Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 26(5): 1045-1053, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31757875

RESUMO

PURPOSE: Patients with HER2+ breast cancer benefit from trastuzumab-containing regimens with improved survival. Adaptive immunity, including cytotoxic T-cell and antibody immunity, is critical to clinical efficacy of trastuzumab. Because Th cells are central to the activation of these antitumor effectors, we reason that HER2 patients treated with trastuzumab may benefit by administering vaccines that are designed to stimulate Th-cell immunity. PATIENTS AND METHODS: We developed a degenerate HER2 epitope-based vaccine consisting of four HLA class II-restricted epitopes mixed with GM-CSF that should immunize most (≥84%) patients. The vaccine was tested in a phase I trial. Eligible women had resectable HER2+ breast cancer and had completed standard treatment prior to enrollment and were disease free. Patients were vaccinated monthly for six doses and monitored for safety and immunogenicity. RESULTS: Twenty-two subjects were enrolled and 20 completed all six vaccines. The vaccine was well tolerated. All patients were alive at analysis with a median follow-up of 2.3 years and only two experienced disease recurrence. The percent of patients that responded with augmented T-cell immunity was high for each peptide ranging from 68% to 88%, which led to 90% of the patients generating T cells that recognized naturally processed HER2 antigen. The vaccine also augmented HER2-specific antibody. Immunity was sustained in patients with little sign of diminishing at 2 years following the vaccination. CONCLUSIONS: Degenerate HLA-DR-based HER2 vaccines induce sustainable HER2-specific T cells and antibodies. Future studies, could evaluate whether vaccination during adjuvant treatment with trastuzumab-containing regimens improves patient outcomes.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Vacinas Anticâncer/uso terapêutico , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Peptídeos/imunologia , Receptor ErbB-2/imunologia , Linfócitos T/imunologia , Adulto , Antineoplásicos Imunológicos/uso terapêutico , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Vacinas Anticâncer/imunologia , Feminino , Humanos , Pessoa de Meia-Idade , Segurança do Paciente , Peptídeos/química , Receptor ErbB-2/antagonistas & inibidores , Trastuzumab/uso terapêutico , Resultado do Tratamento
2.
Cancer Res ; 77(23): 6667-6678, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28993412

RESUMO

Ligation of programmed cell death-1 (PD-1) in the tumor microenvironment is known to inhibit effective adaptive antitumor immunity. Blockade of PD-1 in humans has resulted in impressive, durable regression responses in select tumor types. However, durable responses have been elusive in ovarian cancer patients. PD-1 was recently shown to be expressed on and thereby impair the functions of tumor-infiltrating murine and human myeloid dendritic cells (TIDC) in ovarian cancer. In the present work, we characterize the regulation of PD-1 expression and the effects of PD-1 blockade on TIDC. Treatment of TIDC and bone marrow-derived dendritic cells (DC) with IL10 led to increased PD-1 expression. Both groups of DCs also responded to PD-1 blockade by increasing production of IL10. Similarly, treatment of ovarian tumor-bearing mice with PD-1 blocking antibody resulted in an increase in IL10 levels in both serum and ascites. While PD-1 blockade or IL10 neutralization as monotherapies were inefficient, combination of these two led to improved survival and delayed tumor growth; this was accompanied by augmented antitumor T- and B-cell responses and decreased infiltration of immunosuppressive MDSC. Taken together, our findings implicate compensatory release of IL10 as one of the adaptive resistance mechanisms that undermine the efficacy of anti-PD-1 (or anti-PD-L1) monotherapies and prompt further studies aimed at identifying such resistance mechanisms. Cancer Res; 77(23); 6667-78. ©2017 AACR.


Assuntos
Interleucina-10/metabolismo , Interleucina-10/farmacologia , Neoplasias Ovarianas/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/biossíntese , Animais , Linfócitos B/imunologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/tratamento farmacológico , Interferência de RNA , RNA Interferente Pequeno/genética , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Linfócitos T/imunologia , Microambiente Tumoral/imunologia
4.
Cancer Res ; 76(2): 239-50, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26567141

RESUMO

The PD-1:PD-L1 immune signaling axis mediates suppression of T-cell-dependent tumor immunity. PD-1 expression was recently found to be upregulated on tumor-infiltrating murine (CD11c(+)CD11b(+)CD8(-)CD209a(+)) and human (CD1c(+)CD19(-)) myeloid dendritic cells (TIDC), an innate immune cell type also implicated in immune escape. However, there is little knowledge concerning how PD-1 regulates innate immune cells. In this study, we examined the role of PD-1 in TIDCs derived from mice bearing ovarian tumors. Similar to lymphocytes, TIDC expression of PD-1 was associated with expression of the adapter protein SHP-2, which signals to NF-κB; however, in contrast to its role in lymphocytes, we found that expression of PD-1 in TIDC tonically paralyzed NF-κB activation. Further mechanistic investigations showed that PD-1 blocked NF-κB-dependent cytokine release in a SHP-2-dependent manner. Conversely, inhibition of NF-κB-mediated antigen presentation by PD-1 occurred independently of SHP-2. Collectively, our findings revealed that PD-1 acts in a distinct manner in innate immune cells compared with adaptive immune cells, prompting further investigations of the signaling pathways controlled by this central mediator of immune escape in cancer.


Assuntos
Células Dendríticas/imunologia , NF-kappa B/metabolismo , Neoplasias Ovarianas/imunologia , Receptor de Morte Celular Programada 1/imunologia , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/patologia , Transdução de Sinais
5.
J Immunol ; 194(7): 2985-91, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25795789

RESUMO

Dendritic cells (DCs) play a pivotal role in the tumor microenvironment, which is known to affect disease progression in many human malignancies. Infiltration by mature, active DCs into the tumors confers an increase in immune activation and recruitment of disease-fighting immune effector cells and pathways. DCs are the preferential target of infiltrating T cells. However, tumor cells have means of suppressing DC function or of altering the tumor microenvironment in such a way that immune-suppressive DCs are recruited. Advances in understanding these changes have led to promising developments in cancer-therapeutic strategies targeting tumor-infiltrating DCs to subdue their immunosuppressive functions and enhance their immune-stimulatory capacity.


Assuntos
Células Dendríticas/imunologia , Neoplasias/etiologia , Imunidade Adaptativa , Animais , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Humanos , Tolerância Imunológica , Imunoterapia , Neoplasias/patologia , Neoplasias/terapia , Fenótipo , Fatores de Transcrição/metabolismo
6.
Cancer Metastasis Rev ; 34(1): 53-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25544369

RESUMO

Clinical outcomes, such as recurrence-free survival and overall survival, in ovarian cancer are quite variable, independent of common characteristics such as stage, response to therapy, and grade. This disparity in outcomes warrants further exploration and therapeutic targeting into the interaction between the tumor and host. One compelling host characteristic that contributes both to the initiation and progression of ovarian cancer is the immune system. Hundreds of studies have confirmed a prominent role for the immune system in modifying the clinical course of the disease. Recent studies also show that anti-tumor immunity is often negated by immune regulatory cells present in the tumor microenvironment. Regulatory immune cells also directly enhance the pathogenesis through the release of various cytokines and chemokines, which together form an integrated pathological network. Thus, in the future, research into immunotherapy targeting ovarian cancer will probably become increasingly focused on combination approaches that simultaneously augment immunity while preventing local immune suppression. In this article, we summarize important immunological targets that influence ovarian cancer outcome as well as include an update on newer immunotherapeutic strategies.


Assuntos
Imunoterapia/métodos , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Microambiente Tumoral/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Feminino , Humanos , Imunoterapia Adotiva/métodos , Modelos Imunológicos , Terapia de Alvo Molecular/métodos
7.
Cancer Res ; 74(11): 2974-85, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24728077

RESUMO

Immunosuppression in the tumor microenvironment blunts vaccine-induced immune effectors. PD-1/B7-H1 is an important inhibitory axis in the tumor microenvironment. Our goal in this study was to determine the effect of blocking this inhibitory axis during and following vaccination against breast cancer. We observed that using anti-PD-1 antibody and a multipeptide vaccine (consisting of immunogenic peptides derived from breast cancer antigens, neu, legumain, and ß-catenin) as a combination therapy regimen for the treatment of breast cancer-bearing mice prolonged the vaccine-induced progression-free survival period. This prolonged survival was associated with increase in number of Tc1 and Tc2 CD8 T cells with memory precursor phenotype, CD27+IL-7RhiT-betlo, and decrease in number of PD-1+ dendritic cells (DC) in regressing tumors and enhanced antigen reactivity of tumor-infiltrating CD8 T cells. It was also observed that blockade of PD-1 on tumor DCs enhanced IL-7R expression on CD8 T cells. Taken together, our results suggest that PD-1 blockade enhances breast cancer vaccine efficacy by altering both CD8 T cell and DC components of the tumor microenvironment. Given the recent success of anti-PD-1 monotherapy, our results are encouraging for developing combination therapies for the treatment of patients with cancer in which anti-PD-1 monotherapy alone may be ineffective (i.e., PD-L1-negative tumors).


Assuntos
Anticorpos/imunologia , Anticorpos/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Memória Imunológica/imunologia , Receptor de Morte Celular Programada 1/imunologia , Animais , Antígenos de Neoplasias/imunologia , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Intervalo Livre de Doença , Feminino , Memória Imunológica/efeitos dos fármacos , Mastocitoma/imunologia , Mastocitoma/terapia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Interleucina-7/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
8.
Mol Ther ; 21(5): 1087-95, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23568262

RESUMO

Picornaviruses have been developed as potential therapies for gene delivery and vaccination. One drawback to their use is the potential for recombination and viral persistence. Therefore, the engineering strategies used must take into account the possibility for virus escape. We have developed Theiler's murine encephalomyelitis virus (TMEV) as a potential vaccine vector for use in immunotherapy. This study shows that insertion of a vaccine epitope at a unique site within the TMEV leader protein can dramatically increase the type I interferon (IFN) response to infection and promote rapid viral clearance. This live virus vaccine maintains its ability to drive antigen-specific CD8(+) T-cell responses to a model antigen as well as to the weakly immunogenic tumor antigen Her2/neu. Furthermore, the epitope integration site does not affect the efficacy of this vaccine as cancer immunotherapy for treating models of melanoma and breast cancer as demonstrated by delayed tumor outgrowth and increased survival in animals implanted with these tumors. These findings show that an attenuated virus retaining limited ability to replicate nonetheless can effectively mobilize CD8(+) cellular immunity and will be important for the design of picornavirus vectors used as immunotherapy in clinical settings.


Assuntos
Antígenos/imunologia , Vacinas Anticâncer/imunologia , Epitopos/imunologia , Neoplasias/imunologia , Theilovirus/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/genética , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/mortalidade , Infecções por Cardiovirus/virologia , Linhagem Celular Tumoral , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Imunoterapia , Interferon Tipo I/imunologia , Camundongos , Dados de Sequência Molecular , Mutagênese Insercional , Neoplasias/patologia , Neoplasias/terapia , Receptor ErbB-2/imunologia , Theilovirus/genética , Carga Tumoral/efeitos dos fármacos , Vacinas Atenuadas , Proteínas Virais/química , Proteínas Virais/imunologia
9.
J Immunol ; 186(12): 6905-13, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21551365

RESUMO

Within the ovarian cancer microenvironment, there are several mechanisms that suppress the actions of antitumor immune effectors. Delineating the complex immune microenvironment is an important goal toward developing effective immune-based therapies. A dominant pathway of immune suppression in ovarian cancer involves tumor-associated and dendritic cell (DC)-associated B7-H1. The interaction of B7-H1 with PD-1 on tumor-infiltrating T cells is a widely cited theory of immune suppression involving B7-H1 in ovarian cancer. Recent studies suggest that the B7-H1 ligand, programmed death receptor-1 (PD-1), is also expressed on myeloid cells, complicating interpretations of how B7-H1 regulates DC function in the tumor. In this study, we found that ovarian cancer-infiltrating DCs progressively expressed increased levels of PD-1 over time in addition to B7-H1. These dual-positive PD-1(+) B7-H1(+) DCs have a classical DC phenotype (i.e., CD11c(+)CD11b(+)CD8(-)), but are immature, suppressive, and respond poorly to danger signals. Accumulation of PD-1(+)B7-H1(+) DCs in the tumor was associated with suppression of T cell activity and decreased infiltrating T cells in advancing tumors. T cell suppressor function of these DCs appeared to be mediated by T cell-associated PD-1. In contrast, ligation of PD-1 expressed on the tumor-associated DCs suppressed NF-κB activation, release of immune regulatory cytokines, and upregulation of costimulatory molecules. PD-1 blockade in mice bearing ovarian cancer substantially reduced tumor burden and increased effector Ag-specific T cell responses. Our results reveal a novel role of tumor infiltrating PD-1(+)B7-H1(+) DCs in mediating immune suppression in ovarian cancer.


Assuntos
Antígenos CD/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Células Dendríticas/imunologia , Neoplasias Ovarianas/imunologia , Animais , Antígeno B7-H1 , Células Cultivadas , Células Dendríticas/química , Feminino , Imunofenotipagem , Terapia de Imunossupressão , Linfócitos do Interstício Tumoral , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1 , Carga Tumoral/imunologia
10.
Cancer Biomark ; 6(5-6): 291-305, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20938089

RESUMO

Monoclonal antibodies are currently being used as potential therapeutics against cancer. So far, eight monoclonal antibodies have been approved by FDA for human use and all these antibodies are effective in the treatment of patients with variety of cancers. Also, there is a long list of several antibodies which are in late stage clinical trials awaiting potential approval by the FDA for use in the clinic. Monoclonal antibodies exert their effects on tumor cells either directly or indirectly thereby eliminating or reducing the tumor burden in cancer patients. In this review we will focus our discussion on different monoclonal antibodies that are currently being used for immunotherapy of different types of cancers, their mechanism of action, several strategies that are being used to improve the effectiveness of these monoclonal antibodies and their potential use in combination therapy approaches along with different chemotherapeutic agents. The field of antibody therapy has ushered in a new and vast research paradigm that is definitely impacting public health.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Ensaios Clínicos como Assunto , Humanos , Neoplasias/imunologia
11.
Clin Cancer Res ; 16(3): 825-34, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20103660

RESUMO

PURPOSE: Over the past two decades, there has been significant interest in targeting HER-2/neu in immune-based approaches for the treatment of HER-2/neu+ cancers. For example, peptide vaccination using a CD8 T cell-activating HER-2/neu epitope (amino acids 369-377) is an approach that is being considered in advanced phase clinical trials. Studies have suggested that the persistence of HER-2/neu-specific CD8 T cells could be improved by incorporating human leukocyte antigen (HLA) class II epitopes in the vaccine. Our goal in this study was to identify broad coverage HLA-DR epitopes of HER-2/neu, an antigen that is highly expressed in a variety of carcinomas. EXPERIMENTAL DESIGN: A combination of algorithms and HLA-DR-binding assays was used to identify HLA-DR epitopes of HER-2/neu antigen. Evidence of preexistent immunity in cancer patients against the identified epitopes was determined using IFN-gamma enzyme-linked immunosorbent spot (ELIspot) assay. RESULTS: Eighty-four HLA-DR epitopes of HER-2/neu were predicted, 15 of which had high binding affinity for > or =11 common HLA-DR molecules. A degenerate pool of four HLA-DR-restricted 15-amino acid epitopes (p59, p88, p422, and p885) was identified, against which >58% of breast and ovarian cancer patients had preexistent T-cell immunity. All four epitopes are naturally processed by antigen-presenting cells. Hardy-Weinberg analysis showed that the pool is useful in approximately 84% of population. Lastly, in this degenerate pool, we identified a novel in vivo immunodominant HLA-DR epitope, HER-2/neu(88-102) (p88). CONCLUSION: The broad coverage and natural immunity to this epitope pool suggests potential usefulness in HER-2/neu-targeting, immune-based therapies such as vaccines.


Assuntos
Antígenos HLA-DR/imunologia , Epitopos Imunodominantes/análise , Receptor ErbB-2/imunologia , Algoritmos , Neoplasias da Mama/imunologia , Linfócitos T CD4-Positivos/imunologia , Feminino , Humanos , Neoplasias Ovarianas/imunologia
12.
Cancer Immunol Immunother ; 59(1): 161-71, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19621224

RESUMO

CD4 T cells are important for anti-tumor immune responses. Aside from their role in the activation of CD8 T cells, CD4 T cells also mediate anti-tumor immune responses by recruiting innate immune effectors into the tumor microenvironment. Thus, the search for strategies to boost CD4 T cell immunity is an active area of research. Our goal in this study was to identify HLA-DR epitopes of carcinoembryonic antigen (CEA), a commonly over-expressed tumor antigen. HLA-DR epitopes of CEA were identified using the epitope prediction program, PIC (predicted IC(50)) and tested using in vitro HLA-DR binding assays. Following CEA epitope confirmation, IFN-gamma ELIspot assays were used to detect existing immunity against the HLA-DR epitope panel of CEA in breast and ovarian cancer patients. In vitro generated peptide-specific CD4 T cells were used to determine whether the epitopes are naturally processed from CEA protein. Forty-three epitopes of CEA were predicted, 15 of which had high binding affinity for 8 or more common HLA-DR molecules. A degenerate pool of four, HLA-DR restricted 15 amino acid epitopes (CEA.24, CEA.176/354, CEA.488, and CEA.653) consisting of two novel epitopes (CEA.24 and CEA.488) was identified against which 40% of breast and ovarian cancer patients had pre-existent T cell immunity. All four epitopes are naturally processed by antigen-presenting cells. Hardy-Weinberg analysis showed that the pool is useful in approximately 94% of patients. Patients with breast or ovarian cancer demonstrate pre-existent immune responses to the tumor antigen CEA. The degenerate pool of CEA peptides may be useful for augmenting CD4 T cell immunity.


Assuntos
Antígeno Carcinoembrionário/imunologia , Antígenos HLA-DR/imunologia , Adulto , Neoplasias da Mama/imunologia , Linfócitos T CD4-Positivos/imunologia , Epitopos , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/imunologia , Software
13.
Cell Immunol ; 254(1): 74-80, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18715553

RESUMO

Mycobacterial HSP70 protein coupled with ovalbumin is known to elicit antigen-specific CD8(+) T-cell response. We investigated whether anti-idiotype immunity can likewise be enhanced using a conjugate of recombinant mycHSP70 and the Ig Id in a murine lymphoma model, A20. Plasmids were constructed of A20 tumor to generate A20Id-HSP70 (scFv-H), unconjugated A20Id (scFv), and mycHSP70 (H) recombinant proteins. We evaluated their relative efficacy in activating anti-tumor immunity that can reduce the mortality of tumor-challenged BALB/c mice; significantly, a longer term protection (>50% of the population) was observed in mice vaccinated with scFv-H compared to those receiving the scFv or H proteins. Concomitantly a much higher-level activation in anti-A20 cytotoxic T-cell activity, IFN-gamma secretion and predominantly anti-A20 IgG2a response was also observed with the scFv-H group. Thus, conjugating HSP70 with the A20Id renders the latter significantly immunogenic and affords longer protection against A20 tumor progression.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/imunologia , Proteínas de Choque Térmico HSP70/imunologia , Idiótipos de Imunoglobulinas/imunologia , Linfoma/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium/imunologia , Proteínas Recombinantes de Fusão/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...