Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Immunol ; 212(8): 1381-1391, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38416029

RESUMO

Granzymes are a family of proteases used by CD8 T cells to mediate cytotoxicity and other less-defined activities. The substrate and mechanism of action of many granzymes are unknown, although they diverge among the family members. In this study, we show that mouse CD8+ tumor-infiltrating lymphocytes (TILs) express a unique array of granzymes relative to CD8 T cells outside the tumor microenvironment in multiple tumor models. Granzyme F was one of the most highly upregulated genes in TILs and was exclusively detected in PD1/TIM3 double-positive CD8 TILs. To determine the function of granzyme F and to improve the cytotoxic response to leukemia, we constructed chimeric Ag receptor T cells to overexpress a single granzyme, granzyme F or the better-characterized granzyme A or B. Using these doubly recombinant T cells, we demonstrated that granzyme F expression improved T cell-mediated cytotoxicity against target leukemia cells and induced a form of cell death other than chimeric Ag receptor T cells expressing only endogenous granzymes or exogenous granzyme A or B. However, increasing expression of granzyme F also had a detrimental impact on the viability of the host T cells, decreasing their persistence in circulation in vivo. These results suggest a unique role for granzyme F as a marker of terminally differentiated CD8 T cells with increased cytotoxicity, but also increased self-directed cytotoxicity, suggesting a potential mechanism for the end of the terminal exhaustion pathway.


Assuntos
Leucemia , Receptores de Antígenos Quiméricos , Animais , Camundongos , Linfócitos T CD8-Positivos , Granzimas , Leucemia/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Microambiente Tumoral , Citotoxicidade Imunológica
2.
Nat Cancer ; 4(7): 928-929, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37248396
3.
Cytotherapy ; 25(1): 94-102, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36253252

RESUMO

BACKGROUND AIMS: Vector copy number (VCN), an average quantification of transgene copies unique to a chimeric antigen receptor (CAR) T-cell product, is a characteristic that must be reported prior to patient administration, as high VCN increases the risk of insertional mutagenesis. Historically, VCN assessment in CAR T-cell products has been performed via quantitative polymerase chain reaction (qPCR). qPCR is reliable along a broad range of concentrations, but quantification requires use of a standard curve and precision is limited. Digital PCR (dPCR) methods were developed for absolute quantification of target sequences by counting nucleic acid molecules encapsulated in discrete, volumetrically defined partitions. Advantages of dPCR compared with qPCR include simplicity, reproducibility, sensitivity and lack of dependency on a standard curve for definitive quantification. In the present study, the authors describe a dPCR assay developed for analysis of the novel bicistronic CD19 × CD22 CAR T-cell construct. METHODS: The authors compared the performance of the dPCR assay with qPCR on both the QX200 droplet dPCR (ddPCR) system (Bio-Rad Laboratories, Inc, Hercules, CA, USA) and the QIAcuity nanoplate-based dPCR (ndPCR) system (QIAGEN Sciences, Inc, Germantown, MD, USA). The primer-probe assay was validated with qPCR, ndPCR and ddPCR using patient samples from pre-clinical CAR T-cell manufacturing production runs as well as Jurkat cell subclones, which stably express this bicistronic CAR construct. RESULTS: ddPCR confirmed the specificity of this assay to detect only the bicistronic CAR product. Additionally, the authors' assay gave accurate, precise and reproducible CAR T-cell VCN measurements across qPCR, ndPCR and ddPCR modalities. CONCLUSIONS: The authors demonstrate that dPCR strategies can be utilized for absolute quantification of CAR transgenes and VCN measurements, with improved test-retest reliability, and that specific assays can be developed for detection of unique constructs.


Assuntos
Receptores de Antígenos Quiméricos , Humanos , Reprodutibilidade dos Testes , Receptores de Antígenos Quiméricos/genética , Variações do Número de Cópias de DNA , Linfócitos T , Reação em Cadeia da Polimerase/métodos , Reação em Cadeia da Polimerase em Tempo Real
4.
Haematologica ; 108(2): 457-471, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35950535

RESUMO

Chimeric antigen receptor (CAR) T-cell immunotherapies targeting CD19 or CD22 induce remissions in the majority of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (ALL), although relapse due to target antigen loss or downregulation has emerged as a major clinical dilemma. Accordingly, great interest exists in developing CAR T cells directed against alternative leukemia cell surface antigens that may help to overcome immunotherapeutic resistance. The fms-like tyrosine kinase 3 receptor (FLT3) is constitutively activated via FLT3 mutation in acute myeloid leukemia (AML) or wild-type FLT3 overexpression in KMT2A (lysine-specific methyltransferase 2A)-rearranged ALL, which are associated with poor clinical outcomes in children and adults. We developed monovalent FLT3-targeted CAR T cells (FLT3CART) and bispecific CD19xFLT3CART and assessed their anti-leukemia activity in preclinical models of FLT3-mutant AML and KMT2A-rearranged infant ALL. We report robust in vitro FLT3CART-induced cytokine production and cytotoxicity against AML and ALL cell lines with minimal cross-reactivity against normal hematopoietic and non-hematopoietic tissues. We also observed potent in vivo inhibition of leukemia proliferation in xenograft models of both FLT3-mutant AML and KMT2A-rearranged ALL, including a post-tisagenlecleucel ALL-to-AML lineage switch patient-derived xenograft model pairing. We further demonstrate significant in vitro and in vivo activity of bispecific CD19xFLT3CART against KMT2Arearranged ALL and posit that this additional approach might also diminish potential antigen escape in these high-risk leukemias. Our preclinical data credential FLT3CART as a highly effective immunotherapeutic strategy for both FLT3- mutant AML and KMT2A-rearranged ALL which is poised for further investigation and clinical translation.


Assuntos
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Antígenos Quiméricos , Adulto , Criança , Humanos , Receptores de Antígenos Quiméricos/genética , Leucemia Mieloide Aguda/genética , Imunoterapia , Linfócitos T/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética
5.
Res Sq ; 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38196657

RESUMO

Chimeric antigen receptor T cells are an effective therapy for B-lineage malignancies. However, many patients relapse and this therapeutic has yet to show strong efficacy in other hematologic or solid tumors. One opportunity for improvement lies in the ability to generate T cells with desirable functional characteristics. Here, we dissect the biology of CD8+ CAR T cells (CAR8) by controlling whether the T cell has encountered cognate TCR antigen prior to CAR generation. We find that prior antigen experience influences multiple aspects of in vitro and in vivo CAR8 functionality, resulting in superior effector function and leukemia clearance in the setting of limiting target antigen density compared to antigen-inexperienced T cells. However, this comes at the expense of inferior proliferative capacity, susceptibility to phenotypic exhaustion and dysfunction, and inability to clear wildtype leukemia in the setting of limiting CAR+ cell dose. Epigenomic and transcriptomic comparisons of these cell populations identified overexpression of the Runx2 transcription factor as a novel strategy to enhance CAR8 function, with a differential impact depending on prior cell state. Collectively, our data demonstrate that prior antigen experience determines functional attributes of a CAR T cell, as well as amenability to functional enhancement by transcription factor modulation.

6.
Pediatr Crit Care Med ; 23(12): e595-e600, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36194016

RESUMO

OBJECTIVES: Cytokine release syndrome (CRS) is a potentially lethal toxicity associated with chimeric antigen receptor T cell therapy for pediatric acute lymphoblastic leukemia (ALL). Outcomes after critical illness due to severe CRS are poorly described. Our aim was to characterize critical illness outcomes across a multicenter cohort of PICU patients with ALL and CRS. DESIGN: Multicenter retrospective cohort study. SETTING: Twenty-one PICUs contributing data to Virtual Pediatric Systems, LLC (January 2020-December 2021). PATIENTS: PICU patients with ALL or unclassified leukemia and CRS. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: We identified 55 patients; 34 (62%) were 12 years or older, 48 (87%) were admitted from a hospital inpatient ward, and 23 (42%) received advanced organ failure support or monitoring. Fifty-one survived to PICU discharge (93%) including 19 of 23 (83%) who received advanced organ failure support or monitoring defined as receipt of noninvasive or invasive ventilation, cardiopulmonary resuscitation, extracorporeal membrane oxygenation, continuous renal replacement therapy, or placement of a tracheostomy, arterial catheter, hemodialysis catheter, or intracranial catheter. Twelve patients (22%) received invasive ventilation, nine of whom survived to PICU discharge. Two of four patients who received continuous renal replacement therapy and one of three patients who required cardiopulmonary resuscitation survived to PICU discharge. Lengths of PICU stay were median 3.0 days (interquartile range, 1.4-7.8 d) among PICU survivors, 7.8 (5.4-11.1) among those receiving advanced organ failure support or monitoring, and 7.2 days (interquartile range, 2.9-14.7 d) among nonsurvivors. Of the 51 patients who survived to PICU discharge, 48 (94%) survived the hospitalization. CONCLUSIONS: PICU patients with CRS frequently received a high level of support, and the majority survived their PICU stay and hospitalization. Additional multicenter investigations of severe CRS are necessary to inform evidence-based practice.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Antígenos Quiméricos , Criança , Humanos , Lactente , Estado Terminal/terapia , Unidades de Terapia Intensiva Pediátrica , Estudos Retrospectivos , Síndrome da Liberação de Citocina , Estudos de Coortes , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Terapia Baseada em Transplante de Células e Tecidos
8.
Exp Hematol ; 100: 1-11, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34298117

RESUMO

The clinical success of engineered, CD19-directed chimeric antigen receptor (CAR) T cells in relapsed, refractory B-cell acute lymphoblastic leukemia (B-ALL) has generated great enthusiasm for the use of CAR T cells in patients with cytogenetics that portend a poor prognosis with conventional cytotoxic therapies. One such group includes infants and children with mixed lineage leukemia (MLL1, KMT2A) rearrangements (MLL-r), who fare much worse than patients with low- or standard-risk B-ALL. Although early clinical trials using CD19 CAR T cells for MLL-r B-ALL produced complete remission in most patients, relapse with CD19-negative disease was a common mechanism of treatment failure. Whereas CD19neg relapse has been observed across a broad spectrum of B-ALL patients treated with CD19-directed therapy, patients with MLL-r have manifested the emergence of AML, often clonally related to the B-ALL, suggesting that the inherent heterogeneity or lineage plasticity of MLL-r B-ALL may predispose patients to a myeloid relapse. Understanding the factors that enable and drive myeloid relapse may be important to devise strategies to improve durability of remissions. In this review, we summarize clinical observations to date with MLL-r B-ALL and generally discuss lineage plasticity as a mechanism of escape from immunotherapy.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Imunoterapia Adotiva , Proteína de Leucina Linfoide-Mieloide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Animais , Antígenos CD19/genética , Antígenos CD19/imunologia , Rearranjo Gênico , Histona-Lisina N-Metiltransferase/imunologia , Humanos , Imunoterapia Adotiva/métodos , Proteína de Leucina Linfoide-Mieloide/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Resultado do Tratamento , Evasão Tumoral
9.
J Clin Invest ; 130(10): 5425-5443, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32925169

RESUMO

Late-onset inflammatory toxicities resembling hemophagocytic lymphohistiocytosis (HLH) or macrophage activation syndrome (MAS) occur after chimeric antigen receptor T cell (CAR T cell) infusion and represent a therapeutic challenge. Given the established link between perforin deficiency and primary HLH, we investigated the role of perforin in anti-CD19 CAR T cell efficacy and HLH-like toxicities in a syngeneic murine model. Perforin contributed to both CD8+ and CD4+ CAR T cell cytotoxicity but was not required for in vitro or in vivo leukemia clearance. Upon CAR-mediated in vitro activation, perforin-deficient CAR T cells produced higher amounts of proinflammatory cytokines compared with WT CAR T cells. Following in vivo clearance of leukemia, perforin-deficient CAR T cells reexpanded, resulting in splenomegaly with disruption of normal splenic architecture and the presence of hemophagocytes, which are findings reminiscent of HLH. Notably, a substantial fraction of patients who received anti-CD22 CAR T cells also experienced biphasic inflammation, with the second phase occurring after the resolution of cytokine release syndrome, resembling clinical manifestations of HLH. Elevated inflammatory cytokines such as IL-1ß and IL-18 and concurrent late CAR T cell expansion characterized the HLH-like syndromes occurring in the murine model and in humans. Thus, a murine model of perforin-deficient CAR T cells recapitulated late-onset inflammatory toxicities occurring in human CAR T cell recipients, providing therapeutically relevant mechanistic insights.


Assuntos
Imunoterapia Adotiva/efeitos adversos , Perforina/deficiência , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Citocinas/biossíntese , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Mediadores da Inflamação/metabolismo , Linfo-Histiocitose Hemofagocítica/etiologia , Linfo-Histiocitose Hemofagocítica/imunologia , Linfo-Histiocitose Hemofagocítica/patologia , Síndrome de Ativação Macrofágica/etiologia , Síndrome de Ativação Macrofágica/imunologia , Síndrome de Ativação Macrofágica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Perforina/genética , Linfócitos T/patologia
11.
Immunol Rev ; 290(1): 100-113, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31355496

RESUMO

Advances in the development of immunotherapies have offered exciting new options for the treatment of malignant diseases that are refractory to conventional cytotoxic chemotherapies. The adoptive transfer of T cells expressing chimeric antigen receptors (CARs) has demonstrated dramatic results in clinical trials and highlights the promise of novel immune-based approaches to the treatment of cancer. As experience with CAR T cells has expanded with longer follow-up and to a broader range of diseases, new obstacles have been identified which limit the potential lifelong benefits of CAR T cell therapy. These obstacles highlight not only the gaps in knowledge of the optimal clinical application of this "living drug", but also gaps in our understanding of the fundamental biology of CAR T cells themselves. In this review, we discuss the obstacles facing CAR T cell therapy, how these relate to our current understanding of CAR T cell biology and approaches to enhance the clinical efficacy of this therapy.


Assuntos
Engenharia Genética , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Humanos , Imunomodulação , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Ativação Linfocitária/imunologia , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Evasão Tumoral/genética , Evasão Tumoral/imunologia
12.
Blood ; 132(18): 1899-1910, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30209120

RESUMO

Adoptive transfer of patient-derived T cells modified to express chimeric antigen receptors (CARTs) has demonstrated dramatic success in relapsed/refractory pre-B-cell acute lymphoblastic leukemia (ALL), but response and durability of remission requires exponential CART expansion and persistence. Tumors are known to affect T-cell function, but this has not been well studied in ALL and in the context of chimeric antigen receptor (CAR) expression. Using TCF3/PBX1 and MLL-AF4-driven murine ALL models, we assessed the impact of progressive ALL on T-cell function in vivo. Vaccines protect against TCF3/PBX1.3 but were ineffective when administered after leukemia injection, suggesting immunosuppression induced early during ALL progression. T cells from leukemia-bearing mice exhibited increased expression of inhibitory receptors, including PD1, Tim3, and LAG3, and were dysfunctional following adoptive transfer in a model of T-cell receptor (TCR)-dependent leukemia clearance. Although expression of inhibitory receptors has been linked to TCR signaling, pre-B-cell ALL induced inhibitory receptor expression, at least in part, in a TCR-independent manner. Finally, introduction of a CAR into T cells generated from leukemia-bearing mice failed to fully reverse poor in vivo function.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Células Precursoras de Linfócitos B/patologia , Linfócitos T/patologia , Transferência Adotiva/métodos , Animais , Vacinas Anticâncer/uso terapêutico , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Camundongos Endogâmicos C57BL , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos Quiméricos/análise
13.
Sci Transl Med ; 9(417)2017 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-29167392

RESUMO

Chimeric antigen receptor (CAR)-expressing T cells induce durable remissions in patients with relapsed/refractory B cell malignancies. CARs are synthetic constructs that, when introduced into mature T cells, confer a second, non-major histocompatibility complex-restricted specificity in addition to the endogenous T cell receptor (TCR). The implications of TCR activation on CAR T cell efficacy has not been well defined. Using an immunocompetent, syngeneic murine model of CD19-targeted CAR T cell therapy for pre-B cell acute lymphoblastic leukemia in which the CAR is introduced into T cells with known TCR specificity, we demonstrate loss of CD8 CAR T cell efficacy associated with T cell exhaustion and apoptosis when TCR antigen is present. CD4 CAR T cells demonstrate equivalent cytotoxicity to CD8 CAR T cells and, in contrast, retain in vivo efficacy despite TCR stimulation. Gene expression profiles confirm increased exhaustion and apoptosis of CD8 CAR T cells upon dual receptor stimulation compared to CD4 CAR T cells and indicate inherent differences between CD4 and CD8 CAR T cells in the use of T cell-associated signaling pathways. These results provide insights into important aspects of CAR T cell immune biology and indicate opportunities to rationally design CAR constructs to optimize clinical efficacy.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular , Feminino , Citometria de Fluxo , Ativação Linfocitária/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos/metabolismo
14.
Nat Med ; 21(6): 581-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25939063

RESUMO

Chimeric antigen receptors (CARs) targeting CD19 have mediated dramatic antitumor responses in hematologic malignancies, but tumor regression has rarely occurred using CARs targeting other antigens. It remains unknown whether the impressive effects of CD19 CARs relate to greater susceptibility of hematologic malignancies to CAR therapies, or superior functionality of the CD19 CAR itself. We show that tonic CAR CD3-ζ phosphorylation, triggered by antigen-independent clustering of CAR single-chain variable fragments, can induce early exhaustion of CAR T cells that limits antitumor efficacy. Such activation is present to varying degrees in all CARs studied, except the highly effective CD19 CAR. We further determine that CD28 costimulation augments, whereas 4-1BB costimulation reduces, exhaustion induced by persistent CAR signaling. Our results provide biological explanations for the antitumor effects of CD19 CARs and for the observations that CD19 CAR T cells incorporating the 4-1BB costimulatory domain are more persistent than those incorporating CD28 in clinical trials.


Assuntos
Neoplasias Hematológicas/imunologia , Receptores de Antígenos/imunologia , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Antígenos CD19/imunologia , Antígenos CD19/metabolismo , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Linhagem Celular Tumoral , Neoplasias Hematológicas/patologia , Neoplasias Hematológicas/terapia , Humanos , Imunoterapia Adotiva , Interleucina-2/imunologia , Ativação Linfocitária/imunologia , Receptores de Antígenos/metabolismo , Linfócitos T/patologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/biossíntese
15.
Cell Immunol ; 265(1): 65-73, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20692654

RESUMO

Using a mouse neuroblastoma cell line, we have demonstrated that vaccination of tumor-free mice with a cell-based vaccine leads to productive immunity and resistance to tumor challenge, while vaccination of tumor-bearing mice does not. The T cell immunity induced by this vaccine, as measured by in vitro assays, is amplified by the depletion of Treg. Our goal is to understand this barrier to the development of protective cellular immunity. mRNA microarray analyses of CD8(+) T cells from naïve or tumor-bearing mice undergoing vaccination were carried out with or without administering anti-CD25 antibody. Gene-expression pathway analysis revealed the presence of CD8(+) T cells expressing stem cell-associated genes early after induction of productive anti-tumor immunity in tumor-free mice, prior to any phenotypic changes, but not in tumor-bearing mice. These data demonstrate that early after the induction of productive immune response, cells within the CD8(+) T cell compartment adopt a stem cell-related genetic phenotype that correlates with increased anti-tumor function.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Neuroblastoma/imunologia , Células-Tronco/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Vacinas Anticâncer/genética , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Camundongos , Neuroblastoma/terapia , RNA Neoplásico/química , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Análise de Sobrevida , Vacinação
16.
Cancer Sci ; 101(11): 2316-24, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20718755

RESUMO

The identification of tumor antigens remains a major objective in tumor immunology, especially in pediatric malignancies where solid tumors often do not express a single dominant antigen. Methods such as the Serological Screening of Recombinant cDNA Expression Libraries (SEREX) have been used in the discovery of tumor-expressed proteins by virtue of their ability to induce an antibody response. To focus and accelerate this approach, we first identified candidate antigens by gene expression profiling data from clinical neuroblastoma specimens and then used an animal model to generate an antibody response to an engineered cell-based vaccine. Candidate tumor antigens were expressed as recombinant proteins in a mammalian system and screened for antibody recognition using serum from mice vaccinated with a neuroblastoma cell-based vaccine engineered to express CD80 and CD86, with or without Treg depletion. Through this procedure, the never in mitosis A (NIMA)-related kinase NEK2 was identified as a tumor-associated antigen. Direct testing of serum from patients newly diagnosed with neuroblastoma showed specific serological responses in two of 20 patients. Although NEK2 was not universally recognized, it may serve as a tumor antigen for some patients.


Assuntos
Antígenos de Neoplasias/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neuroblastoma/genética , Animais , Antígenos de Neoplasias/sangue , Antígenos de Neoplasias/metabolismo , Antígeno B7-1/genética , Antígeno B7-2/genética , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/imunologia , Western Blotting , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Análise por Conglomerados , Humanos , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos , Quinases Relacionadas a NIMA , Neuroblastoma/imunologia , Neuroblastoma/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vacinação/métodos
17.
BMC Immunol ; 8: 4, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17397536

RESUMO

BACKGROUND: We have developed a cell-based vaccine that features the expression of both CD80 and CD86 on the surface of a murine neuroblastoma cell line. The cellular immunity induced by this vaccine is enhanced by treatment with antibody that interferes with T-regulatory cell (Treg) function and we report here that immunization combined with interfering with Treg function also produces a profound serological effect. Serum from mice immunized with our cell-based vaccine in the context of Treg blockade was used to screen a cDNA expression library constructed from the parental neuroblastoma tumor cell line, AGN2a. RESULTS: Serum from mice vaccinated in the context of Treg blockade identified a number of potentially oncogenic transcripts that may serve as important immune targets in a tumor-derived cDNA library screen. This novel approach identified far more candidates than could be seen with serum derived from vaccine-treated only, Treg-depleted only, or tumor-bearing mice. The most commonly identified tumor-associated antigen, using serum from immunized and Treg-depleted mice, was the DEK oncogene. Altered expression of the DEK oncogene has been implicated in a number of human cancers. Importantly, we were able to demonstrate that the DEK oncogene also induces a T cell response. CONCLUSION: The use of post-vaccine immune serum in this report differs from previous approaches where serum collected at the time of cancer onset or diagnosis and was used for tumor antigen identification. We hypothesize that the use of diagnostic serum samples may be inadequate for the clinical translation of this approach, and that identification of protective immunogenic tumor antigens may require the use of serum from post-treatment or vaccinated subjects. The identification of DEK as a tumor-associated antigen capable of eliciting a T cell response validates our experimental approach and argues for the antigens we have identified here to be evaluated as targets of effector immunity and as vaccine candidates.


Assuntos
Antígenos de Neoplasias/sangue , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/farmacologia , Proteínas de Ligação a DNA/sangue , Proteínas de Ligação a DNA/imunologia , Neuroblastoma/terapia , Proteínas Oncogênicas/sangue , Proteínas Oncogênicas/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Antígeno B7-1/imunologia , Antígeno B7-2/imunologia , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , DNA Complementar , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Biblioteca Gênica , Imunidade Celular , Imunização , Camundongos , Neuroblastoma/genética , Neuroblastoma/imunologia , Neuroblastoma/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Ligação a Poli-ADP-Ribose , Linfócitos T Reguladores/metabolismo
18.
Semin Cancer Biol ; 16(2): 137-49, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16376101

RESUMO

Suppressor/regulatory T cells were first shown to have an impact on cancer progression in experimental tumor models during the 1970s. However, the lack of specific markers hindered mechanistic investigations, and skepticism grew in the scientific community due to variability in cell populations and reported functions. The identification of regulatory CD4(+)CD25(+) T cells has generated a great deal of renewed interest in cells that have immune regulatory properties. This article will provide a brief historical review of suppressor T cells and cancer, experimental and clinical evidence that CD4(+)CD25(+) natural regulatory T cells play a role in cancer progression, and briefly discuss current strategies to inhibit these cells in an effort to enhance cancer immunotherapy.


Assuntos
Tolerância Imunológica , Terapia de Imunossupressão , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Reguladores/imunologia , Antígenos CD4/análise , Humanos , Receptores de Interleucina-2/análise
19.
AIDS Res Hum Retroviruses ; 19(10): 837-46, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14585215

RESUMO

Using the simian-human immunodeficiency virus (SHIV), we have investigated whether the blood-brain barrier (BBB) is compromised during the early stages of infection. Five macaques were inoculated with pathogenic SHIV(50OLNV) for 2 weeks at which time macaques were anesthetized, perfused with saline, and sacrificed. The brains were removed and examined for the disruption of the blood-brain barrier by immunohistochemical staining for the plasma protein fibrinogen in the neural parenchyma. Our results indicate a disruption of the BBB in the five of five macaques inoculated with SHIV(50OLNV) for 2 weeks. Zonula occludens 1 (ZO-1), which is a marker for the tight junctions formed by brain vascular endothelial cells, was largely absent in areas that showed fibrinogen deposition in all five macaques. To determine if the BBB integrity correlated with the initial stages of infection, the brains from two macaques were analyzed that had progressed to end-stage disease following inoculation with pathogenic SHIV(50OLNV) but developed no neuropathology and from two macaques that were inoculated with a gene-deleted, nonpathogenic virus (novpuSHIV(KU-1bMC33)) for over 1 year. Our results indicate that unlike the macaques sacrificed during the acute phase of infection, immunohistochemical staining for fibrinogen in the neural parenchyma was negative and ZO-1 staining was readily detected in the endothelial cells of the blood vessels. The results of this study indicate that the transient loss of BBB integrity is a function of the high level of virus replication that occurs during the acute phase of infection and provides important information on the early stages of lentivirus neuroinvasion.


Assuntos
Barreira Hematoencefálica , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Encéfalo/citologia , Imuno-Histoquímica , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...