Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(7): e0236175, 2020.
Article in English | MEDLINE | ID: mdl-32697798

ABSTRACT

Adenoviruses cause upper respiratory infections, conjunctivitis, keratitis, and gastrointestinal illness. These can be fatal in immunocompromised individuals. Adenoviruses have also been engineered into viral vectors to deliver therapeutic genes or induce immunity as vaccine carriers. The success of ocular gene therapy is driven partly by the immunologic and biochemical influences of the intraocular environment. We have shown that versican and hyaluronan modulate adenoviral vector transgene expression through CD44 signaling. Herein we explored the role of these pathways on virus replication and viral protein expression of wild type adenovirus. We report that the addition of vitreous humor (which contains both versican and hyaluronan) increases viral hexon protein levels. Vitreous humor also increased wild type adenovirus DNA replication in vitro. Metalloproteinase and γ-secretase inhibitors, which inhibit CD44 proteolytic activation, blocked adenoviral replication in vitro. Similarly, protein kinase C and RhoA kinase inhibitors, both proteins associated with CD44 mediated pathways, also inhibited wild type adenoviral replication in vitro. Application of metalloproteinase and γ-secretase inhibitors to human conjunctival explants sharply decreased adenoviral vector gene expression. Our results demonstrate that pharmacologic delivery of these inhibitors is easily achievable. The inhibition of these enzymes should be explored as potential therapies of wild type adenoviral infections.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviridae/drug effects , Antiviral Agents/pharmacology , Genetic Vectors/drug effects , Virus Replication/drug effects , Adenoviridae/physiology , Adenoviridae Infections/virology , Administration, Ophthalmic , Amides/pharmacology , Amides/therapeutic use , Antiviral Agents/therapeutic use , Conjunctiva/metabolism , DNA, Viral/genetics , DNA, Viral/isolation & purification , Diamines/pharmacology , Diamines/therapeutic use , Dipeptides/pharmacology , Dipeptides/therapeutic use , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , HeLa Cells , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Maleimides/pharmacology , Maleimides/therapeutic use , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Permeability , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proteolysis/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Signal Transduction/drug effects , Thiazoles/pharmacology , Thiazoles/therapeutic use , Versicans/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Vitreous Body/metabolism , rho-Associated Kinases/metabolism
2.
J Biol Chem ; 292(35): 14381-14390, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28684419

ABSTRACT

To examine the biochemical influences that may contribute to the success of gene therapy for ocular disorders, the role of versican, a vitreous component, in adenoviral-mediated transgene expression was examined. Versican is a large chondroitin sulfate-containing, hyaluronic acid-binding proteoglycan present in the extracellular matrix and in ocular vitreous body. Y79 retinoblastoma cells and CD44-negative SK-N-DZ neuroblastoma cells transduced with adenoviral vectors in the presence of versican respond with an activation of transgene expression. Proteolysis of versican generates a hyaluronan-binding G1 domain. The addition of recombinant versican G1 to SK-N-DZ cells results in a similar activation of transgene expression, and treatment with dasatinib, an inhibitor of Src family kinases, also mimics the effects of versican. Enhancement is accompanied by an increase in signal transducer and activator of transcription 5 (STAT5) phosphorylation and is abrogated by treatment with C188-9, a STAT3/5 inhibitor, or with ruxolitinib, a Janus kinase 1/2 (JAK1/2) inhibitor. These data implicate versican G1 in enhancing adenoviral vector transgene expression in a hyaluronic acid-CD44 independent manner that is down-regulated by inhibitors of the JAK/STAT pathway and enhanced by inhibitors of the Src kinase pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/therapy , Protein Kinase Inhibitors/pharmacology , Versicans/metabolism , Adenoviridae/growth & development , Adenoviridae/physiology , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , DNA, Recombinant/metabolism , DNA, Viral/metabolism , Genes, Reporter/drug effects , Genetic Vectors , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasms/metabolism , Neoplasms/virology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Versicans/chemistry , Versicans/genetics , Virus Replication/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
3.
PLoS One ; 8(6): e63519, 2013.
Article in English | MEDLINE | ID: mdl-23826078

ABSTRACT

Verification that cell lines used for cancer research are derived from malignant cells in primary tumors is imperative to avoid invalidation of study results. Retinoblastoma is a childhood ocular tumor that develops from loss of functional retinoblastoma protein (pRb) as a result of genetic or epigenetic changes that affect both alleles of the RB1 gene. These patients contain unique identifiable genetic signatures specifically present in malignant cells. Primary cultures derived from retinoblastoma tumors can be established as non-adherent tumorspheres when grown in defined media or as attached monolayers when grown in serum-containing media. While the RB1 genotypes of tumorspheres match those of the primary tumor, adherent cultures have the germline RB1 genotype. Tumorspheres derived from pRb-negative tumors do not express pRb and express the neuroendocrine tumor markers synaptophysin and microtubule-associated protein 2 (MAP2). Adherent cells are synaptophysin-negative and express pRb, the epithelial cell marker cytokeratin that is expressed in the retinal pigmented epithelium and the vascular endothelial cell marker CD34. While tumorspheres are of malignant origin, our results cast doubt on the assumption that adherent tumor-derived cultures are always valid in vitro models of malignant cells and emphasize the need for validation of primary tumor cultures.


Subject(s)
Cell Adhesion , Eye Neoplasms/pathology , Retinoblastoma/pathology , Cell Differentiation , Eye Neoplasms/genetics , Genes, Retinoblastoma , Genotype , Humans , Mutation , Retinoblastoma/genetics , Tumor Cells, Cultured
4.
J Biol Chem ; 287(39): 32697-707, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22865879

ABSTRACT

The success of gene therapy in the ocular environment is partly due to the presence of hyaluronan in vitreous. Here we explore the mechanism of hyaluronan-mediated enhancement of adenoviral vector transgene expression. Introduction of hyaluronan receptor CD44 into CD44-negative cells followed by transduction in the presence of vitreous with an adenoviral vector containing an IL-12-coding transgene increases IL-12 secretion. We demonstrate that sequential CD44 proteolysis is responsible for hyaluronan-mediated enhancement. Metalloproteinase or γ-secretase inhibitors decrease adenoviral-mediated transgene expression. Deletion of these proteolytic sites in CD44 also inhibits transgene expression. Expression of CD44 with a mutation to prevent phosphorylation of serine 325 inhibits the response to vitreous. Expression of the CD44 intracellular domain enhances transgene expression in the absence of vitreous. CD44-mediated enhancement of gene expression was observed with vectors using different promoters and appears because of an increase in mRNA production, not because of an increase in vector transduction as determined by quantitative RT-PCR and quantitative PCR, respectively. These data fit a model where the interaction of hyaluronan in vitreous and CD44 modulates transgene expression by initiating CD44 proteolysis and release of the cytoplasmic domain, resulting in increased transgene transcription.


Subject(s)
Adenoviridae , Gene Expression , Genetic Vectors , Hyaluronan Receptors/metabolism , Proteolysis , Transgenes , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Humans , Hyaluronan Receptors/genetics , Hyaluronic Acid/genetics , Hyaluronic Acid/metabolism , Interleukin-12/biosynthesis , Interleukin-12/genetics , Mutation , Promoter Regions, Genetic , Protein Structure, Tertiary , Transcription, Genetic
5.
Invest Ophthalmol Vis Sci ; 53(7): 3454-62, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22562503

ABSTRACT

PURPOSE: Human retinoblastomas form during the proliferative phase of retina development and are caused by mutations that result in absent or functionally defective Rb protein. Similar tumors occur in mice only when multiple Rb gene family members are absent. We asked if retinal tumors can arise from an undifferentiated retinal cell. The tumor-initiating cells isolated from these tumors that formed in early embryonic murine retinas were characterized. METHODS: Transgenic mice were created using a Pax6 promoter to target expression of SV40 large T-antigen (T-Ag) in the undifferentiated murine embryonic retina. T-Ag, which sequesters all Rb family proteins and p53, is expressed in the retina and lens by murine embryonic day 10 (E10) and tumors are observed by E12.5. A cell line that is adherent in serum-containing media and forms neurospheres in supplemented serum-free media was developed from retinal tumors isolated on postnatal day 7. RESULTS: In all, 1.5% of attached cells form neurospheres when transferred to serum-free medium. All cultured cells express T-Ag, confirming that they derive from the original tumors; 0.5% of adherent cells express detectable levels of CD133. CD133+ FACS-sorted cells cultured in serum-free medium form 3-fold more neurospheres than do CD133- cells. Six of seven mice injected with CD133+ cells and one of seven mice injected with CD133- cells formed tumors during a 6-month period. Unlike primary adherent cells, primary and secondary tumors heterogeneously express markers of stem cells and differentiation similar to human retinoblastoma. CONCLUSIONS: CD133+ tumor-initiating cells can originate from proliferating undifferentiated precursor cells.


Subject(s)
Antigens, CD/metabolism , Antigens, Polyomavirus Transforming/genetics , Cell Transformation, Neoplastic/pathology , Glycoproteins/metabolism , Peptides/metabolism , Retina/embryology , Retinal Neoplasms/embryology , Retinoblastoma/embryology , AC133 Antigen , Animals , Disease Models, Animal , Eye Proteins/genetics , Flow Cytometry , Homeodomain Proteins/genetics , Humans , Immunophenotyping , Mice , Mice, Transgenic , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/metabolism , Retinoblastoma/pathology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
6.
J Vis Exp ; (54)2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21847079

ABSTRACT

Culturing retinoblastoma tumor cells in defined stem cell media gives rise to primary tumorspheres that can be grown and maintained for only a limited time. These cultured tumorspheres may exhibit markedly different cellular phenotypes when compared to the original tumors. Demonstration that cultured cells have the capability of forming new tumors is important to ensure that cultured cells model the biology of the original tumor. Here we present a protocol for propagating human retinoblastoma tumors in vivo using Rag2(-/-) immune deficient mice. Cultured human retinoblastoma tumorspheres of low passage or cells obtained from freshly harvested human retinoblastoma tumors injected directly into the vitreous cavity of murine eyes form tumors within 2-4 weeks. These tumors can be harvested and either further passaged into murine eyes in vivo or grown as tumorspheres in vitro. Propagation has been successfully carried out for at least three passages thus establishing a continuing source of human retinoblastoma tissue for further experimentation.


Subject(s)
Neoplasm Transplantation/methods , Retinal Neoplasms/immunology , Retinal Neoplasms/pathology , Retinoblastoma/immunology , Retinoblastoma/pathology , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Humans , Mice , Neoplasm Transplantation/immunology , Transplantation, Heterologous , Tumor Cells, Cultured
7.
Mol Ther ; 18(10): 1885-90, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20606647

ABSTRACT

The ocular environment has been shown to induce tolerance to locally administered antigens. We therefore investigated whether there was a systemic immune response against adenoviral vectors injected into the vitreous of retinoblastoma patients enrolled in a phase 1 clinical trial of adenoviral-mediated thymidine kinase gene transfer. Sections of enucleated eyes were immunostained with antibodies against inflammatory cells. A trend toward increasing numbers of plasma cells, T cells, macrophages, and antigen-presenting cells was observed in the injected subjects' eyes, but systemically, there was no significant increase in the number of adenovirus-specific cytotoxic T lymphocytes (CTLs) or in adenovirus neutralizing antibodies. Therefore, in contrast to studies showing significant immunogenicity of Ad-RSVtk following injection into extraocular tumors, injection into the eye produces only a mild local inflammatory response without evidence of systemic cellular or humoral immune responses to adenovirus.


Subject(s)
Adenoviridae/genetics , Adenoviridae/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Retinoblastoma/immunology , Retinoblastoma/therapy , Antigen-Presenting Cells/immunology , Eye/immunology , Eye/metabolism , Genetic Therapy/methods , Humans , Immunohistochemistry , In Vitro Techniques , Macrophages/immunology , Retinoblastoma/genetics , Retinoblastoma/metabolism , T-Lymphocytes/immunology , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
8.
Cancer Res ; 67(22): 10653-6, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006805

ABSTRACT

Retinoblastoma, the most common intraocular malignancy of childhood, metastasizes by initial invasion of the choroid and the optic nerve. There is no effective treatment for metastatic retinoblastoma, especially when the central nervous system (CNS) is involved, and prevention of this complication is a treatment priority. Seneca Valley Virus (SVV-001) is a conditionally replication-competent picornavirus that is not pathogenic to normal human cells but can kill human retinoblastoma cells in vitro with an IC(50) of <1 viral particle (vp) per cell. A xenograft murine model of metastatic retinoblastoma was used to examine the therapeutic potential of SVV-001. Histopathologic analysis of ocular and brain tissues after a single tail vein injection of SVV-001 (1 x 10(13) vp/kg) showed effective treatment of choroid and ocular nerve tumor invasion (1 of 20 animals with invasive disease in the treated group versus 7 of 20 animals with invasive disease in the control group; P = 0.017) and prevention of CNS metastasis (0 of 20 animals with CNS metastatic disease in the treated group versus 4 of 20 animals with CNS disease in the control group; P = 0.036). There were no observed adverse events due to the virus in any of the treated animals. SVV-001 may be effective as a treatment of locally invasive and metastatic retinoblastoma.


Subject(s)
Central Nervous System Neoplasms/therapy , Genetic Therapy/methods , Oncolytic Viruses/genetics , Picornaviridae/genetics , Retinoblastoma/therapy , Animals , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Treatment Outcome
9.
Mol Ther ; 15(3): 566-70, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17180120

ABSTRACT

Adenovirus infection is a significant cause of ocular, respiratory, and gastrointestinal illness and can spread rapidly. Morbidity is considerable in immune-suppressed individuals and there is significant mortality. There are no effective therapies. During preclinical studies of adenoviral-mediated gene therapy for ocular disorders, we noticed a significant increase in transduction when the target cells were exposed to adenovirus in the presence of ocular vitreous. The vitreous is mainly comprised of water, collagen, and the large polysaccharide hyaluronan. In this paper, we report data that implicate hyaluronan in the adenoviral infectious process and show that interference with the interaction between hyaluronan and its cellular receptor CD44 can block adenovirus transduction in vitro and in vivo.


Subject(s)
Adenoviridae/genetics , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Transduction, Genetic , Animals , Cell Line , Genetic Vectors/genetics , Humans , Hyaluronan Receptors/genetics , Mice , Mice, Inbred C57BL , Mutation/genetics
10.
Arch Pathol Lab Med ; 130(11): 1669-72, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17076529

ABSTRACT

CONTEXT: Retinoblastoma is the most common malignant intraocular tumor in children. It has been shown that adjuvant therapy following enucleation in patients with high-risk histopathologic features significantly decreases the mortality. We describe the association of extensive necrosis of tumor and intraocular structures with 2 of the major risk factors: optic nerve invasion and choroidal invasion. This may alert the pathologist who makes the observation of extensive necrosis to carefully search for histologic features associated with adverse outcome. OBJECTIVE: To determine whether extensively necrotic retinoblastoma is associated with high-risk histologic prognostic factors for metastatic disease and patient survival. DESIGN: Retrospective case series. Forty-three eyes of 43 patients with retinoblastoma who underwent enucleation between 1990 and 2001 were evaluated. Medical records, histopathology specimens, pathology reports, and clinical photographs were reviewed. Tumors were designated as exhibiting extensive necrosis if more than 95% of tumor cells and intraocular tissues were necrotic. The main outcome measure was the association of extensive tumor necrosis with 3 high-risk histopathologic features: extraocular extension, optic nerve invasion, or choroidal invasion. Metastatic disease, patient survival, and associations with pathologic findings were also analyzed. RESULTS: Optic nerve head invasion (P < .001), post-lamina-cribrosal invasion (P < .001), and choroidal invasion by tumor (P = .004) were observed more frequently in eyes with extensive necrosis compared with eyes without extensive necrosis. Two of the 11 patients with extensively necrotic intraocular retinoblastoma died from metastatic disease (P = .06). None of the 32 patients without extensive necrosis developed metastatic disease or died. CONCLUSIONS: Extensive ocular tissue and tumor necrosis is associated with histologic high-risk prognostic factors for tumor metastasis and mortality.


Subject(s)
Choroid/pathology , Optic Nerve/pathology , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Chemotherapy, Adjuvant , Eye Enucleation , Female , Humans , Infant , Infant, Newborn , Male , Necrosis , Neoplasm Invasiveness , Prognosis , Radiotherapy, Adjuvant , Retinal Neoplasms/mortality , Retinal Neoplasms/surgery , Retinoblastoma/mortality , Retinoblastoma/secondary , Retinoblastoma/surgery , Risk Factors
11.
J Clin Oncol ; 23(31): 7927-35, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16258092

ABSTRACT

PURPOSE: To evaluate the feasibility and safety of adenovirus-mediated gene therapy as a treatment for tumor seeds in the vitreous of children with retinoblastoma. PATIENTS AND METHODS: An Institutional Biosafety Committee-, Institutional Review Board-, Recombinant DNA Advisory Committee-, and US Food and Drug Administration-approved phase I study used intrapatient dose escalation of adenoviral vector containing a herpes simplex thymidine kinase gene (AdV-TK) followed by systemic administration of ganciclovir to treat bilateral retinoblastoma with vitreous tumor seeding refractory to standard therapies. Vitreous tumor seeds were treated by intravitreous injection of AdV-TK adjacent to disease sites. Each injection was followed by ganciclovir delivered intravenously every 12 hours for 7 days. RESULTS: Eight patients with vitreous tumor seeds were enrolled. One patient who was treated with 10(8) viral particles (vp) had resolution of the tumor seeds around the injection site. The seven patients who were treated with doses > or = 10(10) vp had resolution of their vitreous tumor seeds documented by fundoscopy. Toxicity included mild inflammation at 10(10) vp and moderate inflammation, corneal edema, and increased intraocular pressure at 10(11) vp. One patient was free of active vitreous tumor seeds 38 months after therapy. There has been no evidence of extraocular spread of tumor along the needle tract in any patient. CONCLUSION: AdV-TK followed by ganciclovir can be administered safely to children with retinoblastoma. Suicide gene therapy may contribute to the treatment of children with retinoblastoma tumor seeds in the vitreous, a resistant complication of retinoblastoma.


Subject(s)
Adenoviruses, Human/genetics , Antiviral Agents/therapeutic use , Ganciclovir/therapeutic use , Neoplasm Seeding , Retinal Neoplasms/therapy , Retinoblastoma/therapy , Thymidine Kinase/therapeutic use , Vitreous Body/pathology , Child , Combined Modality Therapy , Feasibility Studies , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Humans , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/therapy , Retinal Neoplasms/enzymology , Retinal Neoplasms/pathology , Retinoblastoma/enzymology , Retinoblastoma/pathology , Thymidine Kinase/genetics , Vitreous Body/drug effects
12.
Invest Ophthalmol Vis Sci ; 45(6): 1680-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161826

ABSTRACT

PURPOSE: A number of ocular diseases are potentially amenable to gene therapy interventions if appropriate vectors for the targeted administration of therapeutic genes can be identified. In vitro and in vivo transduction efficiency of a Group C serotype 5 adenoviral vector containing the fiber domain derived from a Group B serotype 35 adenovirus and the gene encoding green fluorescent protein (AdV5/F35-GFP) was compared to an AdV5-GFP vector for transgene delivery to human retinoblastoma and to human and murine retinas. METHODS: The distribution of the adenoviral receptors CAR and CD46 on normal and malignant retinal tissues was determined using immunohistochemistry. Human retinoblastoma cells were incubated with either AdV5-GFP or AdV5/F35-GFP, and the expression of the reporter protein was compared using quantitative fluorescence and fluorescent-activated cell sorting. Mice were given a single subretinal injection of either viral vector, and eyes were enucleated at specified times after injection for histopathologic examination. Human cadaver eyes were similarly examined ex vivo. RESULTS: CAR was expressed in retina except in photoreceptor outer segments. CD46 was expressed in photoreceptor inner and outer segments. Both vectors efficiently transduced the human retinoblastoma cells in vitro. However, the amount of the transgene expressed using AdV5/F35-GFP was more than sixfold greater than that when AdV5-GFP was used. In vivo, AdV5/F35-GFP at doses as low as 10(5) infectious units (IU) transduced cells in all layers of the retina especially photoreceptors and occasional neuronal cells, and Müller cells as well as retinal pigment epithelial cells, whereas AdV5-GFP transduced only retinal pigment epithelial cells and occasional photoreceptors and Müller cells. CONCLUSIONS: AdV5/F35 chimeric vectors may be superior to AdV5 for gene therapy applications targeting the photoreceptor.


Subject(s)
Adenoviruses, Human/genetics , Antigens, CD/metabolism , Gene Transfer Techniques , Genetic Vectors , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Retina/metabolism , Retinoblastoma/metabolism , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Flow Cytometry , Gene Expression , Green Fluorescent Proteins , Humans , Immunoenzyme Techniques , Integrin alphaV/metabolism , Luminescent Proteins/genetics , Membrane Cofactor Protein , Mice , Mice, Inbred C57BL , Retinoblastoma/pathology , Tumor Cells, Cultured
13.
Expert Rev Mol Med ; 5(1): 1-14, 2003 Jan 07.
Article in English | MEDLINE | ID: mdl-14987394

ABSTRACT

Retinoblastoma (Rb) is the most common primary ocular malignancy of children and is caused by a mutation in the gene RB1. Approximately 40% of cases are associated with one or more constitutional mutations, and are therefore heritable, whereas the other 60% are sporadic. Rb is exclusively found in young children. In some cases, Rb tumours metastasise to extraocular organs including bone, lung and brain. Although there is no effective treatment for metastatic disease, non-metastatic cases can be cured by removal of the eye(enucleation). Newer treatment strategies emphasise salvaging the affected eye whenever possible. Animal models of Rb have been developed with xenograft and transgenic techniques. Each model has both strengths and weaknesses for exploring the mechanisms of disease development and progression and the efficacy of new treatment strategies.


Subject(s)
Retinoblastoma/therapy , Animals , Child , Disease Models, Animal , Eye/pathology , Eye/transplantation , Eye Enucleation , Humans , Organ Transplantation/methods , Retinoblastoma/diagnosis , Retinoblastoma/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...