesis results, at least in part, in transient populations of leaky microvessels and phagocytic macrophages. This suggests that a major role of this angiogenesis is for the removal of necrotic brain tissue.
Collapse
Affiliation(s)
- Sung Wook Yu
- Department of Neurosciences, UCSD School of Medicine, Veterans Administration Medical Center, San Diego, California 92161, USA
| | | | | | | |
Collapse
|
570
|
Wong E, Rosen LS, Mulay M, Vanvugt A, Dinolfo M, Tomoda C, Sugawara M, Hershman JM. Sunitinib induces hypothyroidism in advanced cancer patients and may inhibit thyroid peroxidase activity. Thyroid 2007; 17:351-5. [PMID: 17465866 DOI: 10.1089/thy.2006.0308] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Sunitinib is a novel tyrosine kinase inhibitor with antitumor and antiangiogenic effects. An observed higher than expected rate of hypothyroidism in sunitinib-treated patients prompted assessment of the incidence of hypothyroidism. DESIGN Patients taking sunitinib had their thyroid function tests (TFTs) assessed via chart review. To explore potential effects on the thyroid, we examined the antiperoxidase activity of sunitinib by in vitro testing its effect on guaiacol oxidation and protein iodination by lactoperoxidase. MAIN OUTCOME Of the 89 patients who took sunitinib, 49 patients were excluded from analysis for several reasons. Of the remaining 40 patients, 21 (53%, 24% of the original 89) developed elevated thyrotropin (TSH) after a median of 5 months (range 1-36 months). Median TSH was 21.4 mU/L (range 4.6-174 mU/L). In vitro, sunitinib had antiperoxidase activity that was about one-fourth the potency of propylthiouracil. CONCLUSIONS Of the 40 patients who had TFTs assessed after starting sunitinib, 53% developed elevated TSH. We recommend that all patients treated with sunitinib be monitored for hypothyroidism. The mechanism of the antithyroid effect appears to be inhibition of peroxidase activity. Further research is needed to confirm the mechanism by which sunitinib induces hypothyroidism.
Collapse
Affiliation(s)
- Elaine Wong
- Endocrinology and Diabetes Division, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
571
|
Red-Horse K, Crawford Y, Shojaei F, Ferrara N. Endothelium-microenvironment interactions in the developing embryo and in the adult. Dev Cell 2007; 12:181-94. [PMID: 17276337 DOI: 10.1016/j.devcel.2007.01.013] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The vascular endothelium is best known for its role in oxygen and nutrient delivery to the various tissues. Growing evidence supports a far more complex role in tissue homeostasis. In particular, reciprocal interactions between endothelial cells and the local microenvironment may regulate organ development and pattern formation. Such interactions appear to be important also in the adult, in normal and pathological conditions.
Collapse
|
572
|
Saif MW, Elfiky A, Salem RR. Gastrointestinal perforation due to bevacizumab in colorectal cancer. Ann Surg Oncol 2007; 14:1860-9. [PMID: 17356952 DOI: 10.1245/s10434-006-9337-9] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2006] [Accepted: 12/07/2006] [Indexed: 12/17/2022]
Abstract
Bevacizumab is the first U.S. Food and Drug Association-approved vascular endothelial growth factor-targeted agent that greatly increases progression-free and overall survival in combination with standard chemotherapy regimens in patients with metastatic colorectal cancer. Although bevacizumab is generally well tolerated, some serious adverse events have occurred in some patients in clinical trials, including arterial thromboembolism and gastrointestinal (GI) perforation. GI perforation was first observed in the pivotal phase 3 trial, in which six events occurred in bevacizumab group (1.5%), compared with no events in the control group. Since then, similar rates of GI perforation have been observed in other large trials. Typical presentation was abdominal pain associated with constipation and vomiting. Such events occurred throughout treatment and were not correlated with duration of exposure. No difference in rate of GI perforations was found in patients who did and did not have a baseline history of peptic ulcer disease, diverticulosis, and history of chronic use of nonsteroidal anti-inflammatory drugs. However, the incidence of GI perforation seemed to be higher in patients with primary tumor intact, recent history of sigmoidoscopy or colonoscopy, or previous adjuvant radiotherapy, but it is necessary to confirm these preliminary findings by multivariate analyses. The mechanism responsible for causing GI perforation is not known and may be multifactorial. Bevacizumab should be permanently discontinued in patients who develop GI perforation. This article reviews the incidence, presentation, pathogenesis, risk factors, and management of GI perforation in patients with colorectal cancer who are treated with bevacizumab.
Collapse
Affiliation(s)
- Muhammad Wasif Saif
- Yale Cancer Center, Yale University School of Medicine, Divisions of Medical and Surgical Oncology, 333 Cedar Street, FMP 116, New Haven, Connecticut 06520, USA.
| | | | | |
Collapse
|
573
|
Grothey A. Future directions in vascular endothelial growth factor-targeted therapy for metastatic colorectal cancer. Semin Oncol 2007; 33:S41-9. [PMID: 17145524 DOI: 10.1053/j.seminoncol.2006.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bevacizumab, the first approved vascular endothelial growth factor (VEGF)-targeted agent for metastatic colorectal cancer, continues to be developed in phase III trials in other tumor types. Its use is being explored not only in advanced disease, but also in earlier-stage disease in the adjuvant setting. Preclinical and clinical research is also addressing several potential strategies for maximizing the benefits of bevacizumab and other VEGF-targeted agents, including (1) dual inhibition of VEGF and platelet-derived growth factor signaling to target both the endothelial and the pericyte components of tumor vasculature; (2) combining VEGF-targeted agents with other targeted agents, such as inhibitors of HER2 or epidermal growth factor receptor signaling, which affect several angiogenic pathways; and (3) combining VEGF-targeted agents with low-dose, metronomic chemotherapy. The optimal dose and schedule of VEGF-targeted agents is another unanswered question. Further investigation of the mechanism of action and vascular effects of VEGF-targeted agents in humans will help to address these questions. Mechanistic studies in humans will be aided by the development and validation of surrogate clinical end points such as noninvasive assessment of hemodynamics and vascular changes within tumors, using imaging studies.
Collapse
Affiliation(s)
- Axel Grothey
- Division of Medical Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA.
| |
Collapse
|
574
|
Roskoski R. Vascular endothelial growth factor (VEGF) signaling in tumor progression. Crit Rev Oncol Hematol 2007; 62:179-213. [PMID: 17324579 DOI: 10.1016/j.critrevonc.2007.01.006] [Citation(s) in RCA: 434] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2006] [Revised: 01/01/2007] [Accepted: 01/29/2007] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial cells are ordinarily quiescent in adult humans and divide less than once per decade. When tumors reach a size of about 0.2-2.0mm in diameter, they become hypoxic and limited in size in the absence of angiogenesis. There are about 30 endogenous pro-angiogenic factors and about 30 endogenous anti-angiogenic factors. In order to increase in size, tumors undergo an angiogenic switch where the action of pro-angiogenic factors predominates, resulting in angiogenesis and tumor progression. One mechanism for driving angiogenesis results from the increased production of vascular endothelial growth factor (VEGF) following up-regulation of the hypoxia-inducible transcription factor. The human VEGF family consists of VEGF (VEGF-A), VEGF-B, VEGF-C, VEGF-D, and placental growth factor (PlGF). The VEGF family of receptors consists of three protein-tyrosine kinases and two non-protein kinase receptors (neuropilin-1 and -2). Owing to the importance of angiogenesis in tumor progression, inhibition of VEGF signaling represents an attractive cancer treatment.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 116A, Box 19, Horse Shoe, NC 28742, USA.
| |
Collapse
|
575
|
Gerber HP, Wu X, Yu L, Wiesmann C, Liang XH, Lee CV, Fuh G, Olsson C, Damico L, Xie D, Meng YG, Gutierrez J, Corpuz R, Li B, Hall L, Rangell L, Ferrando R, Lowman H, Peale F, Ferrara N. Mice expressing a humanized form of VEGF-A may provide insights into the safety and efficacy of anti-VEGF antibodies. Proc Natl Acad Sci U S A 2007; 104:3478-83. [PMID: 17360669 PMCID: PMC1805509 DOI: 10.1073/pnas.0611492104] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
VEGF-A is important in tumor angiogenesis, and a humanized anti-VEGF-A monoclonal antibody (bevacizumab) has been approved by the FDA as a treatment for metastatic colorectal and nonsquamous, non-small-cell lung cancer in combination with chemotherapy. However, contributions of both tumor- and stromal-cell derived VEGF-A to vascularization of human tumors grown in immunodeficient mice hindered direct comparison between the pharmacological effects of anti-VEGF antibodies with different abilities to block host VEGF. Therefore, by gene replacement technology, we engineered mice to express a humanized form of VEGF-A (hum-X VEGF) that is recognized by many anti-VEGF antibodies and has biochemical and biological properties comparable with WT mouse and human VEGF-A. The hum-X VEGF mouse model was then used to compare the activity and safety of a panel of VEGF Mabs with different affinities for VEGF-A. Although in vitro studies clearly showed a correlation between binding affinity and potency at blocking endothelial cell proliferation stimulated by VEGF, in vivo experiments failed to document any consistent correlation between antibody affinity and the ability to inhibit tumor growth and angiogenesis in most animal models. However, higher-affinity antibodies were more likely to result in glomerulosclerosis during long-term treatment.
Collapse
Affiliation(s)
| | - Xiumin Wu
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Lanlan Yu
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | | | | | | | - Germaine Fuh
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | | | - Lisa Damico
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - David Xie
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Y. Gloria Meng
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | | | - Racquel Corpuz
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Bing Li
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Linda Hall
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Linda Rangell
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Ron Ferrando
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Henry Lowman
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Franklin Peale
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Napoleone Ferrara
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
576
|
Iwashita N, Uchida T, Choi JB, Azuma K, Ogihara T, Ferrara N, Gerber H, Kawamori R, Inoue M, Watada H. Impaired insulin secretion in vivo but enhanced insulin secretion from isolated islets in pancreatic beta cell-specific vascular endothelial growth factor-A knock-out mice. Diabetologia 2007; 50:380-9. [PMID: 17180351 DOI: 10.1007/s00125-006-0512-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Accepted: 09/22/2006] [Indexed: 10/23/2022]
Abstract
AIMS/HYPOTHESIS Endothelial cells are considered to be essential for normal pancreatic beta cell function. However, there have been no reports showing their importance for beta cell function. MATERIALS AND METHODS Using mice with disrupted vascular endothelial growth factor-A gene specifically in beta cells, we investigated the relation between islet vascular structure and beta cell function. RESULTS Mice with disrupted vascular endothelial growth factor-A gene specifically in beta cells had reduced islet vascular density with impaired formation of endothelial fenestration. While their fasting glucose and body weight were comparable with control mice, their glucose- and tolbutamide-induced rapid insulin release were impaired, thus resulting in glucose intolerance. On the other hand, glucose and KCl enhanced the levels of insulin secreted from islets isolated from these mice. In addition, the production of soluble N-ethylmaleimide-sensitive factor attachment protein receptors in the islets was increased. Insulin content and expression of insulin I and pancreas duodenum homeobox 1 mRNA in the islets were also increased. CONCLUSIONS/INTERPRETATION Our results indicate that an abnormal quality and quantity of blood vessels due to reduced expression of vascular endothelial growth factor-A in beta cells could be a cause of impaired insulin secretion without impairment of beta cell function.
Collapse
Affiliation(s)
- N Iwashita
- Department of Medicine, Metabolism and Endocrinology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
577
|
Abstract
Since the discovery of vascular-specific growth factors with angiogenic activity, there has been a significant effort to develop cancer drugs that restrict tumorigenesis by targeting the blood supply. In this issue of the JCI, Mancuso et al. use mouse models to better understand the plasticity of the tumor vasculature in the face of antiangiogenic therapy (see the related article beginning on page 2610). They describe a rapid regrowth of the tumor vasculature following withdrawal of VEGFR inhibitors, emphasizing the importance of fully understanding the function of these and similar treatments used in the clinic at the cellular and molecular level.
Collapse
|
578
|
Mancuso MR, Davis R, Norberg SM, O’Brien S, Sennino B, Nakahara T, Yao VJ, Inai T, Brooks P, Freimark B, Shalinsky DR, Hu-Lowe DD, McDonald DM. Rapid vascular regrowth in tumors after reversal of VEGF inhibition. J Clin Invest 2007; 116:2610-21. [PMID: 17016557 PMCID: PMC1578604 DOI: 10.1172/jci24612] [Citation(s) in RCA: 632] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Accepted: 07/18/2006] [Indexed: 01/09/2023] Open
Abstract
Inhibitors of VEGF signaling can block angiogenesis and reduce tumor vascularity, but little is known about the reversibility of these changes after treatment ends. In the present study, regrowth of blood vessels in spontaneous RIP-Tag2 tumors and implanted Lewis lung carcinomas in mice was assessed after inhibition of VEGF receptor signaling by AG-013736 or AG-028262 for 7 days. Both agents caused loss of 50%-60% of tumor vasculature. Empty sleeves of basement membrane were left behind. Pericytes also survived but had less alpha-SMA immunoreactivity. One day after drug withdrawal, endothelial sprouts grew into empty sleeves of basement membrane. Vessel patency and connection to the bloodstream followed close behind. By 7 days, tumors were fully revascularized, and the pericyte phenotype returned to baseline. Importantly, the regrown vasculature regressed as much during a second treatment as it did in the first. Inhibition of MMPs or targeting of type IV collagen cryptic sites by antibody HUIV26 did not eliminate the sleeves or slow revascularization. These results suggest that empty sleeves of basement membrane and accompanying pericytes provide a scaffold for rapid revascularization of tumors after removal of anti-VEGF therapy and highlight their importance as potential targets in cancer therapy.
Collapse
MESH Headings
- Actins/metabolism
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antibodies, Monoclonal/pharmacology
- Axitinib
- Basement Membrane/drug effects
- Basement Membrane/metabolism
- Basement Membrane/pathology
- Blood Vessels/drug effects
- Blood Vessels/metabolism
- Blood Vessels/pathology
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/pathology
- Collagen Type IV/immunology
- Collagen Type IV/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Imidazoles/pharmacology
- Imidazoles/therapeutic use
- Indazoles/pharmacology
- Indazoles/therapeutic use
- Insulinoma/blood supply
- Insulinoma/drug therapy
- Insulinoma/pathology
- Matrix Metalloproteinase Inhibitors
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms/blood supply
- Neoplasms/drug therapy
- Neoplasms/pathology
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Organic Chemicals/pharmacology
- Pericytes/drug effects
- Pericytes/metabolism
- Pericytes/pathology
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
- Treatment Outcome
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-2/metabolism
Collapse
Affiliation(s)
- Michael R. Mancuso
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Rachel Davis
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Scott M. Norberg
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Shaun O’Brien
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Barbara Sennino
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Tsutomu Nakahara
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Virginia J. Yao
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Tetsuichiro Inai
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Peter Brooks
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Bruce Freimark
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - David R. Shalinsky
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Dana D. Hu-Lowe
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Donald M. McDonald
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, UCSF, San Francisco, California, USA.
Departments of Radiation Oncology and Cell Biology, New York University Cancer Institute, New York, New York, USA.
Cell-Matrix Inc., a subsidiary of CancerVax, Carlsbad, California, USA.
Department of Research Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| |
Collapse
|
579
|
Hughes MS, Sang DN. Safety and efficacy of intravitreal bevacizumab followed by pegaptanib maintenance as a treatment regimen for age-related macular degeneration. Ophthalmic Surg Lasers Imaging Retina 2007; 37:446-54. [PMID: 17152537 DOI: 10.3928/15428877-20061101-01] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND OBJECTIVE Vascular endothelial growth factor (VEGF)-A, both necessary and sufficient in promoting ocular neovascularization, is an attractive therapeutic target. Combining nonselective and selective VEGF blockade may provide clinical benefit with minimal risks in the treatment of neovascular age-related macular degeneration (AMD). PATIENTS AND METHODS Twenty patients with all subtypes of neovascular AMD and a broad range of baseline vision were treated with intravitreal bevacizumab followed by pegaptanib sodium for 54 weeks. Visual acuity measurements, biomicroscopy, funduscopy, fluorescein angiography, optical coherence tomography, and adverse event assessments were performed. RESULTS Mean visual acuity improved from approximately 20/200 at baseline to 20/80. All patients experienced an improvement in retinal thickness, ranging from -47 to -297 microns. Adverse events were limited to transient irritation or redness. No significant elevation in intraocular pressure occurred following either bevacizumab or pegaptanib injections. CONCLUSIONS Nonselective VEGF blockade with bevacizumab induction and selective VEGF165 blockade with pegaptanib as maintenance therapies may offer clinically meaningful outcomes with acceptable safety profiles in patients with AMD.
Collapse
Affiliation(s)
- Mark S Hughes
- Schepens Eye Research Institute and the Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
580
|
Endothelial Biology and the Bone Marrow. Biol Blood Marrow Transplant 2007. [DOI: 10.1016/j.bbmt.2006.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
581
|
Galambos C, deMello DE. Molecular mechanisms of pulmonary vascular development. Pediatr Dev Pathol 2007; 10:1-17. [PMID: 17378630 DOI: 10.2350/06-06-0122.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Accepted: 10/04/2006] [Indexed: 11/20/2022]
Abstract
In this era of rapidly advancing vascular biology research, a vast array of growth factors and signaling molecules have been recognized as key players in the mechanisms that control lung vascular development. In the lung, vascular development is a complex, multistep process that includes specialization of primitive cells to vascular progenitors; formation of primitive vascular networks; remodeling with local regression and branching; specialization toward arteries, veins, and lymphatics; stabilization of vessels by matrix production and recruitment of supporting cells; and maintenance of the vascular structure. This complex, highly organized process requires exquisite orchestration of the regulatory activity of multiple molecules in a specific temporospatial order. Most of these molecules are members of 3 major growth factor families that have been recently identified. They are the vascular endothelial growth factor, angiopoietin, and ephrin families. Understanding the functional reach of several members of these growth factor families is integral to an appreciation of the etiology and pathogenesis of developmental lung vascular disorders affecting newborns. This review summarizes recent advances in the molecular bases of lung vascular development and some of the pulmonary diseases resulting from aberrant vascular growth, including bronchopulmonary dysplasia, alveolar capillary dysplasia, congenital cystic pulmonary disorders, congenital pulmonary hemangiomatosis, and lung hypoplasia.
Collapse
Affiliation(s)
- Csaba Galambos
- Department of Pathology, Children's Hospital of Pittsburgh, University of Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
582
|
Abstract
Extensive experience with chronic peritoneal dialysis has identified a series of functional and anatomical pathologic changes in the peritoneal membrane thought to be the result of repeated insults from bioincompatible solutions. Laboratory and clinical findings from recent investigations often conflict and are difficult to interpret due to variations in methodologies, animal models, study designs, and data analyses. The principal pathophysiologic mechanisms identified thus far are oxidative stress, inflammation, and their consequences. Many substances used to neutralize the action of these insults, prevent formation of toxic compounds, or directly alter solute and water transport to improve peritoneal membrane performance have been studied. We herein review the most promising of these substances or those that deserve attention because their use has contributed to better understanding of peritoneal pathophysiology. Most peritoneal solution additives have proved useless due to their toxicity and undesirable effects, ineffectiveness, or manufacturing limitations. A few substances deserve more attention, particularly those capable of restoring negatively charged membrane sites, those that somehow improve permselectivity, scavengers of oxidants, and advanced glycation end-product inhibitors and breakers. Recent publications on clinical experience with neutral pH, low glucose degradation product (GDP) peritoneal solutions, although few and preliminary, are most encouraging. The virtual elimination of GDPs in these novel solutions will probably preclude the need for GDP scavengers and inhibitors. Nonetheless, there is room for further significant improvement in solution biocompatibility and for compounds that may restore peritoneal function.
Collapse
Affiliation(s)
- Jose A Diaz-Buxo
- Home Therapies Development, Fresenius Medical Care North America, Lexington Massachusetts, USA.
| | | |
Collapse
|
583
|
Nikolova G, Strilic B, Lammert E. The vascular niche and its basement membrane. Trends Cell Biol 2006; 17:19-25. [PMID: 17129728 DOI: 10.1016/j.tcb.2006.11.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 10/20/2006] [Accepted: 11/15/2006] [Indexed: 12/13/2022]
Abstract
Over the past few years, scientists have realized that many cellular and developmental processes, including pancreatic beta-cell growth and differentiation, stem cell and progenitor cell proliferation and cancer cell metastasis, occur in what are known as 'vascular niches'. Despite increasing numbers of reports on these niches, few common mechanisms have been identified to explain their various effects. Here, we define the term 'vascular niche' and suggest that a common and conserved feature of this niche is to provide a basement membrane to cells that are unable to form their own. We further propose that these cells require a vascular niche when they retain a high degree of plasticity.
Collapse
Affiliation(s)
- Ganka Nikolova
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstr. 108, D-01307 Dresden, Germany
| | | | | |
Collapse
|
584
|
Ferrara N, Damico L, Shams N, Lowman H, Kim R. Development of ranibizumab, an anti-vascular endothelial growth factor antigen binding fragment, as therapy for neovascular age-related macular degeneration. Retina 2006; 26:859-70. [PMID: 17031284 DOI: 10.1097/01.iae.0000242842.14624.e7] [Citation(s) in RCA: 599] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Angiogenesis is a key aspect of the wet form of age-related neovascular (AMD), the leading cause of blindness in the elderly population. Substantial evidence indicated that vascular endothelial growth factor (VEGF)-A is a major mediator of angiogenesis and vascular leakage in wet AMD. VEGF-A is the prototype member of a gene family that includes also PlGF, VEGF-B, VEGF-C, VEGF-D and the orf virus-encoded VEGF-E. Several isoforms of VEGF-A can be generated due to alternative mRNA splicing. Various VEGF inhibitors have been clinically developed. Among these, ranibizumab is a high affinity recombinant Fab that neutralizes all isoforms of VEGF-A. The article briefly reviews the biology of VEGF and then focuses on the path that led to clinical development of ranibizumab. RESULTS The safety and efficacy of ranibizumab in the treatment of neovascular AMD have been evaluated in two large phase III, multicenter, randomized, double-masked, controlled pivotal trials in different neovascular AMD patient populations. Combined, the trial results indicate that ranibizumab results not only in a slowing down of vision loss but also in a significant proportion of patients experiencing a clinically meaningful vision gain. The visual acuity benefit over control was observed regardless of CNV lesion type. Furthermore, the benefit was associated with a low rate of serious adverse events. CONCLUSIONS Ranibizumab represents a novel therapy that, for the first time, appears to have the potential to enable many AMD patients to obtain a meaningful and sustained gain of vision. On June 30 2006, ranibizumab was approved by the US Food and Drug Administration for the treatment of wet AMD.
Collapse
|
585
|
Ioannidou S, Deinhardt K, Miotla J, Bradley J, Cheung E, Samuelsson S, Ng YS, Shima DT. An in vitro assay reveals a role for the diaphragm protein PV-1 in endothelial fenestra morphogenesis. Proc Natl Acad Sci U S A 2006; 103:16770-5. [PMID: 17075074 PMCID: PMC1636530 DOI: 10.1073/pnas.0603501103] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Fenestrae are small pores in the endothelium of renal glomerular, gastrointestinal, and endocrine gland capillaries and are involved in the bidirectional exchange of molecules between blood and tissues. Although decades of studies have characterized fenestrae at the ultrastructural level, little is known on the mechanisms by which fenestrae form. We present the development of an in vitro assay in which rapid and abundant fenestra induction enables a detailed study of their biogenesis. Through the use of agents that stabilize or disassemble actin microfilaments, we show that actin microfilament remodeling is part of fenestra biogenesis in this model. Furthermore, by using a loss-of-function approach, we show that the diaphragm protein PV-1 is necessary for fenestral pore architecture and the ordered arrangement of fenestrae in sieve plates. Together, these data provide insight into the cell biology of fenestra formation and open up the future study of the fenestra to a combined morphological and biochemical analysis.
Collapse
Affiliation(s)
- Sofia Ioannidou
- *Eyetech Research Center, OSI Eyetech, 35 Hartwell Avenue, Lexington, MA 02420; and
- Endothelial Cell Biology Laboratory, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | - Katrin Deinhardt
- Endothelial Cell Biology Laboratory, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | - Jadwiga Miotla
- Endothelial Cell Biology Laboratory, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | - John Bradley
- *Eyetech Research Center, OSI Eyetech, 35 Hartwell Avenue, Lexington, MA 02420; and
| | - Eunice Cheung
- *Eyetech Research Center, OSI Eyetech, 35 Hartwell Avenue, Lexington, MA 02420; and
| | - Steven Samuelsson
- *Eyetech Research Center, OSI Eyetech, 35 Hartwell Avenue, Lexington, MA 02420; and
| | - Yin-Shan Ng
- *Eyetech Research Center, OSI Eyetech, 35 Hartwell Avenue, Lexington, MA 02420; and
| | - David T. Shima
- *Eyetech Research Center, OSI Eyetech, 35 Hartwell Avenue, Lexington, MA 02420; and
- Endothelial Cell Biology Laboratory, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
586
|
Fischer C, Schneider M, Carmeliet P. Principles and therapeutic implications of angiogenesis, vasculogenesis and arteriogenesis. Handb Exp Pharmacol 2006:157-212. [PMID: 16999228 DOI: 10.1007/3-540-36028-x_6] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The vasculature is the first organ to arise during development. Blood vessels run through virtually every organ in the body (except the avascular cornea and the cartilage), assuring metabolic homeostasis by supplying oxygen and nutrients and removing waste products. Not surprisingly therefore, vessels are critical for organ growth in the embryo and for repair of wounded tissue in the adult. Notably, however, an imbalance in angiogenesis (the growth of blood vessels) contributes to the pathogenesis of numerous malignant, inflammatory, ischaemic, infectious and immune disorders. During the last two decades, an explosive interest in angiogenesis research has generated the necessary insights to develop the first clinically approved anti-angiogenic agents for cancer and blindness. This novel treatment is likely to change the face of medicine in the next decade, as over 500 million people worldwide are estimated to benefit from pro- or anti-angiogenesis treatment. In this following chapter, we discuss general key angiogenic mechanisms in health and disease, and highlight recent developments and perspectives of anti-angiogenic therapeutic strategies.
Collapse
Affiliation(s)
- C Fischer
- Centre for Transgene Technology and Gene Therapy, Flanders Interuniversity Institute for Biotechnology, KULeuven, Campus Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | | | | |
Collapse
|
587
|
|
588
|
Griffin RJ, Molema G, Dings RPM. Angiogenesis treatment, new concepts on the horizon. Angiogenesis 2006; 9:67-72. [PMID: 16964528 DOI: 10.1007/s10456-006-9031-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2006] [Indexed: 10/24/2022]
Affiliation(s)
- Robert J Griffin
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
589
|
Abstract
Vascular endothelial growth factor (VEGF) plays a pivotal role for embryo- and organogenesis. By regulating haemodynamics, (lymphoid) vessel architecture, haematopoiesis and immune system, endocrinology and reparative processes in adults, inhibited VEGF can cause multiple adverse events. Although the intravitreal administration of smaller doses can drastically reduce the systemic exposure, possible local side effects on retinal perfusion and survival of neuronal tissue must be taken into consideration. Before experience has been gained with VEGF inhibitors for longer than several years, individual discussion before the use, extensive informed consent and careful follow-up are necessary. From the ethical point of view, the available drugs should not be used without hesitation despite the clear benefit. First experiences with bevacizumab (Avastin), the first available (off-label) drug in Germany, are reported. In future, direct comparison of the different available drugs has to assess possible differences in the risk-benefit profile.
Collapse
Affiliation(s)
- F Ziemssen
- Augenklinik, Eberhard-Karls-Universität, Schleichstrasse 12, 72076 Tübingen.
| | | | | |
Collapse
|
590
|
Karpanen T, Wirzenius M, Mäkinen T, Veikkola T, Haisma HJ, Achen MG, Stacker SA, Pytowski B, Ylä-Herttuala S, Alitalo K. Lymphangiogenic growth factor responsiveness is modulated by postnatal lymphatic vessel maturation. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:708-18. [PMID: 16877368 PMCID: PMC1764216 DOI: 10.2353/ajpath.2006.051200] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Lymphatic vessel plasticity and stability are of considerable importance when attempting to treat diseases associated with the lymphatic vasculature. Development of lymphatic vessels during embryogenesis is dependent on vascular endothelial growth factor (VEGF)-C but not VEGF-D. Using a recombinant adenovirus encoding a soluble form of their receptor VEGFR-3 (AdVEGFR-3-Ig), we studied lymphatic vessel dependency on VEGF-C and VEGF-D induced VEGFR-3 signaling in postnatal and adult mice. Transduction with AdVEGFR-3-Ig led to regression of lymphatic capillaries and medium-sized lymphatic vessels in mice under 2 weeks of age without affecting collecting lymphatic vessels or the blood vasculature. No effect was observed after this period. The lymphatic capillaries of neonatal mice also regressed partially in response to recombinant VEGFR-3-Ig or blocking antibodies against VEGFR-3, but not to adenovirus-encoded VEGFR-2-Ig. Despite sustained inhibitory VEGFR-3-Ig levels, lymphatic vessel regrowth was observed at 4 weeks of age. Interestingly, whereas transgenic expression of VEGF-C in the skin induced lymphatic hyperplasia even during embryogenesis, similar expression of VEGF-D resulted in lymphangiogenesis predominantly after birth. These results indicate considerable plasticity of lymphatic vessels during the early postnatal period but not thereafter, suggesting that anti-lymphangiogenic therapy can be safely applied in adults.
Collapse
Affiliation(s)
- Terhi Karpanen
- Molecular/Cancer Biology Laboratory, Biomedicum Helsinki, P.O.B. 63 (Haartmaninkatu 8), FI-00014 University of Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
591
|
Phung TL, Ziv K, Dabydeen D, Eyiah-Mensah G, Riveros M, Perruzzi C, Sun J, Monahan-Earley RA, Shiojima I, Nagy JA, Lin MI, Walsh K, Dvorak AM, Briscoe DM, Neeman M, Sessa WC, Dvorak HF, Benjamin LE. Pathological angiogenesis is induced by sustained Akt signaling and inhibited by rapamycin. Cancer Cell 2006; 10:159-70. [PMID: 16904613 PMCID: PMC2531257 DOI: 10.1016/j.ccr.2006.07.003] [Citation(s) in RCA: 340] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 02/19/2006] [Accepted: 07/18/2006] [Indexed: 01/23/2023]
Abstract
Endothelial cells in growing tumors express activated Akt, which when modeled by transgenic endothelial expression of myrAkt1 was sufficient to recapitulate the abnormal structural and functional features of tumor blood vessels in nontumor tissues. Sustained endothelial Akt activation caused increased blood vessel size and generalized edema from chronic vascular permeability, while acute permeability in response to VEGF-A was unaffected. These changes were reversible, demonstrating an ongoing requirement for Akt signaling for the maintenance of these phenotypes. Furthermore, rapamycin inhibited endothelial Akt signaling, vascular changes from myrAkt1, tumor growth, and tumor vascular permeability. Akt signaling in the tumor vascular stroma was sensitive to rapamycin, suggesting that rapamycin may affect tumor growth in part by acting as a vascular Akt inhibitor.
Collapse
Affiliation(s)
- Thuy L. Phung
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Keren Ziv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Donnette Dabydeen
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Godfred Eyiah-Mensah
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Marcela Riveros
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Carole Perruzzi
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Jingfang Sun
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Rita A. Monahan-Earley
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Ichiro Shiojima
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Janice A. Nagy
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Michelle I. Lin
- Department of Pharmacology and Program in Vascular Cell Signaling and Therapeutics, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, Connecticut 06536
| | - Kenneth Walsh
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Ann M. Dvorak
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - David M. Briscoe
- Transplantation Research Center, Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Michal Neeman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - William C. Sessa
- Department of Pharmacology and Program in Vascular Cell Signaling and Therapeutics, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, Connecticut 06536
| | - Harold F. Dvorak
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Laura E. Benjamin
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
- Correspondence:
| |
Collapse
|
592
|
Jakobsson L, Kreuger J, Holmborn K, Lundin L, Eriksson I, Kjellén L, Claesson-Welsh L. Heparan sulfate in trans potentiates VEGFR-mediated angiogenesis. Dev Cell 2006; 10:625-34. [PMID: 16678777 DOI: 10.1016/j.devcel.2006.03.009] [Citation(s) in RCA: 172] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Revised: 02/03/2006] [Accepted: 03/20/2006] [Indexed: 01/04/2023]
Abstract
Several receptor tyrosine kinases require heparan sulfate proteoglycans (HSPGs) as coreceptors for efficient signal transduction. We have studied the role of HSPGs in the development of blood capillary structures from embryonic stem cells, a process strictly dependent on signaling via vascular endothelial growth factor receptor-2 (VEGFR-2). We show, by using chimeric cultures of embryonic stem cells defective in either HS production or VEGFR-2 synthesis, that VEGF signaling in endothelial cells is fully supported by HS expressed in trans by adjacent perivascular smooth muscle cells. Transactivation of VEGFR-2 leads to prolonged and enhanced signal transduction due to HS-dependent trapping of the active VEGFR-2 signaling complex. Our data imply that direct signaling via HSPG core proteins is dispensable for a functional VEGF response in endothelial cells. We propose that transactivation of tyrosine kinase receptors by HSPGs constitutes a mechanism for crosstalk between adjacent cells.
Collapse
Affiliation(s)
- Lars Jakobsson
- Department of Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjöldsv. 20, SE-75185 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
593
|
Abstract
After over 30 years of preclinical and clinical development, antiangiogenic agents have recently entered the clinic as attractive targeted therapeutics for the treatment of cancer. Fueled by exciting new developments in the field, the AACR Special Conference was designed to broadly survey critical scientific advances in the antiangiogenic therapy of cancer. Because these advances have come primarily with the use of combinations of antiangiogenic agents with chemotherapy, or with antiangiogenic agents that also directly target the cancer cells, the central theme included the issue of drug delivery to tumors. These two major issues were addressed in concert, from basic mechanisms of action of antiangiogenic agents to new combination approaches to cancer treatment. Nearly 300 participants from 20 countries registered for the conference, drawn both from academia and industry, with a wide range in experience and background. Dr. Rakesh Jain, along with conference co-chairs, Drs. Lee Ellis and Luisa Iruela-Arispe, assembled an outstanding lineup of speakers for this conference that included many of the pioneers in the fields of angiogenesis and drug delivery from the U.S. and abroad. This resulted in an excellent overview of the advances in our understanding of cellular and molecular aspects of tumor angiogenesis and antiangiogenic therapy of cancer in combination with conventional therapy.
Collapse
Affiliation(s)
- Dan G Duda
- Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.
| |
Collapse
|
594
|
Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling - in control of vascular function. Nat Rev Mol Cell Biol 2006; 7:359-71. [PMID: 16633338 DOI: 10.1038/nrm1911] [Citation(s) in RCA: 2338] [Impact Index Per Article: 123.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vascular endothelial growth-factor receptors (VEGFRs) regulate the cardiovascular system. VEGFR1 is required for the recruitment of haematopoietic precursors and migration of monocytes and macrophages, whereas VEGFR2 and VEGFR3 are essential for the functions of vascular endothelial and lymphendothelial cells, respectively. Recent insights have shed light onto VEGFR signal transduction and the interplay between different VEGFRs and VEGF co-receptors in development, adult physiology and disease.
Collapse
Affiliation(s)
- Anna-Karin Olsson
- Department of Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldv. 20, 751 85 Uppsala, Sweden
| | | | | | | |
Collapse
|
595
|
Nakahara T, Norberg SM, Shalinsky DR, Hu-Lowe DD, McDonald DM. Effect of inhibition of vascular endothelial growth factor signaling on distribution of extravasated antibodies in tumors. Cancer Res 2006; 66:1434-45. [PMID: 16452199 DOI: 10.1158/0008-5472.can-05-0923] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antibodies and other macromolecular therapeutics can gain access to tumor cells via leaky tumor vessels. Inhibition of vascular endothelial growth factor (VEGF) signaling can reduce the vascularity of tumors and leakiness of surviving vessels, but little is known about how these changes affect the distribution of antibodies within tumors. We addressed this issue by examining the distribution of extravasated antibodies in islet cell tumors of RIP-Tag2 transgenic mice and implanted Lewis lung carcinomas using fluorescence and confocal microscopic imaging. Extravasated nonspecific immunoglobulin G (IgG) and antibodies to fibrin or E-cadherin accumulated in irregular patchy regions of stroma. Fibrin also accumulated in these regions. Anti-E-cadherin antibody, which targets epitopes on tumor cells of RIP-Tag2 adenomas, was the only antibody to achieve detectable levels within tumor cell clusters at 6 hours after i.v. injection. Treatment for 7 days with AG-013736, a potent inhibitor of VEGF signaling, reduced the tumor vascularity by 86%. The overall area density of extravasated IgG/antibodies decreased after treatment but the change was less than the reduction in vascularity and actually increased when expressed per surviving tumor vessel. Accumulation of anti-E-cadherin antibody in tumor cell clusters was similarly affected. The patchy pattern of antibodies in stroma after treatment qualitatively resembled untreated tumors and surprisingly coincided with sleeves of basement membrane left behind after pruning of tumor vessels. Together, the findings suggest that antibody transport increases from surviving tumor vessels after normalization by inhibition of VEGF signaling. Basement membrane sleeves may facilitate this transport. Antibodies preferentially distribute to tumor stroma but also accumulate on tumor cells if binding sites are accessible.
Collapse
MESH Headings
- Adenoma, Islet Cell/blood
- Adenoma, Islet Cell/blood supply
- Adenoma, Islet Cell/immunology
- Animals
- Antibodies, Neoplasm/blood
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/metabolism
- Axitinib
- Cadherins/immunology
- Carcinoma, Lewis Lung/blood
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/immunology
- Fibrin/immunology
- Fibrin/metabolism
- Imidazoles/pharmacology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Indazoles/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microspheres
- Neovascularization, Pathologic/blood
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/metabolism
- Signal Transduction
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Tsutomu Nakahara
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, University of California-San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0130, USA
| | | | | | | | | |
Collapse
|