Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
PLoS One ; 15(2): e0228112, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32040512

RESUMO

Neoantigens can be predicted and in some cases identified using the data obtained from the whole exome sequencing and transcriptome sequencing of tumor cells. These sequencing data can be coupled with single-cell RNA sequencing for the direct interrogation of the transcriptome, surfaceome, and pairing of αß T-cell receptors (TCRαß) from hundreds of single T cells. Using these 2 large datasets, we established a platform for identifying antigens recognized by TCRαßs obtained from single T cells. Our approach is based on the rapid expression of cloned TCRαß genes as Sleeping Beauty transposons and the determination of the introduced TCRαßs' antigen specificity and avidity using a reporter cell line. The platform enables the very rapid identification of tumor-reactive TCRs for the bioengineering of T cells with redirected specificity.


Assuntos
Engenharia Celular/métodos , Clonagem Molecular/métodos , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/citologia , Linfócitos T/metabolismo , Expressão Gênica , Biblioteca Gênica , Genes MHC Classe I/genética , Genes MHC da Classe II/genética , Células HEK293 , Humanos , Cinética , Receptores de Antígenos de Linfócitos T alfa-beta/genética
2.
Oncoimmunology ; 5(11): e1232220, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27999743

RESUMO

The B-cell receptor (BCR) expressed by a clonal B cell tumor is a tumor specific antigen (idiotype). However, the T-cell epitopes within human BCRs which stimulate protective immunity still lack detailed characterization. In this study, we identified 17 BCR peptide-specific CD4+ T-cell epitopes derived from BCR heavy and light chain variable region sequences. Detailed analysis revealed these CD4+ T-cell epitopes stimulated normal donors' and patients' Th1 CD4+ T cells to directly recognize the autologous tumors by secretion of IFNγ, indicating the epitopes are processed and presented by tumor cells. One BCR peptide-specific CD4+ T cell line was also cytotoxic and lysed autologous tumor cells through the perforin pathway. Sequence analysis of the epitopes revealed that 10 were shared by multiple primary patients' tumors, and 16 had the capacity to bind to more than one HLA DRB1 allele. T cells stimulated by shared epitopes recognized primary tumors expressing the same sequences on multiple HLA DRB1 alleles. In conclusion, we identified 17 BCR-derived CD4+ T-cell epitopes with promiscuous HLA DRB1 binding affinity that are shared by up to 36% of patients, suggesting a strategy to overcome the requirement for individual preparation of therapeutic agents targeting idiotype.

3.
PLoS One ; 11(8): e0159477, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27548616

RESUMO

Adoptive immunotherapy infusing T cells with engineered specificity for CD19 expressed on B- cell malignancies is generating enthusiasm to extend this approach to other hematological malignancies, such as acute myelogenous leukemia (AML). CD123, or interleukin 3 receptor alpha, is overexpressed on most AML and some lymphoid malignancies, such as acute lymphocytic leukemia (ALL), and has been an effective target for T cells expressing chimeric antigen receptors (CARs). The prototypical CAR encodes a VH and VL from one monoclonal antibody (mAb), coupled to a transmembrane domain and one or more cytoplasmic signaling domains. Previous studies showed that treatment of an experimental AML model with CD123-specific CAR T cells was therapeutic, but at the cost of impaired myelopoiesis, highlighting the need for systems to define the antigen threshold for CAR recognition. Here, we show that CARs can be engineered using VH and VL chains derived from different CD123-specific mAbs to generate a panel of CAR+ T cells. While all CARs exhibited specificity to CD123, one VH and VL combination had reduced lysis of normal hematopoietic stem cells. This CAR's in vivo anti-tumor activity was similar whether signaling occurred via chimeric CD28 or CD137, prolonging survival in both AML and ALL models. Co-expression of inducible caspase 9 eliminated CAR+ T cells. These data help support the use of CD123-specific CARs for treatment of CD123+ hematologic malignancies.


Assuntos
Engenharia Genética/métodos , Imunoterapia Adotiva/métodos , Subunidade alfa de Receptor de Interleucina-3/imunologia , Leucemia Mieloide Aguda/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/transplante , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Antígenos CD28/genética , Antígenos CD28/imunologia , Caspase 9/genética , Caspase 9/imunologia , Citotoxicidade Imunológica , Modelos Animais de Doenças , Expressão Gênica , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Humanos , Subunidade alfa de Receptor de Interleucina-3/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Terapia de Alvo Molecular , Plasmídeos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Recombinantes de Fusão/genética , Anticorpos de Domínio Único/genética , Linfócitos T/citologia , Linfócitos T/imunologia , Transfecção , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
4.
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
5.
J Immunother ; 39(5): 205-17, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27163741

RESUMO

Potential for on-target, but off-tissue toxicity limits therapeutic application of genetically modified T cells constitutively expressing chimeric antigen receptors (CARs) from tumor-associated antigens expressed in normal tissue, such as epidermal growth factor receptor (EGFR). Curtailing expression of CAR through modification of T cells by in vitro-transcribed mRNA species is one strategy to mitigate such toxicity. We evaluated expression of an EGFR-specific CAR coded from introduced mRNA in human T cells numerically expanded ex vivo to clinically significant numbers through coculture with activating and propagating cells (AaPC) derived from K562 preloaded with anti-CD3 antibody. The density of AaPC could be adjusted to affect phenotype of T cells such that reduced ratio of AaPC resulted in higher proportion of CD8 and central memory T cells that were more conducive to electrotransfer of mRNA than T cells expanded with high ratios of AaPC. RNA-modified CAR T cells produced less cytokine, but demonstrated similar cytolytic capacity as DNA-modified CAR T cells in response to EGFR-expressing glioblastoma cells. Expression of CAR by mRNA transfer was transient and accelerated by stimulation with cytokine and antigen. Loss of CAR abrogated T-cell function in response to tumor and normal cells expressing EGFR. We describe a clinically applicable method to propagate and modify T cells to transiently express EGFR-specific CAR to target EGFR-expressing tumor cells that may be used to limit on-target, off-tissue toxicity to normal tissue.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Vacinas Anticâncer/imunologia , Glioblastoma/terapia , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/genética , Subpopulações de Linfócitos T/fisiologia , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Receptores ErbB/imunologia , Engenharia Genética , Glioblastoma/imunologia , Humanos , Memória Imunológica , Ativação Linfocitária , RNA Mensageiro/genética , Especificidade do Receptor de Antígeno de Linfócitos T
6.
Mol Ther ; 24(7): 1178-86, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27203439

RESUMO

Chimeric antigen receptor (CAR) endows specificity to T-cells independent of human leukocyte antigen (HLA). This enables one immunoreceptor to directly target the same surface antigen on different subsets of tumor cells from multiple HLA-disparate recipients. Most approaches manufacture individualized CAR(+)T-cells from the recipient or HLA-compatible donor, which are revealing promising clinical results. This is the impetus to broaden the number of patients eligible to benefit from adoptive immunotherapy such as to infuse third-party donor derived CAR(+)T-cells. This will overcome issues associated with (i) time to manufacture T-cells, (ii) cost to generate one product for one patient, (iii) inability to generate a product from lymphopenic patients or patient's immune cells fail to complete the manufacturing process, and (iv) heterogeneity of T-cell products produced for or from individual recipients. Establishing a biobank of allogeneic genetically modified immune cells from healthy third-party donors, which are cryopreserved and validated in advance of administration, will facilitate the centralizing manufacturing and widespread distribution of CAR(+)T-cells to multiple points-of-care in a timely manner. To achieve this, it is necessary to engineer an effective strategy to avoid deleterious allogeneic immune responses leading to toxicity and rejection. We review the strategies to establish "off-the-shelf" donor-derived biobanks for human application of CAR(+)T-cells as a drug.


Assuntos
Expressão Gênica , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão , Linfócitos T/imunologia , Linfócitos T/metabolismo , Pesquisa Translacional Biomédica , Animais , Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Neoplasias/terapia
7.
Sci Rep ; 6: 21757, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26902653

RESUMO

Mismatch of human leukocyte antigens (HLA) adversely impacts the outcome of patients after allogeneic hematopoietic stem-cell transplantation (alloHSCT). This translates into the clinical requirement to timely identify suitable HLA-matched donors which in turn curtails the chances of recipients, especially those from a racial minority, to successfully undergo alloHSCT. We thus sought to broaden the existing pool of registered unrelated donors based on analysis that eliminating the expression of the HLA-A increases the chance for finding a donor matched at HLA-B, -C, and -DRB1 regardless of a patient's race. Elimination of HLA-A expression in HSC was achieved using artificial zinc finger nucleases designed to target HLA-A alleles. Significantly, these engineered HSCs maintain their ability to engraft and reconstitute hematopoiesis in immunocompromised mice. This introduced loss of HLA-A expression decreases the need to recruit large number of donors to match with potential recipients and has particular importance for patients whose HLA repertoire is under-represented in the current donor pool. Furthermore, the genetic engineering of stem cells provides a translational approach to HLA-match a limited number of third-party donors with a wide number of recipients.


Assuntos
Desoxirribonucleases/genética , Deleção de Genes , Antígenos HLA-A/genética , Transplante de Células-Tronco Hematopoéticas/etnologia , Células-Tronco Hematopoéticas/imunologia , Alelos , Animais , Desoxirribonucleases/metabolismo , Seleção do Doador/ética , Expressão Gênica , Engenharia Genética/métodos , Antígenos HLA-A/imunologia , Antígenos HLA-B/genética , Antígenos HLA-B/imunologia , Antígenos HLA-C/genética , Antígenos HLA-C/imunologia , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Acessibilidade aos Serviços de Saúde/ética , Transplante de Células-Tronco Hematopoéticas/ética , Células-Tronco Hematopoéticas/citologia , Teste de Histocompatibilidade , Humanos , Camundongos , Grupos Raciais , Transplante Heterólogo , Transplante Homólogo , Doadores não Relacionados , Dedos de Zinco
8.
Blood ; 122(8): 1341-9, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23741009

RESUMO

Long-term engraftment of allogeneic cells necessitates eluding immune-mediated rejection, which is currently achieved by matching for human leukocyte antigen (HLA) expression, immunosuppression, and/or delivery of donor-derived cells to sanctuary sites. Genetic engineering provides an alternative approach to avoid clearance of cells that are recognized as "non-self" by the recipient. To this end, we developed designer zinc finger nucleases and employed a "hit-and-run" approach to genetic editing for selective elimination of HLA expression. Electro-transfer of mRNA species coding for these engineered nucleases completely disrupted expression of HLA-A on human T cells, including CD19-specific T cells. The HLA-A(neg) T-cell pools can be enriched and evade lysis by HLA-restricted cytotoxic T-cell clones. Recognition by natural killer cells of cells that had lost HLA expression was circumvented by enforced expression of nonclassical HLA molecules. Furthermore, we demonstrate that zinc finger nucleases can eliminate HLA-A expression from embryonic stem cells, which broadens the applicability of this strategy beyond infusing HLA-disparate immune cells. These findings establish that clinically appealing cell types derived from donors with disparate HLA expression can be genetically edited to evade an immune response and provide a foundation whereby cells from a single donor can be administered to multiple recipients.


Assuntos
Desoxirribonucleases/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Transplante de Células-Tronco/métodos , Transplante Homólogo , Antígenos CD19/metabolismo , Sequência de Bases , Diferenciação Celular , Citotoxicidade Imunológica/imunologia , Eletroporação , Células-Tronco Embrionárias/citologia , Técnicas de Transferência de Genes , Células HEK293 , Humanos , Leucócitos Mononucleares/citologia , Dados de Sequência Molecular , Engenharia de Proteínas , Linfócitos T/imunologia , Dedos de Zinco
9.
J Immunother ; 36(2): 112-23, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23377665

RESUMO

The Sleeping Beauty (SB) transposon/transposase DNA plasmid system is used to genetically modify cells for long-term transgene expression. We adapted the SB system for human application and generated T cells expressing a chimeric antigen receptor (CAR) specific for CD19. Electrotransfer of CD19-specific SB DNA plasmids in peripheral blood mononuclear cells and propagation on CD19 artificial antigen presenting cells was used to numerically expand CD3 T cells expressing CAR. By day 28 of coculture, >90% of expanded CD3 T cells expressed CAR. CAR T cells specifically killed CD19 target cells and consisted of subsets expressing biomarkers consistent with central memory, effector memory, and effector phenotypes. CAR T cells contracted numerically in the absence of the CD19 antigen, did not express SB11 transposase, and maintained a polyclonal TCR Vα and TCR Vß repertoire. Quantitative fluorescence in situ hybridization revealed that CAR T cells preserved the telomere length. Quantitative polymerase chain reaction and fluorescence in situ hybridization showed CAR transposon integrated on average once per T-cell genome. CAR T cells in peripheral blood can be detected by quantitative polymerase chain reaction at a sensitivity of 0.01%. These findings lay the groundwork as the basis of our first-in-human clinical trials of the nonviral SB system for the investigational treatment of CD19 B-cell malignancies (currently under 3 INDs: 14193, 14577, and 14739).


Assuntos
Antígenos CD19/imunologia , Imunoterapia Adotiva , Linfócitos T/transplante , Transposases/genética , Animais , Antígenos CD19/genética , Antígenos CD19/metabolismo , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Eletroporação , Técnicas de Transferência de Genes , Glioblastoma/imunologia , Glioblastoma/terapia , Humanos , Camundongos , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão/biossíntese
10.
PLoS One ; 7(10): e47126, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071732

RESUMO

The detailed mechanisms responsible for processing tumor-associated antigens and presenting them to CTLs remain to be fully elucidated. In this study, we demonstrate a unique CTL epitope generated from the ubiquitous protein puromycin-sensitive aminopeptidase, which is presented via HLA-A24 on leukemic and pancreatic cancer cells but not on normal fibroblasts or EBV-transformed B lymphoblastoid cells. The generation of this epitope requires proteasomal digestion and transportation from the endoplasmic reticulum to the Golgi apparatus and is sensitive to chloroquine-induced inhibition of acidification inside the endosome/lysosome. Epitope liberation depends on constitutively active autophagy, as confirmed with immunocytochemistry for the autophagosome marker LC3 as well as RNA interference targeting two different autophagy-related genes. Therefore, ubiquitously expressed proteins may be sources of specific tumor-associated antigens when processed through a unique mechanism involving autophagy.


Assuntos
Autofagia/imunologia , Epitopos/imunologia , Antígeno HLA-A24/imunologia , Linfócitos T Citotóxicos/imunologia , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Aminopeptidases/genética , Aminopeptidases/metabolismo , Antígenos de Neoplasias/imunologia , Autofagia/genética , Linhagem Celular Tumoral , Epitopos/efeitos dos fármacos , Epitopos/genética , Fibroblastos/imunologia , Antígeno HLA-A24/genética , Humanos , Leucemia/imunologia , Leucemia/patologia , Mimetismo Molecular , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Interferência de RNA , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Vacúolos/metabolismo
11.
Blood ; 119(24): 5697-705, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22535661

RESUMO

Clinical-grade T cells are genetically modified ex vivo to express a chimeric antigen receptor (CAR) to redirect specificity to a tumor associated antigen (TAA) thereby conferring antitumor activity in vivo. T cells expressing a CD19-specific CAR recognize B-cell malignancies in multiple recipients independent of major histocompatibility complex (MHC) because the specificity domains are cloned from the variable chains of a CD19 monoclonal antibody. We now report a major step toward eliminating the need to generate patient-specific T cells by generating universal allogeneic TAA-specific T cells from one donor that might be administered to multiple recipients. This was achieved by genetically editing CD19-specific CAR(+) T cells to eliminate expression of the endogenous αß T-cell receptor (TCR) to prevent a graft-versus-host response without compromising CAR-dependent effector functions. Genetically modified T cells were generated using the Sleeping Beauty system to stably introduce the CD19-specific CAR with subsequent permanent deletion of α or ß TCR chains with designer zinc finger nucleases. We show that these engineered T cells display the expected property of having redirected specificity for CD19 without responding to TCR stimulation. CAR(+)TCR(neg) T cells of this type may potentially have efficacy as an off-the-shelf therapy for investigational treatment of B-lineage malignancies.


Assuntos
Antígenos CD19/imunologia , Epitopos/imunologia , Engenharia Genética , Imunoterapia/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes/imunologia , Linfócitos T/imunologia , Adulto , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/imunologia , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Células Cultivadas , Endonucleases/metabolismo , Técnicas de Inativação de Genes , Humanos , Células K562 , Ativação Linfocitária/imunologia , Dedos de Zinco
12.
Cancer Sci ; 102(7): 1281-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21466613

RESUMO

Partial human leukocyte antigen (HLA)-mismatched hematopoietic stem cell transplantation (HSCT) is often performed when an HLA-matched donor is not available. In these cases, CD8(+) or CD4(+) T cell responses are induced depending on the mismatched HLA class I or II allele(s). Herein, we report on an HLA-DRB1*08:03-restricted CD8(+) CTL clone, named CTL-1H8, isolated from a patient following an HLA-DR-mismatched HSCT from his brother. Lysis of a patient Epstein-Barr virus-transformed B cell line (B-LCL) by CTL-1H8 was inhibited after the addition of blocking antibodies against HLA-DR and CD8, whereas antibodies against pan-HLA class I or CD4 had no effect. The 1H8-CTL clone did not lyse the recipient dermal fibroblasts whose HLA-DRB1*08:03 expression was upregulated after 1 week cytokine treatment. Engraftment of HLA-DRB1*08:03-positive primary leukemic stem cells in non-obese diabetic/severe combined immunodeficient/γc-null (NOG) mice was completely inhibited by the in vitro preincubation of cells with CTL-1H8, suggesting that HLA-DRB1*08:03 is expressed on leukemic stem cells. Finally, analysis of the precursor frequency of CD8(+) CTL specific for recipient antigens in post-HSCT peripheral blood T cells revealed a significant fraction of the total donor CTL responses towards the individual mismatched HLA-DR antigen in two patients. These findings underscore unexpectedly significant CD8 T cell responses in the context of HLA class II.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Efeito Enxerto vs Leucemia/imunologia , Antígenos HLA-D/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Teste de Histocompatibilidade , Animais , Doença Enxerto-Hospedeiro/etiologia , Antígenos HLA-DR/imunologia , Subtipos Sorológicos de HLA-DR , Cadeias HLA-DRB1 , Humanos , Masculino , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/cirurgia , Transplante Homólogo
13.
Biomed Microdevices ; 12(5): 855-63, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20574820

RESUMO

It has been demonstrated that a chimeric antigen receptor (CAR) can directly recognize the CD19 molecule expressed on the cell surface of B-cell malignancies independent of major histocompatibility complex (MHC). Although T-cell therapy of tumors using CD19-specific CAR is promising, this approach relies on using expression vectors that stably integrate the CAR into T-cell chromosomes. To circumvent the potential genotoxicity that may occur from expressing integrating transgenes, we have expressed the CD19-specific CAR transgene from mRNA using a high throughput microelectroporation device. This research was accomplished using a microelectroporator to achieve efficient and high throughput non-viral gene transfer of in vitro transcribed CAR mRNA into human T cells that had been numerically expanded ex vivo. Electro-transfer of mRNA avoids the potential genotoxicity associated with vector and transgene integration and the high throughput capacity overcomes the expected transient CAR expression, as repeated rounds of electroporation can replace T cells that have lost transgene expression. We fabricated and tested a high throughput microelectroporator that can electroporate a stream of 2 x 10(8) primary T cells within 10 min. After electroporation, up to 80% of the passaged T cells expressed the CD19-specific CAR. Video time-lapse microscopy (VTLM) demonstrated the redirected effector function of the genetically manipulated T cells to specifically lyse CD19+ tumor cells. Our biomedical microdevice, in which T cells are transiently and safely modified to be tumor-specific and then can be re-infused, offers a method for redirecting T-cell specificity, that has implications for the development of adoptive immunotherapy.


Assuntos
Eletroporação/instrumentação , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/imunologia , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Antígenos CD19/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Humanos , RNA Mensageiro/genética , Receptores de Antígenos/genética , Receptores de Antígenos/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/citologia
14.
Blood ; 113(21): 5041-8, 2009 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-18809759

RESUMO

Minor histocompatibility antigens (mHags) are molecular targets of allo-immunity associated with hematopoietic stem cell transplantation (HSCT) and involved in graft-versus-host disease, but they also have beneficial antitumor activity. mHags are typically defined by host SNPs that are not shared by the donor and are immunologically recognized by cytotoxic T cells isolated from post-HSCT patients. However, the number of molecularly identified mHags is still too small to allow prospective studies of their clinical importance in transplantation medicine, mostly due to the lack of an efficient method for isolation. Here we show that when combined with conventional immunologic assays, the large data set from the International HapMap Project can be directly used for genetic mapping of novel mHags. Based on the immunologically determined mHag status in HapMap panels, a target mHag locus can be uniquely mapped through whole genome association scanning taking advantage of the unprecedented resolution and power obtained with more than 3 000 000 markers. The feasibility of our approach could be supported by extensive simulations and further confirmed by actually isolating 2 novel mHags as well as 1 previously identified example. The HapMap data set represents an invaluable resource for investigating human variation, with obvious applications in genetic mapping of clinically relevant human traits.


Assuntos
Mapeamento Cromossômico/métodos , Antígenos de Histocompatibilidade Menor/genética , Mapeamento de Epitopos/métodos , Marcadores Genéticos , Genoma Humano , Genótipo , Humanos , Neoplasias/imunologia , Polimorfismo de Nucleotídeo Único , Linfócitos T Citotóxicos/imunologia , Imunologia de Transplantes
15.
Int J Hematol ; 87(5): 467-473, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18414982

RESUMO

It has been shown that allogeneic hematopoietic stem cell transplantation (HSCT) can be one of the therapeutic options for patients with metastatic solid tumors, such as renal cancer. However, the development of relatively severe GVHD seems to be necessary to achieve tumor regression in the current setting. Thus, it is crucial to identify minor histocompatibility antigens (mHags) only expressed in tumor cells but not GVHD target organs. In this study, we examined whether three mHags: ACC-1 and ACC-2 encoded by BCL2A1, and HA-1 encoded by HMHA1, could serve as such targets for melanoma. Real-time PCR and immunohistochemical analysis revealed that the expression of both BCL2A1and HMHA1 in melanoma cell lines and primary melanoma cells was comparable to that of hematopoietic cells. Indeed, melanoma cell lines were efficiently lysed by cytotoxic T lymphocytes specific for ACC-1, ACC-2, and HA-1. Our data suggest that targeting mHags encoded not only by HMHA1, whose aberrant expression in solid tumors has been reported, but also BCL2A1 may bring about beneficial selective graft-versus-tumor effects in a population of melanoma patients for whom these mHags are applicable.


Assuntos
Regulação Neoplásica da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Melanoma/metabolismo , Melanoma/terapia , Antígenos de Histocompatibilidade Menor/biossíntese , Oligopeptídeos/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Linhagem Celular Transformada , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Efeito Enxerto vs Tumor/imunologia , Humanos , Imunidade Celular , Melanoma/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , Oligopeptídeos/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Transplante Homólogo
16.
Rinsho Ketsueki ; 49(1): 55-8, 2008 Jan.
Artigo em Japonês | MEDLINE | ID: mdl-18277598

RESUMO

Pyomyositis is a purulent infection of skeletal muscle characterized by fever, localized muscle pain and stiffness, swelling and tenderness. Hematological disorder is one of the predisposing conditions for the development of pyomyositis. A 54-year-old man developed methicillin-resistant Staphylococcus aureus pyomyositis during drug-induced pancytopenia. Debridement of the infection foci combined with antimicrobial agents proved effective even in the advanced stage of the disease. In patients with hematological disorders, pyomyositis should be considered when evaluating local myalgia and high fever because this disease can be very difficult to identify and can become rapidly progressive under a myelosuppressive condition.


Assuntos
Hospedeiro Imunocomprometido , Pancitopenia/induzido quimicamente , Pancitopenia/complicações , Piomiosite/microbiologia , Infecções Estafilocócicas , Antibacterianos/uso terapêutico , Desbridamento , Humanos , Masculino , Resistência a Meticilina , Pessoa de Meia-Idade , Piomiosite/etiologia , Piomiosite/terapia , Fatores de Risco , Staphylococcus aureus , Resultado do Tratamento , Vancomicina/uso terapêutico
18.
Blood ; 111(6): 3286-94, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18178869

RESUMO

Minor histocompatibility (H) antigens are the molecular targets of allo-immunity responsible both for the development of antitumor effects and for graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, despite their potential clinical use, our knowledge of human minor H antigens is largely limited by the lack of efficient methods of their characterization. Here we report a robust and efficient method of minor H gene discovery that combines whole genome association scans (WGASs) with cytotoxic T-lymphocyte (CTL) assays, in which the genetic loci of minor H genes recognized by the CTL clones are precisely identified using pooled-DNA analysis of immortalized lymphoblastoid cell lines with/without susceptibility to those CTLs. Using this method, we have successfully mapped 2 loci: one previously characterized (HMSD encoding ACC-6), and one novel. The novel minor H antigen encoded by BCL2A1 was identified within a 26 kb linkage disequilibrium block on chromosome 15q25, which had been directly mapped by WGAS. The pool size required to identify these regions was no more than 100 individuals. Thus, once CTL clones are generated, this method should substantially facilitate discovery of minor H antigens applicable to targeted allo-immune therapies and also contribute to our understanding of human allo-immunity.


Assuntos
DNA/genética , Antígenos de Histocompatibilidade Menor/genética , Antígenos de Histocompatibilidade Menor/imunologia , Células Cultivadas , Epitopos de Linfócito T/imunologia , Marcadores Genéticos/genética , Genoma Humano/genética , Genótipo , Humanos , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
19.
Int J Hematol ; 87(1): 83-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18224419

RESUMO

Histiocytic sarcoma of the spleen, in which the malignant cells display morphologic and immunophenotypic features similar to those of mature tissue histiocytes, is a rare but potentially lethal condition that can remain asymptomatic or only mildly symptomatic for a long period of time. We studied a case of histiocytic sarcoma of the spleen in an 82-year-old woman with prolonged chronic thrombocytopenia that was non-responsive to steroid therapy. Ultrasonography, computed tomography, and magnetic resonance imaging showed a characteristically enlarged spleen and liver. Palliative irradiation therapy was clinically effective; however, disease progression proved lethal. Autopsy revealed the proliferation of tumor cells within the splenic sinus and the liver sinusoids, which displayed extreme hemophagocytosis and strong expression of the histiocytic markers CD68 (KP1 and PG-M1) and CD163. The postmortem diagnosis showed histiocytic sarcoma of the spleen with liver infiltration. This and previous reports indicate that early detection (facilitated by imaging and clinical features) and management may improve patient prognosis and survival. Histiocytic sarcoma of the spleen should be considered as a differential diagnosis in therapeutically unresponsive patients with chronic thrombocytopenia.


Assuntos
Sarcoma Histiocítico/patologia , Cuidados Paliativos , Neoplasias Esplênicas/patologia , Idoso de 80 Anos ou mais , Diagnóstico por Imagem , Feminino , Sarcoma Histiocítico/complicações , Sarcoma Histiocítico/radioterapia , Humanos , Neoplasias Esplênicas/complicações , Neoplasias Esplênicas/radioterapia , Trombocitopenia/etiologia
20.
Blood ; 110(3): 1055-63, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17409267

RESUMO

Here we report the identification of a novel human leukocyte antigen (HLA)-B44-restricted minor histocompatibility antigen (mHA) with expression limited to hematopoietic cells. cDNA expression cloning studies demonstrated that the cytotoxic T lymphocyte (CTL) epitope of interest was encoded by a novel allelic splice variant of HMSD, hereafter designated as HMSD-v. The immunogenicity of the epitope was generated by differential protein expression due to alternative splicing, which was completely controlled by 1 intronic single-nucleotide polymorphism located in the consensus 5' splice site adjacent to an exon. Both HMSD-v and HMSD transcripts were selectively expressed at higher levels in mature dendritic cells and primary leukemia cells, especially those of myeloid lineage. Engraftment of mHA(+) myeloid leukemia stem cells in nonobese diabetic/severe combined immunodeficient (NOD/SCID)/gammac(null) mice was completely inhibited by in vitro preincubation with the mHA-specific CTL clone, suggesting that this mHA is expressed on leukemic stem cells. The patient from whom the CTL clone was isolated demonstrated a significant increase of the mHA-specific T cells in posttransplantation peripheral blood, whereas mHA-specific T cells were undetectable in pretransplantation peripheral blood and in peripheral blood from his donor. These findings suggest that the HMSD-v-encoded mHA (designated ACC-6) could serve as a target antigen for immunotherapy against hematologic malignancies.


Assuntos
Processamento Alternativo/genética , Epitopos de Linfócito T/genética , Leucemia Mieloide/genética , Antígenos de Histocompatibilidade Menor/genética , Polimorfismo de Nucleotídeo Único , Processamento Alternativo/imunologia , Animais , Linhagem Celular Tumoral , DNA Complementar/genética , DNA Complementar/imunologia , Epitopos de Linfócito T/imunologia , Antígenos HLA-B/genética , Antígenos HLA-B/imunologia , Antígeno HLA-B44 , Humanos , Imunoterapia , Leucemia Mieloide/imunologia , Leucemia Mieloide/terapia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Antígenos de Histocompatibilidade Menor/imunologia , Células-Tronco Neoplásicas , Sítios de Splice de RNA , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...