Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
1.
Esophagus ; 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38990441

RÉSUMÉ

BACKGROUND: S-588410, a cancer peptide vaccine (CPV), comprises five HLA-A*24:02-restricted peptides from five cancer-testis antigens. In a phase 2 study, S-588410 was well-tolerated and exhibited antitumor efficacy in patients with urothelial cancer. Therefore, we aimed to evaluate the efficacy, immune response, and safety of S-588410 in patients with completely resected esophageal squamous cell carcinoma (ESCC). METHODS: This phase 3 study involved patients with HLA-A*24:02-positive and lymph node metastasis-positive ESCC who received neoadjuvant therapy followed by curative resection. After randomization, patients were administered S-588410 and placebo (both emulsified with Montanide™ ISA 51VG) subcutaneously. The primary endpoint was relapse-free survival (RFS). The secondary endpoints were overall survival (OS), cytotoxic T-lymphocyte (CTL) induction, and safety. Statistical significance was tested using the one-sided weighted log-rank test with the Fleming-Harrington class of weights. RESULTS: A total of 276 patients were randomized (N = 138/group). The median RFS was 84.3 and 84.1 weeks in the S-588410 and placebo groups, respectively (P = 0.8156), whereas the median OS was 236.3 weeks and not reached, respectively (P = 0.6533). CTL induction was observed in 132/134 (98.5%) patients who received S-588410 within 12 weeks. Injection site reactions (137/140 patients [97.9%]) were the most frequent treatment-emergent adverse events in the S-588410 group. Prolonged survival was observed in S-588410-treated patients with upper thoracic ESCC, grade 3 injection-site reactions, or high CTL intensity. CONCLUSIONS: S-588410 induced immune response and had acceptable safety but failed to reach the primary endpoint. A high CTL induction rate and intensity may be critical for prolonging survival during future CPV development.

2.
J Clin Oncol ; 42(21): 2578-2587, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-38776485

RÉSUMÉ

PURPOSE: The humanized antivascular endothelial growth factor (VEGF) antibody bevacizumab (Bev) is efficacious for the treatment of NF2-related schwannomatosis (NF2), previously known as neurofibromatosis type 2. This study evaluated the safety and efficacy of a VEGF receptor (VEGFR) vaccine containing VEGFR1 and VEGFR2 peptides in patients with NF2 with progressive schwannomas (jRCTs031180184). MATERIALS AND METHODS: VEGFR1 and VEGFR2 peptides were injected subcutaneously into infra-axillary and inguinal regions, once a week for 4 weeks and then once a month for 4 months. The primary end point was safety. Secondary end points included tolerability, hearing response, imaging response, and immunologic response. RESULTS: Sixteen patients with NF2 with progressive schwannomas completed treatment and were assessed. No severe vaccine-related adverse events occurred. Among the 13 patients with assessable hearing, word recognition score improved in five patients at 6 months and two at 12 months. Progression of average hearing level of pure tone was 0.168 dB/mo during the year of treatment period, whereas long-term progression was 0.364 dB/mo. Among all 16 patients, a partial response was observed in more than one schwannoma in four (including one in which Bev had not been effective), minor response in 5, and stable disease in 4. Both VEGFR1-specific and VEGFR2-specific cytotoxic T lymphocytes (CTLs) were induced in 11 patients. Two years after vaccination, a radiologic response was achieved in nine of 20 assessable schwannomas. CONCLUSION: This study demonstrated the safety and preliminary efficacy of VEGFR peptide vaccination in patients with NF2. Memory-induced CTLs after VEGFR vaccination may persistently suppress tumor progression.


Sujet(s)
Vaccins anticancéreux , Neurinome , Récepteur-1 au facteur croissance endothéliale vasculaire , Récepteur-2 au facteur croissance endothéliale vasculaire , Humains , Mâle , Femelle , Adulte , Adulte d'âge moyen , Récepteur-2 au facteur croissance endothéliale vasculaire/immunologie , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/effets indésirables , Vaccins anticancéreux/administration et posologie , Neurinome/immunologie , Récepteur-1 au facteur croissance endothéliale vasculaire/immunologie , Tumeurs cutanées/immunologie , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/anatomopathologie , Neurofibromatose de type 2 , Jeune adulte , Sujet âgé , Neurofibromatoses
4.
J Hum Genet ; 67(7): 411-419, 2022 Jul.
Article de Anglais | MEDLINE | ID: mdl-35110673

RÉSUMÉ

Since severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19) was first reported in China in December 2019, various variants have been identified in different areas of the world such as United Kingdom (alpha), South Africa (beta and omicron), Brazil (gamma), and India (delta). Some of SARS-CoV-2 variants, each of which is characterized by a unique mutation(s) in spike protein, are concerned due to their high infectivity and the capability to escape from neutralizing antibodies elicited by vaccinations. To identify peptide epitopes that are derived from SARS-CoV-2 viral proteins and possibly induce CD8+ T cell immunity, we investigated SARS-CoV-2-derived peptides that are likely to bind to major histocompatibility complex (MHC) class I molecules. We identified a total of 15 peptides that bind to human leukocyte antigen (HLA)-A*24:02, HLA-A*02:01, or HLA-A*02:06, and possibly induce cytotoxic T lymphocytes (CTLs); thirteen of them corresponded to ORF1ab polyprotein, one peptide to spike protein and the remaining one to membrane glycoprotein. CD8+ T cells that recognize these peptides were detected in peripheral blood samples in three individuals recovered from COVID-19 as well as non-infected individuals. Since most of these peptides are commonly conserved among other coronaviruses including SARS-CoV and/or MERS-CoV, these might be useful to maintain T cell responses to coronaviruses that are pandemic at present and will become the future threat. We could define pairs of TRA and TRB sequences of nine CTL clones that recognize SARS-CoV-2-derived peptides. We might use these SARS-CoV-2-derived peptide-reactive TCR sequences for investigating the history of SARS-CoV-2 infection.


Sujet(s)
COVID-19 , SARS-CoV-2 , Lymphocytes T CD8+ , Déterminants antigéniques des lymphocytes T/génétique , Antigènes HLA-A , Humains , Peptides/composition chimique , Récepteurs aux antigènes des cellules T , Glycoprotéine de spicule des coronavirus/génétique , Lymphocytes T cytotoxiques
5.
Anticancer Res ; 41(3): 1485-1496, 2021 Mar.
Article de Anglais | MEDLINE | ID: mdl-33788741

RÉSUMÉ

BACKGROUND: As the prognosis of biliary tract cancer (BTC) is extremely poor and treatment options are limited, new treatment modalities are urgently needed. We designed a phase II clinical trial to investigate the immune responses and clinical benefits of OCV-C01, an HLA-A*24:02-restricted three-peptide cancer vaccine targeting VEGFR1, VEGFR2, and KIF20A. PATIENTS AND METHODS: Participants were patients with advanced BTC who had unresectable tumours and were refractory to standard chemotherapy. OCV-C01 was injected weekly until the discontinuance criteria were met. RESULTS: Six participants, including four patients positive for HLA-A*24:02, were enrolled in this study for assessment of efficacy. Four out of six patients exhibited vaccine-specific T-cell responses to one or more of three antigens. Log-rank tests revealed that vaccine-specific T cell responses contributed significantly to overall survival. CONCLUSION: The cancer vaccine had positive effects on survival, indicating that this approach warrants further clinical studies.


Sujet(s)
Tumeurs des voies biliaires/traitement médicamenteux , Vaccins anticancéreux/administration et posologie , Kinésine/antagonistes et inhibiteurs , Vaccins sous-unitaires/administration et posologie , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Sujet âgé , Sujet âgé de 80 ans ou plus , Tumeurs des voies biliaires/immunologie , Tumeurs des voies biliaires/métabolisme , Vaccins anticancéreux/effets indésirables , Vaccins anticancéreux/immunologie , Survie sans rechute , Femelle , Fièvre/induit chimiquement , Céphalée/induit chimiquement , Humains , Kinésine/immunologie , Mâle , Adulte d'âge moyen , Thérapie moléculaire ciblée/méthodes , Pronostic , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/métabolisme , Vaccins sous-unitaires/effets indésirables , Vaccins sous-unitaires/immunologie , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Facteur de croissance endothéliale vasculaire de type A/immunologie , Récepteur-2 au facteur croissance endothéliale vasculaire/immunologie
6.
Immunol Invest ; 50(5): 562-579, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-32660279

RÉSUMÉ

Chemorefractory ovarian cancer has limited therapeutic options. Hence, new types of treatment including neoantigen-specific immunotherapy need to be investigated. Neoantigens represent promising targets for personalized cancer immunotherapy. We here describe the clinical and immunological effects of a neoantigen peptide-loaded DC-based immunotherapy in a patient with recurrent and chemoresistant ovarian cancer. A 71-year-old female patient with chemorefractory ovarian cancer and malignant ascites received intranodal vaccination of DCs loaded with four neoantigen peptides that were predicted by our immunogenomic pipeline. Following four rounds of vaccinations with this therapy, CA-125 levels were remarkably declined and tumor cells in the ascites were also decreased. Concordantly, the tumor-related symptoms such as respiratory discomfort improved without any adverse reactions. The reactivity against one HLA-A2402-restricted neoantigen peptide derived from a mutated PPM1 F protein was detected in lymphocytes from peripheral blood by IFN-γ ELISPOT assay. Furthermore, the neoantigen (PPM1 F mutant)-specific TCRs were detected in the tumor-infiltrating T lymphocytes post-vaccination. Our results showed that vaccination with intranodal injection of neoantigen peptide-loaded DCs may have clinical and immunological impacts on cancer treatment.


Sujet(s)
Ascites/thérapie , Vaccins anticancéreux/immunologie , Cellules dendritiques/immunologie , Immunothérapie/méthodes , Lymphocytes TIL/immunologie , Tumeurs de l'ovaire/thérapie , Noeud lymphatique sentinelle/immunologie , Lymphocytes T/immunologie , Sujet âgé , Présentation d'antigène , Antigènes néoplasiques/immunologie , Ascites/immunologie , Antigènes CA-125/sang , Résistance aux médicaments antinéoplasiques , Test ELISpot , Déterminants antigéniques des lymphocytes T/immunologie , Femelle , Humains , Tumeurs de l'ovaire/immunologie , Peptides/immunologie , Charge tumorale , Vaccination
7.
Vaccines (Basel) ; 8(3)2020 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-32887369

RÉSUMÉ

Anti-angiogenic therapy, targeting vascular endothelial growth factor (VEGF)-A/VEGF receptors (VEGFRs), is beneficial for tumor growth prevention in a malignant glioma. A simultaneous blockade using both bevacizumab (Bev), which targets circulating VEGF-A, and a multi-kinase inhibitor on VEGFRs was more effective for advanced solid cancers, including melanoma and renal cell carcinoma. However, previous clinical trials demonstrated a high adverse event rate. Additionally, no studies previously assessed treatment efficacy and safety using both VEGF-A and VEGFR-targeted agents for malignant gliomas. We had conducted clinical trials investigating VEGFRs peptide vaccination in patients with malignant gliomas, in which the treatment exhibited safety and yielded therapeutic effects in some patients. The combined use of Bev and VEGFRs vaccination may enhance the anti-tumor effect in malignant gliomas. In this pilot study, the adverse event profile in patients treated with Bev after the vaccination was investigated to establish this treatment strategy, in comparison to those treated with Bev collected from the published data or treated with the vaccination alone. In our previous clinical studies on patients with malignant gliomas, Bev was administered to 13 patients after VEGFRs vaccinations. One patient had a Grade 4 pulmonary embolism. Two patients had Grade 2 cerebral infarctions. There were no significant differences in the adverse event rates among patients treated with Bev, with the vaccination, or with Bev after the vaccination. Although careful observation is imperative for patients after this combination treatment strategy, VEGFRs-targeted vaccination may coexist with Bev for malignant gliomas.

10.
BMC Cancer ; 20(1): 196, 2020 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-32164575

RÉSUMÉ

BACKGROUND: The expression of vascular endothelial growth factor (VEGF)-A/ VAGF receptors (VEGFRs) signaling plays a pivotal role in the tumor angiogenesis and the development of the immunosuppressive tumor microenvironment in glioblastomas. We have previously conducted exploratory clinical studies investigating VEGFRs peptide vaccination with and without multiple glioma oncoantigens in patients with recurrent high-grade gliomas. Recently, an exploratory clinical investigation of VEGFRs peptide vaccination was conducted in patients with progressive neurofibromatosis type 2. Those studies suggested that cytotoxic T lymphocytes (CTLs) induced by the vaccination can directly kill a wide variety of cells associated with tumor growth, including tumor vessels, tumor cells, and immunosuppressive cells expressing VEGFR1 and/or 2. In the present study, synergistic activity of the combination of VEGFRs peptide vaccination with chemotherapy was evaluated. METHODS: We performed the first clinical trial to assess VEGFR1 and 2 vaccination along with temozolomide (TMZ) -based chemoradiotherapy for the patients with primary glioblastomas. Furthermore, histopathological changes after the vaccination were evaluated using paired pre- and post- vaccination specimens. RESULTS: The disappearance of radiographically enhanced lesion was observed in 2 patients after the vaccination, including one in which the methylation of the O6-methylguanine-DNA methyltransferase (MGMT) promoter was not observed. The histopathological findings of pre- and post-vaccination specimens demonstrated that tumor vessels showed negative or slight VEGFRs expressions after the vaccination and most endothelial cells were covered with PDGFR-ß-positive pericytes. Notably, CTLs induced by VEGFRs peptide vaccination attacked not only tumor vessels but also tumor cells and regulatory T cells expressing VEGFRs even in recurrent tumors. CONCLUSIONS: VEGFR1 and 2 vaccination may have a preliminary synergistic effect when administered with TMZ. The limitation of the present study was the paucity of the number of the samples. Further studies involving more patients are warranted to confirm the findings of this study. TRIAL REGISTRATION: This study was registered as UMIN000013381 (University Hospital Medical Information Network-Clinical Trial Registry: UMIN-CTR) on 5 March, 2014 and with the Japan Registry of Clinical Trials (jRCT) as jRCTs031180170 on 1 March, 2019.


Sujet(s)
Antinéoplasiques alcoylants/administration et posologie , Tumeurs du cerveau/traitement médicamenteux , Vaccins anticancéreux/administration et posologie , Glioblastome/traitement médicamenteux , Fragments peptidiques/administration et posologie , Témozolomide/administration et posologie , Adulte , Sujet âgé , Antinéoplasiques alcoylants/usage thérapeutique , Vaccins anticancéreux/pharmacologie , Synergie des médicaments , Études de faisabilité , Femelle , Humains , Mâle , Adulte d'âge moyen , Fragments peptidiques/usage thérapeutique , Analyse de survie , Témozolomide/usage thérapeutique , Résultat thérapeutique , Récepteur-1 au facteur croissance endothéliale vasculaire/composition chimique , Récepteur-2 au facteur croissance endothéliale vasculaire/composition chimique
11.
Nat Commun ; 10(1): 5758, 2019 12 17.
Article de Anglais | MEDLINE | ID: mdl-31848332

RÉSUMÉ

The anti-VEGF antibody bevacizumab has shown efficacy for the treatment of neurofibromatosis type 2 (NF2). Theoretically, vascular endothelial growth factor receptors (VEGFRs)-specific cytotoxic T lymphocytes (CTLs) can kill both tumor vessel cells and tumor cells expressing VEGFRs. Here we show an exploratory clinical study of VEGFRs peptide vaccine in seven patients with progressive NF2-derived schwannomas. Hearing improves in 2/5 assessable patients (40%) as determined by international guidelines, with increases in word recognition scores. Tumor volume reductions of ≥20% are observed in two patients, including one in which bevacizumab had not been effective. There are no severe adverse events related to the vaccine. Both VEGFR1-specific and VEGFR2-specific CTLs are induced in six patients. Surgery is performed after vaccination in two patients, and significant reductions in the expression of VEGFRs in schwannomas are observed. Therefore, this clinical immunotherapy study demonstrates the safety and preliminary efficacy of VEGFRs peptide vaccination in patients with NF2.


Sujet(s)
Tumeurs du cerveau/thérapie , Vaccins anticancéreux/administration et posologie , Neurofibromatose de type 2/thérapie , Récepteur-1 au facteur croissance endothéliale vasculaire/immunologie , Récepteur-2 au facteur croissance endothéliale vasculaire/immunologie , Adolescent , Adulte , Bévacizumab/usage thérapeutique , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Encéphale/chirurgie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/anatomopathologie , Vaccins anticancéreux/effets indésirables , Vaccins anticancéreux/immunologie , Association thérapeutique/effets indésirables , Association thérapeutique/méthodes , Femelle , Humains , Imagerie par résonance magnétique , Mâle , Neurofibromatose de type 2/immunologie , Neurofibromatose de type 2/anatomopathologie , Radiochirurgie , Tomodensitométrie , Résultat thérapeutique , Charge tumorale/immunologie , Vaccination/effets indésirables , Vaccination/méthodes , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/effets indésirables , Vaccins sous-unitaires/immunologie , Récepteur-1 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Jeune adulte
12.
Oncotarget ; 9(13): 11009-11019, 2018 Feb 16.
Article de Anglais | MEDLINE | ID: mdl-29541393

RÉSUMÉ

Neoantigens are the main targets of tumor-specific T cells reactivated by immune checkpoint-blocking antibodies or when using tumor-infiltrating T cells for adoptive therapy. While cancers often accumulate hundreds of mutations and harbor several immunogenic neoantigens, the repertoire of mutation-specific T cells in patients might be restricted. To bypass suboptimal conditions, which impede the reactivation of existing T cells or the priming of neoantigen-specific T cells in a patient, we employ T cells of healthy donors with an overlapping HLA repertoire to target cancer neoantigens. In this study, we focus on streamlining the process of in vitro-induction of neoantigen-specific T cells and isolating their T cell receptors (TCRs) to establish a time-efficient protocol that will allow the patient to benefit from subsequent therapy. We first optimized the priming of T cells to omit multiple restimulations and extended culturing. Neoantigen-specific T cells were enriched using specific dextramers and next-generation sequencing was applied to determine the TCR repertoire. This allowed us to circumvent the laborious process of expanding T cell clones. Using this protocol, we successfully identified HLA-A-restricted TCRs specific for neoantigens found in an esophageal cancer cell line (TE-8) and a primary ovarian cancer. To verify TCR specificity, we generated TCR-engineered T cells and confirmed recognition of the tumor-derived neoantigens. Our results also emphasize the importance of neoepitope selection in order to avoid cross-reactivity to corresponding wild-type peptide sequences. In conclusion, we established a 2-week protocol for generating and identifying neoantigen-specific TCRs from third-party donors making this strategy applicable for clinical use.

13.
PLoS One ; 9(1): e85267, 2014.
Article de Anglais | MEDLINE | ID: mdl-24416375

RÉSUMÉ

We herein report the identification of an HLA-A2 supertype-restricted epitope peptide derived from hypoxia-inducible protein 2 (HIG2), which is known to be a diagnostic marker and a potential therapeutic target for renal cell carcinoma. Among several candidate peptides predicted by the HLA-binding prediction algorithm, HIG2-9-4 peptide (VLNLYLLGV) was able to effectively induce peptide-specific cytotoxic T lymphocytes (CTLs). The established HIG2-9-4 peptide-specific CTL clone produced interferon-γ (IFN-γ) in response to HIG2-9-4 peptide-pulsed HLA-A*02:01-positive cells, as well as to cells in which HLA-A*02:01 and HIG2 were exogenously introduced. Moreover, the HIG2-9-4 peptide-specific CTL clone exerted cytotoxic activity against HIG2-expressing HLA-A*02:01-positive renal cancer cells, thus suggesting that the HIG2-9-4 peptide is naturally presented on HLA-A*02:01 of HIG-2-expressing cancer cells and is recognized by CTLs. Furthermore, we found that the HIG2-9-4 peptide could also induce CTLs under HLA-A*02:06 restriction. Taken together, these findings indicate that the HIG2-9-4 peptide is a novel HLA-A2 supertype-restricted epitope peptide that could be useful for peptide-based immunotherapy against cancer cells with HIG2 expression.


Sujet(s)
Antigènes néoplasiques/immunologie , Épitopes/immunologie , Antigène HLA-A2/immunologie , Protéines tumorales/immunologie , Peptides/immunologie , Lymphocytes T cytotoxiques/immunologie , Séquence d'acides aminés , Antigènes néoplasiques/composition chimique , Antigènes néoplasiques/génétique , Néphrocarcinome/génétique , Néphrocarcinome/immunologie , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Épitopes/composition chimique , Épitopes/génétique , Expression des gènes/immunologie , Antigène HLA-A2/composition chimique , Antigène HLA-A2/génétique , Humains , Interféron gamma/biosynthèse , Interféron gamma/immunologie , Tumeurs du rein/génétique , Tumeurs du rein/immunologie , Tumeurs du rein/anatomopathologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Données de séquences moléculaires , Protéines tumorales/composition chimique , Protéines tumorales/génétique , Peptides/composition chimique , Peptides/génétique , Peptides/pharmacologie , Liaison aux protéines , Lymphocytes T cytotoxiques/cytologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques
14.
Exp Cell Res ; 272(1): 84-91, 2002 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-11740868

RÉSUMÉ

We found that the convergently epidermal growth factor (EGF)-induced signal and the collagen-induced signal activate mitogen-activated protein kinase (MAPK), which induces migration. We examined the signaling mechanisms of EGF-induced cell migration on collagen using the A431 carcinoma cell. EGF (10 ng/ml) induced migration on collagen, but inhibited proliferation. Using a MAPK cascade inhibitor, PD98059, it was shown that EGF-induced migration on collagen was mediated by MAPK whereas EGF-induced migration on fibronectin and vitronectin was not. PD98059 also showed that activation of MAPK induced by EGF enhanced the adhesiveness of A431 cells to collagen. By Western blotting analysis, the kinetics of MAPK phosphorylation induced by EGF and collagen was examined separately, and convergently. First of all, EGF without collagen caused transient MAPK phosphorylation. Collagen without EGF caused MAPK to be immediately and transiently dephosphorylated, and rephosphorylated followed by sustained hyperphosphorylation. EGF together with collagen caused an immediate, and sustained, hyperphosphorylation. These facts suggest that the transient MAPK dephosphorylation induced by collagen is required for migration in order to maintain an appropriate level of sustained phosphorylation. Furthermore, we found that adhesion of A431 cells to collagen was blocked by the anti-beta1 integrin antibody or by the mixed antibodies composed of anti-alpha1, -alpha2, and -alpha3 antibodies, indicating that collagen-induced MAPK phosphorylation was mediated through alpha1beta1, alpha2beta1, and alpha3beta1 integrins.


Sujet(s)
Mouvement cellulaire/physiologie , Facteur de croissance épidermique/pharmacologie , Antigènes CD29/physiologie , Mitogen-Activated Protein Kinases/physiologie , Collagène , Fibronectines , Humains , Système de signalisation des MAP kinases/physiologie , Cellules cancéreuses en culture , Vitronectine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE