Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2265: 645-654, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33704745

RESUMO

Expression of chimeric antigen receptors can redirect T cell specificity and allow for MHC-independent recognition of melanoma associated antigens. Retroviral transduction is used to express chimeric antigen receptor constructs in murine CD4+ and CD8+ T cells. Here we describe the production of retroviral supernatants and the activation, transduction, expansion, and selection of murine T cells expressing the chimeric-PD1 receptor. This protocol can be modified for any murine chimeric antigen receptor construct.


Assuntos
Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Imunoterapia Adotiva , Melanoma Experimental , Receptores de Antígenos Quiméricos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Humanos , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Camundongos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia
2.
Immunology ; 160(3): 280-294, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32144940

RESUMO

Adoptive transfer of T-cells is a promising therapy for many cancers. To enhance tumour recognition by T-cells, chimeric antigen receptors (CARs) consisting of signalling domains fused to receptors that recognize tumour-associated antigens can be expressed in T-cells. While CAR T-cells have shown clinical success for treating haematopoietic malignancies, using CAR T-cells to treat solid tumours remains a challenge. We developed a chimeric PD1 (chPD1) receptor that recognizes the ligands for the PD1 receptor that are expressed on many types of solid cancer. To determine if this novel CAR could target a wide variety of tumour types, the anti-tumour efficacy of chPD1 T-cells against syngeneic murine models of melanoma, renal, pancreatic, liver, colon, breast, prostate and bladder cancer was measured. Of the 14 cell lines tested, all expressed PD1 ligands on their cell surface, making them potential targets for chPD1 T-cells. ChPD1 T-cells lysed the tumour cells and secreted pro-inflammatory cytokines [interferon (IFN)γ, tumour necrosis factor (TNF)α, interleukin (IL)-2, granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-17 and IL-21], but did not secrete the anti-inflammatory cytokine IL-10. Furthermore, T-cells expressing chPD1 receptors reduced an established tumour burden and led to long-term tumour-free survival in all types of solid tumours tested. ChPD1 T-cells did not survive longer than 14 days in vivo; however, treatment with chPD1 T-cells induced protective host anti-tumour memory responses in tumour-bearing mice. Therefore, adoptive transfer of chPD1 T-cells could be a novel therapeutic strategy to treat multiple types of solid cancer.


Assuntos
Complexo CD3/genética , Vacinas Anticâncer/imunologia , Imunoterapia Adotiva/métodos , Melanoma/terapia , Receptor de Morte Celular Programada 1/genética , Receptores de Antígenos Quiméricos/genética , Neoplasias Cutâneas/terapia , Linfócitos T/fisiologia , Animais , Modelos Animais de Doenças , Humanos , Melanoma/imunologia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Cutâneas/imunologia , Transplante Isogênico , Carga Tumoral
3.
Cell Immunol ; 351: 104069, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32106933

RESUMO

Chimeric antigen receptors (CAR) utilize costimulatory domains to enhance anti-tumor efficacy. However, it is unclear which costimulatory domain is preferable. Therefore, the intracellular domains of CD28, Dap10, 41BB, GITR, ICOS, or OX40 were compared in a murine chimeric PD1 (chPD1) receptor that targets tumor-associated PD1 ligands. Upon antigen restimulation, T cells expressing chPD1-CD28 receptors had reduced lytic capacity. While most of the chPD1 T cell receptors secreted pro-inflammatory (IFNγ, TNFα, IL-2, GM-CSF, IL-17, and IL-21) and anti-inflammatory cytokines (IL-10), chPD1-Dap10 did not secrete IL-10. Furthermore, chPD1-Dap10 and -41BB receptors induced a memory precursor phenotype, had enhanced persistence in vivo, and superior therapeutic efficacy in murine models of T cell lymphoma and melanoma compared to chPD1-CD28 or chPD1-GITR expressing T cells. Therefore, each costimulatory domain induces differential effects in CAR-expressing T cells and inclusion of Dap10 or 4-1BB costimulatory domains may induce a preferential cytokine profile and differentiation for cancer therapy.


Assuntos
Ligante 4-1BB/imunologia , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores Imunológicos/imunologia , Animais , Modelos Animais de Doenças , Feminino , Imunoterapia Adotiva/métodos , Linfoma/imunologia , Masculino , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1
4.
J Vis Exp ; (147)2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31132062

RESUMO

MicRoboCop is a biosensor that has been designed for a unique application in forensic chemistry. MicRoboCop is a system made up of three devices that, when used together, can indicate the presence of gunshot residue (GSR) by producing a fluorescence signal in the presence of three key analytes (antimony, lead, and organic components of GSR). The protocol describes the synthesis of the biosensors using Escherichia coli (E. coli), and the analytical chemistry methods used to evaluate the selectivity and sensitivity of the sensors. The functioning of the system is demonstrated by using GSR collected from the inside of a spent cartridge casing. Once prepared, the biosensors can be stored until needed and can be used as a test for these key analytes. A positive response from all three analytes provides a presumptive positive test for GSR, while each individual device has applications for detecting the analytes in other samples (e.g., a detector for lead contamination in drinking water). The main limitation of the system is the time required for a positive signal; future work may involve studying different organisms to optimize the response time.


Assuntos
Técnicas Biossensoriais , Escherichia coli/metabolismo , Armas de Fogo , Antimônio/análise , Bário/análise , Espectrometria de Fluorescência
5.
ACS Med Chem Lett ; 9(1): 51-55, 2018 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-29348811

RESUMO

Commercially utilized parabens are employed for their antimicrobial properties, but a weak binding to the estrogen receptor alpha (ERα) may lead to breast cancer in some applications. Modification of the paraben scaffold should allow for a disconnection of these observed properties. Toward this goal, various 3,5-substituted parabens were synthesized and assessed for antimicrobial properties against S. aureus as well as competitive binding to the ERα. The minimum inhibitory concentration assay confirmed retention of antimicrobial activity in many of these derivatives, while all compounds exhibited decreased xenoestrogen activity as determined by a combination of competitive enzyme linked immunosorbent assay (ELISA), proliferation, and estrogen receptor binding assay. Thus, these changes to the paraben scaffold have led to a multitude of paraben derivatives with antimicrobial properties up to 16 times more active than the parent paraben and that are devoid or significantly diminished of potential breast cancer causing properties.

6.
Immunology ; 152(3): 472-483, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28670716

RESUMO

Adoptive transfer of T cells is a promising cancer therapy and expression of chimeric antigen receptors can enhance tumour recognition and T-cell effector functions. The programmed death protein 1 (PD1) receptor is a prospective target for a chimeric antigen receptor because PD1 ligands are expressed on many cancer types, including lymphoma. Therefore, we developed a murine chimeric PD1 receptor (chPD1) consisting of the PD1 extracellular domain fused to the cytoplasmic domain of CD3ζ. Additionally, chimeric antigen receptor therapies use various co-stimulatory domains to enhance efficacy. Hence, the inclusion of a Dap10 or CD28 co-stimulatory domain in the chPD1 receptor was compared to determine which domain induced optimal anti-tumour immunity in a mouse model of lymphoma. The chPD1 T cells secreted pro-inflammatory cytokines and lysed RMA lymphoma cells. Adoptive transfer of chPD1 T cells significantly reduced established tumours and led to tumour-free survival in lymphoma-bearing mice. When comparing chPD1 receptors containing a Dap10 or CD28 domain, both receptors induced secretion of pro-inflammatory cytokines; however, chPD1-CD28 T cells also secreted anti-inflammatory cytokines whereas chPD1-Dap10 T cells did not. Additionally, chPD1-Dap10 induced a central memory T-cell phenotype compared with chPD1-CD28, which induced an effector memory phenotype. The chPD1-Dap10 T cells also had enhanced in vivo persistence and anti-tumour efficacy compared with chPD1-CD28 T cells. Therefore, adoptive transfer of chPD1 T cells could be a novel therapy for lymphoma and inclusion of the Dap10 co-stimulatory domain in chimeric antigen receptors may induce a preferential cytokine profile and T-cell differentiation phenotype for anti-tumour therapies.


Assuntos
Transferência Adotiva/métodos , Terapia Genética/métodos , Linfoma de Células T/terapia , Neoplasias Experimentais/terapia , Receptor de Morte Celular Programada 1/imunologia , Receptores Imunológicos/imunologia , Linfócitos T/transplante , Animais , Antígenos CD28/genética , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Complexo CD3/genética , Complexo CD3/imunologia , Complexo CD3/metabolismo , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica , Genótipo , Humanos , Memória Imunológica , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Linfoma de Células T/genética , Linfoma de Células T/imunologia , Linfoma de Células T/metabolismo , Masculino , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Fenótipo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo , Transfecção , Carga Tumoral
7.
Immunology ; 147(3): 305-20, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26661515

RESUMO

Memory CD8+ T cells are an essential component of anti-tumour and anti-viral immunity. Activation of the mammalian/mechanistic target of rapamycin (mTOR) pathway has been implicated in regulating the differentiation of effector and memory T cells. However, the mechanisms that control mTOR activity during immunity to tumours and infections are not well known. Activation of co-stimulatory receptors, including CD28 and natural killer group 2D (NKG2D), activate phosphatidylinositol-3 kinase and subsequently may activate the mTOR pathway in CD8+ T cells. This study compared the activation of the mTOR signalling pathway after co-stimulation through CD28 or NKG2D receptors in murine effector CD8+ T cells. Compared with CD28 co-stimulation, activation through CD3 and NKG2D receptors had weaker activation of mTORc1, as shown by decreased phosphorylation of mTORc1 targets S6K1, ribosomal protein S6 and eukaryotic initiation factor 4E binding protein 1. NKG2D co-stimulation also showed increased gene expression of tuberous sclerosis protein 2, a negative regulator of mTORc1, whereas CD28 co-stimulation increased gene expression of Ras homologue enriched in brain, an activator of mTORc1, and hypoxia-inducible factor-1α and vascular endothelial growth factor-α, pro-angiogenic factors downstream of mTORc1. Strong mTORc1 activation in CD28-co-stimulated cells also increased expression of transcription factors that support effector cell differentiation, namely T-bet, B lymphocyte-induced maturation protein (BLIMP-1), interferon regulatory factor 4, and inhibitor of DNA binding 2, whereas low levels of mTORc1 activation allowed for the expression of Eomes, B-cell lymphoma 6 (BCL6), and inhibitor of DNA binding 3 during NKG2D stimulation, and increased expression of memory markers CD62 ligand and CD127. These data show that compared with CD28, co-stimulation through the NKG2D receptor leads to the differential activation of the mTOR signalling pathway and potentially supports memory CD8+ T-cell differentiation.


Assuntos
Antígenos CD28/imunologia , Linfócitos T CD8-Positivos/imunologia , Ativação Linfocitária/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Serina-Treonina Quinases TOR/imunologia , Animais , Diferenciação Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/imunologia
8.
Mol Immunol ; 63(2): 268-78, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25089028

RESUMO

To induce strong immune responses, naïve CD8(+) T cells require stimulation through the TCR and costimulatory receptors. However, the biological effect of activating costimulatory receptors on effector T cells is still unclear. One costimulatory receptor that is likely to be engaged at the target site is NKG2D. This activating receptor is expressed on human and murine CD8(+) T cells with its ligands expressed on the majority of tumor cells and during some infections. In order to determine how activation of costimulatory receptors alters effector CD8(+) T cell functions, this study compared the activation of the NF-κB signaling pathway by two costimulatory receptors, CD28 and NKG2D. Compared to CD28 costimulation, activation of murine effector CD8(+) T cells through CD3 and NKG2D receptors enhanced activation of NF-κB as shown by increased phosphorylation of IKKα, IκBα, and NF-κB and IκBα degradation. NKG2D costimulation also increased activation, nuclear translocation, and DNA binding of NF-κB p65/p50 dimers. Activation of the NF-κB pathway also lead to increased gene expression and secretion of pro-inflammatory cytokines, including IFNα and IFNγ, and decreased gene expression and secretion of anti-inflammatory cytokines, including IL-10 and CCL2. Altered NF-κB activation also increased expression of the effector molecules TNFα, lymphotoxins α and ß, and Fas ligand, and increased tumor cell killing through FasL. These data show that compared to CD28 costimulation, activation through the NKG2D receptor leads to the differential activation of the NF-κB signaling pathway and potentially enhances the anti-tumor and anti-viral functions of effector CD8(+) T cells.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , NF-kappa B/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Animais , Complexo CD3/metabolismo , Citocinas/genética , Citotoxicidade Imunológica , Proteína Ligante Fas/metabolismo , Humanos , Ligantes , Ativação Linfocitária/imunologia , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia
9.
Oncoimmunology ; 2(4): e23564, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23734311

RESUMO

Conditioning strategies that deplete host lymphocytes have been shown to enhance clinical responses to some adoptive T-cell therapies. However, host T cells are capable of eliminating tumor cells upon the relief of immunosuppression, indicating that lymphodepletion prior to T-cell transfer may reduce optimal tumor protection elicited by cell treatments that are capable of shaping host immunity. In this study, we show that adoptively transferred T cells bearing a chimeric antigen receptor (CAR) harness endogenous T cells for optimal tumor elimination and the development of a tumor-specific memory T cell response. Mice bearing ID8 ovarian cancer cells were treated with T cells transduced with a NKG2D-based CAR. CAR-expressing T cells increased the number of host CD4+ and CD8+ T cells at the tumor site in a CXCR3-dependent manner and increased the number of antigen-specific host CD4+ T cells in the tumor and draining lymph nodes. In addition, the administration of CAR-expressing T cells increased antigen presentation to CD4+ T cells, and this increase was dependent on interferon γ and granulocyte-macrophage colony-stimulating factor produced by the former. Host CD4+ T cells were sufficient for optimal tumor protection mediated by NKG2D CAR-expressing T cells, but they were not necessary if CD4+ T cells were adoptively co-transferred. However, host CD4+ T cells were essential for the development of an antigen-specific memory T-cell response to tumor cells. Moreover, optimal tumor elimination as orchestrated by NKG2D CAR-expressing T cells was dependent on host CD8+ T cells. These results demonstrate that adoptively transferred T cells recruit and activate endogenous T-cell immunity to enhance the elimination of tumor cells and the development of tumor-specific memory responses.

10.
Immunol Cell Biol ; 91(6): 435-40, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23628805

RESUMO

Tumor heterogeneity presents a substantial barrier to increasing clinical responses mediated by targeted therapies. Broadening the immune response elicited by treatments that target a single antigen is necessary for the elimination of tumor variants that fail to express the targeted antigen. In this study, it is shown that adoptive transfer of T cells bearing a chimeric antigen receptor (CAR) inhibited the growth of target-expressing and -deficient tumor cells within ovarian and lymphoma tumors. Mice bearing the ID8 ovarian or RMA lymphoma tumors were treated with T cells transduced with a NKG2D-based CAR (chNKG2D). NKG2D CAR T-cell therapy protected mice from heterogeneous RMA tumors. Moreover, adoptive transfer of chNKG2D T cells mediated tumor protection against highly heterogeneous ovarian tumors in which 50, 20 or only 7% of tumor cells expressed significant amounts of NKG2D ligands. CAR T cells did not mediate an in vivo response against tumor cells that did not express sufficient amounts of NKG2D ligands, and the number of ligand-expressing tumor cells correlated with therapeutic efficacy. In addition, tumor-free surviving mice were protected against a tumor re-challenge with NKG2D ligand-negative ovarian tumor cells. These data indicate that NKG2D CAR T-cell treatment can be an effective therapy against heterogeneous tumors and induce tumor-specific immunity against ligand-deficient tumor cells.


Assuntos
Imunoterapia Adotiva/métodos , Linfoma/terapia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Células T Matadoras Naturais/imunologia , Neoplasias Ovarianas/terapia , Proteínas Recombinantes de Fusão/metabolismo , Animais , Processos de Crescimento Celular/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Feminino , Humanos , Interferon gama/metabolismo , Linfoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Células T Matadoras Naturais/transplante , Neoplasias Ovarianas/imunologia , Proteínas Recombinantes de Fusão/genética
11.
J Immunol ; 188(12): 6389-98, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22586039

RESUMO

The infiltration of suppressive myeloid cells into the tumor microenvironment restrains anti-tumor immunity. However, cytokines may alter the function of myeloid lineage cells to support tumor rejection, regulating the balance between pro- and anti-tumor immunity. In this study, it is shown that effector cytokines secreted by adoptively transferred T cells expressing a chimeric Ag receptor (CAR) shape the function of myeloid cells to promote endogenous immunity and tumor destruction. Mice bearing the ovarian ID8 tumor were treated with T cells transduced with a chimeric NKG2D receptor. GM-CSF secreted by the adoptively transferred T cells recruited peripheral F4/80(lo)Ly-6C(+) myeloid cells to the tumor microenvironment in a CCR2-dependent fashion. T cell IFN-γ and GM-CSF activated local, tumor-associated macrophages, decreased expression of regulatory factors, increased IL-12p40 production, and augmented Ag processing and presentation by host macrophages to Ag-specific T cells. In addition, T cell-derived IFN-γ, but not GM-CSF, induced the production of NO by F4/80(hi) macrophages and enhanced their lysis of tumor cells. The ability of CAR T cell therapy to eliminate tumor was moderately impaired when inducible NO synthase was inhibited and greatly impaired in the absence of peritoneal macrophages after depletion with clodronate encapsulated liposomes. This study demonstrates that the activation of host macrophages by CAR T cell-derived cytokines transformed the tumor microenvironment from immunosuppressive to immunostimulatory and contributed to inhibition of ovarian tumor growth.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Interferon gama/imunologia , Macrófagos/imunologia , Neoplasias Experimentais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Microambiente Tumoral/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/imunologia , Quimera , Citometria de Fluxo , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Células Mieloides/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T/imunologia , Linfócitos T/transplante
12.
Blood ; 117(24): 6571-81, 2011 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-21518928

RESUMO

Although innate immune signals shape the activation of naive T cells, it is unclear how innate signals influence effector T-cell function. This study determined the effects of stimulating the NKG2D receptor in conjunction with the TCR on human effector CD8(+) T cells. Stimulation of CD8(+) T cells through CD3 and NKG2D simultaneously or through a chimeric NKG2D receptor, which consists of NKG2D fused to the intracellular region of CD3ζ, activated ß-catenin and increased expression of ß-catenin-induced genes, whereas T cells stimulated through the TCR or a combination of the TCR and CD28 did not. Activation by TCR and NKG2D prevented expression and production of anti-inflammatory cytokines IL-10, IL-9, IL-13, and VEGF-α in a ß-catenin- and PPARγ- dependent manner. NKG2D stimulation also modulated the cytokine secretion of T cells activated simultaneously through CD3 and CD28. These data indicate that activating CD8(+) T cells through the NKG2D receptor along with the TCR modulates signal transduction and the production of anti-inflammatory cytokines. Thus, human effector T cells alter their function depending on which innate receptors are engaged in conjunction with the TCR complex.


Assuntos
Citocinas/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/fisiologia , Linfócitos T Auxiliares-Indutores/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/fisiologia , Células Cultivadas , Citocinas/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/fisiologia , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/agonistas , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteína Oncogênica v-akt/metabolismo , Proteína Oncogênica v-akt/fisiologia , PPAR gama/metabolismo , PPAR gama/fisiologia , Células U937 , beta Catenina/metabolismo , beta Catenina/fisiologia
13.
J Immunol ; 183(11): 6939-47, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19915047

RESUMO

Adoptive transfer of T cells expressing chimeric NKG2D (chNKG2D) receptors, a fusion of NKG2D and CD3zeta, can lead to long-term, tumor-free survival in a murine model of ovarian cancer. To determine the mechanisms of chNKG2D T cell antitumor efficacy, we analyzed how chNKG2D T cells altered the tumor microenvironment, including the tumor-infiltrating leukocyte populations. chNKG2D T cell treatment of mice bearing ID8 tumor cells increased the number and activation of NK cells and increased the activation of host CD8+ T cells within the tumor. Foxp3+ regulatory T cells at the tumor site decreased more than 300-fold after chNKG2D T cell treatment. Tumor-associated regulatory T cells expressed cell surface NKG2D ligands and were killed by chNKG2D T cells in a perforin-dependent manner. chNKG2D T cells also altered the function of myeloid cells at the tumor site, changing these cells from being immunosuppressive to enhancing T cell responses. Cells isolated from the tumor produced elevated amounts of IFN-gamma, NO, and other proinflammatory cytokines after chNKG2D T cell treatment. ChNKG2D T cells required perforin, IFN-gamma, and GM-CSF to induce a full response at the tumor site. In addition, transfer of chNKG2D T cells into mice bearing tumors that were established for 5 weeks led to long-term survival of the mice. Thus, chNKG2D T cells altered the ovarian tumor microenvironment to eliminate immunosuppressive cells and induce infiltration and activation of antitumor immune cells and production of inflammatory cytokines. This induction of an immune response likely contributes to chNKG2D T cells' ability to eliminate established tumors.


Assuntos
Tolerância Imunológica/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Neoplasias Ovarianas/imunologia , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Transferência Adotiva , Animais , Células Apresentadoras de Antígenos/imunologia , Quimera , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Subpopulações de Linfócitos T/imunologia
14.
J Immunol ; 183(4): 2365-72, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19625653

RESUMO

Treatment of mice bearing established ovarian tumors with T cells expressing chimeric NKG2D receptors (chNKG2D) develop protective host immune responses to tumor Ags. In this study, the mechanisms that chNKG2D T cells require to induce host immunity against ovarian tumors and which of the host immune cells are involved in tumor elimination were determined. Treatment with chNKG2D T cells led to a sustained, increased IFN-gamma production by host NK, CD4(+), and CD8(+) T cells in the spleen and at the tumor site and this continued for many weeks after T cell injection. Tumor Ag presentation was enhanced in chNKG2D T cell-treated mice, and there were greater numbers of tumor-specific T cells at the tumor site and in draining lymph nodes after treatment with chNKG2D T cells. The increase in host cell cytokine secretion and Ag presentation was dependent on chNKG2D T cell-derived perforin, IFN-gamma, and GM-CSF. Host immune mechanisms were involved in tumor elimination because inhibition of tumor growth was limited in mice that lacked perforin, IFN-gamma, NK cells, or T and B cells (Rag1(-/-)). There was no role for host-derived GM-CSF or CD1-dependent NKT cells, because mice deficient in these were able to clear tumors as well as treated wild-type B6 mice. In summary, chNKG2D T cells required both cytotoxicity and cytokine secretion as well as the participation of host immune cells for development of a host antitumor immune response and complete efficacy.


Assuntos
Apresentação de Antígeno/genética , Antígenos de Neoplasias/metabolismo , Citocinas/metabolismo , Imunoterapia Adotiva , Proteínas Mutantes Quiméricas/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Perforina/biossíntese , Subpopulações de Linfócitos T/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/genética , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Imunidade Inata/genética , Imunoterapia Adotiva/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Mutantes Quiméricas/administração & dosagem , Subfamília K de Receptores Semelhantes a Lectina de Células NK/administração & dosagem , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Perforina/genética , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/transplante , Regulação para Cima/genética , Regulação para Cima/imunologia
15.
J Clin Invest ; 119(8): 2231-44, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19620771

RESUMO

The success of clinically relevant immunotherapies requires reversing tumor-induced immunosuppression. Here we demonstrated that linear polyethylenimine-based (PEI-based) nanoparticles encapsulating siRNA were preferentially and avidly engulfed by regulatory DCs expressing CD11c and programmed cell death 1-ligand 1 (PD-L1) at ovarian cancer locations in mice. PEI-siRNA uptake transformed these DCs from immunosuppressive cells to efficient antigen-presenting cells that activated tumor-reactive lymphocytes and exerted direct tumoricidal activity, both in vivo and in situ. PEI triggered robust and selective TLR5 activation in vitro and elicited the production of hallmark TLR5-inducible cytokines in WT mice, but not in Tlr5-/- littermates. Thus, PEI is a TLR5 agonist that, to our knowledge, was not previously recognized. In addition, PEI-complexed nontargeting siRNA oligonucleotides stimulated TLR3 and TLR7. The nonspecific activation of multiple TLRs (specifically, TLR5 and TLR7) reversed the tolerogenic phenotype of human and mouse ovarian tumor-associated DCs. In ovarian carcinoma-bearing mice, this induced T cell-mediated tumor regression and prolonged survival in a manner dependent upon myeloid differentiation primary response gene 88 (MyD88; i.e., independent of TLR3). Furthermore, gene-specific siRNA-PEI nanocomplexes that silenced immunosuppressive molecules on mouse tumor-associated DCs elicited discernibly superior antitumor immunity and enhanced therapeutic effects compared with nontargeting siRNA-PEI nanocomplexes. Our results demonstrate that the intrinsic TLR5 and TLR7 stimulation of siRNA-PEI nanoparticles synergizes with the gene-specific silencing activity of siRNA to transform tumor-infiltrating regulatory DCs into DCs capable of promoting therapeutic antitumor immunity.


Assuntos
Células Dendríticas/imunologia , Nanopartículas/administração & dosagem , Neoplasias Ovarianas/terapia , Polietilenoimina/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Receptor 5 Toll-Like/fisiologia , Animais , Apresentação de Antígeno , Antígeno B7-1/fisiologia , Antígeno B7-H1 , Citotoxicidade Imunológica , Células Dendríticas/fisiologia , Feminino , Humanos , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/fisiologia , Neoplasias Ovarianas/imunologia , Peptídeos/fisiologia , Linfócitos T/imunologia
16.
Exp Hematol ; 36(10): 1318-28, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18599182

RESUMO

OBJECTIVE: Most myeloma tumor cells from patients express NKG2D ligands. We have reported the development of a chimeric NKG2D receptor (chNKG2D), which consists of the NKG2D receptor fused to the CD3zeta chain. T cells expressing this receptor kill and produce cytokines in response to NKG2D-ligand+ tumor cells. Therefore, we investigated whether human chNKG2D T cells respond against human myeloma cells. MATERIALS AND METHODS: ChNKG2D T cells were generated from healthy donors and myeloma patients. The effector phase of chNKG2D T cells was analyzed by cell-surface marker expression and human myeloma cell lines were tested for expression of NKG2D ligands. Lysis of myeloma cell lines and cytokine secretion by chNKG2D T cells was determined. ChNKG2D T cells grown in serum-free media, or cyropreserved, were assessed for effector cell functions. RESULTS: Myeloma cell lines expressed NKG2D ligands. ChNKG2D T cells from healthy donors and myeloma patients lysed myeloma cells, and secreted proinflammatory cytokines when cultured with myeloma cells or patient bone marrow, but not with peripheral blood mononuclear cells or normal bone marrow. Lysis of myeloma cells was dependent on chNKG2D T-cell expression of NKG2D and perforin. Additionally, chNKG2D T cells upregulated CD45RO, did not express CD57, and maintained expression of CD27, CD62L, and CCR7, indicating that the T cells were at an early effector stage. Finally, we showed that chNKG2D T cells generated with serum-free media, or when cryopreserved, maintained effector functions. CONCLUSION: ChNKG2D T cells respond to human myeloma cells and can be generated using clinically applicable cell culture techniques.


Assuntos
Mieloma Múltiplo/microbiologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Proteínas de Fusão Oncogênica/uso terapêutico , Linfócitos T/imunologia , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/patologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Imunoterapia/métodos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Immunol ; 180(1): 72-8, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18097006

RESUMO

Ovarian cancer is one of the leading causes of cancer death in women and the development of novel therapies is needed to complement the standard treatment options such as chemotherapy and radiation. In this study, we show that treatment with T cells expressing a chimeric NKG2D receptor (chNKG2D) was able to lead to long-term, tumor-free survival in mice bearing established ovarian tumors. Tumor-free mice were able to reject a rechallenge with ovarian tumor cells 225 days after original tumor injection. In addition, chNKG2D T cell treatment induced specific host immune responses to ovarian tumor cells, including the development of both CD8+ and CD4+ T cell tumor-specific memory responses. The chNKG2D T cells reduced the ovarian tumor burden using both cytotoxic and cytokine-dependent pathways. Specifically, chNKG2D T cell expression of perforin, GM-CSF, and IFN-gamma were essential for complete antitumor efficacy.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias Ovarianas/terapia , Receptores Imunológicos/genética , Linfócitos T/transplante , Animais , Intervalo Livre de Doença , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interferon gama/metabolismo , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Perforina/metabolismo , Receptores Imunológicos/metabolismo , Receptores de Células Matadoras Naturais , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/imunologia , Carga Tumoral
18.
Cancer Res ; 67(22): 11029-36, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006849

RESUMO

In this study, the efficacy and mechanisms of chimeric NKG2D receptor (chNKG2D)-modified T cells in eliminating NKG2D ligand-positive RMA/Rae1 lymphoma cells were evaluated. Intravenous injection of RMA/Rae1 cells led to significant tumor formation in spleens and lymph nodes within 2 weeks. Adoptive transfer of chNKG2D-modified T cells after tumor injection significantly reduced tumor burdens in both spleens and lymph nodes, and prolonged the survival of tumor-bearing mice. Multiple treatments with chNKG2D T cells resulted in long-term tumor-free survival. Moreover, these long-term survivors were resistant to rechallenge with RMA tumor cells (NKG2D ligand-negative), and their spleen and lymph node cells produced IFN-gamma in response to RMA but not to other tumors in vitro, indicating immunity against RMA tumor antigens. ChNKG2D T cell-derived IFN-gamma and granulocyte-macrophage colony-stimulating factor, but not perforin (Pfp), tumor necrosis factor-related apoptosis-inducing ligand, or Fas ligand (FasL) alone were critical for in vivo efficacy. T cells deficient in both Pfp and FasL did not kill NKG2D ligand-positive RMA cells in vitro. Adoptive transfer of Pfp(-/-)FasL(-/-) chNKG2D T cells had reduced in vivo efficacy, indicating that chNKG2D T cells used both mechanisms to attack RMA/Rae1 cells. Taken together, these results indicate that chNKG2D T-cell-mediated therapeutic effects are mediated by both cytokine-dependent and cytotoxic mechanisms in vivo.


Assuntos
Linfoma de Células T/metabolismo , Linfoma de Células T/terapia , Receptores Imunológicos/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Interferon gama/metabolismo , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores de Células Matadoras Naturais , Baço/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Resultado do Tratamento
19.
Cancer Res ; 67(10): 5003-8, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17510432

RESUMO

Despite advancements in the treatment of ovarian cancer, this disease continues to be a leading cause of cancer death in women. Adoptive transfer of tumor-reactive T cells is a promising antitumor therapy for many cancers. We designed a chimeric receptor linking NKG2D, a natural killer (NK) cell-activating receptor, to the CD3zeta chain of the T-cell receptor to target ovarian tumor cells. Engagement of chimeric NKG2D receptors (chNKG2D) with ligands for NKG2D, which are commonly expressed on tumor cells, leads to T-cell secretion of proinflammatory cytokines and tumor cytotoxicity. In this study, we show that >80% of primary human ovarian cancer samples expressed ligands for NKG2D on the cell surface. The tumor samples expressed MHC class I-related protein A, MICB, and UL-16 binding proteins 1 and 3. ChNKG2D-expressing T cells lysed ovarian cancer cell lines. We show that T cells from ovarian cancer patients that express chNKG2D secreted proinflammatory cytokines when cultured with autologous tumor cells. In addition, we show that chNKG2D T cells can be used therapeutically in a murine model of ovarian cancer. These data indicate that treatment with chNKG2D-expressing T cells is a potential immunotherapy for ovarian cancer.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Receptores Imunológicos/imunologia , Linfócitos T/imunologia , Animais , Processos de Crescimento Celular/imunologia , Linhagem Celular Tumoral , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Proteínas Associadas à Matriz Nuclear/biossíntese , Proteínas Associadas à Matriz Nuclear/imunologia , Proteínas de Transporte Nucleocitoplasmático/biossíntese , Proteínas de Transporte Nucleocitoplasmático/imunologia , Receptores de Células Matadoras Naturais , Linfócitos T/metabolismo
20.
Adv Cancer Res ; 95: 249-92, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16860660

RESUMO

Natural killer (NK) cells were identified 30 years ago based on their ability to "spontaneously" kill tumor cells. The basis for NK cell recognition and activation is due to a variety of receptors that bind to specific ligands on tumor cells and normal cells. Some of these receptors have the ability to inhibit NK cell function, and other receptors activate NK cell function. Therapeutic strategies for cancer therapy are being developed based on preventing NK cell inhibition or using NK cell receptors to activate NK cells or T cells. There are intriguing clinical data from studies of bone marrow transplantation that support the idea that preventing NK cell inhibition by human leukocyte antigen (HLA) class I molecules can be a means to promote graft-versus-leukemia (GvL) effects and limit graft-versus-host disease (GvHD) in acute myeloid leukemia (AML) and chronic myeloid leukemia (CML) patients. Experimental findings also support the blockade of NK cell inhibitory receptors as a way to protect against leukemia relapse. It may be possible to use our knowledge of NK cell activating receptors and their ligands to immunize patients with modified tumor cells to promote beneficial NK cell responses and development of host antitumor cytotoxic T lymphocytes (CTLs). Finally, new data support the idea of using modified NK cell receptors as a means to target patients' T cells against their own tumor cells and induce long-term immunity against them. Tumors are essentially tissues that have overcome normal regulation mechanisms, and therefore the ability to distinguish normal cells from abnormal cells is a key part of selectively attacking tumor cells. NK cells have various receptor systems designed to recognize infected and abnormal cells. Understanding NK cell receptors and their recognition mechanisms provides new tools for the development of immunotherapies against cancer.


Assuntos
Imunoterapia/métodos , Células Matadoras Naturais/citologia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Humanos , Sistema Imunitário/fisiologia , Células Matadoras Naturais/imunologia , Ligantes , Complexo Principal de Histocompatibilidade , Receptores Imunológicos/biossíntese , Receptores de Células Matadoras Naturais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...