Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 13(7)2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916194

RESUMO

BACKGROUND: Tumors can be separated into immunogenic/hot and non-immunogenic/cold on the basis of the presence of tumor-infiltrating lymphocytes (TILs), the expression of PD-L1 and the tumor mutation burden (TMB). In immunogenic tumors, TILs become unable to control tumor growth because their activity is suppressed by different inhibitory pathways, including PD-1/PD-L1. We hypothesized that tumor vaccines may not be active in the immunosuppressive microenvironment of immunogenic/hot tumors while they could be efficient in the immune naïve microenvironment of non-immunogenic/cold tumors. METHODS: The randomized phase II Vx-001-201 study investigated the effect of the Vx-001 vaccine as maintenance treatment in metastatic non-small cell lung cancer (NSCLC) patients. Biopsies from 131 (68 placebo and 63 Vx-001) patients were retrospectively analyzed for PD-L1 expression and TIL infiltration. TILs were measured as tumor-associated immune cells (TAICs), CD3-TILs, CD8-TILs and granzyme B-producing TILs (GZMB-TILs). Patients were distinguished into PD-L1(+) and PD-L1(-) and into TIL high and TIL low. FINDINGS: There was no correlation between PD-L1 expression and Vx-001 clinical activity. In contrast, Vx-001 showed a significant improvement of overall survival (OS) vs. placebo in TAIC low (21 vs. 8.1 months, p = 0.003, HR = 0.404, 95% CI 0.219-0.745), CD3-TIL low (21.6 vs. 6.6 months, p < 0.001, HR = 0.279, 95% CI 0.131-0.595), CD8-TIL low (21 vs. 6.6 months, p < 0.001; HR = 0.240, 95% CI 0.11-0.522) and GZMB-TIL low (20.7 vs. 11.1 months, p = 0.011, HR = 0.490, 95% CI 0.278-0.863). Vx-001 did not offer any clinical benefit in patients with TAIC high, CD3-TIL high, CD8-TIL high or GZMB-TIL high tumors. CD3-TIL, CD8-TIL and GZMB-TIL were independent predictive factors of Vx-001 efficacy. CONCLUSIONS: These results support the hypothesis that Vx-001 may be efficient in patients with non-immunogenic/cold but not with immunogenic/hot tumors.

2.
Br J Cancer ; 122(10): 1461-1466, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32210365

RESUMO

BACKGROUND: The cancer vaccine Vx-001, which targets the universal tumour antigen TElomerase Reverse Transcriptase (TERT), can mount specific Vx-001/TERT572 CD8 + cytotoxic T cells; this immune response is associated with improved overall survival (OS) in patients with advanced/metastatic non-small cell lung cancer (NSCLC). METHODS: A randomised, double blind, phase 2b trial, in HLA-A*201-positive patients with metastatic, TERT-expressing NSCLC, who did not progress after first-line platinum-based chemotherapy were randomised to receive either Vx-001 or placebo. The primary endpoint of the trial was OS. RESULTS: Two hundred and twenty-one patients were randomised and 190 (101 and 89 patients in the placebo and the Vx-001 arm, respectively) were analysed for efficacy. There was not treatment-related toxicity >grade 2. The study did not meet its primary endpoint (median OS 11.3 and 14.3 months for the placebo and the Vx-001, respectively; p = 0.86) whereas the median Time to Treatment Failure (TTF) was 3.5 and 3.6 months, respectively. Disease control for >6months was observed in 30 (33.7%) and 26 (25.7%) patients treated with Vx-001 and placebo, respectively. There was no documented objective CR or PR. Long lasting TERT-specific immune response was observed in 29.2% of vaccinated patients who experienced a significantly longer OS compared to non-responders (21.3 and 13.4 months, respectively; p = 0.004). CONCLUSION: Vx-001 could induce specific CD8+ immune response but failed to meet its primary endpoint. Subsequent studies have to be focused on the identification and treatment of subgroups of patients able to mount an effective immunological response to Vx-001. CLINICAL TRIAL REGISTRATION: NCT01935154.


Assuntos
Vacinas Anticâncer/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/terapia , Imunidade/efeitos dos fármacos , Telomerase/administração & dosagem , Idoso , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Feminino , Humanos , Imunoterapia/efeitos adversos , Quimioterapia de Manutenção/métodos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Efeito Placebo , Telomerase/antagonistas & inibidores , Telomerase/genética , Telomerase/imunologia
3.
Ann Transl Med ; 4(14): 266, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27563653

RESUMO

The very impressive clinical results recently obtained in cancer patients treated with immune response checkpoint inhibitors boosted the interest in immunotherapy as a therapeutic choice in cancer treatment. However, these inhibitors require a pre-existing tumor specific immune response and the presence of tumor infiltrating T cells to be efficient. This immune response can be triggered by cancer vaccines. One of the main issues in tumor vaccination is the choice of the right antigen to target. All vaccines tested to date targeted tumor associated antigens (TAA) that are self-antigens and failed to show a clinical efficacy because of the immune self-tolerance to TAA. A new class of tumor antigens has recently been described, the neo-antigens that are created by point mutations of tumor expressing proteins and are recognized by the immune system as non-self. Neo-antigens exhibit two main properties: they are not involved in the immune self-tolerance process and are immunogenic. However, the majority of the neo-antigens are patient specific and their use as cancer vaccines requires their previous identification in each patient individualy that can be done only in highly specialized research centers. It is therefore evident that neo-antigens cannot be used for patient vaccination worldwide. This raises the question of whether we can find neo-antigen like vaccines, which would not be patient specific. In this review we show that optimized cryptic peptides from TAA are neo-antigen like peptides. Optimized cryptic peptides are recognized by the immune system as non-self because they target self-cryptic peptides that escape self-tolerance; in addition they are strongly immunogenic because their sequence is modified in order to enhance their affinity for the HLA molecule. The first vaccine based on the optimized cryptic peptide approach, Vx-001, which targets the widely expressed tumor antigen telomerase reverse transcriptase (TERT), has completed a large phase I clinical study and is currently being tested in a randomized phase II trial in non-small cell lung cancer (NSCLC) patients.

4.
Oncotarget ; 7(37): 59417-59428, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27506946

RESUMO

Tumor Associated Antigens (TAAs) are the privileged targets of almost all the cancer vaccines tested to date. Unfortunately all these vaccines failed to show a clinical efficacy. The main reason for this failure is the immune tolerance to TAAs that are self-proteins expressed by normal and cancer cells. Self-tolerance to TAAs is directed against their dominant rather than against their cryptic epitopes. The best way to overcome self-tolerance to TAAs would therefore be to target their cryptic epitopes. However, because of their low HLA-I affinity, cryptic peptides are non-immunogenic and cannot be used to stimulate an antitumor immune response unless their immunogenicity has been previously enhanced. In this paper we describe a general approach to enhance immunogenicity of almost all the HLA-B*0702 restricted cryptic peptides derived from TAAs. It consists in substituting residues at position 1 or 9 of low HLA-B*0702 affinity cryptic peptides by an Alanine or a Leucine respectively. These substitutions increase affinity of peptides for HLA-B*0702. These optimized cryptic peptides are strongly immunogenic and very importantly CTL they stimulate recognize their native counterparts.TAAs derived optimized cryptic peptides can be considered as universal antitumor vaccine since they escape self-tolerance, are immunogenic and are not patient specific.


Assuntos
Vacinas Anticâncer/imunologia , Epitopos de Linfócito T/metabolismo , Imunoterapia/métodos , Neoplasias/terapia , Linfócitos T Citotóxicos/imunologia , Animais , Apresentação de Antígeno , Antígenos de Neoplasias/genética , Autoantígenos/genética , Biologia Computacional , Epitopos de Linfócito T/genética , Antígeno HLA-B7/metabolismo , Humanos , Ativação Linfocitária , Neoplasias/imunologia , Peptídeos/genética , Ligação Proteica , Linfócitos T Citotóxicos/transplante
5.
Clin Lung Cancer ; 14(4): 461-5, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23647738

RESUMO

We present the treatment rationale and study design of a multicenter, open-label, randomized, 2-arm, phase IIb study. Patients with stage IV or recurrent stage I to III non-small-cell lung cancer (NSCLC) whose disease does not progress after 4 cycles of first-line platinum-based chemotherapy will be randomized in a 1:1 ratio to 1 of 2 study arms. Patients will receive the cancer vaccine Vx-001 + Montanide ISA51 VG (Seppic, Paris, France) adjuvant subcutaneously, at a dose of 2 mg, or placebo + Montanide ISA51 VG adjuvant subcutaneously. The vaccination protocol comprises 2 injections with the TYR-Vx001 or placebo (1 at day 0 and another at week 3) and 4 injections with the ARG-Vx001 or placebo, at weeks 6, 9, 12, and 15. After the treatment assessment at week 18, patients will receive the ARG-Vx001 or placebo every 12 weeks starting from week 27 until disease progression, unacceptable toxicity, withdrawal of informed consent, or death. The primary end point of this study is the survival rate at 12 months. Secondary end points include time-to-event comparison of overall survival and comparison of time to treatment failure. Exploratory objectives include comparison of disease control rate after the end of subsequent second-line treatments, comparisons of vaccine immune responses, comparison of survival rate at 12 months in patients with vaccine-induced immune response detected after the second and sixth injections, identification of biomarkers on lymphocytes and on tumors, and comparison of safety and tolerability.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/terapia , Recidiva Local de Neoplasia/terapia , Fragmentos de Peptídeos/uso terapêutico , Telomerase/uso terapêutico , Adolescente , Carcinoma Pulmonar de Células não Pequenas/patologia , Método Duplo-Cego , Seguimentos , Humanos , Agências Internacionais , Neoplasias Pulmonares/patologia , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Prognóstico
6.
Cancer Immunol Immunother ; 61(2): 157-168, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21858533

RESUMO

Vx-001, an HLA-A*0201 restricted telomerase (TERT)-specific anti-tumor vaccine, is composed of the 9-mer cryptic TERT(572) peptide and its optimized variant TERT(572Y). We have previously shown that Vx-001 is non-toxic, highly immunogenic and in vaccinated NSCLC patients early specific immune response is associated with prolonged survival. The aim of the present study was to investigate the specific T-cell immune response against Vx-001. Fifty-five patients with chemo-resistant advanced solid tumors were vaccinated with TERT(572Y) (2 subcutaneous injections) followed by TERT(572) peptide (4 subcutaneous injections) every 3 weeks. Specific immune response was evaluated by IFN-γ and perforin ELISpot and intracellular cytokine staining assays. TERT-reactive T cells were detected in 27 (51%) out of 53 evaluable patients after the 2nd vaccination and in 22 (69%) out of 32 evaluable patients after the completion of 6 vaccinations. Immune responses developed irrespective of the stage of disease and disease status before vaccination. Patients with disease progression at study entry who developed a post-vaccination-induced immunological response had a significant overall survival benefit compared to the post-vaccination non-responders. The Vx-001 vaccine is a promising candidate for cancer immunotherapy since it can induce a TERT-specific T-cell immune response that is associated with prolonged survival.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer , Carcinoma Pulmonar de Células não Pequenas/imunologia , Imunoterapia , Neoplasias Pulmonares/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Citotoxicidade Imunológica , ELISPOT , Feminino , Antígeno HLA-A2/imunologia , Antígeno HLA-A2/metabolismo , Humanos , Interferon gama/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Análise de Sobrevida , Telomerase , Vacinação
7.
J Immunother ; 34(9): 641-50, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21989412

RESUMO

The aim of this study was to investigate the best administration of telomerase reverse transcriptase (TERT572), an human leukocyte antigen-A*0201-restricted cryptic epitope of telomerase, and its optimized variant TERT(572Y) to elicit specific T cell immune responses in cancer patients. Forty-eight cancer patients with chemo-resistant tumors received 2 subcutaneous injections of TERT(572Y) at 2 mg followed at random by 4 subcutaneous injections of either TERT572 or TERT(572Y) peptides at 2 mg every 3 weeks. Specific immune response was evaluated by interferon-γ enzyme-linked immunosorbent spot. T cell responses after the sixth vaccination were detected more frequently (44% vs. 17%), and with higher number of peptide-specific reactive T cells (60 T cells/2 × 10(5) peripheral blood mononuclear cell vs. 10 T cells/2 × 10(5) peripheral blood mononuclear cell, P=0.04), and higher avidity in the patients who received 4 more vaccinations with the TERT572 peptide compared with patients who received only TERT(572Y) vaccinations. These results demonstrate that the best vaccination schedule involves first the administration of the optimized TERT(572Y) followed by the native TERT572 peptides in patients who are candidates for cancer immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Fragmentos de Peptídeos/imunologia , Linfócitos T/metabolismo , Telomerase/imunologia , Idoso , Idoso de 80 Anos ou mais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Células Cultivadas , Protocolos Clínicos , Resistencia a Medicamentos Antineoplásicos , ELISPOT , Epitopos de Linfócito T/metabolismo , Feminino , Antígeno HLA-A2/metabolismo , Humanos , Interferon gama/metabolismo , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Mutação/genética , Neoplasias/imunologia , Neoplasias/mortalidade , Neoplasias/patologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/patologia , Telomerase/genética , Telomerase/metabolismo , Vacinação
8.
IDrugs ; 12(2): 98-102, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19204883

RESUMO

Specific immunotherapy is based on the use of tumor-specific antigens to induce an efficient antitumor immune response. Although tumors are known to be weakly immunogenic and therefore capable of escaping immune surveillance, the objective of tumor vaccination is to induce a frequent, strong and long-lasting antitumor immune response based mainly on the activation of cytotoxic T-lymphocytes. However, as widely expressed tumor antigens (universal tumor antigens) often correspond to normal proteins expressed not only by tumor cells but also by normal cells and tissues, these antigens are generally tolerated by the immune system. Thus, circumventing self tolerance to universal tumor antigens is a major goal of cancer vaccine research. Disappointing results obtained to date with most tumor vaccines has led to a shift in research toward determining ways of stimulating the immune response through the use of new adjuvants, immunostimulants and delivery vectors. However, although these aspects are clearly crucial to vaccine development, breakthroughs in the field may lie in the use of strong antigens as optimized cryptic peptides derived from universal tumor antigens, combined with a potent adjuvant. Targeting cryptic tumor peptides/antigens is an efficient way of overcoming tolerance. Indeed, the first vaccine based on an optimized cryptic peptide induced strong antitumor immunity and demonstrated promising clinical activity.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia/métodos , Peptídeos/imunologia , Animais , Antígenos de Neoplasias/imunologia , Ensaios Clínicos como Assunto , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Peptídeos/administração & dosagem , Tolerância a Antígenos Próprios/imunologia
9.
J Immunol ; 179(8): 5090-8, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17911594

RESUMO

Multiple sclerosis (MS) is a demyelinating inflammatory disease of the CNS. Though originally believed to be CD4-mediated, additional immune effector mechanisms, including myelin-specific CD8(+) T cells, are now proposed to participate in the pathophysiology of MS. To study the immunologic and encephalitogenic behavior of HLA-A*0201-binding myelin-derived epitopes in vivo, we used a humanized HLA-A*0201-transgenic mouse model. Eight HLA-A*0201-binding peptides derived from myelin oligodendrocyte glycoprotein (MOG), an immunodominant myelin self-Ag, were identified in silico. After establishing their relative affinity for HLA-A*0201 and their capacity to form stable complexes with HLA-A*0201 in vitro, their immunological characteristics were studied in HLA-A*0201-transgenic mice. Five MOG peptides, which bound stably to HLA-A*0201 exhibited strong immunogenicity by inducing a sizeable MOG-specific HLA-A*0201-restricted CD8(+) T cell response in vivo. Of these five candidate epitopes, four were processed by MOG-transfected RMA target cells and two peptides proved immunodominant in vivo in response to a plasmid-encoding native full-length MOG. One of the immunodominant MOG peptides (MOG(181)) generated a cytotoxic CD8(+) T cell response able to aggravate CD4(+)-mediated EAE. Therefore, this detailed in vivo characterization provides a hierarchy of candidate epitopes for MOG-specific CD8(+) T cell responses in HLA-A*0201 MS patients identifying the encephalitogenic MOG(181) epitope as a primary candidate.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Glicoproteínas/administração & dosagem , Glicoproteínas/imunologia , Antígenos HLA-A/genética , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/imunologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Citotoxicidade Imunológica/genética , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Glicoproteínas/metabolismo , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Epitopos Imunodominantes/administração & dosagem , Epitopos Imunodominantes/imunologia , Epitopos Imunodominantes/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Glicoproteína Mielina-Oligodendrócito , Proteínas do Tecido Nervoso/administração & dosagem , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Fragmentos de Peptídeos/metabolismo , Ligação Proteica/genética , Ligação Proteica/imunologia
10.
Cancer Res ; 67(14): 7028-36, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17638916

RESUMO

A cDNA vaccine (pVax1/pet-neu) was designed to encode 12 different Her-2/ErbB-2-derived, HLA-A*0201-restricted dominant and high-affinity heteroclitic cryptic epitopes. Vaccination with pVax1/pet-neu triggered multiple and ErbB-2-specific CTL responses in HLA-A*0201 transgenic HHD mice and in HLA-A*0201 healthy donors in vitro. Human and murine CTL specific for each one of the 12 ErbB-2 peptides recognized in vitro both human and murine tumor cells overexpressing endogenous ErbB-2. Furthermore, vaccination of HHD mice with pVax1/pet-neu significantly delayed the in vivo growth of challenged ErbB-2-expressing tumor (EL4/HHD/neu murine thymoma) more actively when compared with vaccination with the empty vector (pVax1) or vehicle alone. These data indicate that the pVax1/pet-neu cDNA vaccine coding for a poly-ErbB-2 epitope is able to generate simultaneous ErbB-2-specific antitumor responses against dominant and cryptic multiple epitopes.


Assuntos
Vacinas Anticâncer/química , Epitopos/química , Neoplasias/terapia , Receptor ErbB-2/química , Linfócitos T Citotóxicos/metabolismo , Vacinas de DNA/química , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Transplante de Neoplasias , Peptídeos/química , Receptor ErbB-2/metabolismo , Timoma/metabolismo
11.
J Clin Oncol ; 25(19): 2727-34, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17602077

RESUMO

PURPOSE: To evaluate the immunological and clinical response as well as the safety of the optimized peptide telomerase reverse transcriptase p572Y (TERT572Y) presented by HLA-A*0201 in patients with advanced non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: Twenty-two patients with advanced NSCLC and residual (n = 8) or progressive disease (PD; n = 14) following chemotherapy and/or radiotherapy received two subcutaneous injections of the optimized TERT572Y peptide followed by four injections of the native TERT572 peptide administered every 3 weeks. Peptide-specific immune responses were monitored by enzyme-linked immunosorbent spot assay and/or TERT572Y pentamer staining. RESULTS: Twelve (54.5%) of 22 patients completed the vaccination program. Toxicity consisted primarily of local skin reactions. TERT572-specific CD8+ cells were detected in 16 (76.2%) of 21 patients after the second vaccination, and 10 (90.9%) of 11 patients after the sixth vaccination. Stable disease (SD) occurred in eight (36.4%) of 22 vaccinated patients, with three (13.6%) having had PD before entering the study. The median duration of SD was 11.2 months. After a median follow-up of 10.0 months, patients with early developed immunological response (n = 16) had a significantly longer time to progression and overall survival (OS) than nonresponders (n = 5; log-rank tests P = .046 and P = .012, respectively). The estimated median OS was 30.0 months (range, 2.8 to 40.0 months) and 4.1 months (range, 2.4 to 10.9 months) for responders and nonresponders, respectively. CONCLUSION: TERT572Y peptide vaccine is well tolerated and effective in eliciting a specific T cell immunity. Immunological response is associated with prolonged survival. These results are encouraging and warrant further evaluation in a randomized study.


Assuntos
Vacinas Anticâncer , Carcinoma Pulmonar de Células não Pequenas/terapia , Antígenos HLA-A/genética , Imunoterapia/métodos , Neoplasias Pulmonares/terapia , Peptídeos/química , Telomerase/química , Telomerase/genética , Idoso , Ensaio de Imunoadsorção Enzimática , Feminino , Antígeno HLA-A2 , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Telomerase/uso terapêutico , Resultado do Tratamento
12.
Oncol Rep ; 17(3): 679-85, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17273751

RESUMO

The inducible heat shock protein Hsp70 has been described as a tumour antigen being frequently overexpressed in tumours of various histologic origins, with a role in tumourigenicity, as a critical event in tumour progression. A strategy to enhance the immune response to an antigen is the identification of multiple epitopes and the induction of a polyspecific response. Applied to tumour vaccination, such a polyspecific response should lead to a more robust antitumour efficacy. The long peptide Hsp70380-402 encompasses three nonamer peptides with a high affinity for HLA-A *0201. In a previous paper, we have shown that two of these nonamer peptides, p391 and p393, can raise CTL to recognize tumour cells overexpressing Hsp70. In the present paper, we demonstrate that the third nonamer peptide, p380, is a new epitope efficient in raising an antitumour immune response. The p380-402 polypeptide was able to induce an immune response against each of the three constituent epitopes both in vivo in HLA-A *0201 transgenic mice and in vitro with human PBMC. This polypeptide therefore constitutes an interesting candidate for the induction of multiple HLA-A *0201-restricted anti-Hsp70 antitumour CTL responses.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Proteínas de Choque Térmico HSP70/imunologia , Animais , Antígenos de Neoplasias/química , Western Blotting , Células COS , Chlorocebus aethiops , Epitopos de Linfócito T/química , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Proteínas de Choque Térmico HSP70/química , Humanos , Camundongos , Camundongos Transgênicos , Peptídeos/química , Peptídeos/imunologia , Transfecção
13.
Cancer Immunol Immunother ; 55(12): 1515-23, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16622681

RESUMO

STEAP is a recently identified protein shown to be particularly overexpressed in prostate cancer and also present in numerous human cancer cell lines from prostate, pancreas, colon, breast, testicular, cervical, bladder and ovarian carcinoma, acute lymphocytic leukemia and Ewing sarcoma. This expression profile renders STEAP an appealing candidate for broad cancer immunotherapy. In order to investigate if STEAP is a tumor antigen that can be targeted by specific CD8(+) T cells, we identified two high affinity HLA-A*0201 restricted peptides (STEAP(86-94) and STEAP(262-270)). These peptides were immunogenic in vivo in HLA-A*0201 transgenic HHD mice. Peptide specific murine CD8 T cells recognized COS-7 cells co-transfected with HHD (HLA-A*0201) and STEAP cDNA constructs and also HLA-A*0201(+) STEAP(+) human tumor cells. Furthermore, STEAP(86-94) and STEAP(262-270) stimulated specific CD8(+) T cells from HLA-A*0201(+) healthy donors, and these peptide specific CD8(+) T cells recognized STEAP positive human tumor cells in an HLA-A*0201-restricted manner. Importantly, STEAP(86-94)-specific T cells were detected and reactive in the peripheral blood mononuclear cells in NSCLC and prostate cancer patients ex vivo. These results show that STEAP can be a target of anti-tumor CD8(+) T cells and that STEAP peptides can be used for a broad-spectrum-tumor immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Sequência de Aminoácidos , Animais , Carcinoma Pulmonar de Células não Pequenas/imunologia , Linhagem Celular Tumoral , Antígenos HLA-A/genética , Antígeno HLA-A2 , Humanos , Neoplasias Pulmonares/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Oxirredutases
14.
Vaccine ; 24(12): 2102-9, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16455166

RESUMO

Polyspecific tumor vaccination should offer broad control of tumor cells and reduce the risk of emergence of immune escape variants. Here, we evaluated the capacity of a polypeptide composed of optimized cryptic peptides derived from three different universal tumor antigens (TERT988Y, HER-2/neu402Y and MAGE-A248V9) to induce a polyspecific CD8 cell response both in vivo in HHD mice and in vitro in humans. A mixture of TERT988Y, HER-2/neu402Y and MAGE-A248V9 peptides failed to induce a trispecific response. In contrast, a polypeptide composed of the three peptides stimulated a trispecific immune response. Interestingly, the capacity of the polypeptide to induce a trispecific response depended on its internal organization. Six different polypeptide variants corresponding to all possible combinations of the three peptides were tested. Only one variant, named Poly-6, elicited an immune response simultaneously targeting all three peptides.


Assuntos
Vacinas Anticâncer/imunologia , Fragmentos de Peptídeos/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/administração & dosagem , Linhagem Celular , Camundongos , Proteínas de Neoplasias/administração & dosagem , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/síntese química , Vacinas Combinadas/administração & dosagem , Vacinas Combinadas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
15.
Vaccine ; 24(11): 1880-8, 2006 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-16300869

RESUMO

The use of synthetic peptides derived from tumor-associated Ags is attractive for the development of antitumoral vaccines as far as strong adjuvants are found to render them immunogenic. Here, we investigated the possibility to enhance the CD8 response against the human and mouse shared TERT(572Y) HLA-A*0201 restricted modified cryptic peptide by using ODN-CpG as adjuvant. Humanized transgenic mice were immunized with the TERT(572Y) modified cryptic peptide in the presence of ODN-CpG and compared to mice immunized in IFA. By contrast with IFA, we first showed that, in vivo, ODN-CpG leads to the recruitment of dendritic cells in the lymph nodes draining the injection site. Those cells and especially the CD11c+ CD11b- CD8a+ lymphoid and the CD11c+ B220+ plasmacytoid dendritic cells were activated as shown by up-regulation of CD40 at their cell surface. Immunization against TERT(572Y) peptide in the presence of ODN-CpG rather than IFA led to a strong CD8 response and can delayed mortality in an induced tumor model. Study of the CD8 response obtained after antigenic challenge suggested that a functional memory response is induced upon vaccination with ODN-CpG. Thus, MHC class I-restricted epitope in combination of ODN-CpG is a promising and rather simple cancer vaccine formulation.


Assuntos
Vacinas Anticâncer/imunologia , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Neoplasias Experimentais/prevenção & controle , Oligodesoxirribonucleotídeos/imunologia , Telomerase/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Células Apresentadoras de Antígenos , Antígenos CD40/análise , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Epitopos , Citometria de Fluxo , Adjuvante de Freund/administração & dosagem , Antígenos HLA-A/imunologia , Humanos , Interferon gama/biossíntese , Lipídeos/administração & dosagem , Linfonodos/citologia , Camundongos , Camundongos Transgênicos , Oligodesoxirribonucleotídeos/administração & dosagem , Análise de Sobrevida
16.
J Immunol ; 173(4): 2578-85, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15294974

RESUMO

Improving vaccine delivery to human APCs is a way to increase the CTL response to vaccines. We report the use of a novel pH-triggered microparticle that exploits the ability of APCs to cross-present MHC I-restricted Ags that have been engulfed in the low pH environment of the phagosome. A model MHC class I-restricted peptide Ag from the influenza A matrix protein was encapsulated in spray-dried microparticles composed of dipalmitoylphosphatidylcholine and the pH-sensitive polymethacrylate Eudragit E100. Release of the peptide from the particle was triggered by a drop in pH to the acidity normally found in the phagosome. The particles were efficiently phagocytosed by human monocytes and dendritic cells with minimal cellular toxicity and no functional impairment. Encapsulation of the peptide in the microparticles resulted in efficient presentation of the peptide to CD8(+) T cells by human dendritic cells in vitro, and was superior to unencapsulated peptide or peptide encapsulated in an analogous pH-insensitive particle. Vaccination of human HLA-A*0201 transgenic mice with peptide encapsulated in pH-triggering microparticles resulted in priming of CTL responses. These microparticles can be modified to coencapsulate a range of adjuvants along with the Ag of interest. Encapsulation of MHC I epitopes in pH-triggered microparticles increases Ag presentation and may improve CD8(+) T cell priming to peptide vaccines against viruses and cancer.


Assuntos
Apresentação de Antígeno/imunologia , Ativação Linfocitária/imunologia , Microesferas , Vacinação/métodos , Vacinas de Subunidades Antigênicas/imunologia , 1,2-Dipalmitoilfosfatidilcolina , Acrilatos , Animais , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Portadores de Fármacos/química , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Concentração de Íons de Hidrogênio , Vírus da Influenza A/imunologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Microscopia de Vídeo , Monócitos/imunologia , Tamanho da Partícula , Fagocitose/fisiologia , Polímeros , Vacinas de Subunidades Antigênicas/administração & dosagem
17.
J Clin Invest ; 113(3): 425-33, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14755339

RESUMO

Most of the human tumor-associated antigens (TAAs) characterized thus far are derived from nonmutated "self"-proteins. Numerous strategies have been developed to break tolerance to TAAs, combining various forms of antigens with different vectors and adjuvants. However, no study has yet determined how to select epitopes within a given TAA to induce the highest antitumor effector response. We addressed this question by evaluating in HLA-A*0201-transgenic HHD mice the antitumor vaccination efficacy of high- and low-affinity epitopes from the naturally expressed murine telomerase reverse transcriptase (mTERT). Immunity against low-affinity epitopes was induced with heteroclitical variants. We show here that the CTL repertoire against high-affinity epitopes is partially tolerized, while that against low-affinity epitopes is composed of frequent CTLs with high avidity. The high-affinity p797 and p545 mTERT epitopes are not able to protect mice from a lethal challenge with the mTERT-expressing EL4-HHD tumor. In contrast, mice developing CTL responses against the p572 and p988 low-affinity epitopes exhibit potent antitumor immunity and no sign of autoimmune reactivity against TERT-expressing normal tissues. Our results strongly argue for new TAA epitope selection and modification strategies in antitumor immunotherapy applications in humans.


Assuntos
Vacinas Anticâncer/imunologia , Epitopos/imunologia , Imunoterapia Ativa , Neoplasias/terapia , Animais , Autoimunidade/imunologia , Proteínas de Ligação a DNA , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Humanos , Camundongos , Neoplasias/imunologia , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Telomerase/imunologia
18.
Int J Cancer ; 108(6): 863-70, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14712489

RESUMO

The design of a broad application tumor vaccine requires the identification of tumor antigens expressed in a majority of tumors of various origins. We questioned whether the major stress-inducible heat shock protein Hsp70 (also known as Hsp72), a protein frequently overexpressed in human tumors of various histological origins, but not in most physiological normal tissues, constitutes a tumor antigen. We selected the p391 and p393 peptides from the sequence of the human inducible Hsp70 that had a high affinity for HLA-A*0201. These peptides were able to trigger a CTL response in vivo in HLA-A*0201-transgenic HHD mice and in vitro in HLA-A*0201+ healthy donors. p391- and p393-specific human and murine CTL recognized human tumor cells overexpressing Hsp70 in a HLA-A*0201-restricted manner. Tetramer analysis of TILs showed that these Hsp70 epitopes are targets of an immune response in many HLA-A*0201+ breast cancer patients. Hsp70 is a tumor antigen and the Hsp70-derived peptides p391 and p393 could be used to raise a cytotoxic response against tumors of various origins.


Assuntos
Antígenos HLA-A/química , Proteínas de Choque Térmico HSP70/metabolismo , Imunoterapia/métodos , Animais , Antígenos de Neoplasias/química , Western Blotting , Linfócitos T CD8-Positivos/metabolismo , Células COS , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Epitopos/química , Antígenos HLA/química , Antígeno HLA-A2 , Humanos , Interferon gama/metabolismo , Camundongos , Camundongos Transgênicos , Peptídeos/química , Plasmídeos/metabolismo , Linfócitos T Citotóxicos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
19.
Cancer Res ; 63(23): 8476-80, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14679012

RESUMO

EphA2 (Eck) is a tyrosine kinase receptor that is overexpressed in several human cancers such as breast, colon, lung, prostate, gastric carcinoma, and metastatic melanoma but not in nonmalignant counterparts. To validate EphA2 as a tumor antigen recognized by CD8+ T lymphocytes, we used reverse immunology approach to identify HLA-A*0201-restricted epitopes. Peptides bearing the HLA-A*0201-specific anchor motifs were analyzed for their capacity to bind and stabilize the HLA-A*0201 molecules. Two peptides, EphA2(58) and EphA2(550), with a high affinity for HLA-A*0201 were selected. Both peptides were immunogenic in the HLA-A*0201-transgenic HHD mice. Interestingly, peptide-specific murine CTLs cell lines responded to COS-7 cells coexpressing HLA-A*0201 and EphA2 and to EphA2-positive human tumor cells of various origin (renal cell, lung, and colon carcinoma and sarcoma). This demonstrates that EphA2(58) and EphA2(550) are naturally processed from endogenous EphA2. In addition, EphA2(58) and EphA2(550) stimulated specific CD8(+) T cells from healthy donor peripheral blood mononuclear cells. These T cells recognized EphA2-positive human tumor cells in an HLA-A*0201-restricted manner. Interestingly, EphA2-specific CD8+ T cells were detected in the peripheral blood mononuclear cells of prostate cancer patients. These results show for the first time that EphA2 is a tumor rejection antigen and lead us to propose EphA2(58) and EphA2(550) peptides for a broad-spectrum-tumor immunotherapy.


Assuntos
Antígenos HLA-A/imunologia , Imunoterapia/métodos , Neoplasias/terapia , Fragmentos de Peptídeos/imunologia , Receptor EphA2/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Mapeamento de Epitopos , Epitopos de Linfócito T/imunologia , Antígeno HLA-A2 , Ativação Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Neoplasias/enzimologia , Neoplasias/imunologia , Fragmentos de Peptídeos/farmacologia , Linfócitos T Citotóxicos/imunologia
20.
J Immunol ; 169(9): 4840-9, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391194

RESUMO

Parathyroid hormone-related protein (PTH-rP), a protein produced by prostate carcinoma and other epithelial cancers, is a key agent in the development of bone metastases. We investigated whether the protein follows the self-tolerance paradigm or can be used as a target Ag for anticancer immunotherapy by investigating the immunogenicity of two HLA-A(*)02.01-binding PTH-rP-derived peptides (PTR-2 and -4) with different affinity qualities. PTH-rP peptide-specific CTL lines were generated from the PBMC of two HLA-A(*)02.01(+) healthy individuals, stimulated in vitro with PTH-rP peptide-loaded autologous dendritic cells and IL-2. The peptide-specific CTLs were able to kill PTH-rP(+)HLA-A(*)02.01(+) breast and prostate carcinoma cell lines. The two peptides were also able to elicit a strong antitumor PTH-rP-specific CTL response in HLA-A(*)02.01 (HHD) transgenic mice. The vaccinated mice did not show any sign of side effects due to cell-mediated autoimmunity or toxicity. In this study we describe two immunogenic and toxic-free PTH-rP peptides as valid candidates for the design of peptide-based vaccination strategies against prostate cancer and bone metastases from the most common epithelial malignancies.


Assuntos
Autoimunidade , Citotoxicidade Imunológica , Antígenos HLA-A/imunologia , Antígeno HLA-A2/imunologia , Neoplasias Experimentais/imunologia , Oligopeptídeos/imunologia , Hormônios Peptídicos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Autoimunidade/genética , Ligação Competitiva/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Células Cultivadas , Testes Imunológicos de Citotoxicidade/métodos , Citotoxicidade Imunológica/genética , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Feminino , Antígenos HLA-A/genética , Antígenos HLA-A/metabolismo , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Humanos , Ativação Linfocitária , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/terapia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Hormônios Peptídicos/administração & dosagem , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo , Ligação Proteica/imunologia , Células Tumorais Cultivadas , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...