Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Clin Invest ; 126(9): 3363-76, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27482888

RESUMO

BACKGROUND: T cells expressing antigen-specific chimeric antigen receptors (CARs) improve outcomes for CD19-expressing B cell malignancies. We evaluated a human application of T cells that were genetically modified using the Sleeping Beauty (SB) transposon/transposase system to express a CD19-specific CAR. METHODS: T cells were genetically modified using DNA plasmids from the SB platform to stably express a second-generation CD19-specific CAR and selectively propagated ex vivo with activating and propagating cells (AaPCs) and cytokines. Twenty-six patients with advanced non-Hodgkin lymphoma and acute lymphoblastic leukemia safely underwent hematopoietic stem cell transplantation (HSCT) and infusion of CAR T cells as adjuvant therapy in the autologous (n = 7) or allogeneic settings (n = 19). RESULTS: SB-mediated genetic transposition and stimulation resulted in 2,200- to 2,500-fold ex vivo expansion of genetically modified T cells, with 84% CAR expression, and without integration hotspots. Following autologous HSCT, the 30-month progression-free and overall survivals were 83% and 100%, respectively. After allogeneic HSCT, the respective 12-month rates were 53% and 63%. No acute or late toxicities and no exacerbation of graft-versus-host disease were observed. Despite a low antigen burden and unsupportive recipient cytokine environment, CAR T cells persisted for an average of 201 days for autologous recipients and 51 days for allogeneic recipients. CONCLUSIONS: CD19-specific CAR T cells generated with SB and AaPC platforms were safe, and may provide additional cancer control as planned infusions after HSCT. These results support further clinical development of this nonviral gene therapy approach. TRIAL REGISTRATION: Autologous, NCT00968760; allogeneic, NCT01497184; long-term follow-up, NCT01492036. FUNDING: National Cancer Institute, private foundations, and institutional funds. Please see Acknowledgments for details.


Assuntos
Antígenos CD19/metabolismo , Elementos de DNA Transponíveis , Linfoma não Hodgkin/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Linfócitos T/citologia , Adulto , Células Apresentadoras de Antígenos/imunologia , Citocinas/metabolismo , Intervalo Livre de Doença , Feminino , Seguimentos , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunoterapia Adotiva/métodos , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Segurança do Paciente , Plasmídeos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transplante Homólogo , Resultado do Tratamento , Adulto Jovem
2.
J Clin Invest ; 126(6): 2341-55, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27183388

RESUMO

Vα24-invariant natural killer T cells (NKTs) localize to tumors and have inherent antitumor properties, making them attractive chimeric antigen receptor (CAR) carriers for redirected cancer immunotherapy. However, clinical application of CAR-NKTs has been impeded, as mechanisms responsible for NKT expansion and the in vivo persistence of these cells are unknown. Here, we demonstrated that antigen-induced expansion of primary NKTs in vitro associates with the accumulation of a CD62L+ subset and exhaustion of CD62L- cells. Only CD62L+ NKTs survived and proliferated in response to secondary stimulation. When transferred to immune-deficient NSG mice, CD62L+ NKTs persisted 5 times longer than CD62L- NKTs. Moreover, CD62L+ cells transduced with a CD19-specific CAR achieved sustained tumor regression in a B cell lymphoma model. Proliferating CD62L+ cells downregulated or maintained CD62L expression when activated via T cell receptor alone or in combination with costimulatory receptors. We generated HLAnull K562 cell clones that were engineered to express CD1d and costimulatory ligands. Clone B-8-2 (HLAnullCD1dmedCD86high4-1BBLmedOX40Lhigh) induced the highest rates of NKT expansion and CD62L expression. B-8-2-expanded CAR-NKTs exhibited prolonged in vivo persistence and superior therapeutic activities in models of lymphoma and neuroblastoma. Therefore, we have identified CD62L as a marker of a distinct NKT subset endowed with high proliferative potential and have developed artificial antigen-presenting cells that generate CD62L-enriched NKTs for effective cancer immunotherapy.


Assuntos
Selectina L/metabolismo , Células T Matadoras Naturais/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Citocinas/biossíntese , Citotoxicidade Imunológica , Humanos , Imunoterapia Adotiva , Ativação Linfocitária , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células T Matadoras Naturais/classificação , Neuroblastoma/imunologia , Neuroblastoma/terapia , Receptores de Antígenos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Immunother ; 37(4): 204-13, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24714354

RESUMO

T cells genetically modified to stably express immunoreceptors are being assessed for therapeutic potential in clinical trials. T cells expressing a chimeric antigen receptor (CAR) are endowed with a new specificity to target tumor-associated antigen (TAA) independent of major histocompatibility complex. Our approach to nonviral gene transfer in T cells uses ex vivo numeric expansion of CAR T cells on irradiated artificial antigen presenting cells (aAPC) bearing the targeted TAA. The requirement for aAPC to express a desired TAA limits the human application of CARs with multiple specificities when selective expansion through coculture with feeder cells is sought. As an alternative to expressing individual TAAs on aAPC, we expressed 1 ligand that could activate CAR T cells for sustained proliferation independent of specificity. We expressed a CAR ligand (designated CARL) that binds the conserved IgG4 extracellular domain of CAR and demonstrated that CARL aAPC propagate CAR T cells of multiple specificities. CARL avoids technical issues and costs associated with deploying clinical-grade aAPC for each TAA targeted by a given CAR. Using CARL enables 1 aAPC to numerically expand all CAR T cells containing the IgG4 domain, and simplifies expansion, testing, and clinical translation of CAR T cells of any specificity.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Receptores de Antígenos de Linfócitos T/genética , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD19/genética , Antígenos CD19/metabolismo , Linhagem Celular Tumoral , Humanos , Imunoterapia Adotiva , Células K562 , Camundongos , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
4.
Curr Hematol Malig Rep ; 9(1): 50-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24488441

RESUMO

T cells that have been genetically modified, activated, and propagated ex vivo can be infused to control tumor progression in patients who are refractory to conventional treatments. Early-phase clinical trials demonstrate that the tumor-associated antigen (TAA) CD19 can be therapeutically engaged through the enforced expression of a chimeric antigen receptor (CAR) on clinical-grade T cells. Advances in vector design, the architecture of the CAR molecule especially as associated with T-cell co-stimulatory pathways, and understanding of the tumor microenvironment, play significant roles in the successful treatment of medically fragile patients. However, some recipients of CAR(+) T cells demonstrate incomplete responses. Understanding the potential for treatment failure provides a pathway to improve the potency of adoptive transfer of CAR(+) T cells. High throughput single-cell analyses to understand the complexity of the inoculum coupled with animal models may provide insight into the therapeutic potential of genetically modified T cells. This review focuses on recent advances regarding the human application of CD19-specific CAR(+) T cells and explores how their success for hematologic cancers can provide a framework for investigational treatment of solid tumor malignancies.


Assuntos
Neoplasias Hematológicas/terapia , Imunoterapia/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Antígenos CD19/imunologia , Ensaios Clínicos como Assunto , Engenharia Genética , Neoplasias Hematológicas/imunologia , Humanos
5.
Blood ; 122(25): 4129-39, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24055823

RESUMO

New treatments are needed for B-cell malignancies persisting after allogeneic hematopoietic stem cell transplantation (alloHSCT). We conducted a clinical trial of allogeneic T cells genetically modified to express a chimeric antigen receptor (CAR) targeting the B-cell antigen CD19. T cells for genetic modification were obtained from each patient's alloHSCT donor. All patients had malignancy that persisted after alloHSCT and standard donor lymphocyte infusions (DLIs). Patients did not receive chemotherapy prior to the CAR T-cell infusions and were not lymphocyte depleted at the time of the infusions. The 10 treated patients received a single infusion of allogeneic anti-CD19-CAR T cells. Three patients had regressions of their malignancies. One patient with chronic lymphocytic leukemia (CLL) obtained an ongoing complete remission after treatment with allogeneic anti-CD19-CAR T cells, another CLL patient had tumor lysis syndrome as his leukemia dramatically regressed, and a patient with mantle cell lymphoma obtained an ongoing partial remission. None of the 10 patients developed graft-versus-host disease (GVHD). Toxicities included transient hypotension and fever. We detected cells containing the anti-CD19-CAR gene in the blood of 8 of 10 patients. These results show for the first time that donor-derived allogeneic anti-CD19-CAR T cells can cause regression of B-cell malignancies resistant to standard DLIs without causing GVHD.


Assuntos
Antígenos CD19 , Transfusão de Linfócitos , Linfoma de Células B/terapia , Receptores de Antígenos de Linfócitos T/biossíntese , Transplante de Células-Tronco , Linfócitos T/metabolismo , Linfócitos T/transplante , Adulto , Idoso , Aloenxertos , Feminino , Humanos , Linfoma de Células B/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/biossíntese , Síndrome de Lise Tumoral/etiologia , Síndrome de Lise Tumoral/terapia
6.
PLoS One ; 8(5): e64138, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23741305

RESUMO

Adoptive transfer of T cells expressing a CD19-specific chimeric antigen receptor (CAR) is being evaluated in multiple clinical trials. Our current approach to adoptive immunotherapy is based on a second generation CAR (designated CD19RCD28) that signals through a CD28 and CD3-ζ endodomain. T cells are electroporated with DNA plasmids from the Sleeping Beauty (SB) transposon/transposase system to express this CAR. Stable integrants of genetically modified T cells can then be retrieved when co-cultured with designer artificial antigen presenting cells (aAPC) in the presence of interleukin (IL)-2 and 21. Here, we reveal how the platform technologies of SB-mediated transposition and CAR-dependent propagation on aAPC were adapted for human application. Indeed, we have initiated clinical trials in patients with high-risk B-lineage malignancies undergoing autologous and allogeneic hematopoietic stem-cell transplantation (HSCT). We describe the process to manufacture clinical grade CD19-specific T cells derived from healthy donors. Three validation runs were completed in compliance with current good manufacturing practice for Phase I/II trials demonstrating that by 28 days of co-culture on γ-irradiated aAPC ∼10(10) T cells were produced of which >95% expressed CAR. These genetically modified and propagated T cells met all quality control testing and release criteria in support of infusion.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Antígenos CD19/genética , Elementos de DNA Transponíveis/genética , Leucemia de Células B/terapia , Linfócitos T/citologia , Linfócitos T/metabolismo , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Antígenos CD19/imunologia , Antígenos CD28/genética , Antígenos CD28/imunologia , Complexo CD3/genética , Complexo CD3/imunologia , Proliferação de Células , Ensaios Clínicos como Assunto , Técnicas de Cocultura , Elementos de DNA Transponíveis/imunologia , Eletroporação , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunoterapia Adotiva/métodos , Interleucina-2/genética , Interleucina-2/imunologia , Células K562 , Leucemia de Células B/imunologia , Leucemia de Células B/patologia , Ativação Linfocitária , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante , Transposases/genética , Transposases/imunologia
7.
PLoS One ; 8(3): e57838, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469246

RESUMO

Clinical trials targeting CD19 on B-cell malignancies are underway with encouraging anti-tumor responses. Most infuse T cells genetically modified to express a chimeric antigen receptor (CAR) with specificity derived from the scFv region of a CD19-specific mouse monoclonal antibody (mAb, clone FMC63). We describe a novel anti-idiotype monoclonal antibody (mAb) to detect CD19-specific CAR(+) T cells before and after their adoptive transfer. This mouse mAb was generated by immunizing with a cellular vaccine expressing the antigen-recognition domain of FMC63. The specificity of the mAb (clone no. 136.20.1) was confined to the scFv region of the CAR as validated by inhibiting CAR-dependent lysis of CD19(+) tumor targets. This clone can be used to detect CD19-specific CAR(+) T cells in peripheral blood mononuclear cells at a sensitivity of 1∶1,000. In clinical settings the mAb is used to inform on the immunophenotype and persistence of administered CD19-specific T cells. Thus, our CD19-specific CAR mAb (clone no. 136.20.1) will be useful to investigators implementing CD19-specific CAR(+) T cells to treat B-lineage malignancies. The methodology described to develop a CAR-specific anti-idiotypic mAb could be extended to other gene therapy trials targeting different tumor associated antigens in the context of CAR-based adoptive T-cell therapy.


Assuntos
Anticorpos Monoclonais/biossíntese , Antígenos CD19/imunologia , Proteínas Mutantes Quiméricas/análise , Receptores de Antígenos de Linfócitos T/análise , Anticorpos de Cadeia Única/biossíntese , Linfócitos T/imunologia , Transferência Adotiva , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Antígenos CD19/genética , Ensaios Clínicos como Assunto , Humanos , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Linfócitos T/citologia , Linfócitos T/transplante
8.
Curr Hematol Malig Rep ; 8(1): 60-70, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23225251

RESUMO

Adoptive transfer of antigen-specific T cells has been adapted by investigators for treatment of chronic lymphocytic leukemia (CLL). To overcome issues of immune tolerance which limits the endogenous adaptive immune response to tumor-associated antigens (TAAs), robust systems for the genetic modification and characterization of T cells expressing chimeric antigen receptors (CARs) to redirect specificity have been produced. Refinements with regards to persistence and trafficking of the genetically modified T cells are underway to help improve potency. Clinical trials utilizing this technology demonstrate feasibility, and increasingly, these early-phase trials are demonstrating impressive anti-tumor effects, particularly for CLL patients, paving the way for multi-center trials to establish the efficacy of CAR(+) T cell therapy.


Assuntos
Imunoterapia Adotiva/métodos , Leucemia Linfocítica Crônica de Células B/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Antígenos de Neoplasias/imunologia , Ensaios Clínicos como Assunto , Humanos , Leucemia Linfocítica Crônica de Células B/imunologia , Proteínas Mutantes Quiméricas/imunologia , Proteínas Mutantes Quiméricas/metabolismo , Receptores de Antígenos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/transplante
9.
Blood ; 119(12): 2700-2, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22442331
10.
Hum Gene Ther ; 23(5): 444-50, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22107246

RESUMO

Limited curative treatment options exist for patients with advanced B-lymphoid malignancies, and new therapeutic approaches are needed to augment the efficacy of hematopoietic stem-cell transplantation (HSCT). Cellular therapies, such as adoptive transfer of T cells that are being evaluated to target malignant disease, use mechanisms independent of chemo- and radiotherapy with nonoverlapping toxicities. Gene therapy is employed to generate tumor-specific T cells, as specificity can be redirected through enforced expression of a chimeric antigen receptor (CAR) to achieve antigen recognition based on the specificity of a monoclonal antibody. By combining cell and gene therapies, we have opened a new Phase I protocol at the MD Anderson Cancer Center (Houston, TX) to examine the safety and feasibility of administering autologous genetically modified T cells expressing a CD19-specific CAR (capable of signaling through chimeric CD28 and CD3-ζ) into patients with high-risk B-lymphoid malignancies undergoing autologous HSCT. The T cells are genetically modified by nonviral gene transfer of the Sleeping Beauty system and CAR(+) T cells selectively propagated in a CAR-dependent manner on designer artificial antigen-presenting cells. The results of this study will lay the foundation for future protocols including CAR(+) T-cell infusions derived from allogeneic sources.


Assuntos
Antígenos CD19/imunologia , Transplante de Células-Tronco Hematopoéticas , Imunoterapia Adotiva/métodos , Linfoma não Hodgkin/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/transplante , Adolescente , Adulto , Células Apresentadoras de Antígenos/imunologia , Linfócitos B/imunologia , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Células Cultivadas , Humanos , Ativação Linfocitária/imunologia , Linfoma não Hodgkin/imunologia , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia
11.
Front Biosci (Schol Ed) ; 4(2): 520-31, 2012 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-22202074

RESUMO

Adoptive transfer of antigen-specific T cells is a compelling tool to treat cancer. To overcome issues of immune tolerance which limits the endogenous adaptive immune response to tumor-associated antigens, robust systems for the genetic modification and characterization of T cells expressing chimeric antigen receptors (CARs) to redirect specificity have been produced. Refinements with regards to persistence and trafficking of the genetically modified T cells are underway to help improve the potency of genetically modified T cells. Clinical trials utilizing this technology demonstrate feasibility, and increasingly, antitumor activity, paving the way for multi-center trials to establish the efficacy of this novel T-cell therapy.


Assuntos
Neoplasias/imunologia , Receptores de Antígenos/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T/imunologia , Antígenos de Neoplasias/imunologia , Humanos , Tolerância Imunológica/imunologia , Imunoterapia Adotiva , Neoplasias/terapia
12.
Blood ; 116(7): 1035-44, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20439624

RESUMO

Infusions of antigen-specific T cells have yielded therapeutic responses in patients with pathogens and tumors. To broaden the clinical application of adoptive immunotherapy against malignancies, investigators have developed robust systems for the genetic modification and characterization of T cells expressing introduced chimeric antigen receptors (CARs) to redirect specificity. Human trials are under way in patients with aggressive malignancies to test the hypothesis that manipulating the recipient and reprogramming T cells before adoptive transfer may improve their therapeutic effect. These examples of personalized medicine infuse T cells designed to meet patients' needs by redirecting their specificity to target molecular determinants on the underlying malignancy. The generation of clinical grade CAR(+) T cells is an example of bench-to-bedside translational science that has been accomplished using investigator-initiated trials operating largely without industry support. The next-generation trials will deliver designer T cells with improved homing, CAR-mediated signaling, and replicative potential, as investigators move from the bedside to the bench and back again.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T/imunologia , Animais , Humanos , Receptores de Antígenos de Linfócitos T/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...