Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Immunother Cancer ; 8(2)2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32727810

RESUMO

Accumulation of extracellular adenosine within the microenvironment is a strategy exploited by tumors to escape detection by the immune system. Adenosine signaling through the adenosine 2A receptor (A2AR) on immune cells elicits a range of immunosuppressive effects which promote tumor growth and limit the efficacy of immune checkpoint inhibitors. Preclinical data with A2AR inhibitors have demonstrated tumor regressions in mouse models by rescuing T cell function; however, the mechanism and role on other immune cells has not been fully elucidated. METHODS: We report here the development of a small molecule A2AR inhibitor including characterization of binding and inhibition of A2AR function with varying amounts of a stable version of adenosine. Functional activity was tested in both mouse and human T cells and dendritic cells (DCs) in in vitro assays to understand the intrinsic role on each cell type. The role of adenosine and A2AR inhibition was tested in DC differentiation assays as well as co-culture assays to access the cross-priming function of DCs. Syngeneic models were used to assess tumor growth alone and in combination with alphaprogrammed death-ligand 1 (αPD-L1). Immunophenotyping by flow cytometry was performed to examine global immune cell changes upon A2AR inhibition. RESULTS: We provide the first report of AZD4635, a novel small molecule A2AR antagonist which inhibits downstream signaling and increases T cell function as well as a novel mechanism of enhancing antigen presentation by CD103+ DCs. The role of antigen presentation by DCs, particularly CD103+ DCs, is critical to drive antitumor immunity providing rational to combine a priming agent AZD4635 with check point blockade. We find adenosine impairs the maturation and antigen presentation function of CD103+ DCs. We show in multiple syngeneic mouse tumor models that treatment of AZD4635 alone and in combination with αPD-L1 led to decreased tumor volume correlating with enhanced CD103+ function and T cell response. We extend these studies into human DCs to show that adenosine promotes a tolerogenic phenotype that can be reversed with AZD4635 restoring antigen-specific T cell activation. Our results support the novel role of adenosine signaling as an intrinsic negative regulator of CD103+ DCs maturation and priming. We show that potent inhibition of A2AR with AZD4635 reduces tumor burden and enhances antitumor immunity. This unique mechanism of action in CD103+ DCs may contribute to clinical responses as AZD4635 is being evaluated in clinical trials with IMFINZI (durvalumab, αPD-L1) in patients with solid malignancies. CONCLUSION: We provide evidence implicating suppression of adaptive and innate immunity by adenosine as a mechanism for immune evasion by tumors. Inhibition of adenosine signaling through selective small molecule inhibition of A2AR using AZD4635 restores T cell function via an internal mechanism as well as tumor antigen cross-presentation by CD103+ DCs resulting in antitumor immunity.


Assuntos
Antígenos CD/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Células Dendríticas/imunologia , Cadeias alfa de Integrinas/metabolismo , Neoplasias/imunologia , Receptor A2A de Adenosina/metabolismo , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Transdução de Sinais
2.
Methods Mol Biol ; 1899: 103-118, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30649768

RESUMO

Alloantigen-specific hyporesponsiveness can be induced in alloreactive T cells contained within the whole peripheral blood mononuclear cell (PBMC) population by stimulating these responder cells ex vivo with HLA-mismatched stimulator PBMC as the antigen presenting cell (APC) source, in the presence of a CD28 costimulation blocking agent. As a result of this approach, specific alloreactivity is markedly decreased (by 1-2 logs), but third-party alloresponses and in vitro responses relying on the activation of pathogen- and tumor-associated antigen T-cell functional activities are not globally impinged upon (Guinan et al. N Engl J Med 340(22):1704-1714, 1999, Davies et al. Transplantation 86(6):854-864, 2008, Davies et al. Cell Transplant 21(9):2047-61, 2012). This method has been used clinically to alloanergize bone marrow and PBMC allografts, creating ex vivo cell therapies for adoptive transfer to blood cancer patients at high risk of disease relapse whose best option was to receive haploidentical hematopoietic cell transplants. These early phase trials consisting of, or containing, alloanergized T-cell infusions show promise in reducing graft-versus-host disease (GvHD), providing more rapid immune reconstitution, and decreasing severe post-transplant infectious complications and disease relapse. Herein, we describe this straightforward technique for generating alloanergized PBMC as it is performed in the research lab setting using belatacept for CD28-mediated costimulatory blockade (CSB) and PBMC isolated by Ficoll Hypaque gradient centrifugation as responders and APC. We also describe methods for evaluating subsequent alloproliferation to first and third party stimulation as well as assessment of cell division, pathogen-specific immunity, or allosuppression. The technique has successfully been transferred to collaborating labs, largely owing to the flexibility of using fresh or frozen PBMC, the lack of a requirement for specially isolated APC populations, and the ability to scale up or scale down the cell numbers that are to be anergized.


Assuntos
Antígenos CD28/imunologia , Tolerância Imunológica , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos/métodos , Linfócitos T Reguladores/imunologia , Células Apresentadoras de Antígenos/imunologia , Medula Óssea/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Isoantígenos/imunologia , Leucócitos Mononucleares/imunologia , Transplante Homólogo
3.
Cell Transplant ; 21(9): 2047-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22507909

RESUMO

Ex vivo alloanergization of human immune cells, via allostimulation in the presence of costimulatory blockade with either a combination of anti-B7.1 and anti-B7.2 antibodies or first-generation cytotoxic T-lymphocyte antigen 4-immunoglobulin (CTLA4-Ig), induces alloantigen-specific hyporesponsiveness and expands alloantigen-specific regulatory T cells (Treg). We have successfully used this approach in the clinical setting of haploidentical hematopoietic stem cell transplantation. Recently, the in vivo use of a new second-generation CTLA4-Ig, belatacept, has shown promise in controlling alloresponses after transplantation of both human kidneys and islet cells. We therefore compared the efficiency of first- and second-generation CTLA4-Ig in alloanergizing human peripheral blood mononuclear cells (PBMCs) and investigated whether ex vivo alloanergization with belatacept could be used to engineer an alloantigen-specific immunoregulatory population of autologous cells suitable for administration to recipients of cellular or solid organ transplant recipients. Alloanergization of HLA-mismatched human PBMCs with belatacept resulted in a greater reduction in subsequent alloresponses than alloanergization with first generation CTLA4-Ig. Moreover, subsequent ex vivo re-exposure of alloanergized cells to alloantigen in the absence of belatacept resulted in a significant expansion of Tregs with enhanced alloantigen-specific suppressive function. Alloanergized PBMCs retained functional Epstein-Barr virus (EBV)-specific T-cell responses, and expanded Tregs did not suppress EBV-specific proliferation of autologous cells. These results suggest that ex vivo alloanergization with belatacept provides a platform to engineer populations of recipient Treg with specificity for donor alloantigens but without nonspecific suppressive capacity. The potential advantages of such cells for solid organ transplantation include (1) reduction of the need for nonspecific immunosuppression, (2) retention of pathogen-specific immunity, and (3) control of graft rejection, if used as an intervention.


Assuntos
Imunoconjugados/administração & dosagem , Imunossupressores/administração & dosagem , Isoantígenos/imunologia , Abatacepte , Epitopos , Antígenos HLA/imunologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Imunologia de Transplantes
4.
Sci Transl Med ; 3(110): 110ra118, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22116933

RESUMO

Identification of safe, effective treatments to mitigate toxicity after extensive radiation exposure has proven challenging. Only a limited number of candidate approaches have emerged, and the U.S. Food and Drug Administration has yet to approve any agent for a mass-casualty radiation disaster. Because patients undergoing hematopoietic stem cell transplantation undergo radiation treatment that produces toxicities similar to radiation-disaster exposure, we studied patients early after such treatment to identify new approaches to this problem. Patients rapidly developed endotoxemia and reduced plasma bactericidal/permeability-increasing protein (BPI), a potent endotoxin-neutralizing protein, in association with neutropenia. We hypothesized that a treatment supplying similar endotoxin-neutralizing activity might replace the BPI deficit and mitigate radiation toxicity and tested this idea in mice. A single 7-Gy radiation dose, which killed 95% of the mice by 30 days, was followed 24 hours later by twice-daily, subcutaneous injections of the recombinant BPI fragment rBPI21 or vehicle alone for 14 or 30 days, with or without an oral fluoroquinolone antibiotic with broad-spectrum antibacterial activity, including that against endotoxin-bearing Gram-negative bacteria. Compared to either fluoroquinolone alone or vehicle plus fluoroquinolone, the combined rBPI21 plus fluoroquinolone treatment improved survival, accelerated hematopoietic recovery, and promoted expansion of stem and progenitor cells. The observed efficacy of rBPI21 plus fluoroquinolone initiated 24 hours after lethal irradiation, combined with their established favorable bioactivity and safety profiles in critically ill humans, suggests the potential clinical use of this radiation mitigation strategy and supports its further evaluation.


Assuntos
Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Proteínas Sanguíneas/uso terapêutico , Medula Óssea/patologia , Fluoroquinolonas/uso terapêutico , Lesões por Radiação/tratamento farmacológico , Técnicas de Ablação , Animais , Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Peptídeos Catiônicos Antimicrobianos/farmacologia , Contagem de Células Sanguíneas , Proteínas Sanguíneas/administração & dosagem , Proteínas Sanguíneas/farmacologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/efeitos da radiação , Citocinas/sangue , Endotoxemia/sangue , Endotoxemia/complicações , Endotoxinas/metabolismo , Enrofloxacina , Fluoroquinolonas/administração & dosagem , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Transplante de Células-Tronco Hematopoéticas , Humanos , Mediadores da Inflamação/sangue , Mucosa Intestinal/patologia , Mucosa Intestinal/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutropenia/sangue , Neutropenia/complicações , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/efeitos da radiação , Lesões por Radiação/sangue , Lesões por Radiação/complicações , Análise de Sobrevida , Irradiação Corporal Total
5.
J Vis Exp ; (49)2011 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-21445041

RESUMO

Allogeneic hematopoietic stem cell transplantation (AHSCT) offers the best chance of cure for many patients with congenital and acquired hematologic diseases. Unfortunately, transplantation of alloreactive donor T cells which recognize and damage healthy patient tissues can result in Graft-versus-Host Disease (GvHD). One challenge to successful AHSCT is the prevention of GvHD without associated impairment of the beneficial effects of donor T cells, particularly immune reconstitution and prevention of relapse. GvHD can be prevented by non-specific depletion of donor T cells from stem cell grafts or by administration of pharmacological immunosuppression. Unfortunately these approaches increase infection and disease relapse. An alternative strategy is to selectively deplete alloreactive donor T cells after allostimulation by recipient antigen presenting cells (APC) before transplant. Early clinical trials of these allodepletion strategies improved immune reconstitution after HLA-mismatched HSCT without excess GvHD. However, some allodepletion techniques require specialized recipient APC production and some approaches may have off-target effects including depletion of donor pathogen-specific T cells and CD4 T regulatory cells .One alternative approach is the inactivation of alloreactive donor T cells via induction of alloantigen-specific hyporesponsiveness. This is achieved by stimulating donor cells with recipient APC while providing blockade of CD28-mediated co-stimulation signals.This "alloanergization" approach reduces alloreactivity by 1-2 logs while preserving pathogen- and tumor-associated antigen T cell responses in vitro. The strategy has been successfully employed in 2 completed and 1 ongoing clinical pilot studies in which alloanergized donor T cells were infused during or after HLA-mismatched HSCT resulting in rapid immune reconstitution, few infections and less severe acute and chronic GvHD than historical control recipients of unmanipulated HLA-mismatched transplantation. Here we describe our current protocol for the generation of peripheral blood mononuclear cells (PBMC) which have been alloanergized to HLA-mismatched unrelated stimulator PBMC. Alloanergization is achieved by allostimulation in the presence of monoclonal antibodies to the ligands B7.1 and B7.1 to block CD28-mediated costimulation. This technique does not require the production of specialized stimulator APC and is simple to perform, requiring only a single and relatively brief ex vivo incubation step. As such, the approach can be easily standardized for clinical use to generate donor T cells with reduced alloreactivity but retaining pathogen-specific immunity for adoptive transfer in the setting of AHSCT to improve immune reconstitution without excessive GvHD.


Assuntos
Anticorpos Monoclonais/imunologia , Antígeno B7-1/imunologia , Antígeno B7-2/imunologia , Isoantígenos/imunologia , Leucócitos Mononucleares/imunologia , Anergia Clonal/imunologia , Técnicas de Cocultura , Epitopos , Antígenos HLA/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Teste de Cultura Mista de Linfócitos
6.
Vaccine ; 28(50): 7852-64, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-20943208

RESUMO

Immunization with plasmid DNA that has been encapsulated in poly lactide-co-glycolide (PLG) microparticles targets the plasmid DNA to antigen presenting cells and elicits immune responses to the encoded antigen(s). Application of a series of electrical pulses (EPT) immediately following unformulated DNA injection enhances expression of the encoded antigen and increases immune responses. The combination of using EPT before or after PLG-encapsulated plasmid DNA immunization was tested to determine if enhanced immune responses would be generated. The results show that the combination lead to both enhanced expression of antigen and more robust T cell responses, even if EPT was applied prior to immunization. The data also demonstrate that recruitment of phagocytes to the injection site was markedly enhanced by EPT, and this resulted in an increase of the antigen expression levels in these cells. Co-administration of microparticles and EPT also effected localized necrosis of muscle fibers, caused persistent Th-1-modulated cytokine production, and lead to the release of two endogenous adjuvants, uric acid and HMGB1. In all, we describe that increased immunogenicity observed with the combination of PLG-encapsulated plasmid DNA microparticle with EPT was caused by an increase in the recruitment of antigen presenting cells which mediated a more robust T cell response than observed with immunization alone.


Assuntos
Eletroporação , Ácido Láctico/farmacologia , Ácido Poliglicólico/farmacologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Células Apresentadoras de Antígenos/imunologia , Hidrocarboneto de Aril Hidroxilases/imunologia , Linhagem Celular Tumoral , Citocromo P-450 CYP1B1 , Citocinas/imunologia , Feminino , Imunidade Inata , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Microesferas , Fagócitos/imunologia , Plasmídeos/imunologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Linfócitos T/imunologia
7.
Cell Immunol ; 262(2): 150-61, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20206921

RESUMO

Cyclophosphamide in combination with immunotherapeutic approaches preferentially impinges on T(reg) activity and allows for robust generation of T cell effectors. Reduced dosages of cyclophosphamide are necessary to restrict its cytotoxic effects to the negative regulatory cell populations while sparing effector lymphocytes. We investigated cyclophosphamide dosing in combination with ZYC300, a PLG-encapsulated plasmid DNA vaccine which encodes the cytochrome P450 family member, CYP1B1, a known human tumor-associated antigen. In mice, three consecutive, low doses of cyclophosphamide comprised a superior regimen in enhancing the magnitude, diversity of epitopes, and avidity to individual epitopes of specific T cell responses when compared to regimens that used either a single low or a single high dose. Consecutive low doses of cyclophosphamide predominantly targeted T(regs) while sparing overall T lymphocyte counts. Thus, we report the synergistic activity of pharmacologic T(reg) depletion with cyclophosphamide on quantitatively and qualitatively increasing T cell responses to a known human tumor-associated antigen.


Assuntos
Ciclofosfamida/farmacologia , Imunossupressores/farmacologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Vacinas de DNA , Animais , Antígenos de Neoplasias/imunologia , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/imunologia , Linhagem Celular , Ciclofosfamida/imunologia , Citocromo P-450 CYP1B1 , Composição de Medicamentos , Epitopos/imunologia , Feminino , Humanos , Imunização , Imunossupressores/imunologia , Camundongos , Camundongos Endogâmicos C3H , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia
8.
Mol Genet Metab ; 97(1): 35-42, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19231265

RESUMO

Systemic administration of recombinant acid sphingomyelinase (rhASM) into ASM deficient mice (ASMKO) results in hydrolysis of the abnormal storage of sphingomyelin in lysosomes of the liver, spleen and lung. However, the efficiency with which the substrate is cleared from the lung, particularly the alveolar macrophages, appears to be lower than from the other visceral tissues. To determine if delivery of rhASM into the air spaces of the lung could enhance clearance of pulmonary sphingomyelin, enzyme was administered to ASMKO mice by intranasal instillation. Treatment resulted in a significant and dose-dependent reduction in sphingomyelin levels in the lung. Concomitant with this reduction in substrate levels was a decrease in the amounts of the pro-inflammatory cytokine, MIP-1alpha, in the bronchoalveolar lavage fluids and an improvement in lung pathology. Maximal reduction of lung sphingomyelin levels was observed at 7 days post-treatment. However, reaccumulation of the substrate was noted starting at day 14 suggesting that repeated treatments will be necessary to effect a sustained reduction in sphingomyelin levels. In addition to reducing the storage abnormality in the lung, intranasal delivery of rhASM also resulted in clearance of the substrate from the liver and spleen. Hence, pulmonary administration of rhASM may represent an alternative route of delivery to address the visceral pathology associated with ASM deficiency.


Assuntos
Pulmão/metabolismo , Lisossomos/metabolismo , Doenças de Niemann-Pick/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Esfingomielina Fosfodiesterase/administração & dosagem , Esfingomielina Fosfodiesterase/uso terapêutico , Esfingomielinas/metabolismo , Administração Intranasal , Animais , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Feminino , Humanos , Cinética , Fígado/metabolismo , Fígado/patologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças de Niemann-Pick/enzimologia , Doenças de Niemann-Pick/metabolismo , Doenças de Niemann-Pick/patologia , Proteínas Recombinantes/administração & dosagem , Esfingomielina Fosfodiesterase/genética , Baço/metabolismo , Baço/patologia
9.
Mol Ther ; 15(3): 492-500, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17191071

RESUMO

The advent of novel adeno-associated virus (AAV) serotype vectors with higher transduction activity has encouraged a re-evaluation of the merits of this delivery platform for a variety of diseases. We report here that administration of a recombinant AAV8-based serotype vector encoding human alpha-galactosidase A into Fabry mice facilitated more rapid and significantly higher levels of production of the enzyme than an AAV2 vector. This translated into improved clearance of globotriaosylceramide, the glycosphingolipid that accumulates in the lysosomes of affected Fabry cells, and to correction of the peripheral neuropathy shown associated with this disease. The higher levels of alpha-galactosidase A expression also allowed for a more rapid induction of immunotolerance to the enzyme. Recombinant AAV8 vectors that facilitated hepatic-restricted expression of high levels of alpha-galactosidase A conferred immunotolerance to the expressed enzyme as early as 30 days post-treatment. Animals expressing lower levels of the hydrolase, such as those treated with an AAV2-based vector or with lower doses of the AAV8-based vector, were also able to develop immunotolerance, but only after a more extended time period. Adoptive transfer of T cells isolated from the spleens of immunotolerized mice suppressed the formation of antibodies in naïve recipient animals, suggesting the possible role of regulatory T cells in effecting this state.


Assuntos
Dependovirus/genética , Doença de Fabry/enzimologia , Expressão Gênica/genética , Fígado/metabolismo , alfa-Galactosidase/metabolismo , Animais , Anticorpos/imunologia , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/imunologia , Doença de Fabry/genética , Doença de Fabry/patologia , Doença de Fabry/terapia , Regulação Enzimológica da Expressão Gênica , Terapia Genética , Vetores Genéticos/genética , Humanos , Tolerância Imunológica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Doenças do Sistema Nervoso Periférico/enzimologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/patologia , Doenças do Sistema Nervoso Periférico/terapia , alfa-Galactosidase/genética , alfa-Galactosidase/imunologia
10.
Mol Ther ; 15(3): 492-500, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28182896

RESUMO

The advent of novel adeno-associated virus (AAV) serotype vectors with higher transduction activity has encouraged a re-evaluation of the merits of this delivery platform for a variety of diseases. We report here that administration of a recombinant AAV8-based serotype vector encoding human α-galactosidase A into Fabry mice facilitated more rapid and significantly higher levels of production of the enzyme than an AAV2 vector. This translated into improved clearance of globotriaosylceramide, the glycosphingolipid that accumulates in the lysosomes of affected Fabry cells, and to correction of the peripheral neuropathy shown associated with this disease. The higher levels of α-galactosidase A expression also allowed for a more rapid induction of immunotolerance to the enzyme. Recombinant AAV8 vectors that facilitated hepatic-restricted expression of high levels of α-galactosidase A conferred immunotolerance to the expressed enzyme as early as 30 days post-treatment. Animals expressing lower levels of the hydrolase, such as those treated with an AAV2-based vector or with lower doses of the AAV8-based vector, were also able to develop immunotolerance, but only after a more extended time period. Adoptive transfer of T cells isolated from the spleens of immunotolerized mice suppressed the formation of antibodies in naïve recipient animals, suggesting the possible role of regulatory T cells in effecting this state.

11.
Mol Ther ; 12(3): 431-40, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16099409

RESUMO

Acid sphingomyelinase deficiency is a lysosomal storage disorder in which the defective lysosomal hydrolase fails to degrade sphingomyelin. The resulting accumulation of substrate in the lysosomes of histiocytic cells leads to hepatosplenomegaly and severe pulmonary inflammation. Administration of a recombinant AAV1 vector encoding human acid sphingomyelinase to acid sphingomyelinase knockout (ASMKO) mice effectively reduced the accumulated substrate in all of the affected visceral organs. However, more complete and rapid clearance of sphingomyelin was observed when an AAV8-based serotype vector was used in lieu of AAV1. Importantly, AAV8-mediated hepatic expression of higher and sustained levels of the enzyme also corrected the abnormal cellularity, cell differentials, and levels of the chemokine MIP-1alpha in the bronchoalveolar lavage fluids of the ASMKO mice. Treatment also reversed the morphological aberrations associated with the alveolar macrophages of ASMKO mice and restored their phagocytic activity. No antibodies to the expressed enzyme were detected when the viral vectors were used in conjunction with a transcription cassette harboring a liver-restricted enhancer/promoter. Together, these data support the continued development of AAV8-mediated hepatic gene transfer as an approach to treat the visceral manifestations observed in individuals with acid sphingomyelinase deficiency.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Fígado/enzimologia , Doenças de Niemann-Pick/terapia , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo , Animais , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Elementos Facilitadores Genéticos , Técnicas de Transferência de Genes , Terapia Genética/instrumentação , Humanos , Cinética , Fígado/metabolismo , Lisossomos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Regiões Promotoras Genéticas , Esfingomielinas/metabolismo , Fatores de Tempo
12.
Mol Ther ; 9(2): 231-40, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14759807

RESUMO

The successful application of gene therapy for the treatment of genetic diseases such as Fabry is reliant on the development of vectors that are safe and that facilitate sustained expression of therapeutic levels of the transgene product. Here, we report that intravenous administration of a recombinant AAV2 vector encoding human alpha-galactosidase A under the transcriptional control of a liver-restricted enhancer/promoter (AAV2/DC190-alphagal) generated significantly higher levels of expression in BALB/c and Fabry mice than could be realized using the ubiquitous CMV promoter (AAV2/CMVHI-alphagal). Moreover, AAV2/DC190-alphagal-mediated hepatic expression of alpha-galactosidase A was sustained for 12 months in BALB/c mice and was associated with a significantly reduced immune response to the expressed enzyme. Subsequent challenge of the AAV2/DC190-alphagal-treated animals with recombinant human alpha-galactosidase A at 6 months failed to elicit the production of anti-alpha-galactosidase A antibodies, suggesting the induction of immune tolerance in these animals. The levels of expression attained with AAV2/DC190-alphagal in the Fabry mice were sufficient to reduce the abnormal accumulation of globotriaosylceramide in the liver, spleen, and heart to basal levels and in the kidney by approximately 40% at 8 weeks. Together, these results demonstrate that AAV2-mediated gene transfer that limits the expression of alpha-galactosidase A to the liver may be a viable strategy for treating Fabry disease.


Assuntos
Dependovirus/genética , Doença de Fabry/terapia , Terapia Genética , Tolerância Imunológica , Fígado/metabolismo , Regiões Promotoras Genéticas/genética , alfa-Galactosidase/uso terapêutico , Animais , DNA Recombinante/genética , Modelos Animais de Doenças , Elementos Facilitadores Genéticos/genética , Doença de Fabry/genética , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA