301
|
Manuel Trigo J, Bellmunt J. Estrategias actuales en el tratamiento del carcinoma de células renales: fármacos dirigidos a dianas moleculares. Med Clin (Barc) 2008; 130:380-92. [DOI: 10.1157/13117476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
302
|
Abstract
BACKGROUND: Renal cell carcinoma is a disease marked by a unique biology which has governed it's long history of poor response to conventional cancer treatments. The discovery of the signaling pathway activated as a result of inappropriate constitutive activation of the hypoxia inducible factors (HIF), transcription factors physiologically and transiently stabilized in response to low oxygen, has provided a primary opportunity to devise treatment strategies to target this oncogenic pathway. OBJECTIVE: A review of the molecular pathogenesis of renal cell cancer as well as molecularly targeted therapies, both those currently available and those in development, will be provided. In addition, trials involving combination or sequential targeted therapy are discussed. METHODS: A detailed review of the literature describing the molecular biology of renal cell cancer and novel therapies was performed and summarized. RESULTS/CONCLUSION: Therapeutics targeting angiogenesis have provided the first class of agents which provide clinical benefit in a large majority of patients and heralded renal cell carcinoma as a solid tumor paradigm for the development of novel therapeutics. Multiple strategies targeting this pathway and now other identified pathways in renal cell carcinoma provide numerous potential opportunities to make major improvements in treating this historically devastating cancer.
Collapse
Affiliation(s)
- C Lance Cowey
- Department of Medicine, Division of Hematology and Oncology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill
| | | |
Collapse
|
303
|
Abstract
The increase in body size of humans and other vertebrates requires a physiological infrastructure to provide adequate delivery of oxygen to tissues and cells to maintain oxygen homeostasis. The heart, lungs and the vasculature are all part of a highly regulated system that ensures the distribution of the precise amount of oxygen needed throughout the mammalian organism. Given its fundamental impact on physiology and pathology, it is no surprise that the response of cells to a lack of oxygen, termed hypoxia, has been the focus of many research groups worldwide for many decades now. The transcriptional complex hypoxia-inducible factor has emerged as a key regulator of the molecular hypoxic response, mediating a wide range of physiological and cellular mechanisms necessary to adapt to reduced oxygen.
Collapse
|
304
|
|
305
|
|
306
|
Patard JJ. European Association of Urology Guidelines for Systemic Therapy in Metastatic Renal Cell Carcinoma: What is Recommended and Why? ACTA ACUST UNITED AC 2008. [DOI: 10.1016/j.eursup.2007.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
307
|
Kapitsinou PP, Haase VH. The VHL tumor suppressor and HIF: insights from genetic studies in mice. Cell Death Differ 2008; 15:650-9. [PMID: 18219317 DOI: 10.1038/sj.cdd.4402313] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The von Hippel-Lindau tumor suppressor gene product, pVHL, functions as the substrate recognition component of an E3-ubiquitin ligase, which targets the oxygen-sensitive alpha-subunit of hypoxia-inducible factor (HIF) for rapid proteasomal degradation under normoxic conditions and as such plays a central role in molecular oxygen sensing. Mutations in pVHL can be found in familial and sporadic clear cell carcinomas of the kidney, hemangioblastomas of the retina and central nervous system, and pheochromocytomas, underscoring its gatekeeper function in the pathogenesis of these tumors. Tissue-specific gene targeting of VHL in mice has demonstrated that efficient execution of pVHL-mediated HIF proteolysis under normoxia is fundamentally important for survival, proliferation, differentiation and normal physiology of many cell types, and has provided novel insights into the biological function of individual HIF transcription factors. In this review, we discuss the role of HIF in the development of the VHL phenotype.
Collapse
Affiliation(s)
- P P Kapitsinou
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
308
|
Liu J, Nussinov R. Allosteric effects in the marginally stable von Hippel-Lindau tumor suppressor protein and allostery-based rescue mutant design. Proc Natl Acad Sci U S A 2008; 105:901-6. [PMID: 18195360 PMCID: PMC2242715 DOI: 10.1073/pnas.0707401105] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Indexed: 12/31/2022] Open
Abstract
Many multifunctional tumor suppressor proteins have low stability, a property linked to cancer development. The von Hippel-Lindau tumor suppressor protein (pVHL) is one of these proteins. pVHL forms part of the E3 ubiquitin ligase complex that regulates the degradation of the hypoxia-inducible factor (HIF). Under native conditions, free pVHL is a molten globule, but it is stabilized in the E3 complex. By using molecular dynamics simulations, we observed that the interface between the two pVHL domains is the least stable region in unbound pVHL. We designed five stable mutants: one with a mutation at the interdomain interface and the others in the alpha- or beta-domains. Experimentally, type 2B pVHL disease mutant Y98N at the HIF binding site was shown to destabilize pVHL and decrease its binding affinity to HIF. Our simulations showed that the decrease in pVHL stability and binding affinity are allosterically regulated. The mutations designed to stabilize unbound wild-type pVHL, which are away from the elongin C and HIF binding sites, successfully stabilized the Y98N pVHL-elongin C complex and lowered the binding free energy of pVHL with HIF. Our results indicated both the enthalpic and dynamic allosteric components between the elongin C and HIF binding sites in pVHL, in the alpha- and beta-domains, respectively, mediated by the interdomain interface and linker. Drugs mimicking the allosteric effects of these mutants may rescue pVHL function in von Hippel-Lindau disease.
Collapse
Affiliation(s)
- Jin Liu
- *Basic Research Program, Science Applications International Corporation (SAIC)-Frederick, Inc., Center for Cancer Research Nanobiology Program, National Cancer Institute, Frederick, MD 21702; and
| | - Ruth Nussinov
- *Basic Research Program, Science Applications International Corporation (SAIC)-Frederick, Inc., Center for Cancer Research Nanobiology Program, National Cancer Institute, Frederick, MD 21702; and
- Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
309
|
Bottaro DP, Tan N, Linehan WM. Regulation of Angiogenesis by von Hippel Lindau Protein and HIF2. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
310
|
Rudge JS, Ioffe E, Cao J, Papadopoulos N, Thurston G, Wiegand SJ, Yancopoulos GD. Clinical Development of VEGF Trap. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
311
|
Gaumann A, Schlitt HJ, Geissler EK. Immunosuppression and tumor development in organ transplant recipients: the emerging dualistic role of rapamycin. Transpl Int 2007; 21:207-17. [PMID: 18069922 DOI: 10.1111/j.1432-2277.2007.00610.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer morbidity and mortality are increasingly apparent risks in transplant recipients, thus reducing life quality and overall survival. These risks have largely been attributed to long-term immunosuppressive drug therapy, which remains necessary to prevent organ allograft rejection. Interestingly, however, recent studies challenge the premise that all immunosuppressive drugs necessarily promote cancer. A particular class of immunosuppressants, referred to as mammalian target of rapamycin (mTOR) inhibitors, has been shown to have potent anti-cancer effects that are presently being tested in clinical studies. The focus of this review is to present current evidence that allows us to understand better the dual immunosuppressive and anti-cancer functions of this class of drugs used to prevent allograft rejection. We will concentrate on the different functions of mTOR that allow it to simultaneously control the immune system and tumor development. We will also discuss results from current clinical studies that either support or refute this potential dualistic role.
Collapse
Affiliation(s)
- Andreas Gaumann
- Institute of Pathology, University Regensburg, Regensburg, Germany
| | | | | |
Collapse
|
312
|
Hainsworth JD, Spigel DR, Sosman JA, Burris HA, Farley C, Cucullu H, Yost K, Hart LL, Sylvester L, Waterhouse DM, Greco FA. Treatment of Advanced Renal Cell Carcinoma with the Combination Bevacizumab/Erlotinib/Imatinib: A Phase I/II Trial. Clin Genitourin Cancer 2007; 5:427-32. [DOI: 10.3816/cgc.2007.n.030] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
313
|
Gautron L. [Gustave Roussy's (1874-1948) contribution to neuroendocrinology]. ANNALES D'ENDOCRINOLOGIE 2007; 68:400-402. [PMID: 17996213 DOI: 10.1016/j.ando.2007.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 09/26/2007] [Accepted: 09/27/2007] [Indexed: 05/25/2023]
Abstract
Gustave Roussy (1874-1948) is remembered as a distinguished French neuropathologist and a leader in the field of oncology. However, his original contribution to the study of hypothalamic functions and neuroendocrinology remains ignored. Among Roussy's discoveries is the first experimental demonstration of the hypothalamic origin of diabetes insipidus and adiposo-genital dystrophy. Also, based on his own histological work, he pioneered the concept of neurosecretion, which was termed by him "neuricrinie".
Collapse
Affiliation(s)
- L Gautron
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9077, USA.
| |
Collapse
|
314
|
Abstract
Renal cell carcinoma (RCC) accounts for approximately 2.6% of all cancers in the United States. While early stage disease is curable by surgery, the median survival of metastatic disease is only 13 months. In the last decade, there has been considerable progress in understanding the genetics of RCC. The VHL tumor suppressor gene is inactivated in the majority of RCC cases. The VHL protein (pVHL) acts as an E3 ligase that targets HIF-1, the hypoxia inducible transcription factor, for degradation by the ubiquitin proteasome system (UPS). In RCC cases with mutant pVHL, HIF-1 is stabilized and aberrantly expressed in normoxia, leading to the activation of pro-survival genes such as vascular endothelial growth factor (VEGF). This review will focus on the defect in the UPS that underlies RCC and describe the development of novel therapies that target the UPS. Publication history: Republished from Current BioData's Targeted Proteins database (TPdb; ).
Collapse
Affiliation(s)
- Paul G Corn
- MD Anderson Cancer Center, GU Medical Oncology, Box 1374, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
315
|
VEGF in physiological process and thyroid disease. ANNALES D'ENDOCRINOLOGIE 2007; 68:438-48. [PMID: 17991452 DOI: 10.1016/j.ando.2007.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 09/26/2007] [Accepted: 09/27/2007] [Indexed: 01/04/2023]
Abstract
Angiogenesis is a physiological process involving the growth of new vessels from pre-existing vasculature. Vascular endothelial growth factor (VEGF) is an important regulator of both benign and malignant disease processes in the thyroid gland. The VEGF family includes seven members respectively named VEGF-A, also known as VPF (vascular permeability factor), VEGF-B, VEGF-C, VEGF-D, all described in mammals, VEGF-E (found in Parapoxviridae), VEGF-F (also called svVEGF, for snake venom VEGF, found in viper venom) and PlGF (placental growth factor). Thyrocytes are able to synthesize and secrete VEGF. VEGF-A is implicated in tumour growth and metastasis via blood vessels while VEGF-C and VEGF-D, involved in lymphangiogenesis, favour metastasis to the cervical lymph nodes in papillary thyroid carcinomas. High levels of VEGF expression in thyroid tumour cells may correlate with a poorer outcome in papillary thyroid carcinomas. Because of its important role in malignant angiogenesis, VEGF is the preferential target of a new variety of therapeutic agents called angiogenesis inhibitors.
Collapse
|
316
|
Abstract
Mutations of the von Hippel-Lindau (VHL) or fumarate hydratase (FH) genes lead to morphologically different renal cell carcinomas with distinct clinical courses and outcomes. The VHL protein is a part of an ubiquitin ligase complex that targets proteins for proteosomal degradation. FH is one of the mitochondrial enzymes of the Kreb's cycle. Despite two different functionalities and cellular locations, loss of either VHL or FH products has been shown to alter expression levels of hypoxia-inducible factors (HIF-1alpha and HIF-2alpha) and their downstream targets. HIF proteins are key regulators of oxygen homeostasis. Tight regulation of HIF allows for cell survival and growth at the time of hypoxic stress. HIF acts via transcriptional regulation of vascular endothelial growth factor, platelet derived growth factor, endothelial growth factor receptor, glucose transporter protein 1, erythropoietin, and transforming growth factor-alpha. Loss of VHL or FH is thought to result in a pseudohypoxic state so that cellular response pathways mediated by HIF are activated despite normal oxygen conditions. Understanding of these pseudohypoxic pathways has provided a better appreciation of the molecular mechanisms of carcinogenesis in addition to providing a rationale for targeted therapeutic approaches.
Collapse
Affiliation(s)
- Gennady Bratslavsky
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892-1107, USA
| | | | | | | |
Collapse
|
317
|
Leary A, Johnston SRD. Small molecule signal transduction inhibitors for the treatment of solid tumors. Cancer Invest 2007; 25:347-65. [PMID: 17661211 DOI: 10.1080/07357900701259694] [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] [Indexed: 02/02/2023]
Abstract
A greater understanding of the pathogenesis of malignancy has led to the development of novel therapies designed to target aberrant molecular pathways that characterize and distinguish cancer cells from normal tissue. Small molecules are being designed to interfere with specific steps along the deregulated signaling cascade from the cytoplasmic membrane to the nucleus. Viable targets include growth factor receptors and their downstream second messengers, modulators of the cell cycle or apoptosis, regulators of protein trafficking and degradation, and transcription regulators. This review will discuss the small molecule signal transduction inhibitors in various stages of development and address the strategic issues relating to clinical trial design with these novel targeted agents.
Collapse
|
318
|
Schlesinger T, Appukuttan B, Hwang T, Atchaneeyakasul LO, Chan CC, Zhuang Z, Stout JT, Wilson DJ. Internal en bloc resection and genetic analysis of retinal capillary hemangioblastoma. ARCHIVES OF OPHTHALMOLOGY (CHICAGO, ILL. : 1960) 2007; 125:1189-93. [PMID: 17846357 PMCID: PMC2140243 DOI: 10.1001/archopht.125.9.1189] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
OBJECTIVE To report the clinical outcomes and molecular genetics of nongermline retinal hemangioblastoma managed by surgical resection. METHODS Retrospective case series of 3 patients aged 16 to 46 years treated at a tertiary care referral center (Casey Eye Institute, Portland, Oregon). Tumors 7 to 9 mm in diameter were removed from 3 consecutive eyes (in 3 patients) via internal en bloc surgical resection using a bimanual technique. Samples of DNA from 2 of 3 tumors were tested for von Hippel-Lindau gene (VHL) mutations as a clue to the molecular basis for spontaneously occurring hemangioblastoma. Main outcome measures were morbidity, visual acuity, resolution of macular exudates, and presence of VHL markers. RESULTS Visual acuity improved or remained stable in all patients. All 3 developed cataracts, extracted in 2 instances. Histopathological findings were typical of retinal hemangioblastoma in all cases. The cells from one patient's DNA sample showed loss of heterozygosity for the VHL gene, while no genetic abnormalities were detected in the other patient's DNA sample. CONCLUSIONS Our patients' favorable outcomes suggest that surgical resection is an option for patients with large retinal hemangioblastomas. In addition, ours may be the first report of retinal hemangioblastoma unassociated with a VHL mutation.
Collapse
Affiliation(s)
- Thomas Schlesinger
- Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | |
Collapse
|
319
|
Godinot C, de Laplanche E, Hervouet E, Simonnet H. Actuality of Warburg’s views in our understanding of renal cancer metabolism. J Bioenerg Biomembr 2007; 39:235-41. [PMID: 17665292 DOI: 10.1007/s10863-007-9088-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
More than 50 years ago, Warburg proposed that the shift in glucose metabolism from oxidative phosphorylation (OXPHOS) to glycolysis occurring in spite of an adequate oxygen supply was at the root of cancer. This hypothesis often disregarded over the following years has recently stirred up much interest due to progress made in cancer genetics and proteomics. Studies related to renal cancers have been particularly informative to understand how abnormal use of glucose and decrease in OXPHOS are linked to cell proliferation in tumors. Indeed, in aggressive tumors such as clear cell renal carcinoma, the von Hippel-Lindau factor invalidation stabilizes the hypoxia-inducible factor (HIF) in the presence of oxygen. HIF stimulating glycolytic gene expression increases the glycolytic flux. Deficiencies in genes involved in oxidative phosphorylation that can explain the down-regulation of OXPHOS components also begin to be identified. These findings are important in the search for novel therapeutic approaches to cancer treatment.
Collapse
Affiliation(s)
- Catherine Godinot
- Center of Molecular and Cellular Genetics (CGMC), UMR 5534, CNRS, University Claude Bernard of Lyon 1, 69622 Villeurbanne, France.
| | | | | | | |
Collapse
|
320
|
Podar K, Richardson PG, Chauhan D, Anderson KC. Targeting the vascular endothelial growth factor pathway in the treatment of multiple myeloma. Expert Rev Anticancer Ther 2007; 7:551-66. [PMID: 17428175 DOI: 10.1586/14737140.7.4.551] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Multiple myeloma is a clonal plasma cell malignancy within the bone marrow associated with bone loss, renal disease and immunodeficiency. Despite new insights into the pathogenesis of multiple myeloma and novel targeted therapies, the median survival remains 3-5 years. It is now well established that the intimate relation between the tumor cells and components of the microenvironment plays a key role in multiple myeloma pathogenesis. Specifically, tumor cells impact the bone marrow and thereby cause immune suppression and lytic bone lesions; conversely, components of the bone marrow provide signals that influence the behavior of multiple myeloma cells, including tumor cell growth, survival, migration and drug resistance. Important contributing effectors are tumor cell-stroma cell and cell-extracellular matrix contacts, the bone marrow vasculature, and a variety of cytokines and growth factors in the bone marrow milieu.
Collapse
Affiliation(s)
- Klaus Podar
- Dana-Farber Cancer Institute, Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
321
|
Abstract
Approved for the treatment of advanced renal cell carcinoma by the US FDA and other regulatory agencies, sorafenib is an agent with multiple targets that may also prove beneficial in other malignancies. Phase III trials are underway in melanoma, hepatocellular carcinoma and non-small-cell lung cancer. Scrutiny of the Phase II data and correlative studies conducted in that context suggests that inhibition of angiogenesis and signaling in tumor cells may play a part in the clinical efficacy of sorafenib. Although the vascular endothelial growth factor receptor inhibitors are the most populated class of targeted agents in cancer clinical trials, sorafenib may prove to have unique properties that distinguish it. A detailed discussion of the clinical trials in renal cell carcinoma, melanoma and hepatocellular carcinoma highlights what is known and what has yet to be understood about this agent.
Collapse
Affiliation(s)
- Keith T Flaherty
- University of Pennsylvania, Abramson Cancer Center, PA 19104, USA.
| |
Collapse
|
322
|
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: 110] [Impact Index Per Article: 6.1] [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
|
323
|
Sudarshan S, Pinto PA, Neckers L, Linehan WM. Mechanisms of disease: hereditary leiomyomatosis and renal cell cancer--a distinct form of hereditary kidney cancer. ACTA ACUST UNITED AC 2007; 4:104-10. [PMID: 17287871 DOI: 10.1038/ncpuro0711] [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] [Received: 10/04/2006] [Accepted: 11/27/2006] [Indexed: 01/10/2023]
Abstract
Renal cell carcinoma (RCC) represents a group of diseases linked by their primary site of origin, the kidney. Studies of families with a genetic predisposition to the development of kidney cancer have revealed that multiple genes are involved in the molecular pathogenesis of RCC. Germline mutations in a gene that encodes a Krebs cycle enzyme have been found to result in a distinct clinical entity referred to as hereditary leiomyomatosis and renal cell cancer (HLRCC). HLRCC is inherited in an autosomal-dominant fashion. Affected individuals in HLRCC families are at risk for the development of leiomyomas of the skin and uterus as well as renal cancers. HLRCC-associated kidney tumors are often biologically aggressive. Linkage analysis has identified germline alterations in the fumarate hydratase (FH) gene associated with HLRCC. While the mechanisms of molecular carcinogenesis are not entirely understood, several lines of evidence derived from clinical and basic research suggest that pseudohypoxia might drive cellular transformation. The role of FH mutations in sporadic tumors seems to be limited. Nevertheless, continued investigation of HLRCC should provide further insight into the mechanisms of kidney cancer development, and could potentially identify targets for new therapeutic approaches to RCC.
Collapse
Affiliation(s)
- Sunil Sudarshan
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1107, USA
| | | | | | | |
Collapse
|
324
|
Linehan WM, Pinto PA, Srinivasan R, Merino M, Choyke P, Choyke L, Coleman J, Toro J, Glenn G, Vocke C, Zbar B, Schmidt LS, Bottaro D, Neckers L. Identification of the genes for kidney cancer: opportunity for disease-specific targeted therapeutics. Clin Cancer Res 2007; 13:671s-679s. [PMID: 17255292 DOI: 10.1158/1078-0432.ccr-06-1870] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent advances in understanding the kidney cancer gene pathways has provided the foundation for the development of targeted therapeutic approaches for patients with this disease. Kidney cancer is not a single disease; it includes a number of different types of renal cancers, each with different histologic features, a different clinical course, a different response to therapy, and different genes causing the defects. Most of what is known about the genetic basis of kidney cancer has been learned from study of the inherited forms of kidney cancer: von Hippel Lindau (VHL gene), hereditary papillary renal carcinoma (c-Met gene), Birt Hogg Dubé (BHD gene), and hereditary leiomyomatosis renal cell cancer (fumarate hydratase gene). These Mendelian single-gene syndromes provide a unique opportunity to evaluate the effectiveness of agents that target the VHL, c-Met, BHD, and fumarate hydratase pathways.
Collapse
Affiliation(s)
- W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Longo R, Gasparini G. Challenges for patient selection with VEGF inhibitors. Cancer Chemother Pharmacol 2007; 60:151-70. [PMID: 17370072 DOI: 10.1007/s00280-006-0403-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Accepted: 12/04/2006] [Indexed: 10/23/2022]
Abstract
As targeted therapies for cancer become increasingly integrated into standard practice, appropriate selection of the patients most likely to benefit from these therapies is now receiving critical scrutiny. Early experience with therapies directed at targets that are definitively overactive (e.g. the bcr-abl tyrosine kinase targeted by imatinib) or over-expressed [e.g. the human epidermal growth factor receptor 2 (HER2) targeted by trastuzumab] has generated the perception that pre-treatment target assessment is a pre-requisite for therapy with all targeted agents. However, emerging evidence suggests that this is not presently feasible for anti-angiogenic agents. Despite considerable evidence for the association of intratumoral and/or plasma vascular endothelial growth factor (VEGF) levels with tumor progression and/or poor prognosis, pre-treatment VEGF levels do not appear to be predictive of response to anti-angiogenic therapy. This may possibly be due to the complexity of the angiogenic pathways and the limitations associated with current methods of VEGF detection and quantification; e.g. low assay sensitivity and lack of standardized methods could prevent detection of very small increases in VEGF, which may be clinically important in patients with tumors that are highly dependent on this growth factor. In addition to a general lack of agreement as to the relative clinical relevance of circulating versus tumor VEGF levels, the absence of a 'gold standard' VEGF detection assay and the lack of a predefined, clinically relevant cut-off pose a significant hindrance to the clinical utility of VEGF measurements for therapy selection. Given the fundamental importance of angiogenesis for tumor growth and progression, and the key role of VEGF in these processes, presently it seems appropriate to view anti-VEGF agents such as bevacizumab (Avastin) as having potential utility, independently of pre-treatment screening. Further research is needed to define the relationship between potential surrogate markers of VEGF pathway activity and clinical outcomes.
Collapse
Affiliation(s)
- R Longo
- Division of Medical Oncology, San Filippo Neri Hospital, Via Martinotti 20, 00135 Rome, Italy.
| | | |
Collapse
|
326
|
Tovar-Castillo LE, Cancino-Díaz JC, García-Vázquez F, Cancino-Gómez FG, León-Dorantes G, Blancas-González F, Jiménez-Zamudio L, García-Latorre E, Cancino-Díaz ME. Under-expression of VHL and over-expression of HDAC-1, HIF-1?, LL-37, and IAP-2 in affected skin biopsies of patients with psoriasis. Int J Dermatol 2007; 46:239-46. [PMID: 17343577 DOI: 10.1111/j.1365-4632.2006.02962.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND A feature of psoriasis is the rapid proliferation of keratinocytes, during which apoptosis is blocked and angiogenesis starts. It is known that tumor hypoxic cells produce histone deacetylase-1 (HDAC-1), which up-regulates hypoxia-inducible factor-1alpha (HIF-1alpha) and down-regulates von Hippel-Lindau (VHL) protein by up-regulating vascular endothelial growth factor (VEGF) expression. It has been reported recently that the porcine peptide PR39 (homologous to human LL-37) has angiogenic and antiapoptotic activity. Thus, LL-37, induced by insulin-like growth factor-1 (IGF-1), could help in the production of VEGF. PR39 also induces the expression of inhibitor of apoptosis protein-2 (IAP-2), which blocks apoptosis. The purpose of this work was to analyze whether these genes and their proteins are expressed in psoriatic biopsies. METHODS Using semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR) messenger RNA (mRNA) expression and immunohistochemical staining, we studied VHL, IAP-2, and related genes in skin biopsies from psoriatic patients and healthy subjects. RESULTS An over-expression of the mRNA for HDAC-1, HIF-1alpha, LL-37, and IGF-1 in psoriatic skin, in comparison with skin from healthy subjects, was found. The antiangiogenic VHL mRNA and protein were under-expressed in psoriatic skin and highly expressed in healthy skin. The antiapoptotic IAP-2 was over-expressed in dermal endothelial cells from psoriatic skin. The pro-apoptotic Bax, Fas, and FasL mRNAs were expressed. CONCLUSIONS These findings suggest that there could be an association of HDAC-1, HIF-1alpha, LL-37, VHL, and IAP-2 with angiogenic and apoptotic mechanisms in psoriasis.
Collapse
Affiliation(s)
- Laura E Tovar-Castillo
- Departamentos de Inmunologia and Microbiologia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Servicio de Dermatología, Hospital General de México, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
327
|
Lane BR, Rini BI, Novick AC, Campbell SC. Targeted molecular therapy for renal cell carcinoma. Urology 2007; 69:3-10. [PMID: 17270598 DOI: 10.1016/j.urology.2006.09.028] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 06/01/2006] [Accepted: 09/12/2006] [Indexed: 11/18/2022]
Affiliation(s)
- Brian R Lane
- Glickman Urological Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
328
|
Affiliation(s)
- William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Howard Hughes Medical Institute, Chevy Chase, Maryland 20815;
| |
Collapse
|
329
|
Rini BI, Rathmell WK. Biological Aspects and Binding Strategies of Vascular Endothelial Growth Factor in Renal Cell Carcinoma. Clin Cancer Res 2007; 13:741s-746s. [PMID: 17255303 DOI: 10.1158/1078-0432.ccr-06-2110] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a key mediator in the pathogenesis of renal cell carcinoma (RCC). VEGF is up-regulated in clear cell RCC as a result of loss of the von Hippel-Lindau tumor suppressor gene and subsequent activation of the hypoxia response pathway. VEGF expression drives the migration and proliferation of endothelial cells to support the extensive angiogenesis in RCC. Strategies have been developed to bind and neutralize VEGF and have been investigated in RCC with promising results. Bevacizumab, a VEGF ligand-binding antibody, has shown prolonged time-to-progression versus placebo in treatment-refractory RCC patients and is being investigated currently in multiple RCC settings. VEGF-Trap is also a VEGF binding molecule with ongoing investigation in RCC.
Collapse
Affiliation(s)
- Brian I Rini
- Department of Solid Tumor Oncology and Urology, Cleveland Clinic Taussig Cancer Center, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | |
Collapse
|
330
|
Abstract
Germ line VHL tumor suppressor gene loss-of-function mutations cause von Hippel-Lindau disease, which is associated with an increased risk of central nervous system hemangioblastomas, clear cell renal carcinomas, and pheochromocytomas. Somatic VHL mutations are also common in sporadic clear cell renal carcinomas. The VHL gene product, pVHL, is part of a ubiquitin ligase complex that targets the alpha-subunits of the heterodimeric transcription factor hypoxia-inducible factor (HIF) for polyubiquitylation, and hence, proteasomal degradation, when oxygen is available. pVHL-defective clear cell renal carcinomas overproduce a variety of mRNAs that are under the control of HIF, including the mRNAs that encode vascular endothelial growth factor, platelet-derived growth factor B, and transforming growth factor alpha. In preclinical models, down-regulation of HIF-alpha, especially HIF-2alpha, is both necessary and sufficient for renal tumor suppression by pVHL. These observations are probably relevant to the demonstrated clinical activity of vascular endothelial growth factor antagonists in clear cell renal carcinoma and form a foundation for the testing of additional agents that inhibit HIF, or HIF-responsive gene products, in this disease.
Collapse
Affiliation(s)
- William G Kaelin
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
331
|
Yan Q, Bartz S, Mao M, Li L, Kaelin WG. The hypoxia-inducible factor 2alpha N-terminal and C-terminal transactivation domains cooperate to promote renal tumorigenesis in vivo. Mol Cell Biol 2007; 27:2092-102. [PMID: 17220275 PMCID: PMC1820491 DOI: 10.1128/mcb.01514-06] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Hypoxia-inducible factor (HIF) is a heterodimeric transcription factor, consisting of an alpha subunit and a beta subunit, that controls cellular responses to hypoxia. HIFalpha contains two transcriptional activation domains called the N-terminal transactivation domain (NTAD) and the C-terminal transactivation domain (CTAD). HIFalpha is destabilized by prolyl hydroxylation catalyzed by EglN family members. In addition, CTAD function is inhibited by asparagine hydroxylation catalyzed by FIH1. Both hydroxylation reactions are linked to oxygen availability. The von Hippel-Lindau tumor suppressor protein (pVHL) is frequently mutated in kidney cancer and is part of the ubiquitin ligase complex that targets prolyl hydroxylated HIFalpha for destruction. Recent studies suggest that HIF2alpha plays an especially important role in promoting tumor formation by pVHL-defective renal carcinoma cells among the three HIFalpha paralogs. Here we dissected the relative contribution of the two HIF2alpha transactivation domains to hypoxic gene activation and renal carcinogenesis and investigated the regulation of the HIF2alpha CTAD by FIH1. We found that the HIF2alpha NTAD is capable of activating both artificial and naturally occurring HIF-responsive promoters in the absence of the CTAD. Moreover, we found that the HIF2alpha CTAD, in contrast to the HIF1alpha CTAD, is relatively resistant to the inhibitory effects of FIH1 under normoxic conditions and that, perhaps as a result, both the NTAD and CTAD cooperate to promote renal carcinogenesis in vivo.
Collapse
Affiliation(s)
- Qin Yan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
332
|
Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007; 356:115-24. [PMID: 17215529 DOI: 10.1056/nejmoa065044] [Citation(s) in RCA: 4469] [Impact Index Per Article: 248.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Since sunitinib malate has shown activity in two uncontrolled studies in patients with metastatic renal-cell carcinoma, a comparison of the drug with interferon alfa in a phase 3 trial is warranted. METHODS We enrolled 750 patients with previously untreated, metastatic renal-cell carcinoma in a multicenter, randomized, phase 3 trial to receive either repeated 6-week cycles of sunitinib (at a dose of 50 mg given orally once daily for 4 weeks, followed by 2 weeks without treatment) or interferon alfa (at a dose of 9 MU given subcutaneously three times weekly). The primary end point was progression-free survival. Secondary end points included the objective response rate, overall survival, patient-reported outcomes, and safety. RESULTS The median progression-free survival was significantly longer in the sunitinib group (11 months) than in the interferon alfa group (5 months), corresponding to a hazard ratio of 0.42 (95% confidence interval, 0.32 to 0.54; P<0.001). Sunitinib was also associated with a higher objective response rate than was interferon alfa (31% vs. 6%, P<0.001). The proportion of patients with grade 3 or 4 treatment-related fatigue was significantly higher in the group treated with interferon alfa, whereas diarrhea was more frequent in the sunitinib group (P<0.05). Patients in the sunitinib group reported a significantly better quality of life than did patients in the interferon alfa group (P<0.001). CONCLUSIONS Progression-free survival was longer and response rates were higher in patients with metastatic renal-cell cancer who received sunitinib than in those receiving interferon alfa (ClinicalTrials.gov numbers, NCT00098657 and NCT00083889 [ClinicalTrials.gov]).
Collapse
Affiliation(s)
- Robert J Motzer
- Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
333
|
Abstract
Immunotherapy with interleukin-2 and interferon-alpha has been the only viable option in metastatic renal cell cancer for almost two decades. In the last several years, significant advances in the understanding of the underlying biological and molecular mechanisms of renal cell carcinoma, particularly the role of tumour angiogenesis, have led to the identification of rational therapeutic targets and permitted the design of molecularly targeted therapeutics. At present, new compounds targeting specific signalling pathways are available and have successfully passed clinical testing. The use of small molecules, such as multitargeted tyrosine kinase inhibitors, the mTOR inhibitors and monoclonal antibodies, is dramatically changing the existing concepts of systemic treatment for metastatic kidney cancer.
Collapse
Affiliation(s)
- Doru T Alexandrescu
- Georgetown University, Washington Cancer Institute, 110 Irving Street, N.W., Washington, DC 20010, USA.
| | | |
Collapse
|
334
|
Hickey MM, Simon MC. Regulation of angiogenesis by hypoxia and hypoxia-inducible factors. Curr Top Dev Biol 2007; 76:217-57. [PMID: 17118268 DOI: 10.1016/s0070-2153(06)76007-0] [Citation(s) in RCA: 198] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Maintenance of oxygen homeostasis is critical for the survival of multicellular organs. As a result, both invertebrates and vertebrates have developed highly specialized mechanisms to sense changes in oxygen levels and to mount adequate cellular and systemic responses to these changes. Hypoxia, or low oxygen tension, occurs in physiological situations such as during embryonic development, as well as in pathological conditions such as ischemia, wound healing, and cancer. A primary effector of the adaptive response to hypoxia in mammals is the hypoxia-inducible factor (HIF) family of transcription regulators. These proteins activate the expression of a broad range of genes that mediate many of the responses to decreased oxygen concentration, including enhanced glucose uptake, increased red blood cell production, and the formation of new blood vessels via angiogenesis. This latter process is dynamic and results in the establishment of a mature vascular system that is indispensable for proper delivery of oxygen and nutrients to all cells in both normal tissue and hypoxic regions. Angiogenesis is essential for normal development and neoplastic disease as tumors must develop mechanisms to stimulate vascularization to meet increasing metabolic demands. The link between hypoxia and the regulation of angiogenesis is an area of intense research and the molecular details of this connection are still being elaborated. This chapter will provide an overview of current knowledge and highlight new insights into the importance of HIF and hypoxia in angiogenesis in both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Michele M Hickey
- Department of Cell and Molecular Biology, Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia 19104, USA
| | | |
Collapse
|
335
|
Abstract
BACKGROUND von Hippel-Lindau Disease (VHL) is an autosomal dominant inherited systemic cancer syndrome. Recently, many advances have contributed to the understanding of VHL pathophysiology. METHODS In this article we review recent developments and summarize our findings in VHL molecular pathology related to retinal and optic nerve diseases. RESULTS Loss of heterozygosity (LOH) within the VHL gene is detected in the stromal cells surrounding the capillary endothelial cells and admixed with glial cells in ocular hemangioblastomas. This finding is in line with similar findings in VHL-associated CNS hemangioblastoma and renal clear cell carcinomas. Increases of vascular endothelial growth factor (VEGF), hypoxia induced factor (HIF), and ubiquitin are found in ocular hemangioblastomas.Interestingly, tumorlet cells, which are composed of poorly differentiated small cells with prominent dark nuclei and little cytoplasm, as well as several stem cell markers, such as erythropoietin (Epo), Epo receptor (EpoR), and CD133, are present in ocular VHL lesions. CXCR4, a CXC chemokine receptor is also expressed in retinal VHL hemangioblastomas. CONCLUSIONS These findings imply that VHL cells with LOH of the tumor suppressor gene, most likely originate from a hematopoietic/vascular lineage. Targeting these proteins and ischemic factors, not VEGF alone, may be a potential therapeutic approach for VHL-associated ocular hemangioblastomas.
Collapse
Affiliation(s)
- Chi-Chao Chan
- National Eye Institute, National Institutes of Health, Bethesda 20892-1857, USA.
| | | | | |
Collapse
|
336
|
Wu XH, Zhou HJ, Lee J. Dihydroartemisinin inhibits angiogenesis induced by multiple myeloma RPMI8226 cells under hypoxic conditions via downregulation of vascular endothelial growth factor expression and suppression of vascular endothelial growth factor secretion. Anticancer Drugs 2006; 17:839-48. [PMID: 16926633 DOI: 10.1097/01.cad.0000224443.85834.32] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Multiple myeloma remains incurable to date; therefore, new biologically target-based therapies are urgently needed. Our previous studies have showed that the antimalarial dihydroartemisinin possessed antiangiogenic activity in solid tumors. The present study evaluated the effect of dihydroartemisinin on human multiple myeloma-induced angiogenesis under hypoxia and elucidated its mechanism of action. An in-vivo chicken chorioallantoic membrane model was used to examine the effect of dihydroartemisinin on multiple myeloma-induced angiogenesis. Compared with conditioned medium of control, conditioned medium from human multiple myeloma RPMI8226 cells pretreated with 3 micromol/l dihydroartemisinin in hypoxia was observed to reduce microvessel growth on chicken chorioallantoic membranes by approximately 28.6% (P<0.05). The level of vascular endothelial growth factor in conditioned medium was determined by enzyme-linked immunosorbent assay. The results confirmed that 3 micromol/l dihydroartemisinin could significantly decrease vascular endothelial growth factor secretion by RPMI8226 cells (P<0.05), which correlated well with the reduction of multiple myeloma-induced angiogenesis on chicken chorioallantoic membranes. Western blot and reverse transcription-polymerase chain reaction results revealed that dihydroartemisinin downregulated the expression of vascular endothelial growth factor in RPMI8226 cells in hypoxia. In addition, we demonstrated that dihydroartemisinin reduced extracellular signal-regulated kinase 1/2 activation and inhibited growth of RPMI8226 cells under hypoxic conditions. Therefore, we concluded that dihydroartemisinin, which is already used to treat malaria and is well tolerated, possesses potential as an antiangiogenic drug in multiple myeloma therapy and thereby may improve patient outcome.
Collapse
Affiliation(s)
- Xiu-Hua Wu
- Institute of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PRC
| | | | | |
Collapse
|
337
|
Santarpia L, Lapa D, Benvenga S. Germline mutation of von Hippel-Lindau (VHL) gene 695 G>A (R161Q) in a patient with a peculiar phenotype with type 2C VHL syndrome. Ann N Y Acad Sci 2006; 1073:198-202. [PMID: 17102087 DOI: 10.1196/annals.1353.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Von Hippel-Lindau (VHL) disease is an autosomal dominant familial neoplastic disorder with an estimated birth incidence of approximately 1:36000 live. VHL has intrafamilial variability expression and it is characterized by the predisposition to develop hemangioblastomas of the central nervous system and retina, pheochromocytomas, clear-cell renal carcinoma, adenomas, and carcinomas of the pancreas, paragangliomas, renal and pancreatic cysts, papillary cystadenomas of the epididymis and, rarely, cystadenomas of the endolymphatic sac tumor and broad ligament. We describe a Sicilian girl with type 2C VHL who showed the apparently de novo mutation R161Q in association with an extra-axial supratentorial frontal meningioma, which can be included as a characteristic sign in VHL.
Collapse
Affiliation(s)
- Libero Santarpia
- Sezione di Endocrinologia del Dipartimento Clinico Sperimentale di Medicina e Farmacologia Policlinico Universitario, Messina, I-98125, Italy
| | | | | |
Collapse
|
338
|
Stockmann C, Fandrey J. Hypoxia-induced erythropoietin production: a paradigm for oxygen-regulated gene expression. Clin Exp Pharmacol Physiol 2006; 33:968-79. [PMID: 17002676 DOI: 10.1111/j.1440-1681.2006.04474.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mechanisms controlling the expression of the gene encoding for the hormone erythropoietin (EPO) are exemplary for oxygen-regulated gene expression. In humans and other mammals, hypoxia modulates EPO levels by increasing expression of the EPO gene. An association between polycythaemia and people living at high altitudes was first reported more than 100 years ago. Since the identification of EPO as the humoral regulator of red blood cell production and the cloning of the EPO gene, considerable progress has been made in understanding the regulation of EPO gene expression. This has finally led to the identification of a widespread cellular oxygen-sensing mechanism. Central to this mechanism is the transcription factor complex hypoxia-inducible factor (HIF)-1. The abundance and activity of HIF-1, a heterodimer of an alpha- and beta-subunit, is predominantly regulated by oxygen-dependent post-translational hydroxylation of the alpha-subunit. Non-heme ferrous iron containing hydroxylases use dioxygen and 2-oxoglutarate to specifically target proline and an asparagine residue in HIF-1alpha. As such, the three prolyl hydroxylases (prolyl hydroxylase domain-containing protein (PHD) 1, PHD2 and PHD3) and the asparagyl hydroxylase (factor inhibiting HIF (FIH)-1) act as cellular oxygen sensors. In addition to erythropoiesis, HIF-1 regulates a broad range of physiologically relevant genes involved in angiogenesis, apoptosis, vasomotor control and energy metabolism. Therefore, the HIF system is implicated in the pathophysiology of many human diseases. In addition to the tight regulation by oxygen tension, temporal and tissue-specific signals limit expression of the EPO gene primarily to the fetal liver and the adult kidney.
Collapse
|
339
|
Pan J, Mestas J, Burdick MD, Phillips RJ, Thomas GV, Reckamp K, Belperio JA, Strieter RM. Stromal derived factor-1 (SDF-1/CXCL12) and CXCR4 in renal cell carcinoma metastasis. Mol Cancer 2006; 5:56. [PMID: 17083723 PMCID: PMC1636662 DOI: 10.1186/1476-4598-5-56] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Accepted: 11/03/2006] [Indexed: 12/31/2022] Open
Abstract
Renal cell carcinoma (RCC) is characterized by organ-specific metastases. The chemokine stromal derived factor-1 (SDF-1/CXCL12) and its receptor CXCR4 have been suggested to regulate organ-specific metastasis in various other cancers. On this basis, we hypothesized that the biological axis of CXCL12 via interaction with its receptor, CXCR4, is a major mechanism for RCC metastasis. We demonstrated that CXCR4 was significantly expressed on circulating cytokeratin+ RCC cells from patients with known metastatic RCC. We detected up-regulation of CXCR4 mRNA and protein levels on a human RCC cell line by either knockdown of the von Hippel-Lindau (VHL) tumor suppressor protein, or incubating the cells under hypoxic conditions. The enhanced CXCR4 expression was mediated through the interaction of the Hypoxia Inducible Factor-1α (HIF-1α) with the promoter region of the CXCR4 gene. Furthermore, the expression of CXCR4 on human RCC directly correlated with their metastatic ability in vivo in both heterotopic and orthotopic SCID mouse models of human RCC. Neutralization of CXCL12 in SCID mice abrogated metastasis of RCC to target organs expressing high levels of CXCL12; without altering tumor cell proliferation, apoptosis, or tumor-associated angiogenesis. Therefore, our data suggest that the CXCL12/CXCR4 biological axis plays an important role in regulating the organ-specific metastasis of RCC.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/secondary
- Cell Hypoxia
- Cell Line, Tumor
- Chemokine CXCL12
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/pharmacology
- Chemokines, CXC/physiology
- Chemotaxis/drug effects
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Keratins/analysis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mice
- Mice, SCID
- Promoter Regions, Genetic
- RNA Interference
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Transcriptional Activation
- Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
Collapse
Affiliation(s)
- Judong Pan
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Javier Mestas
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Marie D Burdick
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Roderick J Phillips
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - George V Thomas
- Department of Pathology, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Karen Reckamp
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - John A Belperio
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| | - Robert M Strieter
- Department of Medicine, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
- Department of Pathology, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, 900 Veteran Ave., Los Angeles, California, USA
| |
Collapse
|
340
|
Abstract
PURPOSE OF REVIEW Sorafenib is an oral, multikinase inhibitor that was recently approved for use in metastatic renal cancer. It is currently undergoing investigation in locally advanced renal cancer and in other tumor types. RECENT FINDINGS Sorafenib was initially developed as an inhibitor of Raf kinase; however, it has broad spectrum activity against multiple tyrosine kinases, including angiogenic factors VEGFR and PDGFR. Common toxicities experienced with sorafenib include hypertension, hand-foot syndrome, rash, diarrhea and fatigue. Early clinical trials suggested that sorafenib acts as a cytostatic agent, as many patients experienced prolonged disease stabilization but insufficient tumor shrinkage to meet RECIST criteria for response. To assess whether sorafenib's growth inhibition translated into a clinical benefit, a phase II randomized discontinuation trial was designed. This trial demonstrated that sorafenib increased progression-free survival in patients with metastatic renal cell cancer; the phase II data were confirmed in a large international phase III trial. SUMMARY In this review, we will discuss the clinical development of sorafenib and its role in the treatment of renal cancer. Additionally, we will highlight critical methods of clinical trial design and biomarker development that contribute to the development of sorafenib.
Collapse
Affiliation(s)
- Olwen Hahn
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Illinois 60637, USA
| | | |
Collapse
|
341
|
Abstract
Adaptation to low oxygen tension (hypoxia) in cells and tissues leads to the transcriptional induction of a series of genes that participate in angiogenesis, iron metabolism, glucose metabolism, and cell proliferation/survival. The primary factor mediating this response is the hypoxia-inducible factor-1 (HIF-1), an oxygen-sensitive transcriptional activator. HIF-1 consists of a constitutively expressed subunit HIF-1beta and an oxygen-regulated subunit HIF-1alpha (or its paralogs HIF-2alpha and HIF-3alpha). The stability and activity of the alpha subunit of HIF are regulated by its post-translational modifications such as hydroxylation, ubiquitination, acetylation, and phosphorylation. In normoxia, hydroxylation of two proline residues and acetylation of a lysine residue at the oxygen-dependent degradation domain (ODDD) of HIF-1alpha trigger its association with pVHL E3 ligase complex, leading to HIF-1alpha degradation via ubiquitin-proteasome pathway. In hypoxia, the HIF-1alpha subunit becomes stable and interacts with coactivators such as cAMP response element-binding protein binding protein/p300 and regulates the expression of target genes. Overexpression of HIF-1 has been found in various cancers, and targeting HIF-1 could represent a novel approach to cancer therapy.
Collapse
Affiliation(s)
- Qingdong Ke
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | | |
Collapse
|
342
|
Gläsker S, Tran MGB, Shively SB, Ikejiri B, Lonser RR, Maxwell PH, Zhuang Z, Oldfield EH, Vortmeyer AO. Epididymal cystadenomas and epithelial tumourlets: effects of VHL deficiency on the human epididymis. J Pathol 2006; 210:32-41. [PMID: 16841375 DOI: 10.1002/path.2029] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although epididymal cystadenomas (ECAs) are among the most frequent VHL disease-associated tumours, fundamental questions about their pathogenesis have remained unanswered. Classification of ECAs is controversial, and the cell of origin is unknown. It is also unknown whether ECAs-like other VHL disease-associated tumours-arise as a result of VHL gene inactivation, and whether ECAs exhibit subsequent activation of hypoxia-inducible factor HIF. Moreover, the morphological spectrum of earliest ECA formation is unknown. In a detailed molecular pathological analysis of a series of epididymides collected from VHL patients at autopsy, we found that ECAs are true neoplasms that arise secondary to inactivation of the wild-type copy of the VHL gene, followed by early and simultaneous activation of HIF1 and HIF2 associated with up-regulation of downstream targets, including CAIX and GLUT-1. The observations also indicate that ECA formation evolves from a variety of microscopic epithelial tumourlets, and that these tumourlets are confined to the efferent ductular system. Although genetic and immunohistochemical analysis of precursor structures consistently revealed VHL gene inactivation and activation of HIF in the precursor lesions, only a small subset appears to progress into frank cystadenoma. Thus, ECA tumorigenesis in VHL disease shares fundamental principles with tumorigenesis in other affected organ systems.
Collapse
Affiliation(s)
- S Gläsker
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
343
|
Hergovich A, Lisztwan J, Thoma CR, Wirbelauer C, Barry RE, Krek W. Priming-dependent phosphorylation and regulation of the tumor suppressor pVHL by glycogen synthase kinase 3. Mol Cell Biol 2006; 26:5784-96. [PMID: 16847331 PMCID: PMC1592755 DOI: 10.1128/mcb.00232-06] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene is linked to the development of tumors of the eyes, kidneys, and central nervous system. VHL encodes two gene products, pVHL30 and pVHL19, of which one, pVHL30, associates functionally with microtubules (MTs) to regulate their stability. Here we report that pVHL30 is a novel substrate of glycogen synthase kinase 3 (GSK3) in vitro and in vivo. Phosphorylation of pVHL on serine 68 (S68) by GSK3 requires a priming phosphorylation event at serine 72 (S72) mediated in vitro by casein kinase I. Functional analysis of pVHL species carrying nonphosphorylatable or phosphomimicking mutations at S68 and/or S72 reveals a central role for these phosphorylation events in the regulation of pVHL's MT stabilization (but not binding) activity. Taken together, our results identify pVHL as a novel priming-dependent substrate of GSK3 and suggest a dual-kinase mechanism in the control of pVHL's MT stabilization function. Since GSK3 is a component of multiple signaling pathways that are altered in human cancer, our results further imply that normal operation of the GSK3-pVHL axis may be a critical aspect of pVHL's tumor suppressor mechanism through the regulation of MT dynamics.
Collapse
|
344
|
Abstract
The physiologic response to changes in cellular oxygen tension is ultimately governed by a heterodimeric transcription factor called hypoxia-inducible factor (HIF), which, in adaptation to compromised oxygen availability, transactivates a myriad of genes, including those responsible for de novo vascularization, production of oxygen-carrying red blood cells, and anaerobic metabolism. Accumulation of HIF is observed in most types of solid tumors and is frequently associated with poor prognosis and disease progression, underscoring the importance and relevance of HIF in cancer. The protein stability and, thereby, the activity of HIF are principally regulated by the von Hippel-Lindau (VHL) tumor suppressor-containing E3 ubiquitin ligase complex (ECV) that targets the catalytic subunit HIFalpha for oxygen-dependent ubiquitin-mediated destruction. Individuals who inherit germline VHL mutation develop VHL disease, which is characterized by the development of hypervascular tumors in multiple yet specific organs. This review will examine recent progress in our understanding of the molecular mechanisms governing the function of ECV and the significance of consequential regulation of HIF in oncogenesis.
Collapse
Affiliation(s)
- Michael Ohh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| |
Collapse
|
345
|
Kim WY, Safran M, Buckley MRM, Ebert BL, Glickman J, Bosenberg M, Regan M, Kaelin WG. Failure to prolyl hydroxylate hypoxia-inducible factor alpha phenocopies VHL inactivation in vivo. EMBO J 2006; 25:4650-62. [PMID: 16977322 PMCID: PMC1589988 DOI: 10.1038/sj.emboj.7601300] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Accepted: 07/26/2006] [Indexed: 12/14/2022] Open
Abstract
Many functions have been assigned to the von Hippel-Lindau tumor suppressor gene product (pVHL), including targeting the alpha subunits of the heterodimeric transcription factor HIF (hypoxia-inducible factor) for destruction. The binding of pVHL to HIFalpha requires that HIFalpha be hydroxylated on one of two prolyl residues. We introduced HIF1alpha and HIF2alpha variants that cannot be hydroxylated on these sites into the ubiquitously expressed ROSA26 locus along with a Lox-stop-Lox cassette that renders their expression Cre-dependent. Expression of the HIF2alpha variant in the skin and liver induced changes that were highly similar to those seen when pVHL is lost in these organs. Dual expression of the HIF1alpha and HIF2alpha variants in liver, however, more closely phenocopied the changes seen after pVHL inactivation than did the HIF2alpha variant alone. Moreover, gene expression profiling confirmed that the genes regulated by HIF1alpha and HIF2alpha in the liver are overlapping but non-identical. Therefore, the pathological changes caused by pVHL inactivation in skin and liver are due largely to dysregulation of HIF target genes.
Collapse
Affiliation(s)
- William Y Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michal Safran
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marshall R M Buckley
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan Glickman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marcus Bosenberg
- Department of Pathology, University of Vermont, Burlington, VT, USA
| | - Meredith Regan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, 44 Binney Street, Mayer 457, Boston, MA 02115, USA. Tel.: +1 617 632 3975; Fax: +1 617 632 4760; E-mail:
| |
Collapse
|
346
|
Abstract
Advances in the knowledge of the genetics of phaeochromocytoma have broadened our understanding about the mechanisms of tumorigenesis. Formerly it was believed that 10% of phaeochromocytomas were associated with familial cancer syndromes, but it is now recognised that up to 30% of these tumours may be familial. In particular, attention has been focused on those patients with apparently sporadic presentations where 12-24% of patients have been shown to carry germline mutations indicating hereditary disease. Consideration of genetic testing is now recommended for all apparently sporadic cases and, following the identification of a mutation-positive carrier, the offering of genetic testing to first degree relatives. There is a need for lifelong follow up of affected individuals and asymptomatic mutation-positive carriers, but validation of screening protocols has yet to be determined.
Collapse
Affiliation(s)
- Diana E Benn
- Cancer Genetics Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards NSW 2065, Australia.
| | | |
Collapse
|
347
|
Jiménez C, Cote G, Arnold A, Gagel RF. Review: Should patients with apparently sporadic pheochromocytomas or paragangliomas be screened for hereditary syndromes? J Clin Endocrinol Metab 2006; 91:2851-8. [PMID: 16735498 DOI: 10.1210/jc.2005-2178] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT The recent identification of germline mutations of the mitochondrial complex II genes in variants of paraganglioma/pheochromocytoma syndrome has enlarged the number of known causative genes for hereditary pheochromocytoma. A question confronting clinicians is whether they should screen patients with apparently sporadic pheochromocytomas for unsuspected germline mutations of some or all of the seven genes known to cause hereditary paraganglioma or pheochromocytoma (NF1, VHL, RET, MEN1, SDHD, SDHC, and SDHB). A positive answer was suggested by a report that placed the estimate of hereditary disease in apparently sporadic pheochromocytoma as high as 24%. EVIDENCE ACQUISITION We applied clinically useful criteria to a review of the literature, defining cases of apparently sporadic pheochromocytoma as those without a suspicious personal or family history, with a focal, unilateral pheochromocytoma, and presenting at age less than 50 yr. EVIDENCE SYNTHESIS We reduced the overall estimate of unsuspected hereditary pheochromocytoma patients with apparently sporadic pheochromocytoma to approximately 17%. Mutations in only three genes (VHL, SDHB, and SDHD) accounted for almost this entire minority, and unsuspected RET mutation was rare. Costs, coverage by insurance, the potential effect on insurability, and deficient information for populations outside of referral centers should be considered before recommending genetic testing in patients with apparently sporadic presentations of pheochromocytomas. CONCLUSION We recommend genetic testing for patients with an apparently sporadic pheochromocytoma under the age of 20 yr with family history or features suggestive of hereditary pheochromocytoma or for patients with sympathetic paragangliomas. For individuals who do not meet these criteria, genetic testing is optional.
Collapse
Affiliation(s)
- Camilo Jiménez
- Instituto Nacional de Cancerología, Fundación Santafé de Bogotá, Colombia, South America
| | | | | | | |
Collapse
|
348
|
Abstract
Metastatic renal cell carcinoma (RCC) is currently one of the most treatment-resistant malignancies. However, significant advances in understanding the molecular mechanisms underlying RCC have led to the development of rationally designed therapies, which are now being tested clinically. To date, the vascular endothelial growth factor receptor (VEGFR) pathway has been the most promising target, and two agents (BAY 43-9006 and SU 11248) that inhibit not only VEGFR but also other receptors, including platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), KIT, and Raf kinase, were recently approved by the FDA for advanced RCC. In addition, a phase III trial investigating the addition of VEGF inhibition to interferon alpha (IFN-alpha) in RCC is also now going on. Although the clinical activity of existing agents is to be further defined in ongoing trials, the exciting clinical response data with VEGF inhibition in RCC have demonstrated a key role in the treatment of this historically resistant malignancy.
Collapse
Affiliation(s)
- Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | | |
Collapse
|
349
|
Hong SB, Furihata M, Baba M, Zbar B, Schmidt LS. Vascular defects and liver damage by the acute inactivation of the VHL gene during mouse embryogenesis. J Transl Med 2006; 86:664-75. [PMID: 16652107 DOI: 10.1038/labinvest.3700431] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene leads to the development of central nervous system hemangioblastomas, pheochromocytomas and renal cell carcinomas. The biological role of the VHL gene during development is poorly understood because of early lethality of VHL-null embryos. To overcome early embryo lethality observed in the conventional knockout mouse, we introduced a tamoxifen-inducible Cre (CreER(TM)) transgene for the stage specific inactivation of the VHL gene. Acute tamoxifen-induced inactivation of the VHL gene at E10.5 resulted in embryonic lethality between E14.5 and E15.0 with extensive hemorrhage and necrosis, while littermate controls showed normal development. Examination of the VHL-inactivated embryos between E10.5 and E14.5 revealed dilated blood vessels, hemorrhage and necrotizing liver damage. Concomitant with severe hemorrhage and abnormal vasculature at E15.0, blood circulation in the yolk sac was impaired in the VHL-inactivated embryos, which may be the cause of embryo death. Placental development looked normal before embryo death (E14.5); however, at E16.5 following embryo death, we observed reduced growth of the placental labyrinthine layer. Inactivation of the VHL gene resulted in hypoxia-inducible factor (HIF)-1alpha stabilization and induction of its target genes, VEGF and CAIX, in mouse embryonic fibroblasts (MEFs). In addition, we observed lactate overproduction and acidification of culture media by the inactivation of the VHL gene. Thus, by using a novel conditional VHL knockout mouse model, we could show that the VHL gene plays an important role in the developing vasculature and liver during embryogenesis through regulation of HIF-1alpha and its target genes. This mouse model will be useful for the screening of anti-HIF or anti-VEGF drugs in vivo. Additionally, this acute VHL inactivation system may provide a useful tool for the in vivo study of genes that cause early embryonic lethality.
Collapse
Affiliation(s)
- Seung-Beom Hong
- Laboratory of Immunobiology, Center for Cancer Research, NCI-Frederick, Frederick, MD 21702, USA
| | | | | | | | | |
Collapse
|
350
|
Affiliation(s)
- Erin A Donovan
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|