Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ophthalmology ; 131(6): 682-691, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38160882

RESUMO

PURPOSE: To report long-term results from a phase 1/2a clinical trial assessment of a scaffold-based human embryonic stem cell-derived retinal pigmented epithelium (RPE) implant in patients with advanced geographic atrophy (GA). DESIGN: A single-arm, open-label phase 1/2a clinical trial approved by the United States Food and Drug Administration. PARTICIPANTS: Patients were 69-85 years of age at the time of enrollment and were legally blind in the treated eye (best-corrected visual acuity [BCVA], ≤ 20/200) as a result of GA involving the fovea. METHODS: The clinical trial enrolled 16 patients, 15 of whom underwent implantation successfully. The implant was administered to the worse-seeing eye with the use of a custom subretinal insertion device. The companion nonimplanted eye served as the control. The primary endpoint was at 1 year; thereafter, patients were followed up at least yearly. MAIN OUTCOME MEASURES: Safety was the primary endpoint of the study. The occurrence and frequency of adverse events (AEs) were determined by scheduled eye examinations, including measurement of BCVA and intraocular pressure and multimodal imaging. Serum antibody titers were collected to monitor systemic humoral immune responses to the implanted cells. RESULTS: At a median follow-up of 3 years, fundus photography revealed no migration of the implant. No unanticipated, severe, implant-related AEs occurred, and the most common anticipated severe AE (severe retinal hemorrhage) was eliminated in the second cohort (9 patients) through improved intraoperative hemostasis. Nonsevere, transient retinal hemorrhages were noted either during or after surgery in all patients as anticipated for a subretinal surgical procedure. Throughout the median 3-year follow-up, results show that implanted eyes were more likely to improve by > 5 letters of BCVA and were less likely to worsen by > 5 letters compared with nonimplanted eyes. CONCLUSIONS: This report details the long-term follow-up of patients with GA to receive a scaffold-based stem cell-derived bioengineered RPE implant. Results show that the implant, at a median 3-year follow-up, is safe and well tolerated in patients with advanced dry age-related macular degeneration. The safety profile, along with the early indication of efficacy, warrants further clinical evaluation of this novel approach for the treatment of GA. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.


Assuntos
Atrofia Geográfica , Epitélio Pigmentado da Retina , Acuidade Visual , Humanos , Atrofia Geográfica/cirurgia , Atrofia Geográfica/fisiopatologia , Epitélio Pigmentado da Retina/transplante , Epitélio Pigmentado da Retina/patologia , Idoso , Acuidade Visual/fisiologia , Feminino , Idoso de 80 Anos ou mais , Masculino , Seguimentos , Tomografia de Coerência Óptica , Células-Tronco Embrionárias Humanas/transplante , Células-Tronco Embrionárias Humanas/citologia , Transplante de Células-Tronco , Resultado do Tratamento
2.
J Neurosurg Spine ; : 1-10, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35364569

RESUMO

OBJECTIVE: The purpose of this study was to evaluate the safety of oligodendrocyte progenitor cells (LCTOPC1) derived from human pluripotent stem cells administered between 7 and 14 days postinjury to patients with T3 to T11 neurologically complete spinal cord injury (SCI). The rationale for this first-in-human trial was based on evidence that administration of LCTOPC1 supports survival and potential repair of key cellular components and architecture at the SCI site. METHODS: This study was a multisite, open-label, single-arm interventional clinical trial. Participants (n = 5) received a single intraparenchymal injection of 2 × 106 LCTOPC1 caudal to the epicenter of injury using a syringe positioning device. Immunosuppression with tacrolimus was administered for a total of 60 days. Participants were followed with annual in-person examinations and MRI for 5 years at the time of this report and will be followed with annual telephone questionnaires for 6 to 15 years postinjection. The primary endpoint was safety, as measured by the frequency and severity of adverse events related to the LCTOPC1 injection, the injection procedure, and/or the concomitant immunosuppression administered. The secondary endpoint was neurological function as measured by sensory scores and lower-extremity motor scores as measured by the International Standards for Neurological Classification of Spinal Cord Injury examinations. RESULTS: No unanticipated serious adverse events related to LCTOPC1 have been reported with 98% follow-up of participants (49 of 50 annual visits) through the first 10 years of the clinical trial. There was no evidence of neurological decline, enlarging masses, further spinal cord damage, or syrinx formation. MRI results during the long-term follow-up period in patients administered LCTOPC1 cells showed that 80% of patients demonstrated T2 signal changes consistent with the formation of a tissue matrix at the injury site. CONCLUSIONS: This study provides crucial first-in-human safety data supporting the pursuit of future human embryonic stem cell-derived therapies. While we cannot exclude the possibility of future adverse events, the experience in this trial provides evidence that this cell type can be well tolerated by patients, with an event-free period of up to 10 years. Based on the safety profile of LCTOPC1 obtained in this study, a cervical dose escalation trial was initiated (NCT02302157).

3.
Transl Vis Sci Technol ; 10(10): 13, 2021 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34613357

RESUMO

Purpose: To report 1-year follow-up of a phase 1/2a clinical trial testing a composite subretinal implant having polarized human embryonic stem cell (hESC)-derived retinal pigment epithelium (RPE) cells on an ultrathin parylene substrate in subjects with advanced non-neovascular age-related macular degeneration (NNAMD). Methods: The phase 1/2a clinical trial included 16 subjects in two cohorts. The main endpoint was safety assessed at 365 days using ophthalmic and systemic exams. Pseudophakic subjects with geographic atrophy (GA) and severe vision loss were eligible. Low-dose tacrolimus immunosuppression was utilized for 68 days in the peri-implantation period. The implant was delivered to the worst seeing eye with a custom subretinal insertion device in an outpatient setting. A data safety monitoring committee reviewed all results. Results: The treated eyes of all subjects were legally blind with a baseline best-corrected visual acuity (BCVA) of ≤ 20/200. There were no unexpected serious adverse events. Four subjects in cohort 1 had serious ocular adverse events, including retinal hemorrhage, edema, focal retinal detachment, or RPE detachment, which was mitigated in cohort 2 using improved hemostasis during surgery. Although this study was not powered to assess efficacy, treated eyes from four subjects showed an increased BCVA of >5 letters (6-13 letters). A larger proportion of treated eyes experienced a >5-letter gain when compared with the untreated eye (27% vs. 7%; P = not significant) and a larger proportion of nonimplanted eyes demonstrated a >5-letter loss (47% vs. 33%; P = not significant). Conclusions: Outpatient delivery of the implant can be performed routinely. At 1 year, the implant is safe and well tolerated in subjects with advanced dry AMD. Translational Relevance: This work describes the first clinical trial, to our knowledge, of a novel implant for advanced dry AMD.


Assuntos
Atrofia Geográfica , Transplante de Células-Tronco Hematopoéticas , Degeneração Macular , Seguimentos , Atrofia Geográfica/terapia , Humanos , Degeneração Macular/terapia , Acuidade Visual
4.
J Neurosurg Spine ; 35(3): 389-397, 2021 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-34243160

RESUMO

OBJECTIVE: This study was conducted as a final proof-of-safety direct injection of oligodendrocyte progenitor cells into the uninjured spinal cord prior to translation to the human clinical trials. METHODS: In this study, 107 oligodendrocyte progenitor cells (LCTOPC1, also known as AST-OPC1 and GRNOPC1) in 50-µL suspension were injected directly into the uninjured spinal cords of 8 immunosuppressed Göttingen minipigs using a specially designed stereotactic delivery device. Four additional Göttingen minipigs were given Hanks' Balanced Salt Solution and acted as the control group. RESULTS: Cell survival and no evidence of histological damage, abnormal inflammation, microbiological or immunological abnormalities, tumor formation, or unexpected morbidity or mortality were demonstrated. CONCLUSIONS: These data strongly support the safety of intraparenchymal injection of LCTOPC1 into the spinal cord using a model anatomically similar to that of the human spinal cord. Furthermore, this research provides guidance for future clinical interventions, including mechanisms for precise positioning and anticipated volumes of biological payloads that can be safely delivered directly into uninjured portions of the spinal cord.

5.
Sci Transl Med ; 10(435)2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29618560

RESUMO

Retinal pigment epithelium (RPE) dysfunction and loss are a hallmark of non-neovascular age-related macular degeneration (NNAMD). Without the RPE, a majority of overlying photoreceptors ultimately degenerate, leading to severe, progressive vision loss. Clinical and histological studies suggest that RPE replacement strategies may delay disease progression or restore vision. A prospective, interventional, U.S. Food and Drug Administration-cleared, phase 1/2a study is being conducted to assess the safety and efficacy of a composite subretinal implant in subjects with advanced NNAMD. The composite implant, termed the California Project to Cure Blindness-Retinal Pigment Epithelium 1 (CPCB-RPE1), consists of a polarized monolayer of human embryonic stem cell-derived RPE (hESC-RPE) on an ultrathin, synthetic parylene substrate designed to mimic Bruch's membrane. We report an interim analysis of the phase 1 cohort consisting of five subjects. Four of five subjects enrolled in the study successfully received the composite implant. In all implanted subjects, optical coherence tomography imaging showed changes consistent with hESC-RPE and host photoreceptor integration. None of the implanted eyes showed progression of vision loss, one eye improved by 17 letters and two eyes demonstrated improved fixation. The concurrent structural and functional findings suggest that CPCB-RPE1 may improve visual function, at least in the short term, in some patients with severe vision loss from advanced NNAMD.


Assuntos
Degeneração Macular/terapia , Células Cultivadas , Feminino , Atrofia Geográfica/terapia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Masculino , Estudos Prospectivos , Epitélio Pigmentado da Retina/citologia , Transplante de Células-Tronco , Tomografia de Coerência Óptica
6.
Stem Cells Transl Med ; 6(10): 1917-1929, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28834391

RESUMO

Cervical spinal cord injury (SCI) remains an important research focus for regenerative medicine given the potential for severe functional deficits and the current lack of treatment options to augment neurological recovery. We recently reported the preclinical safety data of a human embryonic cell-derived oligodendrocyte progenitor cell (OPC) therapy that supported initiation of a phase I clinical trial for patients with sensorimotor complete thoracic SCI. To support the clinical use of this OPC therapy for cervical injuries, we conducted preclinical efficacy and safety testing of the OPCs in a nude rat model of cervical SCI. Using the automated TreadScan system to track motor behavioral recovery, we found that OPCs significantly improved locomotor performance when administered directly into the cervical spinal cord 1 week after injury, and that this functional improvement was associated with reduced parenchymal cavitation and increased sparing of myelinated axons within the injury site. Based on large scale biodistribution and toxicology studies, we show that OPC migration is limited to the spinal cord and brainstem and did not cause any adverse clinical observations, toxicities, allodynia, or tumors. In combination with previously published efficacy and safety data, the results presented here supported initiation of a phase I/IIa clinical trial in the U.S. for patients with sensorimotor complete cervical SCI. Stem Cells Translational Medicine 2017;6:1917-1929.


Assuntos
Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Neurais/transplante , Oligodendroglia/transplante , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/efeitos adversos , Animais , Movimento Celular , Vértebras Cervicais/lesões , Feminino , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Ratos , Transplante de Células-Tronco/métodos
7.
Cancer ; 123(16): 3061-3072, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28411378

RESUMO

BACKGROUND: Telomerase activity in leukemic blasts frequently is increased among patients with high-risk acute myeloid leukemia (AML). In the current study, the authors evaluated the feasibility, safety, immunogenicity, and therapeutic potential of human telomerase reverse transcriptase (hTERT)-expressing autologous dendritic cells (hTERT-DCs) in adult patients with AML. METHODS: hTERT-DCs were produced from patient-specific leukapheresis, electroporated with an mRNA-encoding hTERT and a lysosomal-targeting sequence, and cryopreserved. A total of 22 patients with a median age of 58 years (range, 30-75 years) with intermediate-risk or high-risk AML in first or second complete remission (CR) were enrolled. hTERT-DCs were generated for 24 patients (73%). A median of 17 intradermal vaccinations (range, 6-32 intradermal vaccinations) containing 1×107 cells were administered as 6 weekly injections followed by 6 biweekly injections. A total of 21 patients (16 in first CR, 3 in second CR, and 2 with early disease recurrence) received hTERT-DCs. RESULTS: hTERT-DCs were well tolerated with no severe toxicities reported, with the exception of 1 patient who developed idiopathic thrombocytopenic purpura. Of the 19 patients receiving hTERT-DCs in CR, 11 patients (58%) developed hTERT-specific T-cell responses that primarily were targeted toward hTERT peptides with predicted low human leukocyte antigen (HLA)-binding affinities. With a median follow-up of 52 months, 58% of patients in CR (11 of 19 patients) were free of disease recurrence at the time of their last follow-up visit; 57% of the patients who were aged ≥60 years (4 of 7 patients) also were found to be free of disease recurrence at a median follow-up of 54 months. CONCLUSIONS: The generation of hTERT-DCs is feasible and vaccination with hTERT-DCs appears to be safe and may be associated with favorable recurrence-free survival. Cancer 2017;123:3061-72. © 2017 American Cancer Society.


Assuntos
Vacinas Anticâncer/uso terapêutico , Células Dendríticas/metabolismo , Imunoterapia/métodos , Leucaférese , Leucemia Mieloide Aguda/terapia , Telomerase/genética , Adulto , Idoso , Intervalo Livre de Doença , ELISPOT , Estudos de Viabilidade , Feminino , Humanos , Leucemia Mieloide Aguda/imunologia , Masculino , Pessoa de Meia-Idade , RNA Mensageiro , Indução de Remissão , Linfócitos T/imunologia
8.
Regen Med ; 10(8): 939-58, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26345388

RESUMO

AIM: To characterize the preclinical safety profile of a human embryonic stem cell-derived oligodendrocyte progenitor cell therapy product (AST-OPC1) in support of its use as a treatment for spinal cord injury (SCI). MATERIALS & METHODS: The phenotype and functional capacity of AST-OPC1 was characterized in vitro and in vivo. Safety and toxicology of AST-OPC1 administration was assessed in rodent models of thoracic SCI. RESULTS: These results identify AST-OPC1 as an early-stage oligodendrocyte progenitor population capable of promoting neurite outgrowth in vitro and myelination in vivo. AST-OPC1 administration did not cause any adverse clinical observations, toxicities, allodynia or tumors. CONCLUSION: These results supported initiation of a Phase I clinical trial in patients with sensorimotor complete thoracic SCI.


Assuntos
Células-Tronco Embrionárias Humanas , Oligodendroglia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/métodos , Animais , Xenoenxertos , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/transplante , Humanos , Camundongos , Camundongos Nus , Oligodendroglia/metabolismo , Oligodendroglia/transplante , Traumatismos da Medula Espinal/metabolismo , Transplante de Células-Tronco/efeitos adversos
9.
Regen Med ; 6(3): 303-18, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21548736

RESUMO

AIM: Dendritic cell (DC)-based vaccines are designed to exploit the intrinsic capacity of these highly effective antigen presenting cells to prime and boost antigen-specific T-cell immune responses. Successful development of DC-based vaccines will be dependent on the ability to utilize and harness the full potential of these potent immune stimulatory cells. Recent advances to generate DCs derived from human embryonic stem cells (hESCs) that are suitable for clinical use represent an alternative strategy from conventional approaches of using patient-specific DCs. Although the differentiation of hESC-derived DCs in serum-free defined conditions has been established, the stimulatory potential of these hESC-derived DCs have not been fully evaluated. METHODS: hESC-derived DCs were differentiated in serum-free defined culture conditions. The delivery of antigen into hESC-derived DCs was investigated using mRNA transfection and replication-deficient adenoviral vector transduction. hESC-derived DCs modified with antigen were evaluated for their capacity to stimulate antigen-specific T-cell responses with known HLA matching. Since IL-12 is a key cytokine that drives T-cell function, further enhancement of DC potency was evaluated by transfecting mRNA encoding the IL-12p70 protein into hESC-derived DCs. RESULTS: The transfection of mRNA into hESC-derived DCs was effective for heterologous protein expression. The efficiency of adenoviral vector transduction into hESC-derived DCs was poor. These mRNA-transfected DCs were capable of stimulating human telomerase reverse transcriptase antigen-specific T cells composed of varying degrees of HLA matching. In addition, we observed the transfection of mRNA encoding IL-12p70 enhanced the T-cell stimulation potency of hESC-derived DCs. CONCLUSION: These data provide support for the development and modification of hESC-derived DCs with mRNA as a potential strategy for the induction of T-cell-mediated immunity.


Assuntos
Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Adenoviridae/efeitos dos fármacos , Adenoviridae/genética , Apresentação de Antígeno/efeitos dos fármacos , Linhagem Celular , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Epitopos/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Teste de Histocompatibilidade , Humanos , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/imunologia , Interferon gama/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Peptídeos/farmacologia , Fenótipo , Fosfoproteínas/imunologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Transdução Genética , Transfecção , Proteínas da Matriz Viral/imunologia
10.
Regen Med ; 4(5): 659-61, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19761390

RESUMO

Dr Jane Lebkowski joined Geron Corporation in 1998 and is currently Senior Vice President and Chief Scientific Officer of the Regenerative Medicine Division. Dr Lebkowski heads Geron's human embryonic stem cell program, and is responsible for all research, preclinical development, product development, manufacturing and clinical development activities. Prior to Geron, Dr Lebkowski was Vice President of Research and Development at Applied Immune Sciences. Following the acquisition of Applied Immune Sciences by Rhone Poulenc Rorer (RPR, currently Sanofi-Aventis), Dr Lebkowski remained at RPR as Vice President of Discovery Research. During Dr Lebkowski's tenure at RPR, she coordinated preclinical investigations of gene-therapy approaches for treatment of cancer, cardiovascular disease and nervous system disorders, and directed vector formulations and delivery development. Dr Lebkowski received her PhD in Biochemistry from Princeton University in 1982, and completed a postdoctoral fellowship at the Department of Genetics, Stanford University (CA, USA) in 1986. Dr Lebkowski has published over 70 peer-reviewed papers and has 12 issued US patents. Dr Lebkowski serves as the co-chair of the Industrial Committee of the International Society for Stem Cell Research and serves on the editorial boards of several scientific publications.


Assuntos
Células-Tronco Embrionárias , Medicina Regenerativa , Ensaios Clínicos como Assunto , Humanos , Apoio à Pesquisa como Assunto , Transplante de Células-Tronco/efeitos adversos
11.
Regen Med ; 4(4): 513-26, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19580370

RESUMO

AIM: Dendritic cell (DC)-based vaccines have a potential utility for use in the treatment of malignancy. Human embryonic stem cells (hESCs) may provide a more cost-effective and reliable source of DCs for immunotherapy purposes, providing on-demand access for patients. METHOD: We developed a protocol to generate DCs from hESCs in vitro in the absence of serum and feeder cells. This protocol uses growth factors bone morphogenetic protein-4, granulocyte macrophage-colony stimulating factor (GM-CSF), stem cell factor and VEGF in serum-free media to generate hESC-derived monocytic cells. These cells are further differentiated to hESC-derived immature DCs with GM-CSF and IL-4, and matured to hESC-derived mature DCs with a maturation cocktail consisting of GM-CSF, TNF-alpha, IL-1beta, IFN-gamma and PGE2. RESULTS: This study demonstrates the applicability of our defined differentiation process in generating functional hESC-derived DCs from multiple hESC lines. We show that hESC-derived immature DCs phagocytose, process, and present antigen upon maturation. hESC-derived mature DCs express the maturation marker CD83, produce Th1-directing cytokine IL-12p70, migrate in response to chemokine, and activate both viral and tumor antigen-specific T-cell responses. CONCLUSION: We developed a chemically defined system to generate unlimited numbers of DCs from hESCs. Our results demonstrate that hESC-derived DCs generated from this process are immunogenic and have the potential to be used for DC immunotherapy.


Assuntos
Biotecnologia/métodos , Diferenciação Celular/fisiologia , Células Dendríticas/citologia , Células-Tronco Embrionárias/citologia , Imunoterapia/métodos , Vacinas/biossíntese , Proteína Morfogenética Óssea 4 , Células-Tronco Embrionárias/fisiologia , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Humanos , Fator de Células-Tronco , Linfócitos T/citologia , Linfócitos T/metabolismo , Telomerase/metabolismo , Fator A de Crescimento do Endotélio Vascular
12.
Stem Cells ; 23(3): 315-23, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15749926

RESUMO

Previous studies have shown that prolonged propagation of undifferentiated human embryonic stem cells (hESCs) requires conditioned medium from mouse embryonic feeders (MEF-CM) as well as matrix components. Because hESCs express growth factor receptors, including those for basic fibroblast growth factor (bFGF), stem cell factor (SCF), and fetal liver tyrosine kinase-3 ligand (Flt3L), we evaluated these and other growth factors for their ability to maintain undifferentiated hESCs in the absence of conditioned medium. We found cultures maintained in bFGF alone or in combination with other factors showed characteristics similar to MEF-CM control cultures, including morphology, surface marker and transcription factor expression, telomerase activity, differentiation, and karyotypic stability. In contrast, cells in media containing Flt-3L, thrombopoietin, and SCF, individually or in combination, showed almost complete differentiation after 6 weeks in culture. These data demonstrate that hESCs can be maintained in nonconditioned medium using growth factors.


Assuntos
Proliferação de Células/efeitos dos fármacos , Embrião de Mamíferos/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células-Tronco Pluripotentes/citologia , Animais , Antígenos CD/metabolismo , Antígenos de Superfície , Diferenciação Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Proteínas de Ligação a DNA/genética , Fator de Crescimento Epidérmico/genética , Citometria de Fluxo , Proteínas Ligadas por GPI , Expressão Gênica/genética , Glicoproteínas/metabolismo , Glicoesfingolipídeos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Cariotipagem , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos SCID , Proteínas de Neoplasias/genética , Fator 3 de Transcrição de Octâmero , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Proteoglicanas , Antígenos Embrionários Estágio-Específicos , Telomerase/genética , Telomerase/metabolismo , Teratoma/patologia , Tetraspanina 29 , Fatores de Transcrição/genética
13.
Haematologica ; 89(3): 377-8, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15020288

RESUMO

Overexpression of telomerase reverse transcriptase (hTERT) can immortalize some primary human mesenchymal cells. We investigated whether retrovirally-mediated expression of hTERT in CD34+ umbilical cord blood (UCB) cells can extend the replicative lifespan of human hematopoietic progenitor cells. Overexpression of hTERT did not immortalize these cells but did lead to enhanced survival of mature hematopoietic cells.


Assuntos
Antígenos CD34/sangue , Proteínas de Ligação a DNA/metabolismo , Sangue Fetal/citologia , Sangue Fetal/enzimologia , Células-Tronco Hematopoéticas/citologia , Telomerase/metabolismo , Divisão Celular , Sobrevivência Celular , Sangue Fetal/imunologia , Células-Tronco Hematopoéticas/enzimologia , Humanos , Retroviridae/fisiologia , Transdução Genética
14.
Cancer Gene Ther ; 11(3): 174-85, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14726958

RESUMO

The human telomerase reverse transcriptase (hTERT) promoter is known to selectively drive transgene expression in many human cancer cells expressing hTERT, the catalytic component of the telomerase ribonucleoprotein complex. We have created a conditionally replicative adenovirus where the viral E1A gene, which is required for viral replication, is under the control of the hTERT promoter (AdhTERTp-E1A). In vitro studies with AdhTERTp-E1A virus on a variety of normal and tumor cell lines have shown that viral genome replication and productive infection is primarily restricted to telomerase-positive tumor cells. Lytic replication was not observed in normal primary fibroblast and epithelial cell lines tested. In vivo administration of the virus into nude mice bearing human liver or prostate tumor xenografts produced significant tumor reduction and, in some cases, resulted in complete tumor regression. AdhTERTp-E1A virus did not actively express E1A in normal mouse liver, in contrast to a control oncolytic vector in which the CMV promoter (AdCMVp-E1A) was driving the E1A gene. In addition, AdhTERTp-E1A virus produced no apparent toxicity to the liver in systemically injected mice. The hTERT promoter-driven oncolytic virus also produced significantly less toxicity to freshly cultured human hepatocytes. These studies demonstrate that an oncolytic virus driven by the telomerase promoter can be used to effectively kill a wide variety of cancer cell types and has the potential to treat primary and metastatic cancer of diverse origins.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Vetores Genéticos/toxicidade , Neoplasias/terapia , Telomerase/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Feminino , Hepatócitos/patologia , Humanos , Fígado/patologia , Neoplasias Hepáticas/terapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/patologia , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/terapia , Replicação Viral , Vírus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cancer Gene Ther ; 10(3): 239-49, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12637945

RESUMO

Human telomerase reverse transcriptase (hTERT) is the catalytic component of a functional telomerase complex, which is important in maintaining cell immortality. In most normal human adult cells, the expression of telomerase is very low and/or transient. In contrast, almost 90% of human tumors express a relatively high level of telomerase implying the possibility of using hTERT as a universal candidate tumor antigen. In this study, we show that human monocyte-derived dendritic cells (DCs) lack telomerase activity. Similar to other normal somatic cells, DCs express the RNA (hTR) component but not the catalytic component, hTERT. We also show that telomerase activity could be reconstituted using either lipid-mediated transfection of the hTERT plasmid DNA or transduction with an E1-, E3-deleted adenoviral vector containing the hTERT gene. However, relative to plasmid transfection, adenoviral gene transfer produced higher levels of hTERT expression. Nine of 10 AdhTERT-transduced DCs were able to generate CTL responses, while only three of nine plasmid-transfected DCs did. CTLs primed against hTERT exhibited killing of telomerase positive but not telomerase negative tumor lines of diverse tissue origins. Antigenic specificity of these T cells to telomerase was further determined by introducing hTERT gene into a telomerase negative cell line, U2OS, by adenoviral transduction. Although some antigenic specificity was directed against adenoviral epitopes, the majority of CTLs were targeted against telomerase-derived antigen(s). Thus, the hTERT gene, particularly as delivered via the recombinant adenovirus, may be useful as vaccine to induce specific T-cell-mediated tumor immunity in cancer patients. In addition, our results suggest that telomerase activity and/or telomerase expression after hTERT gene transfer have a predictive value in the success of hTERT/DC-based cancer vaccination.


Assuntos
Células Dendríticas/imunologia , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Telomerase/genética , Adenoviridae/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Imunofluorescência , Vetores Genéticos , Humanos , Neoplasias/terapia , Telomerase/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA