Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Gynecol Oncol ; 145(3): 420-425, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28392127

RESUMO

OBJECTIVES: NY-ESO-1 is a cancer testis antigen and a promising target for immunotherapy. The purpose of this study was to determine the expression frequency, immunogenicity, and clinical impact of NY-ESO-1 in ovarian cancer. METHODS: Immunohistochemistry (IHC), reverse-transcription polymerase chain reaction (RT-PCR), and quantitative-PCR (qRT-PCR) were utilized in an ovarian cancer (including Fallopian tube and primary peritoneal cancers) patient cohort; humoral responses against NY-ESO-1 were determined by ELISA. Clinicopathologic outcomes including progression-free (PFS) and overall (OS) survival were evaluated based on NY-ESO-1 expression. Cohen's kappa (κ) tested agreement between expression tests. RESULTS: NY-ESO-1 expression was detected by any method in 40.7% of 1002 patients' tumors (NY-ESO-1+) and baseline humoral response was identified in 19.0% of 689 tested patients. NY-ESO-1+ patients were older (p<0.001), higher stage (85% stage III/IV vs. 76.4%, p=0.015), less likely to have a complete response to initial therapy (53.9% vs. 68.9%, p=0.002), had more serous histotype (74.5% vs. 66.9%, p=0.011), and had more grade 3 tumors (83.7% vs. 70.8%, p<0.001). There was a trend towards shorter PFS (22.2 vs. 25.0months, p=0.07) and significantly shorter OS (42.9 vs. 50.0months, p=0.003) among NY-ESO-1+ patients. A subset analysis of NY-ESO-1+ patients that received immunotherapy demonstrated improved OS by >2years (52.6 vs. 27.2months, p<0.001). CONCLUSIONS: This study is the first demonstration of an association between NY-ESO-1 expression and an aggressive cancer phenotype. The relatively high expression frequency of NY-ESO-1 in ovarian cancer patients coupled with the poor clinical outcomes in NY-ESO-1+ patients reveals an underappreciated need for targeted therapy against this antigen. In support, our study reveals that NY-ESO-1+ patients enrolled on immunotherapy trials targeting the antigen exhibited an improvement in OS.


Assuntos
Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/imunologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Ensaios Clínicos como Assunto , Feminino , Humanos , Imuno-Histoquímica , Imunoterapia , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Gradação de Tumores , Neoplasias Ovarianas/patologia , Fenótipo , Adulto Jovem
2.
Gynecol Oncol ; 142(1): 158-162, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27103177

RESUMO

OBJECTIVES: To characterize the association between major histocompatibility complex (MHC) types and spontaneous antibody development to the cancer testis (CT) antigen NY-ESO-1. METHODS: Tumor expression of NY-ESO-1 and serum antibodies to NY-ESO-1 were characterized in addition to human leukocyte antigen (HLA) type for patients with epithelial ovarian cancer. HLA types were assigned to structure-based superfamilies and statistical associations were examined. HLA types were compared to existing reference libraries of HLA frequencies in a European-Caucasian American population. RESULTS: Out of 126 patients identified, 81% were expression positive and 48% had spontaneous antibody responses to NY-ESO-1. There was an association between HLA-B superfamily and seropositivity among patients with tumors expressing NY-ESO-1 (p<0.001). The differences in HLA-B superfamily assignment were driven by HLA-B44. Among all patients, the B27 superfamily was over-represented compared with the general population (p<0.001). CONCLUSIONS: HLA type appears to be associated with spontaneous anti-CT antigen antibodies, as well as with the overall risk of ovarian cancer.


Assuntos
Antígenos de Neoplasias/imunologia , Antígeno HLA-B27/imunologia , Antígeno HLA-B44/imunologia , Proteínas de Membrana/imunologia , Neoplasias Ovarianas/imunologia , Idoso , Anticorpos Antineoplásicos/sangue , Anticorpos Antineoplásicos/imunologia , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/sangue , Feminino , Antígeno HLA-B27/biossíntese , Antígeno HLA-B27/sangue , Antígeno HLA-B44/biossíntese , Antígeno HLA-B44/sangue , Haplótipos , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/sangue , Pessoa de Meia-Idade , Neoplasias Ovarianas/sangue
3.
Int J Gynecol Pathol ; 32(6): 536-40, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24071868

RESUMO

Developmentally restricted differentiation antigens or cancer-placental antigens, tastin and bystin, are components of an adhesion molecule that plays a critical role in the implantation of the embryo to the uterus. Cell adhesion molecules have been implicated in the metastasis of carcinomas and could be critical targets for immunotherapy in epithelial ovarian carcinomas (EOCs). Our objectives were to define the expression of tastin and bystin proteins in EOCs. Expression of tastin and bystin mRNA in a panel of human tissues and 70 EOC specimens was investigated using qualitative polymerase chain reaction. Amplification products were confirmed by sequencing. Validation of results was performed using immunohistochemical analysis of tastin and bystin applied on a tissue microarray of 202 EOC tissues. The distribution of tastin and bystin expression and clinicopathologic variables were analyzed. Survival probabilities were estimated using the Kaplan-Meier method and statistical significance was determined by performing the logrank test. Expression of tastin and bystin was restricted to placental and testis tissue by qualitative polymerase chain reaction. Of the 70 EOC specimens tested with polymerase chain reaction, 89% and 94% expressed tastin and bystin, respectively. Immunoexpressions of tastin and bystin protein were observed in 69% and 80 % of the ovarian tumors, respectively. Tastin and bystin expression in Stage I/II disease were 66% and 67% compared with 69% and 81% in Stage III/IV disease, respectively. The tissue-restricted expression of tastin and bystin and their abundant expression in EOCs and advanced-stage disease make these developmentally restricted antigens attractive targets for antigen-specific immunotherapy in EOCs.


Assuntos
Antígenos de Diferenciação/imunologia , Carcinoma/terapia , Imunoterapia/métodos , Neoplasias Ovarianas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma/imunologia , Carcinoma/metabolismo , Moléculas de Adesão Celular/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo
4.
J Immunother Cancer ; 9(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34607898

RESUMO

PURPOSE: Resident memory CD8 T cells, owing to their ability to reside and persist in peripheral tissues, impart adaptive sentinel activity and amplify local immune response, and have beneficial implications for tumor surveillance and control. The current study aimed to clarify the less known chemotactic mechanisms that govern the localization, retention, and residency of memory CD8 T cells in the ovarian tumor microenvironment. EXPERIMENTAL DESIGN: RNA and protein expressions of chemokine receptors in CD8+ resident memory T cells in human ovarian tumor-infiltrating CD8+ T cells and their association with survival were analyzed. The role of CXCR6 on antitumor T cells was investigated using prophylactic vaccine models in murine ovarian cancer. RESULTS: Chemokine receptor profiling of CD8+CD103+ resident memory tumor-infiltrating lymphocytes in patients with ovarian cancer revealed high expression of CXCR6. Analysis of The Cancer Genome Atlas (TCGA) (ovarian cancer database revealed CXCR6 to be associated with CD103 and increased patient survival. Functional studies in mouse models of ovarian cancer revealed that CXCR6 is a marker of resident, but not circulatory, tumor-specific memory CD8+ T cells. CXCR6-deficient tumor-specific CD8+ T cells showed reduced retention in tumor tissues, leading to diminished resident memory responses and poor control of ovarian cancer. CONCLUSIONS: CXCR6, by promoting retention in tumor tissues, serves a critical role in resident memory T cell-mediated immunosurveillance and control of ovarian cancer. Future studies warrant exploiting CXCR6 to promote resident memory responses in cancers.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Monitorização Imunológica/métodos , Neoplasias Ovarianas/genética , Receptores CXCR6/metabolismo , Animais , Feminino , Humanos , Camundongos , Camundongos Knockout , Neoplasias Ovarianas/patologia , Microambiente Tumoral
5.
Mol Ther Oncolytics ; 23: 38-50, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34632049

RESUMO

We have demonstrated that oncolytic vaccinia virus synergizes with doxorubicin (DOX) in inducing immunogenic cell death in platinum-resistant ovarian cancer cells and increases survival in syngeneic and xenograft tumor models. However, the mechanisms underlying the virus- and doxorubicin-mediated cancer cell death remain unknown. In this study, we investigated the effect of the oncolytic virus and doxorubicin used alone or in combination on activation of the cytoplasmic transcription factor CREB3L1 (cyclic AMP [cAMP] response element-binding protein 3-like 1) in ovarian cancer cell lines and clinical specimens. We demonstrated that doxorubicin-mediated cell death in ovarian cancer cell lines was associated with nuclear translocation of CREB3L1 and that the effect was augmented by infection with oncolytic vaccinia virus or treatment with recombinant interferon (IFN)-ß used as a viral surrogate. This combination treatment was also effective in mediating nuclear translocation of CREB3L1 in cancer cells isolated from ovarian tumor biopsies at different stages of disease progression. The measurement of CREB3L1 expression in clinical specimens of ovarian cancer revealed lack of correlation with the stage of disease progression, suggesting that understanding the mechanisms of nuclear accumulation of CREB3L1 after doxorubicin treatment alone or in combination with oncolytic virotherapy may lead to the development of more effective treatment strategies against ovarian cancer.

6.
J Immunother Cancer ; 8(2)2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33177175

RESUMO

BACKGROUND: Immune checkpoint blockers (ICBs) have been approved by the Food and Drug Administration to be used alone in front-line therapies or in combination with other regimens for certain advanced cancers. Since ICB only works in a subset of patients and has limited efficacy in treating ovarian cancer (OVC), developing preclinical models that help to understand which patients may derive benefit from ICB would be of tremendous benefit in OVC. METHODS: Here, we generated preclinical human OVC models from freshly resected tumors, which include six patient-derived xenografts (PDXs) from six different patient tumors, three transplantable OVC PD spheroid lines (PD-sphs), and 3 cell lines (PD-CLs). We tested the therapeutic combination of anti-PD1/CTLA4 antibodies with (1) autologous tumor-associated leukocytes (TALs) on the growth of PD-sphs in a coculture system in vitro, (2) with adoptively transferred autologous peripheral blood mononuclear cells or TALs in patient-derived OVC models using partially humanized mice, NSG-HHDxSGM3 (N-HSGM3). RESULTS: We show that PD-1 and CTLA-4 dual blockade when combined with autologous TALs effectively reduced PD-sph number in a co-culture system and led to regression of established PD-CLs and PDXs in the N-HSGM3 mice. Combinatorial PD-1 and CTLA-4 blockade increased the frequency and function of tumor-specific CD8 T cells. These CD8 T cells persisted in the tumor microenvironment, exhibited memory phenotype and protected animals from tumor growth on tumor rechallenge. Gene expression analysis of tumors resistant to dual PD1/CTLA4 blockade treatment identified upregulation of antigen processing and presentation pathways and downregulation of extracellular matrix organization genes. CONCLUSIONS: These findings describe a novel platform for developing patient-derived preclinical tumor models suitable for rationally testing combinatorial ICB in the context of autologous tumor-reactive T cells. This platform can be further developed for testing additional targeted therapies relevant to OVC.


Assuntos
Imunoterapia/métodos , Neoplasias Ovarianas/tratamento farmacológico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncotarget ; 11(27): 2669-2683, 2020 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-32676168

RESUMO

CD8+ tumor-infiltrating lymphocytes (TILs) are not all specific for tumor antigens, but can include bystander TILs that are specific for cancer-irrelevant epitopes, and it is unknown whether the T-cell repertoire affects prognosis. To delineate the complexity of anti-tumor T-cell responses, we utilized a computational method for de novo assembly of sequences from CDR3 regions of 369 high-grade serous ovarian cancers from TCGA, and then applied deep TCR-sequencing for analyses of paired tumor and peripheral blood specimens from an independent cohort of 99 ovarian cancer patients. Strongly monoclonal T-cell repertoires were associated with favorable prognosis (PFS, HR = 0.65, 0.50-0.84, p = 0.003; OS, HR = 0.61, 0.44-0.83, p = 0.006) in TCGA cohort. In the validation cohort, we discovered that patients with low T-cell infiltration but low diversity or focused repertoires had clinical outcomes almost indistinguishable from highly-infiltrated tumors (median 21.0 months versus 15.9 months, log-rank p = 0.945). We also found that the degree of divergence of the peripheral repertoire from the TIL repertoire, and the presence of detectable spontaneous anti-tumor immune responses are important determinants of clinical outcome. We conclude that the prognostic significance of TILs in ovarian cancer is dictated by T-cell clonality, degree of overlap with peripheral repertoire, and the presence of detectable spontaneous anti-tumor immune response in the patients. These immunological phenotypes defined by the TCR repertoire may provide useful insights for identifying "TIL-low" ovarian cancer patients that may respond to immunotherapy.

8.
Cancer Immun ; 9: 6, 2009 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-19705800

RESUMO

Cancer-embryo antigens or developmentally restricted differentiation antigens (DRDAGs), such as PLAC1 (CT92) and developmental pluripotency associated-2 (DPPA2/CT100), are expressed in pluripotent embryonic cells. They are also recognized as cancer-testis antigens (CT) which are proteins normally expressed only in the human germ line but that are also present in a significant subset of malignant tumors. These antigens may prove to be markers of 'repopulating' cells with stem cell-like characteristics and could be critical targets for immunotherapy in epithelial ovarian cancer (EOC). Our objective was to define the frequency of expression and immunogenicity of PLAC1 and DPPA2 in EOC and correlate expression with clinical outcome. One-step reverse transcriptase PCR was performed on 101 EOC samples and a panel of normal tissues. Expression of PLAC1 and DPPA2 in the EOC specimens was 21/101 (21%) and 31/101 (31%) respectively. In normal tissues, PLAC1 expression was restricted to the placenta while DPPA2 expression was restricted to the placenta and testis. Immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA) were also performed on a subset of specimens. Humoral immunity was demonstrable in 2/12 serum samples from patients whose tumors expressed DPPA2. There was no demonstrable antibody response to PLAC1 in patients with PLAC1 positive tumors. The presence of PLAC1 and DPPA2 did not have a statistically significant effect on recurrence-free and overall survival. The tissue-restricted expression of PLAC1 and DPPA2, their expression in a significant proportion of EOC patients, and their potential to represent markers of stem cells make DRDAGs attractive targets for antigen-specific immunotherapy in EOC.


Assuntos
Biomarcadores Tumorais/sangue , Proteínas Nucleares/sangue , Proteínas Nucleares/genética , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/genética , Proteínas da Gravidez/sangue , Proteínas da Gravidez/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Neoplasias , Biomarcadores Tumorais/imunologia , Proteínas de Ciclo Celular , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas Nucleares/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Proteínas da Gravidez/imunologia , RNA Mensageiro/genética , Taxa de Sobrevida , Fatores de Transcrição
9.
J Immunother Cancer ; 7(1): 189, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31315674

RESUMO

BACKGROUND: Cancer immunotherapies are emerging as promising treatment strategies for ovarian cancer patients that experience disease relapse following first line therapy. As such, identifying strategies to bolster anti-tumor immunity and limit immune suppression, while recognizing diverse patterns of tumor response to immunotherapy is critical to selecting treatment combinations that lead to durable therapeutic benefit. METHODS: Using a pre-clinical mouse model, we evaluated a heterologous prime/boost vaccine in combination with checkpoint blockade to treat metastatic intraperitoneal ovarian cancer. Vaccine-elicited CD8+ T cell responses and changes in the tumor microenvironment following treatment were analyzed and compared to treatment outcome. Kinetics of intraperitoneal tumor growth were assessed using non-invasive magnetic resonance imaging (MRI). RESULTS: Vaccine priming followed by antigen-armed oncolytic Maraba virus boosting elicited robust tumor-specific CD8+ T cell responses that improved tumor control and led to unique immunological changes in the tumor, including a signature that correlated with improved clinical outcome of ovarian cancer patients. However, this treatment was not curative and T cells in the tumor microenvironment (TME) were functionally suppressed. Combination PD-1 blockade partially overcame the adaptive resistance in the tumor observed in response to prime/boost vaccination, restoring CD8+ T cell function in the TME and enhancing the therapeutic response. Non-invasive MRI of tumors during the course of combination treatment revealed heterogeneous radiologic response patterns following treatment, including pseudo-progression, which was associated with improved tumor control prior to relapse. CONCLUSIONS: Our findings point to a key hierarchical role for PD-1 signaling and adaptive immune resistance in the ovarian TME in determining the functional fate of tumor-specific CD8+ T cells, even in the context of robust therapy mediated anti-tumor immunity, as well as the ability of multiple unique patterns of therapeutic response to result in durable tumor control.


Assuntos
Antígenos de Neoplasias/genética , Vacinas Anticâncer/administração & dosagem , Oxirredutases Intramoleculares/genética , Ovalbumina/genética , Neoplasias Ovarianas/terapia , Vesiculovirus/fisiologia , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Oxirredutases Intramoleculares/imunologia , Camundongos , Metástase Neoplásica , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Ovalbumina/imunologia , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/imunologia , Resultado do Tratamento , Microambiente Tumoral , Vesiculovirus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Immunother Cancer ; 7(1): 156, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31221207

RESUMO

BACKGROUND: Efficient identification of neoantigen-specific T-cell responses in epithelial ovarian cancer (EOC) remains a challenge. Existing investigations of spontaneous T-cell response to tumor neoepitope in EOC have taken the approach of comprehensive screening all neoantigen candidates, with a validation rate of 0.5-2%. METHODS: Whole-exome and transcriptome sequencing analysis of treatment-naive EOC patients were performed to identify neoantigen candidates, and the immunogenicity of prioritized neoantigens was evaluated by analyzing spontaneous neoantigen-specfic CD4+ and CD8+ T-cell responses in the tumor and/or peripheral blood. The biological relevance of neoantigen-specific T-cell lines and clones were analyzed by evaluating the capacity of autologous ovarian tumor recognition. Genetic transfer of T-cell receptor (TCR) from these neoantigen-specific T-cell clones into peripheral blood T-cells was conducted to generate neoepitope-specific T-cells. The molecular signature associated with positive neoantigen T-cell responses was investigated, and the impacts of expression level and lymphocyte source on neoantigen identification were explored. RESULTS: Using a small subset of prioritized neoantigen candidates, we were able to detect spontaneous CD4+ and/or CD8+ T-cell responses against neoepitopes from autologous lymphocytes in half of treatment-naïve EOC patients, with a significantly improved validation rate of 19%. Tumors from patients exhibiting neoantigen-specific T-cell responses exhibited a signature of upregulated antigen processing and presentation machinery, which was also associated with favorable patient survival in the TCGA ovarian cohort. T-cells specific against two mutated cancer-associated genes, NUP214 and JAK1, recognized autologous tumors. Gene-engineering with TCR from these neoantigen-specific T-cell clones conferred neoantigen-reactivity to peripheral T-cells. CONCLUSIONS: Our study demonstrated the feasibility of efficiently identifying both CD4+ and CD8+ neoantigen-specific T-cells in EOC. Autologous lymphocytes genetically engineered with tumor antigen-specific TCR can be used to generate cells for use in the personalized adoptive T-cell transfer immunotherapy.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Epitelial do Ovário/imunologia , Imunoterapia/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Feminino , Humanos
11.
Cancer Immunol Res ; 6(5): 594-604, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29588318

RESUMO

T cells genetically engineered with tumor antigen-specific T-cell receptor (TCR) genes have demonstrated therapeutic potential in patients with solid tumors. In order to achieve broader application, an efficient method to identify TCR genes for an array of tumor antigens and HLA restriction elements is required. Here, we have developed a method to construct a TCR-expression library from specimens, including frozen tumor biopsies, that contain antigen-specific T cells. TCR-expressing cassettes were constructed and cloned in a retroviral plasmid vector within 24 hours by unbiased PCR amplification of TCR α and ß chain variable regions assembled with TCR constant regions. The method was validated by constructing TCR-expressing vectors from tumor antigen-specific T-cell clones and functionally assessing TCR gene-transduced T cells. We applied this method to frozen ovarian tumor specimens that were infiltrated by tumor antigen-specific T cells. The tumor-derived TCR libraries were expressed in peripheral T cells from healthy volunteers and screened for tumor antigen-specific TCR pairs with the use of an MHC/peptide tetramer reagent. Tumor antigen-specific TCR-expressing transgenes were recovered from isolated tetramer-positive T cells. Peripheral T cells that were engineered with library-derived TCR gene showed potent therapeutic antitumor effect in a tumor xenograft model. Our method can efficiently and rapidly provide tumor-specific TCR-expressing viral vectors for the manufacture of therapeutic and personalized antitumor T-cell products. Cancer Immunol Res; 6(5); 594-604. ©2018 AACR.


Assuntos
Engenharia Celular/métodos , Biblioteca Gênica , Terapia Genética/métodos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/transplante , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Células Cultivadas , Clonagem Molecular/métodos , Feminino , Secções Congeladas , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Receptores de Antígenos de Linfócitos T/metabolismo , Retroviridae/genética , Linfócitos T/metabolismo , Transdução Genética
12.
Oncoimmunology ; 6(1): e1249561, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28197366

RESUMO

Tumor-associated or -infiltrating lymphocytes (TALs or TILs) co-express multiple immune inhibitory receptors that contribute to immune suppression in the ovarian tumor microenvironment (TME). Dual blockade of PD-1 along with LAG-3 or CTLA-4 has been shown to synergistically enhance T-cell effector function, resulting in a delay in murine ovarian tumor growth. However, the mechanisms underlying this synergy and the relative contribution of other inhibitory receptors to immune suppression in the ovarian TME are unknown. Here, we report that multiple immune checkpoints are expressed in TALs and TILs isolated from ovarian tumor-bearing mice. Importantly, blockade of PD-1, LAG-3, or CTLA-4 alone using genetic ablation or blocking antibodies conferred a compensatory upregulation of the other checkpoint pathways, potentiating their capacity for local T-cell suppression that, in turn, could be overcome through combinatorial blockade strategies. Whereas single-agent blockade led to tumor outgrowth in all animals, dual antibody blockade against PD-1/CTLA-4 or triple blockade against PD-1/LAG-3/CTLA-4 resulted in tumor-free survival in 20% of treated mice. In contrast, dual blockade of LAG-3 and CTLA-4 pathways using PD-1 knockout mice led to tumor-free survival in 40% of treated mice, suggesting a hierarchical ordering of checkpoint function. Durable antitumor immunity was most strongly associated with increased numbers of CD8+ T cells, the frequency of cytokine-producing effector T cells, reduced frequency of Tregs and arginine-expressing monocytic myeloid-derived suppressor cells in the peritoneal TME. These data provide a basis for combinatorial checkpoint blockade in clinical intervention for ovarian cancer.

13.
PLoS One ; 9(8): e104099, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25101620

RESUMO

The MAGE cancer-testis antigens (CTA) are attractive candidates for immunotherapy. The aim of this study was to determine the frequency of expression, humoral immunity and prognostic significance of MAGE CTA in human epithelial ovarian cancer (EOC). mRNA or protein expression frequencies were determined for MAGE-A1, -A3, -A4, -A10 and -C1 (CT7) in tissue samples obtained from 400 patients with EOC. The presence of autologous antibodies against the MAGE antigens was determined from 285 serum samples. The relationships between MAGE expression, humoral immunity to MAGE antigens, and clinico-pathologic characteristics were studied. The individual frequencies of expression were as follows: A1: 15% (42/281), A3: 36% (131/390), A4: 47% (186/399), A10: 52% (204/395), C1: 16% (42/267). Strong concordant expression was noted with MAGE-A1:-A4, MAGE-A1:-C1 and MAGE-A4:-A10 (p<0.0005). Expression of MAGE-A1 or -A10 antigens resulted in poor progression free survival (PFS) (OR 1.44, CI 1.01-2.04, p = 0.044 and OR 1.3, CI 1.03-1.64, p = 0.03, respectively); whereas, MAGE-C1 expression was associated with improved PFS (OR 0.62, CI 0.42-0.92, p = 0.016). The improved PFS observed for MAGE-C1 expression, was diminished by co-expression of MAGE-A1 or -A10. Spontaneous humoral immunity to the MAGE antigens was present in 9% (27/285) of patients, and this predicted poor overall survival (log-rank test p = 0.0137). These findings indicate that MAGE-A1, MAGE-A4, MAGE-A3, and MAGE-A10 are priority attractive targets for polyvalent immunotherapy in ovarian cancer patients.


Assuntos
Antígenos de Neoplasias/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias Ovarianas/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Neoplasias/metabolismo , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunidade Humoral , Imuno-Histoquímica , Imunoterapia , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Cancer Immunol Res ; 2(1): 37-49, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24535937

RESUMO

The cancer-testis/cancer-germline antigen NY-ESO-1 is a vaccine target in epithelial ovarian cancer (EOC), but its limited expression is a barrier to vaccine efficacy. As NY-ESO-1 is regulated by DNA methylation, we hypothesized that DNA methyltransferase (DNMT) inhibitors may augment NY-ESO-1 vaccine therapy. In agreement, global DNA hypomethylation in EOC was associated with the presence of circulating antibodies to NY-ESO-1. Pre-clinical studies using EOC cell lines showed that decitabine treatment enhanced both NY-ESO-1 expression and NY-ESO-1-specific CTL-mediated responses. Based on these observations, we performed a phase I dose-escalation trial of decitabine, as an addition to NY-ESO-1 vaccine and doxorubicin liposome (doxorubicin) chemotherapy, in 12 patients with relapsed EOC. The regimen was safe, with limited and clinically manageable toxicities. Both global and promoter-specific DNA hypomethylation occurred in blood and circulating DNAs, the latter of which may reflect tumor cell responses. Increased NY-ESO-1 serum antibodies and T cell responses were observed in the majority of patients, and antibody spreading to additional tumor antigens was also observed. Finally, disease stabilization or partial clinical response occurred in 6/10 evaluable patients. Based on these encouraging results, evaluation of similar combinatorial chemo-immunotherapy regimens in EOC and other tumor types is warranted.


Assuntos
Antígenos de Neoplasias/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Vacinas Anticâncer/imunologia , Epigênese Genética/efeitos dos fármacos , Proteínas de Membrana/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Adulto , Idoso , Antígenos de Neoplasias/genética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Vacinas Anticâncer/genética , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Metilação de DNA , Decitabina , Doxorrubicina/administração & dosagem , Doxorrubicina/análogos & derivados , Feminino , Expressão Gênica , Humanos , Imunidade Humoral , Imunoterapia Ativa , Elementos Nucleotídeos Longos e Dispersos , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/imunologia , Neoplasias Epiteliais e Glandulares/terapia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Polietilenoglicóis/administração & dosagem , Subpopulações de Linfócitos T/imunologia , Resultado do Tratamento
15.
Int J Oncol ; 41(3): 1139-47, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22684412

RESUMO

SEREX has proven to be a powerful method that takes advantage of the presence of spontaneous humoral immune response in some cancer patients. In this study, immunoscreening of normal testis and two ovarian cancer cell line cDNA expression libraries with sera from ovarian cancer patients led to the isolation of 75 independent antigens, designated KP-OVA-1 through KP-OVA-75. Of these, RT-PCR showed KP-OVA-52 to be expressed strongly in normal testis, in ovarian cancer cell lines (3/9) and in ovarian cancer tissues (1/17). The expression of KP-OVA-52 in cancer cells is also induced by the demethylating agent 5­aza­2'­deoxycytidine (ADC). To test immunogenicity, we used the Serum Antibody Detection Assay (SADA) to analyze anti-IgG antibodies against the 75 antigens that were initially isolated by SEREX. Four of the 75 antigens (KP­OVA­25, KP­OVA­35, KP­OVA­68 and KP­OVA­73) reacted exclusively with sera from cancer patients. However, KP­OVA­52 reacted with 1 of 20 ovarian cancer sera. These data suggest that the KP-OVA-52 can be considered a novel CT antigen that is regulated by DNA methylation.


Assuntos
Antígenos de Neoplasias/metabolismo , Metilação de DNA , Neoplasias Ovarianas/imunologia , Neoplasias Testiculares/imunologia , Anticorpos Antineoplásicos , Antígenos de Neoplasias/sangue , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Linhagem Celular Tumoral , Decitabina , Feminino , Biblioteca Gênica , Humanos , Masculino , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/genética , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/imunologia
16.
PLoS One ; 7(6): e38783, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761704

RESUMO

PURPOSE: NY-ESO-1 cancer testis (CT) antigen is an attractive candidate for immunotherapy as a result of its high immunogenicity. The aim of this study was to explore the potential for NY-ESO-1 antigen directed immunotherapy in triple negative breast cancer (TNBC) by determining the frequency of expression by immunohistochemistry (IHC) and the degree of inherent immunogenicity to NY-ESO-1. EXPERIMENTAL DESIGN: 168 TNBC and 47 ER+/HER2- primary breast cancer specimens were used to determine NY-ESO-1 frequency by IHC. As previous studies have shown that patients with a robust innate humoral immune response to CT antigens are more likely to develop CD8 T-cell responses to NY-ESO-1 peptides, we evaluated the degree to which patients with NY-ESO-1 expression had inherent immunogenicity by measuring antibodies. The relationship between NY-ESO-1 expression and CD8+ T lymphocytes was also examined. RESULTS: The frequency of NY-ESO-1 expression in the TNBC cohort was 16% versus 2% in ER+/HER2- patients. A higher NY-ESO-1 score was associated with a younger age at diagnosis in the TNBC patients with NY-ESO-1 expression (p = 0.026). No differences in OS (p = 0.278) or PFS (p = 0.238) by NY-ESO-1 expression status were detected. Antibody responses to NY-ESO-1 were found in 73% of TNBC patients whose tumors were NY-ESO-1 positive. NY-ESO-1 positive patients had higher CD8 counts than negative patients (p = 0.018). CONCLUSION: NY-ESO-1 is expressed in a substantial subset of TNBC patients and leads to a high humoral immune response in a large proportion of these individuals. Given these observations, patients with TNBC may benefit from targeted therapies directed against NY-ESO-1.


Assuntos
Anticorpos Antineoplásicos/sangue , Antígenos de Neoplasias/imunologia , Neoplasias da Mama/imunologia , Carcinoma Ductal de Mama/imunologia , Carcinoma Lobular/imunologia , Proteínas de Membrana/metabolismo , Recidiva Local de Neoplasia/imunologia , Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Linfócitos T CD8-Positivos , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/mortalidade , Carcinoma Lobular/metabolismo , Carcinoma Lobular/mortalidade , Metilação de DNA , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Proteínas de Membrana/imunologia , Pessoa de Meia-Idade , Gradação de Tumores , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/mortalidade , Estadiamento de Neoplasias , Prognóstico , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Taxa de Sobrevida
17.
PLoS One ; 6(8): e23651, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21858191

RESUMO

Analyses of NY-ESO-1-specific spontaneous immune responses in cancer patients revealed that antibody and both CD4(+) and CD8(+) T cell responses were induced together in cancer patients. To explore whether such integrated immune responses are also spontaneously induced for other tumor antigens, we have evaluated antibody and T cell responses against self/tumor antigen p53 in ovarian cancer patients and healthy individuals. We found that 21% (64/298) of ovarian cancer patients but no healthy donors showed specific IgG responses against wild-type p53 protein. While none of 12 patients with high titer p53 antibody showed spontaneous p53-specific CD8(+) T cell responses following a single in vitro sensitization, significant p53-specific IFN-γ producing CD4(+) T cells were detected in 6 patients. Surprisingly, similar levels of p53-specific CD4(+) T cells but not CD8(+) T cells were also detected in 5/10 seronegative cancer patients and 9/12 healthy donors. Importantly, p53-specific CD4(+) T cells in healthy donors originated from a CD45RA(-) antigen-experienced T cell population and recognized naturally processed wild-type p53 protein. These results raise the possibility that p53-specific CD4(+) T cells reflect abnormalities in p53 occurring in normal individuals and that they may play a role in processes of immunosurveillance or immunoregulation of p53-related neoplastic events.


Assuntos
Antígenos de Neoplasias/imunologia , Autoantígenos/imunologia , Tolerância Imunológica/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Antígenos de Neoplasias/metabolismo , Autoanticorpos/sangue , Autoanticorpos/imunologia , Autoantígenos/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/metabolismo , Masculino , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Mutação , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Peptídeos/genética , Peptídeos/imunologia , Peptídeos/metabolismo , Linfócitos T/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA