Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Semin Immunol ; 49: 101414, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-33011064

RESUMO

Immunotherapies have become the first line of treatment for many cancer types. Unfortunately, only a small fraction of patients benefits from these therapies. This low rate of success can be attributed to 3 main barriers: 1) low frequency of anti-tumor specific T cells; 2) lack of infiltration of the anti-tumor specific T cells into the tumor parenchyma and 3) accumulation of highly suppressive cells in the tumor mass that inhibit the effector function of the anti-tumor specific T cells. Thus, the identification of immunomodulators that can increase the frequency and/or the infiltration of antitumor specific T cells while reducing the suppressive capacity of the tumor microenvironment is necessary to ensure the effectiveness of T cell immunotherapies. In this review, we discuss the potential of poly-ICLC as a multi-functional immune modulator for treating cancer and its impact on the 3 above mentioned barriers. We describe the unique capacity of poly-ICLC in stimulating 2 separate pattern recognition receptors, TLR3 and cytosolic MDA5 and the consequences of these activations on cytokines and chemokines production. We emphasize the role of poly-ICLC as an adjuvant in the setting of peptide-based cancer vaccines and in situ tumor vaccination by mimicking natural immune responses to infections. Finally, we summarize the impact of poly-ICLC in enhancing T infiltration into the tumor parenchyma and address the implication of this finding in the clinic.


Assuntos
Antineoplásicos/farmacologia , Carboximetilcelulose Sódica/análogos & derivados , Fatores Imunológicos/farmacologia , Imunomodulação , Poli I-C/imunologia , Poli I-C/farmacologia , Polilisina/análogos & derivados , Animais , Antineoplásicos/uso terapêutico , Carboximetilcelulose Sódica/farmacologia , Carboximetilcelulose Sódica/uso terapêutico , Citocinas/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Imunomodulação/efeitos dos fármacos , Helicase IFIH1 Induzida por Interferon/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Poli I-C/uso terapêutico , Polilisina/imunologia , Polilisina/farmacologia , Polilisina/uso terapêutico , Receptores de Reconhecimento de Padrão/metabolismo , Receptor 3 Toll-Like/metabolismo
2.
Cancer Sci ; 112(7): 2705-2713, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34009705

RESUMO

Recent studies have revealed that tumor cells decrease their immunogenicity by epigenetically repressing the expression of highly immunogenic antigens to survive in immunocompetent hosts. We hypothesized that these epigenetically hidden "stealth" antigens should be favorable targets for cancer immunotherapy due to their high immunogenicity. To identify these stealth antigens, we treated human lung cell line A549 with DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5Aza) and its prodrug guadecitabine for 3 d in vitro and screened it using cDNA microarray analysis. We found that the gene encoding sperm equatorial segment protein 1 (SPESP1) was re-expressed in cell lines including solid tumors and leukemias treated with 5Aza, although SPESP1 was not detected in untreated tumor cell lines. Using normal human tissue cDNA panels, we demonstrated that SPESP1 was not detected in normal human tissue except for testis and placenta. Moreover, we found using immunohistochemistry SPESP1 re-expression in xenografts in BALB/c-nu/nu mice that received 5Aza treatment. To assess the antigenicity of SPESP1, we stimulated human CD4+ T-cells with a SPESP1-derived peptide designed using a computer algorithm. After repetitive stimulation, SPESP1-specific helper T-cells were obtained; these cells produced interferon-γ against HLA-matched tumor cell lines treated with 5Aza. We also detected SPESP1 expression in freshly collected tumor cells derived from patients with acute myeloid leukemia or lung cancer. In conclusion, SPESP1 can be classified as a stealth antigen, a molecule encoded by a gene that is epigenetically silenced in tumor cells but serves as a highly immunogenic antigen suitable for cancer immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Proteínas de Transporte/imunologia , Epigênese Genética/imunologia , Neoplasias/imunologia , Proteínas de Plasma Seminal/imunologia , Animais , Antígenos de Neoplasias/genética , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Decitabina/farmacologia , Epigênese Genética/efeitos dos fármacos , Epitopos de Linfócito T/imunologia , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/genética , Neoplasias/terapia , Proteínas de Plasma Seminal/genética , Linfócitos T Auxiliares-Indutores/imunologia , Evasão Tumoral/genética
3.
Cancer Immunol Immunother ; 70(12): 3421-3434, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33866408

RESUMO

Identification of immunogenic tumor antigens, their corresponding T cell epitopes and the selection of effective adjuvants are prerequisites for developing effective cancer immunotherapies such as therapeutic vaccines. Murine double minute 2 (MDM2) is an E3 ubiquitin-protein ligase that negatively regulates tumor suppressor p53. Because MDM2 overexpression serves as a poor prognosis factor in various types of tumors, it would be beneficial to develop MDM2-targeted cancer vaccines. In this report, we identified an MDM2-derived peptide epitope (MDM232-46) that elicited antigen-specific and tumor-reactive CD4+ T cell responses. These CD4+ T cells directly killed tumor cells via granzyme B. MDM2 is expressed in head and neck cancer patients with poor prognosis, and the T cells that recognize this MDM2 peptide were present in these patients. Notably, Nutlin-3 (MDM2-p53 blocker), inhibited tumor cell proliferation, was shown to augment antitumor T cell responses by increasing MDM2 expression, HLA-class I and HLA-DR through class II transactivator (CIITA). These results suggest that the use of this MDM2 peptide as a therapeutic vaccine combined with MDM2 inhibitors could represent an effective immunologic strategy to treat cancer.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Proteínas Proto-Oncogênicas c-mdm2/imunologia , Animais , Vacinas Anticâncer/imunologia , Linhagem Celular , Proliferação de Células/fisiologia , Antígenos HLA-DR/imunologia , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Imunoterapia/métodos , Camundongos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/imunologia
4.
FASEB J ; 34(2): 2595-2608, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31919918

RESUMO

Dendritic cells are an important link between innate and adaptive immune response. The role of dendritic cells in bone homeostasis, however, is not understood. Osteoporosis medications that inhibit osteoclasts have been associated with osteonecrosis, a condition limited to the jawbone, thus called medication-related osteonecrosis of the jaw. We propose that disruption of the local immune response renders the oral microenvironment conducive to osteonecrosis. We tested whether zoledronate (Zol) treatment impaired dendritic cell (DC) functions and increased bacterial load in alveolar bone in vivo and whether DC inhibition alone predisposed the animals to osteonecrosis. We also analyzed the role of Zol in impairment of differentiation and function of migratory and tissue-resident DCs, promoting disruption of T-cell activation in vitro. Results demonstrated a Zol induced impairment in DC functions and an increased bacterial load in the oral cavity. DC-deficient mice were predisposed to osteonecrosis following dental extraction. Zol treatment of DCs in vitro caused an impairment in immune functions including differentiation, maturation, migration, antigen presentation, and T-cell activation. We conclude that the mechanism of Zol-induced osteonecrosis of the jaw involves disruption of DC immune functions required to clear bacterial infection and activate T cell effector response.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Osso e Ossos/efeitos dos fármacos , Células Dendríticas/metabolismo , Homeostase/imunologia , Doenças Maxilomandibulares/imunologia , Osteonecrose/tratamento farmacológico , Ácido Zoledrônico/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Homeostase/efeitos dos fármacos , Imidazóis/farmacologia , Doenças Maxilomandibulares/tratamento farmacológico , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteonecrose/imunologia , Extração Dentária/métodos , Cicatrização/efeitos dos fármacos
5.
Cancer Immunol Immunother ; 68(3): 455-466, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30604041

RESUMO

Vaccines consisting of synthetic peptides representing cytotoxic T-lymphocyte (CTL) epitopes have long been considered as a simple and cost-effective approach to treat cancer. However, the efficacy of these vaccines in the clinic in patients with measurable disease remains questionable. We believe that the poor performance of peptide vaccines is due to their inability to generate sufficiently large CTL responses that are required to have a positive impact against established tumors. Peptide vaccines to elicit CTLs in the clinic have routinely been administered in the same manner as vaccines designed to induce antibody responses: injected subcutaneously and in many instances using Freund's adjuvant. We report here that peptide vaccines and poly-ICLC adjuvant administered via the unconventional intravenous route of immunization generate substantially higher CTL responses as compared to conventional subcutaneous injections, resulting in more successful antitumor effects in mice. Furthermore, amphiphilic antigen constructs such as palmitoylated peptides were shown to be better immunogens than long peptide constructs, which now are in vogue in the clinic. The present findings if translated into the clinical setting could help dissipate the wide-spread skepticism of whether peptide vaccines will ever work to treat cancer.


Assuntos
Vacinas Anticâncer/imunologia , Administração Intravenosa , Animais , Vacinas Anticâncer/administração & dosagem , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Vacinação , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
6.
J Transl Med ; 17(1): 207, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31221178

RESUMO

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) originates from squamous epithelium of the upper aerodigestive tract and is the most common malignancy in the head and neck region. Among HNSCCs, oropharynx squamous cell carcinoma (OSCC) has a unique profile and is associated with human papillomavirus infection. Recently, anti-programmed cell death-1 monoclonal antibody has yielded good clinical responses in recurrent and/or metastatic HNSCC patients. Therefore, programmed death-ligand 1 (PD-L1) may be a favorable target molecule for cancer immunotherapy. Although PD-L1-expressing malignant cells could be targeted by PD-L1-specific CD8+ cytotoxic T lymphocytes, it remains unclear whether CD4+ helper T lymphocytes (HTLs) recognize and kill tumor cells in a PD-L1-specific manner. METHODS: The expression levels of PD-L1 and HLA-DR were evaluated using immunohistochemical analyses. MHC class II-binding peptides for PD-L1 were designed based on computer algorithm analyses and added into in vitro culture of HTLs with antigen-presenting cells to evaluate their stimulatory activity. RESULTS: We found that seven of 24 cases of OSCC showed positive for both PD-L1 and HLA-DR and that PD-L1241-265 peptide efficiently activates HTLs, which showed not only cytokine production but also cytotoxicity against tumor cells in a PD-L1-dependent manner. Also, an adoptive transfer of the PD-L1-specific HTLs significantly inhibited growth of PD-L1-expressing human tumor cell lines in an immunodeficient mouse model. Importantly, T cell responses specific for the PD-L1241-265 peptide were detected in the HNSCC patients. CONCLUSIONS: The cancer immunotherapy targeting PD-L1 as a helper T-cell antigen would be a rational strategy for HNSCC patients.


Assuntos
Antígeno B7-H1/metabolismo , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/terapia , Imunoterapia/métodos , Carcinoma de Células Escamosas de Cabeça e Pescoço/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Linfócitos T Auxiliares-Indutores/fisiologia , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Células Cultivadas , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Epitopos de Linfócito T/química , Epitopos de Linfócito T/uso terapêutico , Feminino , Técnicas de Silenciamento de Genes , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imunoterapia/tendências , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/uso terapêutico , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo
7.
Hepatology ; 68(2): 574-589, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29443377

RESUMO

Hepatocellular carcinoma (HCC) is the major form of liver cancer for which there is no effective therapy. Genetic modification with T-cell receptors (TCRs) specific for HCC-associated antigens, such as α-fetoprotein (AFP), can potentially redirect human T cells to specifically recognize and kill HCC tumor cells to achieve antitumor effects. In this study, using lentivector and peptide immunization, we identified a population of cluster of differentiation 8 (CD8) T cells in human leukocyte antigen (HLA)-A2 transgenic AAD mice that recognized AFP158 epitope on human HCC cells. Adoptive transfer of the AFP158 -specific mouse CD8 T cells eradicated HepG2 tumor xenografts as large as 2 cm in diameter in immunocompromised nonobese diabetic severe combined immunodeficient gamma knockout (NSG) mice. We then established T-cell hybridoma clones from the AFP158 -specific mouse CD8 T cells and identified three sets of paired TCR genes out of five hybridomas. Expression of the murine TCR genes redirected primary human T cells to bind HLA-A2/AFP158 tetramer. TCR gene-engineered human T (TCR-T) cells also specifically recognized HLA-A2+ AFP+ HepG2 HCC tumor cells and produced effector cytokines. Importantly, the TCR-T cells could specifically kill HLA-A2+ AFP+ HepG2 tumor cells without significant toxicity to normal primary hepatocytes in vitro. Adoptive transfer of the AFP-specific TCR-T cells could eradicate HepG2 tumors in NSG mice. CONCLUSION: We have identified AFP-specific murine TCR genes that can redirect human T cells to specifically recognize and kill HCC tumor cells, and those AFP158 -specific TCRs have a great potential to engineer a patient's autologous T cells to treat HCC tumors. (Hepatology 2018).


Assuntos
Linfócitos T CD8-Positivos/imunologia , Carcinoma Hepatocelular/imunologia , Imunoterapia/métodos , Neoplasias Hepáticas/imunologia , alfa-Fetoproteínas/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Feminino , Antígeno HLA-A2/imunologia , Células Hep G2 , Humanos , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia
8.
Cancer Immunol Immunother ; 67(7): 1091-1103, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29696308

RESUMO

Cytotoxic T lymphocytes (CTLs) are effective components of the immune system capable of destroying tumor cells. Generation of CTLs using peptide vaccines is a practical approach to treat cancer. We have previously described a peptide vaccination strategy that generates vast numbers of endogenous tumor-reactive CTLs after two sequential immunizations (prime-boost) using poly-ICLC adjuvant, which stimulates endosomal toll-like receptor 3 (TLR3) and cytoplasmic melanoma differentiation antigen 5 (MDA5). Dendritic cells (DCs) play an important role not only in antigen presentation but are critical in generating costimulatory cytokines that promote CTL expansion. Poly-ICLC was shown to be more effective than poly-IC in generating type-I interferon (IFN-I) in various DC subsets, through its enhanced ability to escape the endosomal compartment and stimulate MDA5. In our system, IFN-I did not directly function as a T cell costimulatory cytokine, but enhanced CTL expansion through the induction of IL15. With palmitoylated peptide vaccines, CD8α+ DCs were essential for peptide crosspresentation. For vaccine boosts, non-professional antigen-presenting cells were able to present minimal epitope peptides, but DCs were still required for CTL expansions through the production of IFN-I mediated by poly-ICLC. Overall, these results clarify the roles of DCs, TLR3, MDA5, IFN-I and IL15 in the generation of vast and effective antitumor CTL responses using peptide and poly-IC vaccines.


Assuntos
Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon/fisiologia , Melanoma Experimental/terapia , Linfócitos T Citotóxicos/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Animais , Carboximetilcelulose Sódica/administração & dosagem , Carboximetilcelulose Sódica/análogos & derivados , Células Dendríticas/efeitos dos fármacos , Indutores de Interferon/administração & dosagem , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli I-C/administração & dosagem , Polilisina/administração & dosagem , Polilisina/análogos & derivados , Linfócitos T Citotóxicos/efeitos dos fármacos , Receptor 3 Toll-Like/fisiologia , Células Tumorais Cultivadas , Vacinação
10.
Cancer Immunol Immunother ; 66(2): 203-213, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27052572

RESUMO

The design of efficacious and cost-effective therapeutic vaccines against cancer remains both a research priority and a challenge. For more than a decade, our laboratory has been involved in the development of synthetic peptide-based anti-cancer therapeutic vaccines. We first dedicated our efforts in the identification and validation of peptide epitopes for both CD8 and CD4 T cells from tumor-associated antigens (TAAs). Because of suboptimal immune responses and lack of therapeutic benefit of peptide vaccines containing these epitopes, we have focused our recent efforts in optimizing peptide vaccinations in mouse tumor models using numerous TAA epitopes. In this focused research review, we describe how after taking lessons from the immune system's way of dealing with acute viral infections, we have designed peptide vaccination strategies capable of generating very high numbers of therapeutically effective CD8 T cells. We also discuss some of the remaining challenges to translate these findings into the clinical setting.


Assuntos
Vacinas Anticâncer/imunologia , Melanoma Experimental/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Viroses/imunologia , Animais , Modelos Animais de Doenças , Humanos
11.
Cancer Immunol Immunother ; 66(6): 705-716, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28243692

RESUMO

Stimulator of IFN genes (STING) spontaneously contributes to anti-tumor immunity by inducing type I interferons (IFNs) following sensing of tumor-derived genomic DNAs in the tumor-bearing host. Although direct injection of STING ligands such as cyclic diguanylate monophosphate (c-di-GMP) and cyclic [G(2',5')pA(3',5')p] (cGAMP) into the tumor microenvironment exerts anti-tumor effects through strong induction of type I IFNs and activation of innate and adaptive immunity, the precise events caused by STING in the tumor microenvironment remain to be elucidated. We describe here our finding that a CD45+ CD11bmid Ly6C+ cell subset transiently accumulated in mouse tumor microenvironment of 4T1 breast cancer, squamous cell carcinomas, CT26 colon cancer, or B16F10 melanoma tissue after intratumoral injection of cGAMP. The accumulated cells displayed a macrophage (M ) phenotype since the cells were positive for F4/80 and MHC class II and negative for Ly6G. Intratumoral cGAMP treatment did not induce Mφ accumulation in STING-deficient mice. Depletion of CD8+ T cell using anti-CD8 mAb impaired the anti-tumor effects of cGAMP treatment. Depletion of the Mφ using clodronate liposomes impaired the anti-tumor effects of cGAMP treatment. Functional analysis indicated that the STING-triggered tumor-migrating Mφ exhibited phagocytic activity, production of tumor necrosis factor alpha TNFα), and high expression levels of T cell-recruiting chemokines, Cxcl10 and Cxcl11, IFN-induced molecules, MX dynamin-like GTPase 1 (Mx1) and 2'-5' oligoadenylate synthetase-like 1 (Oasl1), nitric oxide synthase 2 (Nos2), and interferon beta 1 (Ifnb1). These results indicate that the STING-triggered tumor-migrating Mφ participate in the anti-tumor effects of STING-activating compounds.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/imunologia , Carcinoma de Células Escamosas/imunologia , Neoplasias do Colo/imunologia , Macrófagos/imunologia , Melanoma Experimental/imunologia , Nucleotídeos Cíclicos/administração & dosagem , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/prevenção & controle , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Feminino , Imunoterapia , Injeções Intralesionais , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/efeitos dos fármacos , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nucleotídeos Cíclicos/farmacologia , Fagocitose
12.
Cancer Immunol Immunother ; 66(7): 877-890, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28349165

RESUMO

Nasal natural killer/T-cell lymphoma (NNKTL) is an aggressive neoplasm with poor therapeutic responses and prognosis. The programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) pathway plays an important role in immune evasion of tumor cells through T-cell exhaustion. The aim of the present study was to examine the expression of PD-L1 and PD-1 molecules in NNKTL. We detected the expression of PD-L1 in biopsy samples from all of the NNKTL patients studied. PD-L1 was found on both malignant cells and tumor-infiltrating macrophages, while PD-1-positive mononuclear cells infiltrated the tumor tissues in 36% of patients. Most significantly, soluble PD-L1 (sPD-L1) was present in sera of NNKTL patients at higher levels as compared to healthy individuals and the levels of serum sPD-L1 in patients positively correlated with the expression of PD-L1 in lymphoma cells of tumor tissues. In addition, the high-sPD-L1 group of patients showed significantly worse prognosis than the low-sPD-L1 group. Furthermore, we confirmed that membrane and soluble PD-L1 was expressed on the surface and in the culture supernatant, respectively, of NNKTL cell lines. The expression of PD-L1 was observed in tumor tissues and sera from a murine xenograft model inoculated with an NNKTL cell line. Our results suggest that sPD-L1 could be a prognostic predictor for NNKTL and open up the possibility of immunotherapy of this lymphoma using PD-1/PD-L1 axis inhibitors.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Imunoterapia/métodos , Linfoma Extranodal de Células T-NK/terapia , Neoplasias Nasais/terapia , Receptor de Morte Celular Programada 1/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Biópsia , Linhagem Celular Tumoral , Intervalo Livre de Doença , Feminino , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Linfoma Extranodal de Células T-NK/metabolismo , Linfoma Extranodal de Células T-NK/mortalidade , Linfoma Extranodal de Células T-NK/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Neoplasias Nasais/metabolismo , Neoplasias Nasais/mortalidade , Neoplasias Nasais/patologia , Prognóstico
13.
J Immunol ; 192(6): 2920-31, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24554775

RESUMO

Cross-presentation is one of the main features of dendritic cells (DCs), which is critically important for the development of spontaneous and therapy-inducible antitumor immune responses. Patients, at early stages of cancer, have normal presence of DCs. However, the difficulties in the development of antitumor responses in patients with low tumor burden raised the question of the mechanisms of DC dysfunction. In this study, we found that, in differentiated DCs, tumor-derived factors blocked the cross-presentation of exogenous Ags without inhibiting the Ag presentation of endogenous protein or peptides. This effect was caused by intracellular accumulation of different types of oxidized neutral lipids: triglycerides, cholesterol esters, and fatty acids. In contrast, the accumulation of nonoxidized lipids did not affect cross-presentation. Oxidized lipids blocked cross-presentation by reducing the expression of peptide-MHC class I complexes on the cell surface. Thus, this study suggests the novel role of oxidized lipids in the regulation of cross-presentation.


Assuntos
Apresentação de Antígeno/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Lipídeos/imunologia , Neoplasias/imunologia , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Interferon gama/farmacologia , Lipídeos/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Neoplasias/metabolismo , Neoplasias/patologia , Ovalbumina/imunologia , Oxirredução , Fragmentos de Peptídeos/imunologia
14.
Mol Ther ; 23(5): 845-856, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25544599

RESUMO

We used a VSV-cDNA library to treat recurrent melanoma, identifying immunogenic antigens, allowing us to target recurrences with immunotherapy or chemotherapy. Primary B16 melanoma tumors were induced to regress by frontline therapy. Mice with recurrent tumors were treated with VSV-cDNA immunotherapy. A Th17 recall response was used to screen the VSV-cDNA library for individual viruses encoding rejection antigens, subsequently targeted using immunotherapy or chemotherapy. Recurrent tumors were effectively treated with a VSV-cDNA library using cDNA from recurrent B16 tumors. Recurrence-associated rejection antigens identified included Topoisomerase-IIα, YB-1, cdc7 kinase, and BRAF. Fourteen out of 16 recurrent tumors carried BRAF mutations (595-605 region) following frontline therapy, even though the parental B16 tumors were BRAF wild type. The emergence of mutated BRAF-containing recurrences served as an excellent target for BRAF-specific immune-(VSV-BRAF), or chemo-(PLX-4720) therapies. Successful PLX-4720 therapy of recurrent tumors was associated with the development of a broad spectrum of T-cell responses. VSV-cDNA technology can be used to identify recurrence specific antigens. Emergence of mutated BRAF may be a major effector of melanoma recurrence which could serve as a target for chemo or immune therapy. This study suggests a rationale for offering patients with initially wild-type BRAF melanomas an additional biopsy to screen for mutant BRAF upon recurrence.


Assuntos
Melanoma/genética , Melanoma/patologia , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Antígenos de Neoplasias/imunologia , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Sequência de Bases , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Ganciclovir/farmacologia , Expressão Gênica , Humanos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/farmacologia , Masculino , Melanoma/tratamento farmacológico , Melanoma/imunologia , Melanoma Experimental , Camundongos , Dados de Sequência Molecular , Recidiva Local de Neoplasia , Proteínas Proto-Oncogênicas B-raf/química , Timidina Quinase/genética
15.
Eur J Immunol ; 44(12): 3741-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25251667

RESUMO

In the accepted model of T-cell activation, parallel signal-transduction pathways activate the transcription factors NF-κB, NFAT, and AP-1 to drive clonal expansion of T cells in response to Ag. Genome-wide transcriptional profiling following Ag-induced CD8(+) T-cell activation in C57BL/6 mouse T cells revealed that genes regulated by NFAT were also reduced in the absence of NF-κB p50 and cRel subunits. Importantly, p50(-/-) cRel(-/-) CD8(+) T cells had significantly diminished NFAT and AP-1 activation compared with WT or PKCθ(-/-) CD8(+) T cells. Attenuated NFAT activation after TCR engagement was associated with reduced calcium influx, PLCγ and Zap70 activation. Interestingly, pharmacological bypass of PLCγ-regulated pathways largely rescued p50(-/-) cRel(-/-) T-cell proliferative defects. These results indicate a crucial and unexpected requirement for NF-κB p50 and cRel subunits in proximal TCR signaling and calcium responses. They further suggest that key defects in T cells in the absence of NF-κB pathway components may be due to impaired proximal T-cell signaling.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Sinalização do Cálcio/imunologia , Subunidade p50 de NF-kappa B/imunologia , Fatores de Transcrição NFATC/imunologia , Proteínas Proto-Oncogênicas c-rel/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Sinalização do Cálcio/genética , Proliferação de Células/fisiologia , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Fatores de Transcrição NFATC/genética , Fosfolipase C gama/genética , Fosfolipase C gama/imunologia , Proteínas Proto-Oncogênicas c-rel/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Proteína-Tirosina Quinase ZAP-70/genética , Proteína-Tirosina Quinase ZAP-70/imunologia
16.
Cancer Immunol Immunother ; 64(8): 1057-66, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25986168

RESUMO

Therapeutic vaccines to induce anti-tumor CD8 T cells have been used in clinical trials for advanced melanoma patients, but the clinical response rate and overall survival time have not improved much. We believe that these dismal outcomes are caused by inadequate number of antigen-specific CD8 T cells generated by most vaccines. In contrast, huge CD8 T cell responses readily occur during acute viral infections. High levels of type-I interferon (IFN-I) are produced during these infections, and this cytokine not only exhibits anti-viral activity but also promotes CD8 T cell responses. The studies described here were performed to determine whether promoting the production of IFN-I could enhance the potency of a peptide vaccine. We report that cyclic diguanylate monophosphate (c-di-GMP), which activates the stimulator of interferon genes, potentiated the immunogenicity and anti-tumor effects of a peptide vaccine against mouse B16 melanoma. The synergistic effects of c-di-GMP required co-administration of costimulatory anti-CD40 antibody, the adjuvant poly-IC, and were mediated in part by IFN-I. These findings demonstrate that peptides representing CD8 T cell epitopes can be effective inducers of large CD8 T cell responses in vaccination strategies that mimic acute viral infections.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Vacinas Anticâncer/uso terapêutico , GMP Cíclico/análogos & derivados , Imunoterapia/métodos , Melanoma Experimental/terapia , Neoplasias Cutâneas/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Antígenos CD40/imunologia , Linfócitos T CD8-Positivos/imunologia , GMP Cíclico/administração & dosagem , GMP Cíclico/efeitos adversos , Epitopos de Linfócito T/administração & dosagem , Humanos , Interferon Tipo I/imunologia , Ativação Linfocitária/efeitos dos fármacos , Melanoma Experimental/imunologia , Proteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli I-C/administração & dosagem , Receptores de Interferon/genética , Neoplasias Cutâneas/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico
17.
Cancer Immunol Immunother ; 64(6): 697-705, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25754123

RESUMO

Nasal natural killer/T-cell lymphoma (NNKTL) is associated with Epstein-Barr virus and has a poor prognosis because of local invasion and/or multiple dissemination. Various chemokines play a role in tumor proliferation and invasion, and chemokine receptors including the C-C chemokine receptor 4 (CCR4) are recognized as potential targets for treating hematologic malignancies. The aim of the present study was to determine whether specific chemokines are produced by NNKTL. We compared chemokine expression patterns in culture supernatants of NNKTL cell lines with those of other lymphoma or leukemia cell lines using chemokine protein array and ELISA. Chemokine (C-C motif) ligand (CCL) 17 and CCL22 were highly produced by NNKTL cell lines as compared to the other cell lines. In addition, CCL17 and CCL22 were readily observed in the sera of NNKTL patients. The levels of these chemokines were significantly higher in patients than in healthy controls. Furthermore, we detected the expression of CCR4 (the receptor for CCL17 and CCL22) on the surface of NNKTL cell lines and in tissues of NNKTL patients. Anti-CCR4 monoclonal antibody (mAb) efficiently induced antibody-dependent cellular cytotoxicity mediated by natural killer cells against NNKTL cell lines. Our results suggest that CCL17 and CCL22 may be important factors in the development of NNKTL and open up the possibility of immunotherapy of this lymphoma using anti-CCR4 mAb.


Assuntos
Quimiocina CCL17/imunologia , Quimiocina CCL22/imunologia , Imunoterapia/métodos , Linfoma Extranodal de Células T-NK/imunologia , Linfoma Extranodal de Células T-NK/terapia , Receptores CCR4/imunologia , Quimiocinas , Humanos , Linfoma de Células T , Terapia de Alvo Molecular , Transdução de Sinais
18.
J Immunol ; 190(4): 1873-81, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23315072

RESUMO

Adoptive cell transfer (ACT) of ex vivo-activated autologous tumor-reactive T cells is currently one of the most promising approaches for cancer immunotherapy. Recent studies provided some evidence that IL-17-producing CD8(+) (Tc17) cells may exhibit potent antitumor activity, but the specific mechanisms have not been completely defined. In this study, we used a murine melanoma lung-metastasis model and tested the therapeutic effects of gp100-specific polarized type I CD8(+) cytotoxic T (Tc1) or Tc17 cells combined with autologous bone marrow transplantation after total body irradiation. Bone marrow transplantation combined with ACT of antitumor (gp100-specific) Tc17 cells significantly suppressed the growth of established melanoma, whereas Tc1 cells induced long-term tumor regression. After ACT, Tc1 cells maintained their phenotype to produce IFN-γ, but not IL-17. However, although Tc17 cells largely preserved their ability to produce IL-17, a subset secreted IFN-γ or both IFN-γ and IL-17, indicating the plasticity of Tc17 cells in vivo. Furthermore, after ACT, the Tc17 cells had a long-lived effector T cell phenotype (CD127(hi)/KLRG-1(low)) as compared with Tc1 cells. Mechanistically, Tc1 cells mediated antitumor immunity primarily through the direct effect of IFN-γ on tumor cells. In contrast, despite the fact that some Tc17 cells also secreted IFN-γ, Tc17-mediated antitumor immunity was independent of the direct effects of IFN-γ on the tumor. Nevertheless, IFN-γ played a critical role by creating a microenvironment that promoted Tc17-mediated antitumor activity. Taken together, these studies demonstrate that both Tc1 and Tc17 cells can mediate effective antitumor immunity through distinct effector mechanisms, but Tc1 cells are superior to Tc17 cells in mediating tumor regression.


Assuntos
Transferência Adotiva/métodos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Interleucina-17/biossíntese , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/métodos , Transplante de Medula Óssea/patologia , Linfócitos T CD8-Positivos/efeitos da radiação , Citotoxicidade Imunológica/efeitos da radiação , Interleucina-17/efeitos da radiação , Neoplasias Pulmonares/patologia , Ativação Linfocitária/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Irradiação Corporal Total/métodos , Antígeno gp100 de Melanoma/biossíntese , Antígeno gp100 de Melanoma/imunologia
19.
Cancer Immunol Immunother ; 63(5): 469-78, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24633296

RESUMO

Posttranslational modifications regulate the function and stability of proteins, and the immune system is able to recognize some of these modifications. Therefore, the presence of posttranslational modifications increases the diversity of potential immune responses to a determinant antigen. The stimulation of tumor-specific CD4(+) helper T lymphocytes (HTLs) is considered important for the production of anti-tumor antibodies by B cells and for the generation and persistence of CD8(+) cytotoxic T lymphocytes, and in some instances, HTLs can directly reduce tumor cell growth. Identification of MHC class II-restricted peptide epitopes from tumor-associated antigens including those generated from posttranslational protein modifications should enable the improvement of peptide-based cancer immunotherapy. We describe here an MHC class II binding peptide from the tumor protein p53, which possesses an acetylated lysine at position 120 (p53110-124/AcK120) that is effective in eliciting CD4(+) T cell responses specific for the acetylated peptide. Most importantly, the acetylated peptide-reactive CD4 HTLs recognized the corresponding naturally processed posttranslational modified epitope presented by either dendritic cells loaded with tumor cell lysates or directly on tumors expressing p53 and the restricting MHC class II molecules. Treatment of tumor cells with a histone deacetylase inhibitor augmented their recognition by the p53110-124/AcK120-reactive CD4(+) T cells. These findings prove that the epitope p53110-124/AcK120 is immunogenic for anti-tumor responses and is likely to be useful for cancer immunotherapy.


Assuntos
Epitopos de Linfócito T/imunologia , Neoplasias/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Proteína Supressora de Tumor p53/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Western Blotting , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Antígenos de Histocompatibilidade Classe II , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Neoplasias/genética , Processamento de Proteína Pós-Traducional/genética , Proteína Supressora de Tumor p53/genética
20.
J Transl Med ; 12: 265, 2014 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-25240937

RESUMO

BACKGROUND: EGFR-targeted therapy is an attractive option for head and neck squamous cell carcinoma patients. We have recently reported the use of EGFR inhibitors as an adjunct treatment to enhance HLA-DR expression in tumor cells to improve cancer immunotherapy. Nevertheless, we observed that EGFR inhibitors resulted in decreased anti-tumor responses, regardless of upregulation of HLA-DR expression on the tumor cell. In this study, we specifically investigated the mechanisms by which EGFR inhibition modulated anti-tumor responses. METHODS: An EGFR inhibitor erlotinib was used to assess the modulation of anti-tumor responses by tumor antigen-specific helper T cells. We then examined whether administration of the EGFR inhibitor altered tumor cytokine profiles and expression of immune-related molecules on tumor cells. RESULTS: Despite the augmented HLA-DR expression on a gingival cancer cell line by EGFR inhibition, anti-tumor responses of EGFR reactive helper T cell clones against tumor cells were decreased. EGFR inhibition did not change the expression of CD80, CD86, or PD-L1 on the tumor cells. Conversely, production of transforming growth factor beta (TGF-ß) and prostaglandin E2 was increased by EGFR inhibition, indicating that these immunosuppressive molecules were involved in diminishing tumor recognition by T cells. Significantly, attenuation of HTL responses against tumors after EGFR inhibition was reversed by the addition of anti-TGF-ß antibody or COX2 inhibitors. CONCLUSIONS: Targeting TGF-ß and prostaglandin E2 may allow for improved outcomes for cancer patients treated with combination immunotherapy and EGFR inhibitors.


Assuntos
Carcinoma de Células Escamosas/imunologia , Dinoprostona/fisiologia , Receptores ErbB/antagonistas & inibidores , Cloridrato de Erlotinib/uso terapêutico , Neoplasias de Cabeça e Pescoço/imunologia , Fator de Crescimento Transformador beta/fisiologia , Linfócitos T CD4-Positivos/imunologia , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Cloridrato de Erlotinib/farmacologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA