Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell ; 165(1): 35-44, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26997480

RESUMO

PD-1 immune checkpoint blockade provides significant clinical benefits for melanoma patients. We analyzed the somatic mutanomes and transcriptomes of pretreatment melanoma biopsies to identify factors that may influence innate sensitivity or resistance to anti-PD-1 therapy. We find that overall high mutational loads associate with improved survival, and tumors from responding patients are enriched for mutations in the DNA repair gene BRCA2. Innately resistant tumors display a transcriptional signature (referred to as the IPRES, or innate anti-PD-1 resistance), indicating concurrent up-expression of genes involved in the regulation of mesenchymal transition, cell adhesion, extracellular matrix remodeling, angiogenesis, and wound healing. Notably, mitogen-activated protein kinase (MAPK)-targeted therapy (MAPK inhibitor) induces similar signatures in melanoma, suggesting that a non-genomic form of MAPK inhibitor resistance mediates cross-resistance to anti-PD-1 therapy. Validation of the IPRES in other independent tumor cohorts defines a transcriptomic subset across distinct types of advanced cancer. These findings suggest that attenuating the biological processes that underlie IPRES may improve anti-PD-1 response in melanoma and other cancer types.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Melanoma/tratamento farmacológico , Metástase Neoplásica/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos/efeitos adversos , Proteína BRCA2/genética , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/genética , Metástase Neoplásica/genética , Nivolumabe , Transcriptoma
3.
Immunity ; 45(3): 527-539, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27566938

RESUMO

B cell development is often depicted as a linear process initiating in the fetus and continuing postnatally. Using a PU.1 hypomorphic mouse model, we found that B-1 and B-2 lymphopoiesis occurred in distinct fetal and adult waves differentially dependent on the Sfpi1 14 kB upstream regulatory element. The initial wave of fetal B-1 development was absent in PU.1 hypomorphic mice, while subsequent fetal and adult waves emerged. In contrast, B-2 lymphopoiesis occurred in distinct fetal and adult waves. Whole-transcriptome profiling of fetal and adult B cell progenitors supported the existence of three waves of B-1 and two waves of B-2 development and revealed that the network of transcription factors governing B lineage specification and commitment was highly divergent between B-1 and B-2 progenitors. These findings support the view that the B-1 and B-2 lineages are distinct and provide a genetic basis for layering of immune system development.


Assuntos
Subpopulações de Linfócitos B/imunologia , Redes Reguladoras de Genes/imunologia , Linfopoese/imunologia , Animais , Linhagem da Célula/imunologia , Feto/imunologia , Perfilação da Expressão Gênica/métodos , Camundongos , Células Precursoras de Linfócitos B/imunologia , Fatores de Transcrição/imunologia
4.
N Engl J Med ; 375(9): 819-29, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27433843

RESUMO

BACKGROUND: Approximately 75% of objective responses to anti-programmed death 1 (PD-1) therapy in patients with melanoma are durable, lasting for years, but delayed relapses have been noted long after initial objective tumor regression despite continuous therapy. Mechanisms of immune escape in this context are unknown. METHODS: We analyzed biopsy samples from paired baseline and relapsing lesions in four patients with metastatic melanoma who had had an initial objective tumor regression in response to anti-PD-1 therapy (pembrolizumab) followed by disease progression months to years later. RESULTS: Whole-exome sequencing detected clonal selection and outgrowth of the acquired resistant tumors and, in two of the four patients, revealed resistance-associated loss-of-function mutations in the genes encoding interferon-receptor-associated Janus kinase 1 (JAK1) or Janus kinase 2 (JAK2), concurrent with deletion of the wild-type allele. A truncating mutation in the gene encoding the antigen-presenting protein beta-2-microglobulin (B2M) was identified in a third patient. JAK1 and JAK2 truncating mutations resulted in a lack of response to interferon gamma, including insensitivity to its antiproliferative effects on cancer cells. The B2M truncating mutation led to loss of surface expression of major histocompatibility complex class I. CONCLUSIONS: In this study, acquired resistance to PD-1 blockade immunotherapy in patients with melanoma was associated with defects in the pathways involved in interferon-receptor signaling and in antigen presentation. (Funded by the National Institutes of Health and others.).


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Imunoterapia , Janus Quinase 1/genética , Janus Quinase 2/genética , Melanoma/genética , Mutação , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Microglobulina beta-2/genética , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/uso terapêutico , Biópsia , Exoma , Regulação Neoplásica da Expressão Gênica , Genes MHC Classe I , Humanos , Interferon gama/uso terapêutico , Melanoma/tratamento farmacológico , Melanoma/secundário , Receptor de Morte Celular Programada 1/metabolismo , Recidiva , Análise de Sequência de DNA , Transdução de Sinais
5.
Semin Immunol ; 24(5): 303-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22559986

RESUMO

The number of T cell progenitors is significantly reduced in the involuted thymus, and the growth and developmental potential of the few cells that are present is severely attenuated. This review provides an overview of how aging affects T cell precursors before and following entry into the thymus and discusses the age-related genetic changes that may occur in them. Finally, interventions that rejuvenate thymopoiesis in the elderly by targeting T cell progenitors are discussed.


Assuntos
Senescência Celular , Regulação da Expressão Gênica , Células Precursoras de Linfócitos T/imunologia , Timócitos/imunologia , Animais , Humanos , Linfopoese , Células Precursoras de Linfócitos T/citologia , Timócitos/citologia
6.
J Biol Chem ; 289(11): 7682-90, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24478313

RESUMO

A hallmark feature of mast cells is their high content of cytoplasmic secretory granules filled with various preformed compounds, including proteases of tryptase-, chymase-, and carboxypeptidase A3 type that are electrostatically bound to serglycin proteoglycan. Apart from participating in extracellular processes, serglycin proteoglycan and one of its associated proteases, tryptase, are known to regulate cell death by promoting apoptosis over necrosis. Here we sought to outline the underlying mechanism and identify core histones as primary proteolytic targets for the serglycin-tryptase axis. During the cell death process, tryptase was found to relocalize from granules into the cytosol and nucleus, and it was found that the absence of tryptase was associated with a pronounced accumulation of core histones both in the cytosol and in the nucleus. Intriguingly, tryptase deficiency resulted in defective proteolytic modification of core histones even at baseline conditions, i.e. in the absence of cytotoxic agent, suggesting that tryptase has a homeostatic impact on nuclear events. Indeed, tryptase was found in the nucleus of viable cells and was shown to cleave core histones in their N-terminal tail. Moreover, it was shown that the absence of the serglycin-tryptase axis resulted in altered chromatin composition. Together, these findings implicate histone proteolysis through a secretory granule-derived serglycin-tryptase axis as a novel principle for histone modification, during both cell homeostasis and cell death.


Assuntos
Mastócitos/citologia , Proteoglicanas/química , Vesículas Secretórias/metabolismo , Triptases/química , Proteínas de Transporte Vesicular/química , Animais , Apoptose , Células da Medula Óssea/citologia , Morte Celular , Núcleo Celular/metabolismo , Citosol/metabolismo , Histonas/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Estrutura Terciária de Proteína , Proteoglicanas/genética , Proteínas de Transporte Vesicular/genética
7.
Blood ; 119(24): 5715-21, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22555975

RESUMO

Involution of the thymus results in reduced production of naive T cells, and this in turn is thought to contribute to impaired immunity in the elderly. Early T-cell progenitors (ETPs), the most immature intrathymic T-cell precursors, harvested from the involuted thymus exhibit a diminished proliferative potential and increased rate of apoptosis and as a result their number is significantly reduced. In the present study, we show that these age-induced alterations result in part from increased expression of the Ink4a tumor-suppressor gene in ETPs. We also show that repression of Ink4a in aged ETPs results in their partial rejuvenation and that this can be accomplished by in vivo fibroblast growth factor 7 administration. These results define a genetic basis for thymocyte progenitor aging and demonstrate that the senescence-associated gene Ink4a can be pharmacologically repressed in ETPs to partially reverse the effects of aging.


Assuntos
Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Fator 7 de Crescimento de Fibroblastos/farmacologia , Células-Tronco/citologia , Timócitos/citologia , Timócitos/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Senescência Celular/imunologia , Regulação para Baixo/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Imunológicos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Timócitos/efeitos dos fármacos , Timo/citologia , Timo/efeitos dos fármacos , Timo/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
J Immunother Cancer ; 11(5)2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37156551

RESUMO

BACKGROUND: The tumor antigen NY-ESO-1 has been shown to be an effective target for transgenic adoptive cell therapy (ACT) for the treatment of sarcoma and melanoma. However, despite frequent early clinical responses, many patients ultimately develop progressive disease. Understanding the mechanisms underlying treatment resistance is crucial to improve future ACT protocols. Here, we describe a novel mechanism of treatment resistance in sarcoma involving loss of expression of NY-ESO-1 in response to transgenic ACT with dendritic cell (DC) vaccination and programmed cell death protein-1 (PD-1) blockade. METHODS: A HLA-A*02:01-positive patient with an NY-ESO-1-positive undifferentiated pleomorphic sarcoma was treated with autologous NY-ESO-1-specific T-cell receptor (TCR) transgenic lymphocytes, NY-ESO-1 peptide-pulsed DC vaccination, and nivolumab-mediated PD-1 blockade. RESULTS: Peripheral blood reconstitution with NY-ESO-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. There was initial tumor regression, and immunophenotyping of the peripheral transgenic T cells showed a predominantly effector memory phenotype over time. Tracking of transgenic T cells to the tumor sites was demonstrated in on-treatment biopsy via both TCR sequencing-based and RNA sequencing-based immune reconstitution, and nivolumab binding to PD-1 on transgenic T cells was confirmed at the tumor site. At the time of disease progression, the promoter region of NY-ESO-1 was found to be extensively methylated, and tumor NY-ESO-1 expression was completely lost as measured by RNA sequencing and immunohistochemistry. CONCLUSIONS: ACT of NY-ESO-1 transgenic T cells given with DC vaccination and anti-PD-1 therapy resulted in transient antitumor activity. NY-ESO-1 expression was lost in the post-treatment sample in the setting of extensive methylation of the NY-ESO-1 promoter region. BIOLOGICAL/CLINICAL INSIGHT: Antigen loss represents a novel mechanism of immune escape in sarcoma and a new point of improvement in cellular therapy approaches. TRIAL REGISTRATION NUMBER: NCT02775292.


Assuntos
Melanoma , Sarcoma , Humanos , Nivolumabe , Imunoterapia/métodos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo
9.
Cancer Immunol Res ; 11(12): 1589-1597, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37871333

RESUMO

Transgenic T-cell receptor (TCR) T cell-based adoptive cell therapies for solid tumors are associated with dramatic initial response rates, but there remain many instances of treatment failure and disease relapse. The association of infusion product cytokine profiles with clinical response has not been explored in the context of TCR T-cell therapy products. Single-cell antigen-dependent secretomic and proteomic analysis of preinfusion clinical TCR T-cell therapy products revealed that TNFα cytokine functionality of CD8+ T cells and phospho-STAT3 signaling in these cells were both associated with superior clinical responsiveness to therapy. By contrast, CD4+ T-helper 2 cell cytokine profiles were associated with inferior clinical responses. In parallel, preinfusion levels of IL15, Flt3-L, and CX3CL1 were all found to be associated with clinical response to therapy. These results have implications for the development of therapeutic biomarkers and identify potential targets for enrichment in the design of transgenic TCR T-cell therapies for solid tumors.


Assuntos
Neoplasias , Fator de Necrose Tumoral alfa , Animais , Humanos , Camundongos , Proteômica , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Citocinas , Animais Geneticamente Modificados , Terapia Baseada em Transplante de Células e Tecidos , Camundongos Transgênicos , Fator de Transcrição STAT3
10.
Cancer Discov ; 13(3): 580-597, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36416874

RESUMO

To address antigen escape and loss of T-cell functionality, we report a phase I clinical trial (NCT04007029) evaluating autologous naive and memory T (TN/MEM) cells engineered to express a bispecific anti-CD19/CD20 chimeric antigen receptor (CAR; CART19/20) for patients with relapsed/refractory non-Hodgkin lymphoma (NHL), with safety as the primary endpoint. Ten patients were treated with 36 × 106 to 165 × 106 CART19/20 cells. No patient experienced neurotoxicity of any grade or over grade 1 cytokine release syndrome. One case of dose-limiting toxicity (persistent cytopenia) was observed. Nine of 10 patients achieved objective response [90% overall response rate (ORR)], with seven achieving complete remission [70% complete responses (CR) rate]. One patient relapsed after 18 months in CR but returned to CR after receiving a second dose of CART19/20 cells. Median progression-free survival was 18 months and median overall survival was not reached with a 17-month median follow-up. In conclusion, CART19/20 TN/MEM cells are safe and effective in patients with relapsed/refractory NHL, with durable responses achieved at low dosage levels. SIGNIFICANCE: Autologous CD19/CD20 bispecific CAR-T cell therapy generated from TN/MEM cells for patients with NHL is safe (no neurotoxicity, maximum grade 1 cytokine release syndrome) and demonstrates strong efficacy (90% ORR, 70% CR rate) in a first-in-human, phase I dose-escalation trial. This article is highlighted in the In This Issue feature, p. 517.


Assuntos
Linfoma não Hodgkin , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/terapia , Células T de Memória , Linfoma não Hodgkin/terapia , Imunoterapia Adotiva/efeitos adversos , Antígenos CD19
11.
Ann Allergy Asthma Immunol ; 106(3): 239-44, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21354026

RESUMO

BACKGROUND: FAS has been shown to be involved in the regulation of many immune processes by induction of cellular apoptosis. However, accumulated evidence shows that FAS signaling also exhibits nonapoptotic functions, such as induction of cell proliferation and differentiation. FAS is the only death receptor known to be expressed on murine mast cells (MCs). OBJECTIVE: To evaluate the role of FAS on murine MC maturation. METHODS: Mouse bone marrow-derived MCs (BMMCs) or peritoneal MCs were derived from FAS-deficient, FASlpr/lpr, and congenic wild-type strains. The MC degranulation and cytokine release after IgE activation was assessed by measuring ß-hexosaminidase, interleukin 13, and tumor necrosis factor α release. Transmission electron microscopy analysis was performed to evaluate the level of BMMC maturation. The surface markers and intracellular preformed mediators were measured as well. RESULTS: Our data reveal that FAS deficiency has an impact on IgE-dependent activation of BMMCs, resulting in a significant decrease in ß-hexosaminidase, interleukin 13, and tumor necrosis factor α release. The total content of preformed mediators (eg, tryptase and ß-hexosaminidase) was reduced in BMMCs derived from FAS-deficient mice. We also found that the level of FcεRI in peritoneal mast cells from FAS-deficient mice was significantly diminished. FAS deficiency also influenced the kinetics of BMMC maturation as was revealed by transmission electron microscopy analysis. CONCLUSION: Our data show that FAS has an impact on the regulation of mouse MC maturation in vitro.


Assuntos
Mastócitos/imunologia , Receptor fas/imunologia , Animais , Degranulação Celular/efeitos dos fármacos , Degranulação Celular/imunologia , Feminino , Citometria de Fluxo , Interleucina-13/análise , Interleucina-13/imunologia , Mastócitos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/imunologia , beta-N-Acetil-Hexosaminidases/análise , beta-N-Acetil-Hexosaminidases/imunologia
12.
Cell Immunol ; 262(2): 80-3, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20189551

RESUMO

Recently we demonstrated that human mast cells (MC) express functional TRAIL death receptors. Here we assessed the expression of TRAIL on both mRNA and protein level in cord blood derived MC (CBMC) and HMC-1. The TRAIL release either spontaneous or induced by LPS, IFN-gamma and IgE-dependent activation, was evaluated as well. The protein location was restricted to the intracellular compartment in CBMC, but not in HMC-1. The intracellular TRAIL was not localized inside the granules. The treatment with IFN-gamma and LPS up-regulated intracellular TRAIL expression in CBMC, but did not induce its release. These in vitro data show that human MC can produce and express intracellular TRAIL whose location could not be altered by different stimuli.


Assuntos
Mastócitos/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Células Cultivadas , Sangue Fetal/citologia , Sangue Fetal/imunologia , Humanos , Imunoglobulina E/imunologia , Imunoglobulina E/farmacologia , Interferon gama/imunologia , Interferon gama/farmacologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Mastócitos/citologia , Mastócitos/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/genética
13.
Cancer Discov ; 10(11): 1645-1653, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32699033

RESUMO

Transgenic T-cell receptor (TCR) adoptive cell therapies recognizing tumor antigens are associated with robust initial response rates, but frequent disease relapse. This usually occurs in the setting of poor long-term persistence of cells expressing the transgenic TCR, generated using murine stem cell virus (MSCV) γ-retroviral vectors. Analysis of clinical transgenic adoptive cell therapy products in vivo revealed that despite strong persistence of the transgenic TCR DNA sequence over time, its expression was profoundly decreased over time at the RNA and protein levels. Patients with the greatest degrees of expression suppression displayed significant increases in DNA methylation over time within the MSCV promoter region, as well as progressive increases in DNA methylation within the entire MSCV vector over time. These increases in vector methylation occurred independently of its integration site within the host genomes. These results have significant implications for the design of future viral vector gene-engineered adoptive cell transfer therapies. SIGNIFICANCE: Cellular immunotherapies' reliance on retroviral vectors encoding foreign genetic material can be vulnerable to progressive acquisition of DNA methylation and subsequent epigenetic suppression of the transgenic product in TCR adoptive cell therapy. This must be considered in the design of future generations of cellular immunotherapies for cancer.This article is highlighted in the In This Issue feature, p. 1611.


Assuntos
Epigênese Genética/genética , Vetores Genéticos/genética , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução Genética/métodos , Humanos
14.
J Immunother ; 43(9): 273-282, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32925563

RESUMO

Expression of carbonic-anhydrase IX (CAIX) in clear cell renal cell carcinoma (RCC) makes it an attractive vaccine target. We developed a fusion-gene construct, granulocyte-macrophage (GM) colony-stimulating factor+CAIX, delivered by an adenoviral vector (Ad) into autologous dendritic cells (DCs) in this phase 1 study. The injected immature DCs were expected to stimulate an antigen-specific immune response against CAIX expressing RCC. Three dose-escalation cohorts (5, 15, and 50×10 cells/administration) were injected intradermally q2wk×3 doses based on a 3+3 design. The primary objective was the safety of the injections. Secondary objectives were immune responses using enzyme-linked immunosorbent spot, a serum biomarker panel, and clinical response. Fifteen patients with metastatic RCC were enrolled, and 9 patients received all 3 doses. No serious adverse events were seen. There were 3 (33%) patients with grade 1 fatigue, 1 of whom subsequently experienced grade 2 fatigue. One patient (11%) experienced grade 1-2 leukopenia. Only 1 patient (11%) experienced grade 2 flu-like symptoms. Of the 9 patients who received treatment, 1 expired of progressive disease, 2 patients were lost to follow-up and 6 patients are alive. Of the 6 patients, 5 have progressive disease, and 1 has completed treatment with stable disease at 27 months follow-up. Immune response measurements appeared more robust in higher dose cohorts, which appeared to be related to patients with stable disease at 3 months. These early data show that autologous immature DC-AdGMCAIX can be safely given to metastatic RCC patients without any serious adverse events with CAIX-specific immune response elicited by the treatment. These preliminary data support further study of Ad-GMCAIX, particularly with combination therapies that may enhance clinical activity.


Assuntos
Antígenos de Neoplasias/genética , Vacinas Anticâncer/administração & dosagem , Anidrase Carbônica IX/genética , Carcinoma de Células Renais/terapia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neoplasias Renais/terapia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/genética , Vacinas Anticâncer/metabolismo , Anidrase Carbônica IX/imunologia , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/patologia , Células Dendríticas/metabolismo , Gerenciamento Clínico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Resultado do Tratamento
15.
Nat Cancer ; 1(1): 46-58, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34368780

RESUMO

Lack of tumor infiltration by immune cells is the main mechanism of primary resistance to programmed cell death protein 1 (PD-1) blockade therapies for cancer. It has been postulated that cancer cell-intrinsic mechanisms may actively exclude T cells from tumors, suggesting that the finding of actionable molecules that could be inhibited to increase T cell infiltration may synergize with checkpoint inhibitor immunotherapy. Here, we show that p21-activated kinase 4 (PAK4) is enriched in non-responding tumor biopsies with low T cell and dendritic cell infiltration. In mouse models, genetic deletion of PAK4 increased T cell infiltration and reversed resistance to PD-1 blockade in a CD8 T cell-dependent manner. Furthermore, combination of anti-PD-1 with the PAK4 inhibitor KPT-9274 improved anti-tumor response compared with anti-PD-1 alone. Therefore, high PAK4 expression is correlated with low T cell and dendritic cell infiltration and a lack of response to PD-1 blockade, which could be reversed with PAK4 inhibition.


Assuntos
Inibidores de Checkpoint Imunológico , Imunoterapia , Neoplasias , Receptor de Morte Celular Programada 1 , Quinases Ativadas por p21 , Animais , Linfócitos T CD8-Positivos , Camundongos , Neoplasias/tratamento farmacológico , Quinases Ativadas por p21/genética
16.
Cancer Discov ; 10(8): 1140-1157, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32467343

RESUMO

Mechanism-based strategies to overcome resistance to PD-1 blockade therapy are urgently needed. We developed genetic acquired resistant models of JAK1, JAK2, and B2M loss-of-function mutations by gene knockout in human and murine cell lines. Human melanoma cell lines with JAK1/2 knockout became insensitive to IFN-induced antitumor effects, while B2M knockout was no longer recognized by antigen-specific T cells and hence was resistant to cytotoxicity. All of these mutations led to resistance to anti-PD-1 therapy in vivo. JAK1/2-knockout resistance could be overcome with the activation of innate and adaptive immunity by intratumoral Toll-like receptor 9 agonist administration together with anti-PD-1, mediated by natural killer (NK) and CD8 T cells. B2M-knockout resistance could be overcome by NK-cell and CD4 T-cell activation using the CD122 preferential IL2 agonist bempegaldesleukin. Therefore, mechanistically designed combination therapies can overcome genetic resistance to PD-1 blockade therapy. SIGNIFICANCE: The activation of IFN signaling through pattern recognition receptors and the stimulation of NK cells overcome genetic mechanisms of resistance to PD-1 blockade therapy mediated through deficient IFN receptor and antigen presentation pathways. These approaches are being tested in the clinic to improve the antitumor activity of PD-1 blockade therapy.This article is highlighted in the In This Issue feature, p. 1079.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Janus Quinase 1/genética , Janus Quinase 2/genética , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Microglobulina beta-2/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Interferons/farmacologia , Interleucina-2/análogos & derivados , Interleucina-2/imunologia , Interleucina-2/farmacologia , Interleucina-2/uso terapêutico , Células Matadoras Naturais/imunologia , Mutação com Perda de Função , Camundongos Endogâmicos C57BL , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/imunologia , Polietilenoglicóis/farmacologia , Polietilenoglicóis/uso terapêutico , Receptor Toll-Like 9/imunologia
17.
Clin Cancer Res ; 25(7): 2096-2108, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30573690

RESUMO

PURPOSE: Transgenic adoptive cell therapy (ACT) targeting the tumor antigen NY-ESO-1 can be effective for the treatment of sarcoma and melanoma. Preclinical models have shown that this therapy can be improved with the addition of dendritic cell (DC) vaccination and immune checkpoint blockade. We studied the safety, feasibility, and antitumor efficacy of transgenic ACT with DC vaccination, with and without CTLA-4 blockade with ipilimumab. PATIENTS AND METHODS: Freshly prepared autologous NY-ESO-1-specific T-cell receptor (TCR) transgenic lymphocytes were adoptively transferred together with NY-ESO-1 peptide-pulsed DC vaccination in HLA-A2.1-positive subjects alone (ESO, NCT02070406) or with ipilimumab (INY, NCT01697527) in patients with advanced sarcoma or melanoma. RESULTS: Six patients were enrolled in the ESO cohort, and four were enrolled in the INY cohort. Four out of six patients treated per ESO (66%), and two out of four patients treated per INY (50%) displayed evidence of tumor regression. Peripheral blood reconstitution with NY-ESO-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. Tracking of transgenic T cells to the tumor sites was demonstrated in on-treatment biopsies via TCR sequencing. Multiparametric mass cytometry of transgenic cells demonstrated shifting of transgenic cells from memory phenotypes to more terminally differentiated effector phenotypes over time. CONCLUSIONS: ACT of fresh NY-ESO-1 transgenic T cells prepared via a short ex vivo protocol and given with DC vaccination, with or without ipilimumab, is feasible and results in transient antitumor activity, with no apparent clinical benefit of the addition of ipilimumab. Improvements are needed to maintain tumor responses.


Assuntos
Transferência Adotiva , Antineoplásicos Imunológicos/farmacologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Ipilimumab/farmacologia , Neoplasias/imunologia , Neoplasias/terapia , Transferência Adotiva/métodos , Adulto , Animais , Antígeno CTLA-4/antagonistas & inibidores , Linhagem Celular Tumoral , Terapia Combinada , Células Dendríticas/metabolismo , Feminino , Técnicas de Introdução de Genes , Humanos , Imunoterapia , Linfócitos/imunologia , Linfócitos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Neoplasias/patologia , Fenótipo , Projetos Piloto , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Adulto Jovem
18.
Cell Stem Cell ; 25(4): 542-557.e9, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31495780

RESUMO

Invariant natural killer T (iNKT) cells are potent immune cells for targeting cancer; however, their clinical application has been hindered by their low numbers in cancer patients. Here, we developed a proof-of-concept for hematopoietic stem cell-engineered iNKT (HSC-iNKT) cell therapy with the potential to provide therapeutic levels of iNKT cells for a patient's lifetime. Using a human HSC engrafted mouse model and a human iNKT TCR gene engineering approach, we demonstrated the efficient and long-term generation of HSC-iNKT cells in vivo. These HSC-iNKT cells closely resembled endogenous human iNKT cells, could deploy multiple mechanisms to attack tumor cells, and effectively suppressed tumor growth in vivo in multiple human tumor xenograft mouse models. Preclinical safety studies showed no toxicity or tumorigenicity of the HSC-iNKT cell therapy. Collectively, these results demonstrated the feasibility, safety, and cancer therapy potential of the proposed HSC-iNKT cell therapy and laid a foundation for future clinical development.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Imunoterapia Adotiva/métodos , Células T Matadoras Naturais/fisiologia , Neoplasias/terapia , Animais , Células Cultivadas , Engenharia Genética , Humanos , Camundongos , Camundongos SCID , Células T Matadoras Naturais/transplante , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/genética , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Clin Cancer Res ; 25(3): 1000-1011, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30409823

RESUMO

PURPOSE: To improve persistence of adoptively transferred T-cell receptor (TCR)-engineered T cells and durable clinical responses, we designed a clinical trial to transplant genetically-modified hematopoietic stem cells (HSCs) together with adoptive cell transfer of T cells both engineered to express an NY-ESO-1 TCR. Here, we report the preclinical studies performed to enable an investigational new drug (IND) application. EXPERIMENTAL DESIGN: HSCs transduced with a lentiviral vector expressing NY-ESO-1 TCR and the PET reporter/suicide gene HSV1-sr39TK and T cells transduced with a retroviral vector expressing NY-ESO-1 TCR were coadministered to myelodepleted HLA-A2/Kb mice within a formal Good Laboratory Practice (GLP)-compliant study to demonstrate safety, persistence, and HSC differentiation into all blood lineages. Non-GLP experiments included assessment of transgene immunogenicity and in vitro viral insertion safety studies. Furthermore, Good Manufacturing Practice (GMP)-compliant cell production qualification runs were performed to establish the manufacturing protocols for clinical use. RESULTS: TCR genetically modified and ex vivo-cultured HSCs differentiated into all blood subsets in vivo after HSC transplantation, and coadministration of TCR-transduced T cells did not result in increased toxicity. The expression of NY-ESO-1 TCR and sr39TK transgenes did not have a detrimental effect on gene-modified HSC's differentiation to all blood cell lineages. There was no evidence of genotoxicity induced by the lentiviral vector. GMP batches of clinical-grade transgenic cells produced during qualification runs had adequate stability and functionality. CONCLUSIONS: Coadministration of HSCs and T cells expressing an NY-ESO-1 TCR is safe in preclinical models. The results presented in this article led to the FDA approval of IND 17471.


Assuntos
Terapia Genética/métodos , Células-Tronco Hematopoéticas/imunologia , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/genética , Células Cultivadas , Ensaios Clínicos como Assunto , Drogas em Investigação/uso terapêutico , Antígeno HLA-A2/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo
20.
Infect Immun ; 76(10): 4489-97, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18644875

RESUMO

The ability of Staphylococcus aureus to invade and survive within host cells is believed to contribute to its propensity to cause persistent and metastatic infections. In addition, S. aureus infections often are associated with atopic diseases such as dermatitis, rhinitis, and asthma. Mast cells, the key cells of allergic diseases, have a pivotal role in innate immunity and have the capacity of phagocytosis, and they can destroy some pathogenic bacteria. However, little is known about the ability of some other bacteria to survive and overcome mast cell phagocytosis. Therefore, we were interested in evaluating the interplay between mast cells and S. aureus. In this study, we show that human cord blood-derived mast cells (CBMC) can be infected by pathogenic S. aureus. S. aureus displayed a high adherence to mast cells as well as invasive and survival abilities within them. However, when infections were performed in the presence of cytochalasin D or when CBMC were preincubated with anti-Toll-like receptor 2 (TLR2) or anti-CD48 antibodies, the invasiveness and the inflammatory response were abrogated, respectively. Furthermore, we observed an increase of TLR2 and CD48 molecules on CBMC after S. aureus infection. The infection of CBMC with S. aureus also caused the release of tumor necrosis factor alpha (TNF-alpha) and interleukin-8 (IL-8). Both live and killed S. aureus organisms were found to trigger TNF-alpha and IL-8 release by CBMC in a time-dependent manner. Cumulatively, these findings suggest that S. aureus internalizes and survives in mast cells. This may play an important role in infections and in atopic diseases associated with S. aureus.


Assuntos
Antígenos CD/imunologia , Interleucina-8/metabolismo , Mastócitos/imunologia , Mastócitos/microbiologia , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Antígenos CD/biossíntese , Aderência Bacteriana , Antígeno CD48 , Células Cultivadas , Citoplasma/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Viabilidade Microbiana , Receptor 2 Toll-Like/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA