Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Neurosci ; 43(19): 3567-3581, 2023 05 10.
Article in English | MEDLINE | ID: mdl-36977578

ABSTRACT

Metachromatic leukodystrophy (MLD) is a rare, inherited, demyelinating lysosomal storage disorder caused by mutations in the arylsulfatase-A gene (ARSA). In patients, levels of functional ARSA enzyme are diminished and lead to deleterious accumulation of sulfatides. Herein, we demonstrate that intravenous administration of HSC15/ARSA restored the endogenous murine biodistribution of the corresponding enzyme, and overexpression of ARSA corrected disease biomarkers and ameliorated motor deficits in Arsa KO mice of either sex. In treated Arsa KO mice, when compared with intravenously administered AAV9/ARSA, significant increases in brain ARSA activity, transcript levels, and vector genomes were observed with HSC15/ARSA Durability of transgene expression was established in neonate and adult mice out to 12 and 52 weeks, respectively. Levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit was also defined. Finally, we demonstrated blood-nerve, blood-spinal and blood-brain barrier crossing as well as the presence of circulating ARSA enzyme activity in the serum of healthy nonhuman primates of either sex. Together, these findings support the use of intravenous delivery of HSC15/ARSA-mediated gene therapy for the treatment of MLD.SIGNIFICANCE STATEMENT Herein, we describe the method of gene therapy adeno-associated virus (AAV) capsid and route of administration selection leading to an efficacious gene therapy in a mouse model of metachromatic leukodystrophy. We demonstrate the therapeutic outcome of a new naturally derived clade F AAV capsid (AAVHSC15) in a disease model and the importance of triangulating multiple end points to increase the translation into higher species via ARSA enzyme activity and biodistribution profile (with a focus on the CNS) with that of a key clinically relevant biomarker.


Subject(s)
Arylsulfatases , Genetic Therapy , Leukodystrophy, Metachromatic , Animals , Mice , Macaca fascicularis , Arylsulfatases/genetics , Mice, Knockout , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/physiopathology , Leukodystrophy, Metachromatic/therapy , Disease Models, Animal , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Brain/enzymology , Motor Disorders/genetics , Motor Disorders/therapy , Administration, Intravenous , Biomarkers/analysis , Blood-Brain Barrier , Male , Female , Humans
2.
Proc Natl Acad Sci U S A ; 115(31): E7379-E7388, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30018062

ABSTRACT

The precise correction of genetic mutations at the nucleotide level is an attractive permanent therapeutic strategy for human disease. However, despite significant progress, challenges to efficient and accurate genome editing persist. Here, we report a genome editing platform based upon a class of hematopoietic stem cell (HSC)-derived clade F adeno-associated virus (AAV), which does not require prior nuclease-mediated DNA breaks and functions exclusively through BRCA2-dependent homologous recombination. Genome editing is guided by complementary homology arms and is highly accurate and seamless, with no evidence of on-target mutations, including insertion/deletions or inclusion of AAV inverted terminal repeats. Efficient genome editing was demonstrated at different loci within the human genome, including a safe harbor locus, AAVS1, and the therapeutically relevant IL2RG gene, and at the murine Rosa26 locus. HSC-derived AAV vector (AAVHSC)-mediated genome editing was robust in primary human cells, including CD34+ cells, adult liver, hepatic endothelial cells, and myocytes. Importantly, high-efficiency gene editing was achieved in vivo upon a single i.v. injection of AAVHSC editing vectors in mice. Thus, clade F AAV-mediated genome editing represents a promising, highly efficient, precise, single-component approach that enables the development of therapeutic in vivo genome editing for the treatment of a multitude of human gene-based diseases.


Subject(s)
Dependovirus/genetics , Gene Editing , Hematopoietic Stem Cells/metabolism , Homologous Recombination , BRCA2 Protein/physiology , Genetic Vectors , Humans , Interleukin Receptor Common gamma Subunit/genetics , K562 Cells
3.
Cancer Discov ; 3(12): 1404-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24065731

ABSTRACT

UNLABELLED: Patients with non-small cell lung cancer (NSCLC) with activating EGF receptor (EGFR) mutations initially respond to first-generation reversible EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by acquired resistance, frequently driven by the EGFR(T790M) mutation. CO-1686 is a novel, irreversible, and orally delivered kinase inhibitor that specifically targets the mutant forms of EGFR, including T790M, while exhibiting minimal activity toward the wild-type (WT) receptor. Oral administration of CO-1686 as single agent induces tumor regression in EGFR-mutated NSCLC tumor xenograft and transgenic models. Minimal activity of CO-1686 against the WT EGFR receptor was observed. In NSCLC cells with acquired resistance to CO-1686 in vitro, there was no evidence of additional mutations or amplification of the EGFR gene, but resistant cells exhibited signs of epithelial-mesenchymal transition and demonstrated increased sensitivity to AKT inhibitors. These results suggest that CO-1686 may offer a novel therapeutic option for patients with mutant EGFR NSCLC. SIGNIFICANCE: We report the preclinical development of a novel covalent inhibitor, CO-1686, that irreversibly and selectively inhibits mutant EGFR, in particular the T790M drug-resistance mutation, in NSCLC models. CO-1686 is the fi rst drug of its class in clinical development for the treatment of T790M-positive NSCLC, potentially offering potent inhibition of mutant EGFR while avoiding the on-target toxicity observed with inhibition of the WT EGFR.


Subject(s)
Acrylamides/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Acrylamides/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Molecular Targeted Therapy , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Xenograft Model Antitumor Assays
4.
J Med Chem ; 56(3): 712-21, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23360348

ABSTRACT

PI3Kα has been identified as an oncogene in human tumors. By use of rational drug design, a targeted covalent inhibitor 3 (CNX-1351) was created that potently and specifically inhibits PI3Kα. We demonstrate, using mass spectrometry and X-ray crystallography, that the selective inhibitor covalently modifies PI3Kα on cysteine 862 (C862), an amino acid unique to the α isoform, and that PI3Kß, -γ, and -δ are not covalently modified. 3 is able to potently (EC(50) < 100 nM) and specifically inhibit signaling in PI3Kα-dependent cancer cell lines, and this leads to a potent antiproliferative effect (GI(50) < 100 nM). A covalent probe, 8 (CNX-1220), which selectively bonds to PI3Kα, was used to investigate the duration of occupancy of 3 with PI3Kα in vivo. This is the first report of a PI3Kα-selective inhibitor, and these data demonstrate the biological impact of selectively targeting PI3Kα.


Subject(s)
Drug Discovery , Isoenzymes/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Signal Transduction
5.
Microvasc Res ; 82(3): 253-62, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21958527

ABSTRACT

Antiangiogenesis has been validated as a therapeutic strategy to treat cancer, however, a need remains to identify new targets and therapies for specific diseases and to improve clinical benefit from antiangiogenic agents. Tumor endothelial marker 7 (TEM-7) was investigated as a possible target for therapeutic antiangiogenic intervention in cancer. TEM-7 expression was assessed by in situ hybridization or by immunohistochemistry (IHC) in 130 formalin-fixed paraffin-embedded (FFPE) and 410 frozen human clinical specimens of cancer plus 301 normal tissue samples. In vitro TEM-7 expression was evaluated in 4 human endothelial cell models and in 32 human cancer cell lines by RT-PCR and flow cytometry. An anti-TEM-7 antibody was tested in vitro on human SKOV3 ovarian and MDA-MB-231 breast carcinoma cells that expressed TEM-7 in antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis assays. In frozen tumor tissues, TEM-7 mRNA and protein was detected in all but one of the cancer types tested and was infrequently expressed in normal frozen tissues. In FFPE tumor tissues, TEM-7 protein was detected by IHC in colon, breast, lung, bladder, ovarian and endometrial cancers and in sarcomas. TEM-7 protein was not detected in head and neck, prostate or liver cancers. TEM-7 expression was restricted to the vasculature and was absent from tumor cells. In vitro, TEM-7 was not detected in human microvascular endothelial cells (HMVEC) or human umbilical vein endothelial cells (HUVEC) but was induced in endothelial precursor/progenitor cells (EPC) in the presence of the mitogen phorbol ester PMA. An anti-TEM-7 antibody mediated ADCC and phagocytosis in SKOV3 and MDA-MB-231 cell lines infected with an adenovirus expressing TEM-7. These data demonstrate that TEM-7 is a vascular protein associated with angiogenic states. TEM-7 is a novel and attractive target for antiangiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies/pharmacology , Endothelial Cells/drug effects , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Receptors, Cell Surface/antagonists & inhibitors , Animals , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Flow Cytometry , Formaldehyde , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , In Situ Hybridization , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Paraffin Embedding , Phagocytosis , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Tissue Fixation , Transfection
6.
Nat Chem Biol ; 7(1): 22-4, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21113170

ABSTRACT

Designing selective inhibitors of proteases has proven problematic, in part because pharmacophores that confer potency exploit the conserved catalytic apparatus. We developed a fundamentally different approach by designing irreversible inhibitors that target noncatalytic cysteines that are structurally unique to a target in a protein family. We have successfully applied this approach to the important therapeutic target HCV protease, which has broad implications for the design of other selective protease inhibitors.


Subject(s)
Cysteine Proteinase Inhibitors/therapeutic use , Cysteine/antagonists & inhibitors , Drug Design , Oligopeptides/therapeutic use , Biocatalysis , Biochemistry/methods , Crystallography, X-Ray , Cysteine/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Hepacivirus/drug effects , Hepacivirus/enzymology , Hepacivirus/growth & development , Oligopeptides/chemistry , Oligopeptides/pharmacology , Virology/methods
7.
Exp Cell Res ; 315(5): 784-94, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19094984

ABSTRACT

Netrin-4 is a 628 amino acid basement membrane component that promotes neurite elongation at low concentrations but inhibits neurite extension at high concentrations. There is a growing body of literature suggesting that several molecules, including netrins, are regulators of both neuronal and vascular growth. It is believed that molecules that guide neural growth and development are also involved in regulating morphogenesis of the vascular tree. Further, netrins have recently been implicated in controlling epithelial cell branching morphogenesis in the breast, lung and pancreas. Characterization of purified netrin-4 in in vitro angiogenesis assays demonstrated that netrin-4 markedly inhibits HMVEC migration and tube formation. Moreover, netrin-4 inhibits proliferation of a variety of human tumor cells in vitro. Netrin-4 has only modest effects on proliferation of endothelial and other non-transformed cells. Netrin-4 treatment results in phosphorylation changes of proteins that are known to control cell growth. Specifically, Phospho-Akt-1, Phospho-Jnk-2, and Phospho-c-Jun are reduced in tumor cells that have been treated with netrin-4. Together, these data suggest a potential role for netrin-4 in regulating tumor growth.


Subject(s)
Cell Proliferation , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Nerve Growth Factors/physiology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasms/blood supply , Neoplasms/genetics , Nerve Growth Factors/genetics , Nerve Growth Factors/pharmacology , Netrins , Oncogene Protein v-akt/antagonists & inhibitors , RNA, Messenger/metabolism , Signal Transduction/drug effects
8.
Mol Cancer Ther ; 7(8): 2536-46, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18723498

ABSTRACT

Angiogenesis occurs during normal physiologic processes as well as under pathologic conditions such as tumor growth. Serial analysis of gene expression profiling revealed genes [tumor endothelial markers (TEM)] that are overexpressed in tumor endothelial cells compared with normal adult endothelial cells. Because blood vessel development of malignant tumors under certain conditions may include endothelial precursor cells (EPC) recruited from bone marrow, we investigated TEM expression in EPC. The expression of TEM1 or endosialin (CD248) and other TEM has been discovered in a population of vascular endothelial growth factor receptor 2+/CD31+/CD45-/VE-cadherin+ EPC derived from human CD133+/CD34+ cells. EPC share some properties with fully differentiated endothelial cells from normal tissue, yet reverse transcription-PCR and flow cytometry reveal that EPC express higher levels of endosialin at the molecular and protein levels. The elevated expression of endosialin in EPC versus mature endothelial cells suggests that endosialin is involved in the earlier stages of tumor angiogenesis. Anti-endosialin antibodies inhibited EPC migration and tube formation in vitro. In vivo, immunohistochemistry indicated that human EPC continued to express endosialin protein in a Matrigel plug angiogenesis assay established in nude mice. Anti-endosialin antibodies delivered systemically at 25 mg/kg were also able to inhibit circulating murine EPC in nude mice bearing s.c. SKNAS tumors. EPC and bone marrow-derived cells have been shown previously to incorporate into malignant blood vessels in some instances, yet they remain controversial in the field. The data presented here on endothelial genes that are up-regulated in tumor vasculature and in EPC support the hypothesis that the angiogenesis process in cancer can involve EPC.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Endothelium, Vascular/metabolism , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Base Sequence , Cells, Cultured , DNA Primers , Endothelium, Vascular/cytology , Flow Cytometry , Humans , Reverse Transcriptase Polymerase Chain Reaction
9.
Mol Cancer Ther ; 5(2): 219-29, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505094

ABSTRACT

Protein tyrosine phosphatase PRL-3 mRNA was found highly expressed in colon cancer endothelium and metastases. We sought to associate a function with PRL-3 expression in both endothelial cells and malignant cells using in vitro models. PRL-3 mRNA levels were determined in several normal human endothelial cells exposed or unexposed to the phorbol ester phorbol 12-myristate 13-acetate (PMA) and in 27 human tumor cell lines. In endothelial cells, PRL-3 mRNA expression was increased in human umbilical vascular endothelial cells and human microvascular endothelial cells (HMVEC) exposed to PMA. An oligonucleotide microarray analysis revealed that PRL-3 was among the 10 genes with the largest increase in expression on PMA stimulation. Phenotypically, PMA-treated HMVEC showed increased invasion, tube formation, and growth factor-stimulated proliferation. A flow cytometric analysis of cell surface markers showed that PMA-treated HMVEC retained endothelial characteristics. Infection of HMVEC with an adenovirus expressing PRL-3 resulted in increased tube formation. In tumor cells, PRL-3 mRNA levels varied markedly with high expression in SKNAS neuroblastoma, MCF-7 and BT474 breast carcinoma, Hep3B hepatocellular carcinoma, and HCT116 colon carcinoma. Western blotting analysis of a subset of cell line lysates showed a positive correlation between PRL-3 mRNA and protein levels. PRL-3 was stably transfected into DLD-1 colon cancer cells. PRL-3-overexpressing DLD-1 subclones were assessed for doubling time and invasion. Although doubling time was similar among parental, empty vector, and PRL-3 subclones, invasion was increased in PRL-3-expressing subclones. In models of endogenous expression, we observed that the MCF-7 cell line, which expresses high levels of PRL-3, was more invasive than the SKBR3 cell line, which expresses low levels of PRL-3. However, the MDA-MB-231 cell line was highly invasive with low levels of PRL-3, suggesting that in some models invasion is PRL-3 independent. Transfection of a PRL-3 small interfering RNA into MCF-7 cells inhibited PRL-3 expression and cell invasion. These results indicate that PRL-3 is functional in both endothelial cells and malignant cells and further validate PRL-3 as a potentially important molecular target for anticancer therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Neoplasms/enzymology , Salivary Proteins and Peptides/genetics , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Gene Expression/drug effects , Humans , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , RNA, Messenger/metabolism , Salivary Proteins and Peptides/analysis , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation
10.
Cancer Res ; 64(21): 7857-66, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15520192

ABSTRACT

The molecular signature that defines tumor microvasculature will likely provide clues as to how vascular-dependent tumor proliferation is regulated. Using purified endothelial cells, we generated a database of gene expression changes accompanying vascular proliferation in invasive breast cancer. In contrast to normal mammary vasculature, invasive breast cancer vasculature expresses extracellular matrix and surface proteins characteristic of proliferating and migrating endothelial cells. We define and validate the up-regulated expression of VE-cadherin and osteonectin in breast tumor vasculature. In contrast to other tumor types, invasive breast cancer vasculature induced a high expression level of specific transcription factors, including SNAIL1 and HEYL, that may drive gene expression changes necessary for breast tumor neovascularization. We demonstrate the expression of HEYL in tumor endothelial cells and additionally establish the ability of HEYL to both induce proliferation and attenuate programmed cell death of primary endothelial cells in vitro. We also establish that an additional intracellular protein and previously defined metastasis-associated gene, PRL3, appears to be expressed predominately in the vasculature of invasive breast cancers and is able to enhance the migration of endothelial cells in vitro. Together, our results provide unique insights into vascular regulation in breast tumors and suggest specific roles for genes in driving tumor angiogenesis.


Subject(s)
Blood Vessels/metabolism , Breast Neoplasms/blood supply , Neovascularization, Pathologic/etiology , Breast/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/blood supply , Carcinoma, Ductal, Breast/metabolism , Female , GPI-Linked Proteins , Gene Expression , Humans , Immunohistochemistry , Neoplasm Invasiveness , Neuropeptides/genetics , Osteonectin/genetics
11.
Am J Pathol ; 165(2): 601-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277233

ABSTRACT

Malignant gliomas are uniformly lethal tumors whose morbidity is mediated in large part by the angiogenic response of the brain to the invading tumor. This profound angiogenic response leads to aggressive tumor invasion and destruction of surrounding brain tissue as well as blood-brain barrier breakdown and life-threatening cerebral edema. To investigate the molecular mechanisms governing the proliferation of abnormal microvasculature in malignant brain tumor patients, we have undertaken a cell-specific transcriptome analysis from surgically harvested nonneoplastic and tumor-associated endothelial cells. SAGE-derived endothelial cell gene expression patterns from glioma and nonneoplastic brain tissue reveal distinct gene expression patterns and consistent up-regulation of certain glioma endothelial marker genes across patient samples. We define the G-protein-coupled receptor RDC1 as a tumor endothelial marker whose expression is distinctly induced in tumor endothelial cells of both brain and peripheral vasculature. Further, we demonstrate that the glioma-induced gene, PV1, shows expression both restricted to endothelial cells and coincident with endothelial cell tube formation. As PV1 provides a framework for endothelial cell caveolar diaphragms, this protein may serve to enhance glioma-induced disruption of the blood-brain barrier and transendothelial exchange. Additional characterization of this extensive brain endothelial cell gene expression database will provide unique molecular insights into vascular gene expression.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Endothelium, Vascular/metabolism , Glioma/metabolism , Neovascularization, Pathologic/genetics , Biomarkers, Tumor/genetics , Brain/blood supply , Brain Neoplasms/pathology , Endothelium, Vascular/pathology , Glioma/pathology , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Mol Cancer Res ; 1(6): 453-62, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12692265

ABSTRACT

The von Hippel-Lindau tumor suppressor, pVHL, is a key player in one of the best characterized hypoxia signaling pathways, the VHL-hypoxia-inducible factor (VHL-HIF) pathway. To better understand the role of VHL in the hypoxia signaling pathways of tumor cells, we used serial analysis of gene expression (SAGE) to investigate hypoxia-regulated gene expression in renal carcinoma cells (786-0), with and without VHL. The gene expression profiles of the cancer cells were compared to SAGE profiles from normal renal proximal tubule cells grown under both normoxia and hypoxia. The data suggest that the role of VHL as a tumor suppressor may be more complex than previously thought. Further, the data reveal that renal carcinoma cells have evolved an alternative hypoxia signaling pathway(s) compared with normal renal cells. These alternative hypoxia pathways demonstrate VHL-dependent and VHL-independent regulation. The genes involved in such pathways include those with potential importance in the physiological and pathological regulation of tumor growth and angiogenesis. Some of the genes identified as showing overexpression in the cancer cells, particularly those encoding secreted or membrane-bound proteins, could be potential biomarkers for tumors or targets for rational therapeutics that are dependent on VHL status.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hypoxia/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Carcinoma, Renal Cell/pathology , Cell Division , Cell Line, Tumor , Humans , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...