Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
JCI Insight ; 3(13)2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-29997287

RESUMO

BACKGROUND: Immune checkpoint inhibitors provide significant clinical benefit to a subset of patients, but novel prognostic markers are needed to predict which patients will respond. This study was initiated to determine if features of patient T cell repertoires could provide insights into the mechanisms of immunotherapy, while also predicting outcomes. METHODS: We examined T cell receptor (TCR) repertoires in peripheral blood of 25 metastatic pancreatic cancer patients treated with ipilimumab with or without GVAX (a pancreatic cancer vaccine), as well as peripheral blood and tumor biopsies from 32 patients treated with GVAX and mesothelin-expressing Listeria monocytogenes with or without nivolumab. Statistics from these repertoires were then tested for their association with clinical response and treatment group. RESULTS: We demonstrate that, first, the majority of patients receiving these treatments experience a net diversification of their peripheral TCR repertoires. Second, patients receiving ipilimumab experienced larger changes in their repertoires, especially in combination with GVAX. Finally, both a low baseline clonality and a high number of expanded clones following treatment were associated with significantly longer survival in patients who received ipilimumab but not in patients receiving nivolumab. CONCLUSIONS: We show that these therapies have measurably different effects on the peripheral repertoire, consistent with their mechanisms of action, and demonstrate the potential for TCR repertoire profiling to serve as a biomarker of clinical response in pancreatic cancer patients receiving immunotherapy. In addition, our results suggest testing sequential administration of anti-CTLA-4 and anti-PD-1 antibodies to achieve optimal therapeutic benefit. TRIAL REGISTRATION: Samples used in this study were collected from the NCT00836407 and NCT02243371 clinical trials. FUNDING: Research supported by a Stand Up To Cancer Lustgarten Foundation Pancreatic Cancer Convergence Dream Team Translational Research grant (SU2C-AACR-DT14-14). Stand Up To Cancer is a program of the Entertainment Industry Foundation administered by the American Association for Cancer Research (AACR). Additional clinical trial funding was provided by AACR-Pancreatic Cancer Action Network Research Acceleration Network grant (14-90-25-LE), NCI SPORE in GI Cancer (CA062924), Quick-Trials for Novel Cancer Therapies: Exploratory Grants (R21CA126058-01A2), and the US Food and Drug Administration (R01FD004819). Research collaboration and financial support were provided by Adaptive Biotechnologies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Carcinoma Ductal Pancreático/imunologia , Imunoterapia/métodos , Neoplasias Pancreáticas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/sangue , Antígeno CTLA-4/imunologia , Vacinas Anticâncer/uso terapêutico , Carcinoma Ductal Pancreático/terapia , Proteínas Ligadas por GPI/uso terapêutico , Humanos , Ipilimumab/uso terapêutico , Estimativa de Kaplan-Meier , Mesotelina , Nivolumabe/uso terapêutico , Neoplasias Pancreáticas/terapia , Receptor de Morte Celular Programada 1/imunologia , Estados Unidos , United States Food and Drug Administration , Neoplasias Pancreáticas
3.
Cell Rep ; 19(1): 203-217, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28380359

RESUMO

Here, we describe a multiplexed immunohistochemical platform with computational image processing workflows, including image cytometry, enabling simultaneous evaluation of 12 biomarkers in one formalin-fixed paraffin-embedded tissue section. To validate this platform, we used tissue microarrays containing 38 archival head and neck squamous cell carcinomas and revealed differential immune profiles based on lymphoid and myeloid cell densities, correlating with human papilloma virus status and prognosis. Based on these results, we investigated 24 pancreatic ductal adenocarcinomas from patients who received neoadjuvant GVAX vaccination and revealed that response to therapy correlated with degree of mono-myelocytic cell density and percentages of CD8+ T cells expressing T cell exhaustion markers. These data highlight the utility of in situ immune monitoring for patient stratification and provide digital image processing pipelines to the community for examining immune complexity in precious tissue sections, where phenotype and tissue architecture are preserved to improve biomarker discovery and assessment.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Escamosas/imunologia , Neoplasias de Cabeça e Pescoço/imunologia , Citometria por Imagem/métodos , Processamento de Imagem Assistida por Computador , Monitorização Imunológica/métodos , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Estudos de Coortes , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Prognóstico , Estatísticas não Paramétricas , Análise Serial de Tecidos
4.
Nat Rev Cancer ; 17(4): 209-222, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28233802

RESUMO

The past decade of cancer research has been marked by a growing appreciation of the role of immunity in cancer. Mutations in the tumour genome can cause tumours to express mutant proteins that are tumour specific and not expressed on normal cells (neoantigens). These neoantigens are an attractive immune target because their selective expression on tumours may minimize immune tolerance as well as the risk of autoimmunity. In this Review we discuss the emerging evidence that neoantigens are recognized by the immune system and can be targeted to increase antitumour immunity. We also provide a framework for personalized cancer immunotherapy through the identification and selective targeting of individual tumour neoantigens, and present the potential benefits and obstacles to this approach of targeted immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia/métodos , Neoplasias/terapia , Transferência Adotiva , Animais , Humanos , Mutação , Neoplasias/imunologia , Medicina de Precisão , Linfócitos T/imunologia , Evasão Tumoral , Microambiente Tumoral
5.
J Clin Oncol ; 33(12): 1325-33, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25584002

RESUMO

PURPOSE: GVAX pancreas, granulocyte-macrophage colony-stimulating factor-secreting allogeneic pancreatic tumor cells, induces T-cell immunity to cancer antigens, including mesothelin. GVAX is administered with low-dose cyclophosphamide (Cy) to inhibit regulatory T cells. CRS-207, live-attenuated Listeria monocytogenes-expressing mesothelin, induces innate and adaptive immunity. On the basis of preclinical synergy, we tested prime/boost vaccination with GVAX and CRS-207 in pancreatic adenocarcinoma. PATIENTS AND METHODS: Previously treated patients with metastatic pancreatic adenocarcinoma were randomly assigned at a ratio of 2:1 to two doses of Cy/GVAX followed by four doses of CRS-207 (arm A) or six doses of Cy/GVAX (arm B) every 3 weeks. Stable patients were offered additional courses. The primary end point was overall survival (OS) between arms. Secondary end points were safety and clinical response. RESULTS: A total of 90 patients were treated (arm A, n = 61; arm B, n = 29); 97% had received prior chemotherapy; 51% had received ≥ two regimens for metastatic disease. Mean number of doses (± standard deviation) administered in arms A and B were 5.5 ± 4.5 and 3.7 ± 2.2, respectively. The most frequent grade 3 to 4 related toxicities were transient fevers, lymphopenia, elevated liver enzymes, and fatigue. OS was 6.1 months in arm A versus 3.9 months in arm B (hazard ratio [HR], 0.59; P = .02). In a prespecified per-protocol analysis of patients who received at least three doses (two doses of Cy/GVAX plus one of CRS-207 or three of Cy/GVAX), OS was 9.7 versus 4.6 months (arm A v B; HR, 0.53; P = .02). Enhanced mesothelin-specific CD8 T-cell responses were associated with longer OS, regardless of treatment arm. CONCLUSION: Heterologous prime/boost with Cy/GVAX and CRS-207 extended survival for patients with pancreatic cancer, with minimal toxicity.


Assuntos
Vacinas Anticâncer/administração & dosagem , Carcinoma Ductal Pancreático/terapia , Ciclofosfamida/administração & dosagem , Proteínas Ligadas por GPI/biossíntese , Listeria monocytogenes/metabolismo , Neoplasias Pancreáticas/terapia , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/efeitos adversos , Vacinas Anticâncer/efeitos adversos , Carcinoma Ductal Pancreático/imunologia , Terapia Combinada , Ciclofosfamida/efeitos adversos , Feminino , Proteínas Ligadas por GPI/genética , Humanos , Listeria monocytogenes/genética , Masculino , Mesotelina , Pessoa de Meia-Idade , Neoplasias Pancreáticas/imunologia , Taxa de Sobrevida , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
6.
Cancer Prev Res (Phila) ; 7(11): 1072-80, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25245763

RESUMO

Cancer immunotherapy is a rapidly developing field, but limited in its success by a high tumor burden and immune tolerance. In contrast, immunoprevention has the potential to prevent cancer before the development of immune tolerance, and to prevent cancer recurrence in the setting of minimal residual disease. Although immunoprevention for viral-induced cancers has been successful in the setting of hepatitis B and human papillomavirus vaccination, primary prevention of nonviral-induced cancers is in its infancy. In contrast, prevention of cancer recurrence after adjuvant treatment (secondary prevention) is gaining steam. This review provides an overview of the scope of research in cancer immunoprevention over the last three years and directions for future research.


Assuntos
Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Animais , Antígenos de Neoplasias/imunologia , Biomarcadores , Vacinas Anticâncer/imunologia , Transformação Celular Neoplásica , Ensaios Clínicos como Assunto , Humanos , Tolerância Imunológica , Camundongos , Recidiva Local de Neoplasia/imunologia
7.
Cancer Immunol Res ; 2(7): 616-31, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24942756

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is considered a "nonimmunogenic" neoplasm. Single-agent immunotherapies have failed to demonstrate significant clinical activity in PDAC and other "nonimmunogenic" tumors, in part due to a complex tumor microenvironment (TME) that provides a formidable barrier to immune infiltration and function. We designed a neoadjuvant and adjuvant clinical trial comparing an irradiated, granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic PDAC vaccine (GVAX) given as a single agent or in combination with low-dose cyclophosphamide to deplete regulatory T cells (Treg) as a means to study how the TME is altered by immunotherapy. Examination of resected PDACs revealed the formation of vaccine-induced intratumoral tertiary lymphoid aggregates in 33 of 39 patients 2 weeks after vaccine treatment. Immunohistochemical analysis showed these aggregates to be regulatory structures of adaptive immunity. Microarray analysis of microdissected aggregates identified gene-expression signatures in five signaling pathways involved in regulating immune-cell activation and trafficking that were associated with improved postvaccination responses. A suppressed Treg pathway and an enhanced Th17 pathway within these aggregates were associated with improved survival, enhanced postvaccination mesothelin-specific T-cell responses, and increased intratumoral Teff:Treg ratios. This study provides the first example of immune-based therapy converting a "nonimmunogenic" neoplasm into an "immunogenic" neoplasm by inducing infiltration of T cells and development of tertiary lymphoid structures in the TME. Post-GVAX T-cell infiltration and aggregate formation resulted in the upregulation of immunosuppressive regulatory mechanisms, including the PD-1-PD-L1 pathway, suggesting that patients with vaccine-primed PDAC may be better candidates than vaccine-naïve patients for immune checkpoint and other immunomodulatory therapies.


Assuntos
Adenocarcinoma/terapia , Vacinas Anticâncer/uso terapêutico , Neoplasias Pancreáticas/terapia , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/uso terapêutico , Agregação Celular/imunologia , Quimioterapia Adjuvante , Ciclofosfamida/administração & dosagem , Ciclofosfamida/uso terapêutico , Esquema de Medicação , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Interferon gama/biossíntese , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Linfócitos T Reguladores/imunologia , Regulação para Cima/imunologia , Neoplasias Pancreáticas
9.
Oncoimmunology ; 3(11): e962401, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25941589

RESUMO

Single agent immunotherapy is effective against several cancers, but has failed against poorly immunogenic cancers, including pancreatic cancer. Evaluation of pancreatic tumors following treatment with an experimental vaccine (Lutz et al. Cancer Immunology Research 2014) suggests that vaccination primes the tumor microenvironment (TME) for checkpoint-inhibitor immunotherapy, and supports a new platform for evaluating checkpoint-inhibitors in poorly immunogenic cancers.

10.
Blood ; 111(1): 251-9, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909080

RESUMO

Tissue-derived adenosine, acting via the adenosine A(2A) receptor (A(2A)R), is emerging as an important negative regulator of T-cell function. In this report, we demonstrate that A(2A)R stimulation not only inhibits the generation of adaptive effector T cells but also promotes the induction of adaptive regulatory T cells. In vitro, antigen recognition in the setting of A(2A)R engagement induces T-cell anergy, even in the presence of costimulation. T cells initially stimulated in the presence of an A(2A)R agonist fail to proliferate and produce interleukin-2 and interferon (IFN)-gamma when rechallenged in the absence of A(2A)R stimulation. Likewise, in an in vivo model of autoimmunity, tissue-derived adenosine promotes anergy and abrogates tissue destruction. Indeed, A(2A)R stimulation inhibits interleukin-6 expression while enhancing the production of transforming growth factor-beta. Accordingly, treating mice with A(2A)R agonists not only inhibits Th1 and Th17 effector cell generation but also promotes the generation of Foxp3(+) and LAG-3(+) regulatory T cells. In this regard, A(2A)R agonists fail to prevent autoimmunity by LAG-3(-/-) clonotypic T cells, implicating an important role for LAG-3 in adenosine-mediated peripheral tolerance. Overall, our findings demonstrate that extracellular adenosine stimulates the A(2A)R to promote long-term T-cell anergy and the generation of adaptive regulatory T cells.


Assuntos
Tolerância Imunológica/fisiologia , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Linfócitos T Reguladores/imunologia , Adenosina/metabolismo , Transferência Adotiva , Animais , Antígenos CD/metabolismo , Linhagem Celular , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Transcrição NFATC/metabolismo , Proteínas ras/metabolismo , Proteína do Gene 3 de Ativação de Linfócitos
11.
J Immunol ; 176(2): 974-83, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16393983

RESUMO

T cell costimulation via OX40 is known to increase CD4+ T cell expansion and effector function and enhances the development of T cell memory. OX40 costimulation can also prevent, and even reverse, CD4+ T cell anergy. However, the role of OX40 in CD8+ T cell function is less well defined, particularly in the setting of immune tolerance. To determine the effects of OX40 costimulation on the induction of the host CD8+ T cell repertoire to an endogenous tumor Ag, we examined the fate of CD8+ T cells specific for the immunodominant rat HER-2/neu epitope, RNEU420-429, in FVB MMTV-neu (neu-N) mice, which express rat HER-2/neu protein in a predominantly mammary-restricted fashion. We show that the RNEU420-429-specific T cell repertoire in neu-N mice expands transiently after vaccination with a neu-targeted GM-CSF-secreting whole-cell vaccine, but quickly declines to an undetectable level. However, OX40 costimulation, when combined with GM-CSF-secreting tumor-targeted vaccination, can break established CD8+ T cell tolerance in vivo by enhancing the expansion, and prolonging the survival and effector function of CD8+ T cells specific for RNEU420-429. Moreover, we demonstrate that OX40 expression is up-regulated on both CD4+ and CD8+ T cells shortly after administration of a GM-CSF expressing vaccine. These studies highlight the increased efficacy of OX40 costimulation when combined with a GM-CSF-secreting vaccine, and define a new role for OX40 costimulation of CD8+ T cells in overcoming tolerance and boosting antitumor immunity.


Assuntos
Antígenos de Neoplasias , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Glicoproteínas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Receptores do Fator de Necrose Tumoral/administração & dosagem , Animais , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Feminino , Tolerância Imunológica , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Fragmentos de Peptídeos/imunologia , Ratos , Receptor ErbB-2 , Receptores OX40 , Proteínas Recombinantes , Linfócitos T Auxiliares-Indutores/imunologia
12.
J Exp Med ; 200(3): 297-306, 2004 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-15289501

RESUMO

Tumor-specific CD8(+) T cells can potentially be activated by two distinct mechanisms of major histocompatibility complex class I-restricted antigen presentation as follows: direct presentation by tumor cells themselves or indirect presentation by professional antigen-presenting cells (APCs). However, controversy still exists as to whether indirect presentation (the cross-priming mechanism) can contribute to effective in vivo priming of tumor-specific CD8(+) T cells that are capable of eradicating cancer in patients. A clinical trial of vaccination with granulocyte macrophage-colony stimulating factor-transduced pancreatic cancer lines was designed to test whether cross-presentation by locally recruited APCs can activate pancreatic tumor-specific CD8(+) T cells. Previously, we reported postvaccination delayed-type hypersensitivity (DTH) responses to autologous tumor in 3 out of 14 treated patients. Mesothelin is an antigen demonstrated previously by gene expression profiling to be up-regulated in most pancreatic cancers. We report here the consistent induction of CD8(+) T cell responses to multiple HLA-A2, A3, and A24-restricted mesothelin epitopes exclusively in the three patients with vaccine-induced DTH responses. Importantly, neither of the vaccinating pancreatic cancer cell lines expressed HLA-A2, A3, or A24. These results provide the first direct evidence that CD8 T cell responses can be generated via cross-presentation by an immunotherapy approach designed to recruit APCs to the vaccination site.


Assuntos
Células Apresentadoras de Antígenos/fisiologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Glicoproteínas de Membrana/imunologia , Neoplasias Pancreáticas/imunologia , Linhagem Celular , Proteínas Ligadas por GPI , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Antígeno HLA-A2/análise , Antígeno HLA-A3/análise , Antígeno HLA-A3/fisiologia , Humanos , Hipersensibilidade Tardia/etiologia , Mesotelina , Neoplasias Pancreáticas/terapia , Vacinação
13.
J Immunol ; 171(4): 2161-9, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12902523

RESUMO

HER-2/neu is overexpressed in several cancers including 30% of breast carcinomas, and correlates with a poor outcome. HER-2/neu-transgenic (neu-N) mice that overexpress the non-transforming rat neu develop spontaneous mammary carcinomas and demonstrate immunotolerance to the neu protein similar to that observed in patients with neu-expressing cancers. In neu-N mice, neu-targeted vaccination induces weak T cell and negligible Ab responses sufficient to delay but not eradicate transplanted neu-expressing tumor. Here we demonstrate that passive infusion of neu-specific mAbs in sequence with whole cell vaccination significantly improves tumor-free survival over either modality alone. Importantly, treatment of neu-N mice with vaccine in combination with two distinct neu-specific Abs is particularly efficacious, preventing tumor in 70% and eradicating established tumor in 30% of neu-N mice. In vivo lymphocyte subpopulation depletion experiments demonstrate that the efficacy of Ab, alone or combined with vaccine, is dependent on both CD4(+) and CD8(+) T cells. Furthermore, the in vivo antitumor effects of vaccine and Ab are associated with a significant increase in the number and function of neu-specific CD8(+) T cells. Collectively, these observations suggest that similarly increased efficacy could be obtained by combining neu-targeted vaccination and neu-specific Abs such as trastuzumab (Herceptin) in patients with neu-expressing cancers.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Epitopos de Linfócito T/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/genética , Células 3T3 , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/imunologia , Feminino , Esquemas de Imunização , Injeções Subcutâneas , Contagem de Linfócitos , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/mortalidade , Camundongos , Camundongos Transgênicos , Ratos , Receptor ErbB-2/biossíntese , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Análise de Sobrevida , Ubiquitinas/metabolismo , Regulação para Cima/genética , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...