Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Cancer Gene Ther ; 18(8): 553-62, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21566667

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) as an adjuvant in autologous cell-based anti-tumor immunotherapy has recently been approved for clinical application. To avoid the need for individualized processing of autologous cells, we developed a novel strategy based on the encapsulation of GM-CSF-secreting human allogeneic cells. GM-CSF-producing K562 cells showed high, stable and reproducible cytokine secretion when enclosed into macrocapsules. For clinical development, the cryopreservation of these devices is critical. Thawing of capsules frozen at different time points displayed differences in GM-CSF release shortly after thawing. However, similar secretion values to those of non-frozen control capsules were obtained 8 days after thawing at a rate of >1000 ng GM-CSF per capsule every 24 h. For future human application, longer and reinforced capsules were designed. After irradiation and cryopreservation, these capsules produced >300 ng GM-CSF per capsule every 24 h 1 week after thawing. The in vivo implantation of encapsulated K562 cells was evaluated in mice and showed preserved cell survival. Finally, as a proof of principle of biological activity, capsules containing B16-GM-CSF allogeneic cells implanted in mice induced a prompt inflammatory reaction. The ability to reliably achieve high adjuvant release using a standardized procedure may lead to a new clinical application of GM-CSF in cell-based cancer immunization.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Animais , Linhagem Celular Tumoral , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/química , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Células K562 , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/genética
2.
Neurology ; 71(4): 240-7, 2008 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-18525034

RESUMO

BACKGROUND: Alpha-sarcoglycan (alpha-SG) deficiency (limb-girdle muscular dystrophy [LGMD] type 2D) is the most common form of sarcoglycan-LGMD. No treatment is currently available. Prior studies suggest that overexpression of alpha-SG via adeno-associated virus (AAV)-mediated gene transfer results in poorly sustained gene expression related to transgene toxicity. These findings potentially preclude gene therapy as a treatment approach for LGMD2D. METHODS: The human alpha-SG gene (halpha-SG) was directly transferred to the tibialis anterior muscle of 4- to 5-week-old alpha-SG KO mice using AAV, type 1. The gene was placed under control of either the ubiquitously expressed cytomegalovirus (CMV) promoter or muscle specific promoters that included desmin, muscle creatine kinase (MCK), and its further modification, truncated MCK (tMCK). Low (3 x 10(9) vg) and high (3 x 10(10) vg) doses of AAV1.halpha-SG were administered. RESULTS: Sustained gene expression was observed irrespective of promoters at 6 and 12 weeks post gene transfer. Quantitation of alpha-SG gene expression by fiber counts yielded similar levels of myofiber transduction for both MCK promoters (60 to 70%), while 34% of fibers were transduced with the DES promoter. There was a trend toward lower expression at the 12-week time point with the CMV promoter. Western blot analysis revealed alpha-SG overexpression using CMV and both the MCK promoters. CONCLUSION: Our data demonstrate robust and sustained adeno-associated virus type 1 alpha-sarcoglycan gene expression under control of muscle creatine kinase promoters, without evidence of cytotoxicity. These findings support the use of gene therapy as a potential treatment approach for limb-girdle muscular dystrophy type 2D.


Assuntos
Técnicas de Transferência de Genes/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Sarcoglicanas/genética , Animais , Ensaios Clínicos como Assunto/normas , Creatina Quinase/genética , Citomegalovirus/genética , Dependovirus/genética , Expressão Gênica/genética , Terapia Genética/efeitos adversos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Miosite/genética , Miosite/imunologia , Miosite/metabolismo , Regiões Promotoras Genéticas/genética , Sarcoglicanas/biossíntese , Sarcoglicanas/deficiência , Resultado do Tratamento , Regulação para Cima/genética
3.
Gene Ther ; 10(17): 1479-86, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12900763

RESUMO

Peripheral lymph nodes (PLN) are strategic microenvironments where antigen-presenting dendritic cells (DC), loaded with environmental antigens, and naive lymphocytes meet to initiate immune responses. The unique capacity of DC to induce primary immune responses has led to their use in clinical medicine; however, delivering DC to lymph nodes is problematic. Intravenously injected DC cannot access to PLN, while DC injected into tissue migrate inefficiently through lymphatics to PLN. We achieved DC targeting to T-cell areas of PLN by endowing DC with a novel receptor for peripheral node addressin (PNAd), an adhesion molecule present on the lymph node venular endothelium. This novel receptor is a chimeric E/L-selectin (ELS) that, we have previously shown, binds to PNAd. DC were genetically modified by retroviral transduction to express ELS. ELS expression was targeted to tips of microvilli, and mediated rolling of DC on PNAd both in vivo and in vitro. Such genetically engineered DC could extravasate directly from blood through the lymph node endothelium as opposed to nontransduced DC. This study provides evidence that the trafficking of DC can be modified using gene transfer technologies. More efficient delivery of DC to PLN should assist the development of improved vaccination strategies.


Assuntos
Transferência Adotiva/métodos , Células Dendríticas/metabolismo , Terapia Genética/métodos , Linfonodos/imunologia , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Movimento Celular , Células Cultivadas , Selectina E/genética , Vetores Genéticos/administração & dosagem , Humanos , Selectina L/genética , Proteínas de Membrana , Microscopia Imunoeletrônica , Retroviridae/genética , Transdução Genética/métodos
4.
Nat Cell Biol ; 3(10): 897-904, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11584271

RESUMO

The Wiskott-Aldrich syndrome protein (WASP) family of molecules integrates upstream signalling events with changes in the actin cytoskeleton. N-WASP has been implicated both in the formation of cell-surface projections (filopodia) required for cell movement and in the actin-based motility of intracellular pathogens. To examine N-WASP function we have used homologous recombination to inactivate the gene encoding murine N-WASP. Whereas N-WASP-deficient embryos survive beyond gastrulation and initiate organogenesis, they have marked developmental delay and die before embryonic day 12. N-WASP is not required for the actin-based movement of the intracellular pathogen Listeria but is absolutely required for the motility of Shigella and vaccinia virus. Despite these distinct defects in bacterial and viral motility, N-WASP-deficient fibroblasts spread by using lamellipodia and can protrude filopodia. These results imply a crucial and non-redundant role for N-WASP in murine embryogenesis and in the actin-based motility of certain pathogens but not in the general formation of actin-containing structures.


Assuntos
Actinas/metabolismo , Movimento Celular/fisiologia , Extensões da Superfície Celular/metabolismo , Desenvolvimento Embrionário e Fetal , Proteínas do Tecido Nervoso/fisiologia , Animais , Linhagem Celular , Linhagem Celular Transformada , Fibroblastos , Marcação de Genes , Listeria/fisiologia , Camundongos , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/genética , Fator de Crescimento Derivado de Plaquetas/farmacologia , Recombinação Genética , Shigella flexneri/fisiologia , Vaccinia virus/fisiologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich
5.
J Biol Chem ; 276(39): 36695-702, 2001 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-11443115

RESUMO

We have shown that intermediate lobe (IL) pituitary cells can be engineered to produce sufficient amounts of insulin (ins) to cure diabetes in nonobese diabetic mice but, unlike transplanted islets, ILins cells evade immune attack. To confer glucose-sensing capabilities into these cells, they were further modified with recombinant adenoviruses to express high levels of GLUT2 and the beta-cell isoform of glucokinase (GK). Although expression of GLUT2 alone had negligible effects on glucose usage and lactate production, expression of GK alone resulted in approximately 2-fold increase in glycolytic flux within the physiological (3-20 mm) glucose range. GLUT2/GK coexpression further increased glycolytic flux at 20 mm glucose but disproportionately increased flux at 3 mm glucose. Despite enhanced glycolytic fluxes, GLUT2/GK-coexpressing cells showed glucose dose-dependent accumulation of hexose phosphates, depletion of intracellular ATP, and severe apoptotic cell death. These studies demonstrate that glucose-sensing properties can be introduced into non-islet cells by the single expression of GK and that glucose responsiveness can be augmented by the coexpression of GLUT2. However, in the metabolic engineering of surrogate beta cells, it is critical that the levels of the components be closely optimized to ensure their physiological function and to avoid the deleterious consequences of glucose-induced toxicity.


Assuntos
Glucoquinase/biossíntese , Glucose/toxicidade , Insulina/biossíntese , Proteínas de Transporte de Monossacarídeos/biossíntese , Hipófise/metabolismo , Trifosfato de Adenosina/metabolismo , Adenoviridae/genética , Animais , Apoptose , Northern Blotting , Western Blotting , Morte Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Glucose/metabolismo , Transportador de Glucose Tipo 2 , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Microscopia de Fluorescência , Fosforilação , Isoformas de Proteínas , Distribuição Tecidual
6.
Proc Natl Acad Sci U S A ; 98(8): 4605-10, 2001 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-11274374

RESUMO

Although the systemic administration of a number of different gene products has been shown to result in the inhibition of angiogenesis and tumor growth in different animal tumor models, the relative potency of those gene products has not been studied rigorously. To address this issue, recombinant adenoviruses encoding angiostatin, endostatin, and the ligand-binding ectodomains of the vascular endothelial growth factor receptors Flk1, Flt1, and neuropilin were generated and used to systemically deliver the different gene products in several different preexisting murine tumor models. Single i.v. injections of viruses encoding soluble forms of Flk1 or Flt1 resulted in approximately 80% inhibition of preexisting tumor growth in murine models involving both murine (Lewis lung carcinoma, T241 fibrosarcoma) and human (BxPC3 pancreatic carcinoma) tumors. In contrast, adenoviruses encoding angiostatin, endostatin, or neuropilin were significantly less effective. A strong correlation was observed between the effects of the different viruses on tumor growth and the activity of the viruses in the inhibition of corneal micropocket angiogenesis. These data underscore the need for comparative analyses of different therapeutic approaches that target tumor angiogenesis and provide a rationale for the selection of specific antiangiogenic gene products as lead candidates for use in gene therapy approaches aimed at the treatment of malignant and ocular disorders.


Assuntos
Neovascularização Patológica , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Transfecção , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Divisão Celular/genética , Estudos de Avaliação como Assunto , Humanos , Camundongos , Dados de Sequência Molecular , Neoplasias/irrigação sanguínea , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Células Tumorais Cultivadas , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
7.
J Cell Biol ; 152(6): 1233-46, 2001 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11257123

RESUMO

Collagen XVIII (c18) is a triple helical endothelial/epithelial basement membrane protein whose noncollagenous (NC)1 region trimerizes a COOH-terminal endostatin (ES) domain conserved in vertebrates, Caenorhabditis elegans and Drosophila. Here, the c18 NC1 domain functioned as a motility-inducing factor regulating the extracellular matrix (ECM)-dependent morphogenesis of endothelial and other cell types. This motogenic activity required ES domain oligomerization, was dependent on rac, cdc42, and mitogen-activated protein kinase, and exhibited functional distinction from the archetypal motogenic scatter factors hepatocyte growth factor and macrophage stimulatory protein. The motility-inducing and mitogen-activated protein kinase-stimulating activities of c18 NC1 were blocked by its physiologic cleavage product ES monomer, consistent with a proteolysis-dependent negative feedback mechanism. These data indicate that the collagen XVIII NC1 region encodes a motogen strictly requiring ES domain oligomerization and suggest a previously unsuspected mechanism for ECM regulation of motility and morphogenesis.


Assuntos
Proteínas de Bactérias , Movimento Celular/fisiologia , Colágeno/metabolismo , Endotélio Vascular/citologia , Matriz Extracelular/fisiologia , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/metabolismo , Animais , Toxinas Bacterianas/farmacologia , Western Blotting , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Colágeno/química , Colágeno/genética , Colágeno Tipo XVIII , Citotoxinas/farmacologia , Dimerização , Endostatinas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/crescimento & desenvolvimento , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfogênese , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
8.
Nat Biotechnol ; 19(2): 169-72, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11175734

RESUMO

The hormone leptin has been shown to be an afferent signal in a negative-feedback loop regulating body weight, and consequently, the administration of the gene product for the treatment of obesity has recently attracted considerable attention. Leptin is produced by adipocytes in response to increased trigyceride storage, and appears to affect body weight primarily through target cells in the hypothalamus. Although plasma levels of leptin correlate positively with adipose tissue mass in normal humans and animals, recent studies have shown that obese humans and animals appear to be relatively resistant to the increased plasma levels of leptin. Analysis of the levels of leptin in the cerebrospinal fluid suggests that the uptake of leptin across the blood-brain barrier may be saturable. Taken together, these results suggest that therapeutic approaches to deliver leptin through the circulation may prove to be problematic. Although recent clinical trials have suggested that peripherally administered leptin might lead to a reduction in body weight in humans, it is likely that the more effective delivery of leptin to cellular targets within the central nervous system will be necessary in order to fully reveal the therapeutic potential of the gene product. In an effort to provide a means for the delivery of leptin that obviates the need for the gene product to traverse the blood-brain barrier, we have evaluated the use of recombinant adeno-associated vectors to deliver leptin intraventricularly or directly to the hypothalamus.


Assuntos
Encéfalo/citologia , Dependovirus/genética , Terapia Genética/métodos , Leptina/genética , Leptina/uso terapêutico , Proteínas Luminescentes/genética , Obesidade/terapia , Redução de Peso/fisiologia , Animais , Ventrículos Cerebrais/citologia , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde , Humanos , Hipotálamo/citologia , Leptina/administração & dosagem , Proteínas Luminescentes/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Músculo Esquelético/citologia , Obesidade/genética , Recombinação Genética
9.
J Control Release ; 70(1-2): 231-42, 2001 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-11166423

RESUMO

The first step toward hydrophobic polymer-based nanospheres for gene delivery is to encapsulate and release plasmid DNA. However, encapsulating large hydrophilic molecules in very small nanospheres has been difficult, and only a few examples exist in the literature. For example, maximizing protein and peptide as well as small molecule encapsulation requires adjustments in pH or addition of excipients to charge neutralize, and make less hydrophilic, the compound to be encapsulated. Following this model, we have used a cationic lipid to load and release plasmid DNA from nanospheres made by the phase inversion/solvent diffusion method.


Assuntos
DNA/administração & dosagem , Terapia Genética , Ácido Láctico/administração & dosagem , Plasmídeos , Ácido Poliglicólico/administração & dosagem , Polímeros/administração & dosagem , Difusão , Excipientes , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
10.
J Virol ; 75(3): 1571-5, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11152532

RESUMO

Previously, we have demonstrated that bridge proteins comprised of avian leukosis virus (ALV) receptors fused to epidermal growth factor (EGF) can be used to selectively target retroviral vectors with ALV envelope proteins to cells expressing EGF receptors. To determine whether another type of ligand incorporated into an ALV receptor-containing bridge protein can also function to target retroviral infection, the TVA-VEGF110 bridge protein was generated. TVA-VEGF110 consists of the extracellular domain of the TVA receptor for ALV subgroup A (ALV-A), fused via a proline-rich linker peptide to a 110-amino-acid form of vascular endothelial growth factor (VEGF). This bridge protein bound specifically to its cell surface receptor, VEGFR-2, and efficiently mediated the entry of an ALV-A vector into cells. These studies indicate that ALV receptor-ligand bridge proteins may be generally useful tools for retroviral targeting approaches.


Assuntos
Vírus da Leucose Aviária/genética , Fatores de Crescimento Endotelial/fisiologia , Vetores Genéticos , Linfocinas/fisiologia , Receptores Virais/fisiologia , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/fisiologia , Proteínas Aviárias , Fatores de Crescimento Endotelial/genética , Linfocinas/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Receptores Virais/genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
11.
J Virol ; 74(20): 9540-5, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11000224

RESUMO

We have previously described an approach that employs retroviral receptor-ligand bridge proteins to target retroviral vectors to specific cell types. To determine whether targeted retroviral entry can also be achieved using a retroviral receptor-single-chain antibody bridge protein, the TVA-MR1 fusion protein was generated. TVA-MR1 is comprised of the extracellular domain of the TVA receptor for subgroup A avian leukosis viruses (ALV-A), fused to the MR1 single-chain antibody that binds specifically to EGFRvIII, a tumor-specific form of the epidermal growth factor receptor. We show that TVA-MR1 binds specifically to a murine version of EGFRvIII and promotes ALV-A entry selectively into cells that express this cell surface marker. These studies demonstrate that it is possible to target retroviral vectors to specific cell types through the use of a retroviral receptor-single-chain antibody fusion protein.


Assuntos
Vírus da Leucose Aviária/fisiologia , Receptores ErbB/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Aviárias , Linhagem Celular , Vetores Genéticos , Humanos , Antígenos de Histocompatibilidade Menor
12.
Blood ; 96(6): 2125-33, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10979957

RESUMO

A novel Hoechst 33342 dye efflux assay was recently developed that identifies a population of hematopoietic cells termed side population (SP) cells. In the bone marrow of multiple species, including mice and primates, the SP is composed primarily of CD34(-) cells, yet has many of the functional properties of hematopoietic stem cells (HSCs). This report characterizes SP cells from human umbilical cord blood (UCB). The SP in unfractionated UCB was enriched for CD34(+) cells but also contained a large population of CD34(-) cells, many of which were mature lymphocytes. SP cells isolated from UCB that had been depleted of lineage-committed cells (Lin(-) UCB) contained CD34(+) and CD34(-) cells in approximately equivalent proportions. Similar to previous descriptions of human HSCs, the CD34(+)Lin(-) SP cells were CD38(dim)HLA-DR(dim)Thy-1(dim)CD45RA(-)CD71(-) and were enriched for myelo-erythroid precursors. In contrast, the CD34(-)Lin(-) SP cells were CD38(-)HLA-DR(-)Thy-1(-)CD71(-) and failed to generate myelo-erythroid progeny in vitro. The majority of these cells were CD7(+)CD11b(+)CD45RA(+), as might be expected of early lymphoid cells, but did not express other lymphoid markers. The CD7(+)CD34(-)Lin(-) UCB SP cells did not proliferate in simple suspension cultures but did differentiate into natural killer cells when cultured on stroma with various cytokines. In conclusion, the human Lin(-) UCB SP contains both CD34(+) multipotential stem cells and a novel CD7(+)CD34(-)Lin(-) lymphoid progenitor. This observation adds to the growing body of evidence that CD34(-) progenitors exist in humans.


Assuntos
Sangue Fetal , Células-Tronco Hematopoéticas , Linfócitos , Animais , Antígenos CD34 , Antígenos CD7 , Benzimidazóis , Diferenciação Celular , Linhagem da Célula , Corantes Fluorescentes , Células-Tronco Hematopoéticas/citologia , Humanos , Linfócitos/citologia , Camundongos
13.
Cancer Chemother Pharmacol ; 46 Suppl: S67-72, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10950151

RESUMO

When irradiated and administered intradermally as vaccines, cancer cells engineered to secrete high levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) by gene transfer elicit potent anticancer immune responses in a variety of animal tumor models. Upon vaccination, antigens present in the cancer cells are phagocytosed and processed by skin dendritic cells. These dendritic cells then prime anticancer immune responses by presenting antigenic peptides to T cells. The immune responses generated are capable of eradicating small but lethal cancer cell inocula with minimal toxicity in preclinical animal tumor studies. To develop this vaccination strategy for the treatment of human genitourinary cancers, we have conducted phase I clinical trials using human genitourinary cancer cells as sources of cancer cell antigens. In the first human clinical trial of genetically engineered cancer cell vaccines, a phase I clinical trial of kidney cancer cell vaccines (n = 18), kidney cancer cells were removed at surgery, propagated briefly in vitro, and then genetically modified to secrete high levels of GM-CSF via ex vivo transduction with the retrovirus MFG-GM-CSF. After irradiation, the kidney cancer cells were administered as vaccines to 18 patients with advanced kidney cancers. Vaccine treatment, which caused few side effects, nonetheless appeared to trigger anticancer immune responses manifest as conversion of delayed-type hypersensitivity (DTH) skin responses against irradiated autologous cancer cells after vaccination. Biopsies of vaccine sites yielded findings reminiscent of biopsies from preclinical animal model studies, with evidence of vaccine cell recruitment of dendritic cells, T cells, and eosinophils. One patient with measurable kidney cancer metastases treated at the highest vaccine dose level experienced a partial treatment response. The bioactivity of GM-CSF-secreting autologous cancer cell vaccines was confirmed in a phase I clinical trial for prostate cancer (n = 8). Vaccine cells were prepared from surgically harvested prostate tumors by ex vivo transduction with MFG-GM-CSF in a manner similar to that used for the kidney cancer trial. Vaccine treatment was well tolerated and associated with induction of anticancer immunity as assessed using DTH skin testing. In addition, new antiprostate cancer cell antibodies were detected in serum samples from treated men as a consequence of vaccination. These first clinical trials of GM-CSF-secreting cancer cell vaccines for the treatment of genitourinary cancers have demonstrated both safety and bioactivity, in that very few side effects have been seen and anticancer immune responses have been detected. Future clinical studies will be required to assess vaccine treatment efficacy, refine vaccination dose and schedule, define the appropriate clinical context for the use of such vaccines, and ascertain optimal combinations involving vaccines and other local or systemic anticancer treatments.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma de Células Renais/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Neoplasias Renais/imunologia , Neoplasias da Próstata/imunologia , Adulto , Idoso , Antígenos de Neoplasias/sangue , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Renais/terapia , Relação Dose-Resposta Imunológica , Feminino , Técnicas de Transferência de Genes , Engenharia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Hipersensibilidade Tardia/imunologia , Neoplasias Renais/terapia , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/terapia , Retroviridae/genética
14.
J Exp Med ; 191(12): 2031-8, 2000 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-10859328

RESUMO

Lymphocytes deficient in the T cell costimulatory molecule CD28 exhibit defects in cell survival, clonal expansion, and differentiation into effector cells. It is known that CD28-mediated signaling results in the upregulation of the Bcl family member Bcl-X(L). To investigate the role that Bcl-X(L) plays in the various functions of CD28, we expressed Bcl-X(L) in CD28-deficient primary T lymphocytes using retrovirus-mediated gene transfer. T cells were activated in vitro and infected with Bcl-X(L) or control retroviruses; this method allows gene expression in activated, cycling cells. Expression of Bcl-X(L) in naive T cells was achieved by reconstitution of the immune system of lethally irradiated recipient mice with retrovirus-infected purified bone marrow stem cells from CD28(-/)- or wild-type donor mice. Our studies demonstrate that Bcl-X(L) prolongs the survival of CD28(-/)- T cells but does not restore normal proliferation or effector cell development. These results indicate that the various functions of CD28 can be dissociated, and provide an experimental approach for testing the roles of downstream signals in the functions of cellular receptors such as CD28.


Assuntos
Antígenos CD28/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Linfócitos T/imunologia , Animais , Transplante de Medula Óssea , Diferenciação Celular , Divisão Celular , Sobrevivência Celular , Técnicas de Transferência de Genes , Teste de Complementação Genética , Ativação Linfocitária , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Quimera por Radiação , Retroviridae , Transdução de Sinais , Células Th2 , Proteína bcl-X
15.
J Exp Med ; 191(10): 1699-708, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10811863

RESUMO

We have directly compared the efficacy of two immunotherapeutic strategies for the treatment of cancer: "vaccination" of tumor-bearing mice with genetically modified dendritic cells (DCs), and vaccination with genetically modified tumor cells. Using several different preexisting tumor models that make use of B16F10 melanoma cells expressing a target tumor antigen (human melanoma-associated gene [MAGE]-1), we found that vaccination with bone marrow-derived DCs engineered to express MAGE-1 via adenoviral-mediated gene transfer led to a dramatic decrease in the number of metastases in a lung metastasis model, and led to prolonged survival and some long-term cures in a subcutaneous preexisting tumor model. In contrast, vaccination with granulocyte/macrophage colony-stimulating factor (GM-CSF)-transduced tumor cells, previously shown to induce potent antitumor immunity in standard tumor challenge assays, led to a decreased therapeutic effect in the metastasis model and no effect in the subcutaneous tumor model. Further engineering of DCs to express either GM-CSF, tumor necrosis factor alpha, or CD40 ligand via retroviral-mediated gene transfer, led to a significantly increased therapeutic effect in the subcutaneous tumor model. The immunological mechanism, as shown for GM-CSF-transduced DCs, involves MAGE-1-specific CD4(+) and CD8(+) T cells. Expression of GM-CSF by DCs led to enhanced cytotoxic T lymphocyte activity, potentially mediated by increased numbers of DCs in draining lymph nodes. Our results suggest that clinical studies involving the vaccination with genetically modified DCs may be warranted.


Assuntos
Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Células Dendríticas/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos CD4/genética , Antígenos CD4/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Células Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Engenharia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Imunoterapia , Antígenos Específicos de Melanoma , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Transdução Genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
16.
Lab Invest ; 80(1): 99-115, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10653008

RESUMO

Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) is an angiogenic cytokine with potential for the treatment of tissue ischemia. To investigate the properties of the new blood vessels induced by VPF/VEGF, we injected an adenoviral vector engineered to express murine VPF/VEGF164 into several normal tissues of adult nude mice or rats. A dose-dependent angiogenic response was induced in all tissues studied but was more intense and persisted longer (months) in skin and fat than in heart or skeletal muscle (< or =3 weeks). The initial response (within 18 hours) was identical in all tissues studied and was characterized by microvascular hyperpermeability, edema, deposition of an extravascular fibrin gel, and the formation of enlarged, thin-walled pericyte-poor vessels ("mother" vessels). Mother vessels developed from preexisting microvessels after pericyte detachment and basement membrane degradation. Mother vessels were transient structures that evolved variably in different tissues into smaller daughter vessels, disorganized vessel tangles (glomeruloid bodies), and medium-sized muscular arteries and veins. Vascular structures closely resembling mother vessels and each mother vessel derivative have been observed in benign and malignant tumors, in other examples of pathological and physiological angiogenesis, and in vascular malformations. Together these data suggest that VPF/VEGF has a role in the pathogenesis of these entities. They also indicate that the angiogenic response induced by VPF/VEGF is heterogeneous and tissue specific. Finally, the muscular vessels that developed from mother vessels in skin and perimuscle fat have the structure of collaterals and could be useful clinically in the relief of tissue ischemia.


Assuntos
Fatores de Crescimento Endotelial/fisiologia , Linfocinas/fisiologia , Neovascularização Fisiológica/fisiologia , Animais , Permeabilidade Capilar , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ratos , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
17.
Nature ; 401(6751): 390-4, 1999 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-10517639

RESUMO

The development of cell or gene therapies for diseases involving cells that are widely distributed throughout the body has been severely hampered by the inability to achieve the disseminated delivery of cells or genes to the affected tissues or organ. Here we report the results of bone marrow transplantation studies in the mdx mouse, an animal model of Duchenne's muscular dystrophy, which indicate that the intravenous injection of either normal haematopoietic stem cells or a novel population of muscle-derived stem cells into irradiated animals results in the reconstitution of the haematopoietic compartment of the transplanted recipients, the incorporation of donor-derived nuclei into muscle, and the partial restoration of dystrophin expression in the affected muscle. These results suggest that the transplantation of different stem cell populations, using the procedures of bone marrow transplantation, might provide an unanticipated avenue for treating muscular dystrophy as well as other diseases where the systemic delivery of therapeutic cells to sites throughout the body is critical. Our studies also suggest that the inherent developmental potential of stem cells isolated from diverse tissues or organs may be more similar than previously anticipated.


Assuntos
Transplante de Medula Óssea , Distrofina/biossíntese , Transplante de Células-Tronco Hematopoéticas , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Animais , Diferenciação Celular , Núcleo Celular , Separação Celular , Feminino , Terapia Genética , Células-Tronco Hematopoéticas/citologia , Hibridização in Situ Fluorescente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/citologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia
18.
Cancer Res ; 59(20): 5160-8, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10537292

RESUMO

Vaccination with irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting gene-transduced cancer vaccines induces tumoricidal immune responses. In a Phase I human gene therapy trial, eight immunocompetent prostate cancer (PCA) patients were treated with autologous, GM-CSF-secreting, irradiated tumor vaccines prepared from ex vivo retroviral transduction of surgically harvested cells. Expansion of primary cultures of autologous vaccine cells was successful to meet trial specifications in 8 of 11 cases (73%); the yields of the primary culture cell limited the number of courses of vaccination. Side effects were pruritus, erythema, and swelling at vaccination sites. Vaccine site biopsies manifested infiltrates of dendritic cells and macrophages among prostate tumor vaccine cells. Vaccination activated new T-cell and B-cell immune responses against PCA antigens. T-cell responses, evaluated by assessing delayed-type hypersensitivity (DTH) reactions against untransduced autologous tumor cells, were evident in two of eight patients before vaccination and in seven of eight patients after treatment. Reactive DTH site biopsies manifested infiltrates of effector cells consisting of CD45RO+ T-cells, and degranulating eosinophils consistent with activation of both Th1 and Th2 T-cell responses. A distinctive eosinophilic vasculitis was evident near autologous tumor cells at vaccine sites, and at DTH sites. B-cell responses were also induced. Sera from three of eight vaccinated men contained new antibodies recognizing polypeptides of 26, 31, and 150 kDa in protein extracts from prostate cells. The 150-kDa polypeptide was expressed by LNCaP and PC-3 PCA cells, as well as by normal prostate epithelial cells, but not by prostate stromal cells. No antibodies against prostate-specific antigen were detected. These data suggest that both T-cell and B-cell immune responses to human PCA can be generated by treatment with irradiated, GM-CSF gene-transduced PCA vaccines.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neoplasias da Próstata/terapia , Vacinas Sintéticas/imunologia , Linfócitos B/imunologia , Técnicas de Transferência de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Hipersensibilidade Tardia/etiologia , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Linfócitos T/imunologia , Vacinação
19.
Transplantation ; 67(12): 1607-13, 1999 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-10401769

RESUMO

BACKGROUND: Systemic administration of the inhibitor of costimulation, CTLA4Ig, has been shown to prolong islet graft survival. The purpose of this study was to compare local and systemic expression of murine CTLA4Ig in transplants of rat islets into mice. METHODS: Murine CTLA4Ig was made by joining two polymerase chain reaction products, the extracellular portion of CTLA4 and the Fc portion of IgG2a. Recombinant adenovirus expressing CTLA4Ig (AdCTLA4Ig) was generated using the strategy of Cre-lox recombination. Isolated rat islets infected with AdCTLA4Ig at multiplicities of infection (MOIs) ranging from 0.1 to 10 were transplanted into streptozocin diabetic male B6AF1 mice. Control islets were mock infected or infected with AdLacZ or AdsIg, a recombinant adenovirus expressing only the Fc portion of IgG2a. Also, AdCTLA4Ig and control viruses were injected intramuscularly into mouse transplant recipients at the time of islet transplantation to provide CTLA4Ig systemically. RESULTS: Control islets transplanted into diabetic mice were rejected in 13-17 days. Islets infected with AdCTLA4Ig had dose-dependent prolongation of graft survival. Prolonged survival was even found with very low MOIs of 0.1 and 0.5, with survivals of 24+/-4.2 and 25+/-2.2 days, respectively. Survival with an MOI of 10 was 39+/-8.7 days. With intramuscular injection, no prolongation was found at the lowest relative MOIs of 0.2 and 1, but there was dose-dependent prolongation of graft survival with larger doses. At the highest relative MOI of 400, survival was prolonged to 58+/-10 days. CONCLUSIONS: Rat islets infected with AdCTLA4Ig transplanted into mice had prolonged graft survival. Prolonged survival with MOIs as low as 0.1 and 0.5 indicates that only a minority of islet cells need to express CTLA4Ig to exert an effect. Moreover, the results suggest that the improved islet graft survival is due to a local influence of CTLA4Ig.


Assuntos
Infecções por Adenoviridae/genética , Antígenos de Diferenciação/genética , Imunoconjugados , Transplante das Ilhotas Pancreáticas/imunologia , Transplante Heterólogo/imunologia , Abatacepte , Animais , Antígenos CD , Antígenos de Diferenciação/farmacologia , Antígeno CTLA-4 , Expressão Gênica/efeitos dos fármacos , Sobrevivência de Enxerto/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/farmacologia , Imunossupressores/farmacologia , Injeções Intramusculares , Óperon Lac/genética , Masculino , Camundongos , Camundongos Endogâmicos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , beta-Galactosidase/genética
20.
Blood ; 93(7): 2217-24, 1999 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10090930

RESUMO

Retrovirus-mediated gene transfer into long-lived human pluripotent hematopoietic stem cells (HSCs) is a widely sought but elusive goal. A major problem is the quiescent nature of most HSCs, with the perceived requirement for ex vivo prestimulation in cytokines to induce stem cell cycling and allow stable gene integration. However, ex vivo culture may impair stem cell function, and could explain the disappointing clinical results in many current gene transfer trials. To address this possibility, we examined the ex vivo survival of nonobese diabetic/severe combined immune-deficient (NOD/SCID) repopulating cells (SRCs) over 3 days. After 1 day of culture, the SRC number and proliferation declined twofold, and was further reduced by day 3; self-renewal was only detectable in noncultured cells. To determine if the period of ex vivo culture could be shortened, we used a vesicular stomatitis virus G protein (VSV-G) pseudotyped retrovirus vector that was concentrated to high titer. The results showed that gene transfer rates were similar without or with 48 hours prestimulation. Thus, the use of high-titer VSV-G pseudotyped retrovirus may minimize the loss of HSCs during culture, because efficient gene transfer can be obtained without the need for extended ex vivo culture.


Assuntos
Capsídeo/fisiologia , Técnicas de Cultura de Células/métodos , Técnicas de Transferência de Genes , Vetores Genéticos/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Glicoproteínas de Membrana , Vírus da Leucemia Murina de Moloney/fisiologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Proteínas do Envelope Viral/fisiologia , Animais , Capsídeo/genética , Divisão Celular , Sobrevivência Celular , Células Cultivadas/transplante , Vetores Genéticos/genética , Vetores Genéticos/ultraestrutura , Sobrevivência de Enxerto , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Vírus da Leucemia Murina de Moloney/genética , Fatores de Tempo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA