Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cancer Immunol Immunother ; 72(2): 475-491, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-35960332

RÉSUMÉ

A phase II study (PRIMMO) of patients with pretreated persistent/recurrent/metastatic cervical or endometrial cancer is presented. Patients received an immunomodulatory five-drug cocktail (IDC) consisting of low-dose cyclophosphamide, aspirin, lansoprazole, vitamin D, and curcumin starting 2 weeks before radioimmunotherapy. Pembrolizumab was administered three-weekly from day 15 onwards; one of the tumor lesions was irradiated (8Gyx3) on days 15, 17, and 19. The primary endpoint was the objective response rate per immune-related response criteria (irORR) at week 26 (a lower bound of the 90% confidence interval [CI] of > 10% was considered efficacious). The prespecified 43 patients (cervical, n = 18; endometrial, n = 25) were enrolled. The irORR was 11.1% (90% CI 2.0-31.0) in cervical cancer and 12.0% (90% CI 3.4-28.2) in endometrial cancer. Median duration of response was not reached in both cohorts. Median interval-censored progression-free survival was 4.1 weeks (95% CI 4.1-25.7) in cervical cancer and 3.6 weeks (95% CI 3.6-15.4) in endometrial cancer; median overall survival was 39.6 weeks (95% CI 15.0-67.0) and 37.4 weeks (95% CI 19.0-50.3), respectively. Grade ≥ 3 treatment-related adverse events were reported in 10 (55.6%) cervical cancer patients and 9 (36.0%) endometrial cancer patients. Health-related quality of life was generally stable over time. Responders had a significantly higher proportion of peripheral T cells when compared to nonresponders (p = 0.013). In conclusion, PRIMMO did not meet its primary objective in both cohorts; pembrolizumab, radiotherapy, and an IDC had modest but durable antitumor activity with acceptable but not negligible toxicity.Trial registration ClinicalTrials.gov (identifier NCT03192059) and EudraCT Registry (number 2016-001569-97).


Sujet(s)
Tumeurs de l'endomètre , Tumeurs du col de l'utérus , Femelle , Humains , Tumeurs du col de l'utérus/traitement médicamenteux , Qualité de vie , Anticorps monoclonaux humanisés/usage thérapeutique , Tumeurs de l'endomètre/anatomopathologie
2.
Front Oncol ; 11: 688755, 2021.
Article de Anglais | MEDLINE | ID: mdl-34055652

RÉSUMÉ

Vaccines used to prevent infections have long been known to stimulate immune responses to cancer as illustrated by the approval of the Bacillus Calmette-Guérin (BCG) vaccine to treat bladder cancer since the 1970s. The recent approval of immunotherapies has rejuvenated this research area with reports of anti-tumor responses with existing infectious diseases vaccines used as such, either alone or in combination with immune checkpoint inhibitors. Here, we have reviewed and summarized research activities using approved vaccines to treat cancer. Data supporting a cancer therapeutic use was found for 16 vaccines. For 10 (BCG, diphtheria, tetanus, human papillomavirus, influenza, measles, pneumococcus, smallpox, typhoid and varicella-zoster), clinical trials have been conducted or are ongoing. Within the remaining 6, preclinical evidence supports further evaluation of the rotavirus, yellow fever and pertussis vaccine in carefully designed clinical trials. The mechanistic evidence for the cholera vaccine, combined with the observational data in colorectal cancer, is also supportive of clinical translation. There is limited data for the hepatitis B and mumps vaccine (without measles vaccine). Four findings are worth highlighting: the superiority of intravesical typhoid vaccine instillations over BCG in a preclinical bladder cancer model, which is now the subject of a phase I trial; the perioperative use of the influenza vaccine to limit and prevent the natural killer cell dysfunction induced by cancer surgery; objective responses following intratumoral injections of measles vaccine in cutaneous T-cell lymphoma; objective responses induced by human papillomavirus vaccine in cutaneous squamous cell carcinoma. All vaccines are intended to induce or improve an anti-tumor (immune) response. In addition to the biological and immunological mechanisms that vary between vaccines, the mode of administration and sequence with other (immuno-)therapies warrant more attention in future research.

4.
BMC Cancer ; 19(1): 506, 2019 May 28.
Article de Anglais | MEDLINE | ID: mdl-31138229

RÉSUMÉ

BACKGROUND: Immunotherapeutic approaches have revolutionized oncological practice but are less evaluated in gynecological malignancies. PD-1/PD-L1 blockade in gynecological cancers showed objective responses in 13-17% of patients. This could be due to immunosuppressive effects exerted by gynecological tumors on the microenvironment and an altered tumor vasculature. In other malignancies, combining checkpoint blockade with radiation delivers benefit that is believed to be due to the abscopal effect. Addition of immune modulation agents has also shown to enhance immune checkpoint blockade efficacy. Therefore we designed a regimen consisting of PD-1 blockade combined with radiation, and different immune/environmental-targeting compounds: repurposed drugs, metronomic chemotherapy and a food supplement. We hypothesize that these will synergistically modulate the tumor microenvironment and induce and sustain an anti-tumor immune response, resulting in tumor regression. METHODS: PRIMMO is a multi-center, open-label, non-randomized, 3-cohort phase 2 study with safety run-in in patients with recurrent/refractory cervical carcinoma, endometrial carcinoma or uterine sarcoma. Treatment consists of daily intake of vitamin D, lansoprazole, aspirin, cyclophosphamide and curcumin, starting 2 weeks before the first pembrolizumab dose. Pembrolizumab is administered 3-weekly for a total of 6 cycles. Radiation (3 × 8 Gy) is given on days 1, 3 and 5 of the first pembrolizumab dose. The safety run-in consists of 6 patients. In total, 18 and 25 evaluable patients for cervical and endometrial carcinoma respectively are foreseen to enroll. No sample size is determined for uterine sarcoma due to its rarity. The primary objective is objective response rate at week 26 according to immune-related response criteria. Secondary objectives include safety, objective response rate at week 26 according to RECIST v1.1, best overall response, progression-free survival, overall survival and quality of life. Exploratory, translational research aims to evaluate immune biomarkers, extracellular vesicles, cell death biomarkers and the gut microbiome. DISCUSSION: In this study, a combination of PD-1 blockade, radiation and immune/environmental-targeting compounds is tested, aiming to tackle the tumor microenvironment and induce anti-tumor immunity. Translational research is performed to discover biomarkers related to the mode of action of the combination. TRIAL REGISTRATION: EU Clinical Trials Register: EudraCT 2016-001569-97 , registered on 19-6-2017. Clinicaltrials.gov: NCT03192059 , registered on 19-6-2017.


Sujet(s)
Antinéoplasiques/administration et posologie , Tumeurs de l'endomètre/thérapie , Récidive tumorale locale/thérapie , Sarcomes/thérapie , Tumeurs du col de l'utérus/thérapie , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Acide acétylsalicylique/administration et posologie , Acide acétylsalicylique/usage thérapeutique , Chimioradiothérapie , Curcumine/administration et posologie , Curcumine/usage thérapeutique , Cyclophosphamide/administration et posologie , Cyclophosphamide/usage thérapeutique , Repositionnement des médicaments , Femelle , Humains , Immunothérapie , Lansoprazole/administration et posologie , Lansoprazole/usage thérapeutique , Analyse de survie , Résultat thérapeutique , Vitamine D/administration et posologie , Vitamine D/usage thérapeutique
5.
Front Nutr ; 5: 138, 2018.
Article de Anglais | MEDLINE | ID: mdl-30687714

RÉSUMÉ

Curcumin is a botanical with anti-tumor and immunomodulatory properties. We hypothesized that curcumin supplementation might influence inflammatory biomarker levels in endometrial carcinoma (EC). In this open-label, non-randomized phase 2 study (NCT02017353), seven EC patients consumed 2 g/day Curcumin Phytosome (CP) orally for 2 weeks. Blood was taken at baseline, days 1, 7, 14, and 21. The following analytes were measured: curcuminoids and metabolites, 56 inflammatory biomarkers, COX-2, frequencies of myeloid-derived suppressor cells, dendritic cells and NK cells, expression of MHC molecules on leukocytes and monocytes and activation/memory status of T cells. Patients completed quality of life (QoL) questionnaires at baseline and end of treatment. Curcumin metabolites were detectable in plasma upon CP intake. CP downregulated MHC expression levels on leukocytes (P = 0.0313), the frequency of monocytes (P = 0.0114) and ICOS expression by CD8+ T cells (P = 0.0002). However, CP upregulated CD69 levels on CD16- NK cells (P = 0.0313). No differences were observed regarding inflammatory biomarkers, frequencies of other immune cell types, T cell activation and COX-2 expression. A non-significant trend to improved QoL was observed. Overall, CP-induced immunomodulatory effects in EC were modest without significant QoL changes. Given the small population and the observed variability in inter-patient biomarker levels, more research is necessary to explore whether benefits of CP can be obtained in EC by different supplementation regimens. Clinical Trial Registration: www.ClinicalTrials.gov, identifier NCT02017353; www.clinicaltrialsregister.eu, identifier 2013-001737-40.

6.
Cancer Immunol Immunother ; 64(3): 381-8, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25548092

RÉSUMÉ

PURPOSE: Melanoma patients with a high risk of recurrence may benefit from immunotherapy with mRNA-electroporated autologous monocyte-derived dendritic cells (DCs). Further benefit may be found in combining DC-therapy with interferon alfa-2b. PATIENTS AND METHODS: The long-term clinical outcome of AJCC stage III/IV melanoma patients who had no evidence of disease at the time of treatment with autologous mRNA-electroporated DCs in a single-center pilot clinical trial was analyzed. Antigen loading was accomplished by co-electroporation of mRNA encoding a fusion protein between MAGE-A1, -A3, -C2, Tyrosinase, MelanA/MART-1, or gp100, and an HLA class II-targeting sequence. DCs were administered by 4-6 bi-weekly intradermal injections. IFN-α-2b (5 MIU TIW) was initiated either at recurrence (cohort 1), concomitant with DCs (cohorts 2 and 3), or following the fourth DC administration (cohort 4). RESULTS: Thirty melanoma patients were recruited between April 2006 and June 2009. DC-related adverse events included grade 2 local injection site reactions in all patients, grade 2 fever and flu-like symptoms in one patient, and skin depigmentation in seven patients. After a median follow-up of over 6 years, the median relapse-free survival is 22 months (95% CI 12-32 months). Twelve patients have died. The median overall survival has not been reached; the 2-year and 4-year survival rates are 93 and 70%, respectively. CONCLUSIONS: Adjuvant therapy following the resection of melanoma metastases with autologous mRNA-electroporated DCs, combined with interferon alfa-2b, is tolerable and results in encouraging long-term overall survival rates justifying further evaluation in a randomized clinical trial.


Sujet(s)
Vaccins anticancéreux/administration et posologie , Cellules dendritiques/immunologie , Immunothérapie adoptive/méthodes , Mélanome/thérapie , ARN messager/administration et posologie , Tumeurs cutanées/thérapie , Adulte , Sujet âgé , Vaccins anticancéreux/génétique , Vaccins anticancéreux/immunologie , Électroporation , Femelle , Humains , Interféron alpha-2 , Interféron alpha/administration et posologie , Interféron alpha/immunologie , Antigène MART-1/génétique , Mâle , Mélanome/immunologie , Mélanome/chirurgie , Antigènes spécifiques du mélanome/génétique , Adulte d'âge moyen , Monophenol monooxygenase/génétique , Métastase tumorale , Projets pilotes , ARN messager/génétique , Protéines recombinantes/administration et posologie , Protéines recombinantes/immunologie , Tumeurs cutanées/immunologie , Tumeurs cutanées/chirurgie , Résultat thérapeutique ,
7.
Methods Mol Biol ; 1139: 3-15, 2014.
Article de Anglais | MEDLINE | ID: mdl-24619665

RÉSUMÉ

Dendritic cells (DC) are key players in several types of cancer vaccines. Large numbers of DC can easily be generated in closed systems from the monocyte fraction of the peripheral blood. They are the professional antigen-presenting cells, and electroporation of mRNA-encoding tumor antigens is a very efficient and a relatively simple way to load the DC with antigen. The co-electroporation of a tumor antigen of choice and the combination of 3 costimulatory molecules, including CD70, caTLR4, and CD40L (TriMix-DC), leads to fully potent antigen-presenting DC able to generate a broad immune response.Here we describe the in vitro transcription of the mRNA and the subsequent generation and electroporation of autologous DC used for the treatment of melanoma patients.


Sujet(s)
Antigènes néoplasiques/génétique , Antigènes néoplasiques/immunologie , Différenciation cellulaire , Cellules dendritiques/cytologie , Cellules dendritiques/immunologie , Électroporation/méthodes , ARN messager/métabolisme , Antigènes CD70/génétique , Antigènes CD70/immunologie , Ligand de CD40/génétique , Ligand de CD40/immunologie , Cellules dendritiques/transplantation , Humains , Mélanome/immunologie , Mélanome/prévention et contrôle , ARN messager/génétique , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/immunologie , Transcription génétique
8.
J Immunol ; 191(4): 1976-83, 2013 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-23842750

RÉSUMÉ

Regulatory T cells (Tregs) counteract anticancer immune responses through a number of mechanisms, limiting dendritic cell (DC)-based anticancer immunotherapy. In this study, we investigated the influence of various DC activation stimuli on the Treg functionality. We compared DCs activated by electroporation with mRNA encoding constitutively active TLR4 (caTLR4) and CD40 ligand (DiMix-DCs), or these factors together with mRNA encoding the costimulatory molecule CD70 (TriMix-DCs) with DCs maturated in the presence of a mixture of inflammatory cytokines (DCs maturated with a combination of the cytokines IL-1ß, IL-6, TNF-α, and PGE2) for their ability to counteract Tregs on different levels. We first demonstrated that there was no difference in the extent of Treg induction starting from CD4(+)CD25(-) T cells under the influence of the different DC maturation stimuli. Second, we showed that both DiMix- and TriMix-DCs could partly alleviate Treg inhibition of CD8(+) T cells. Third, we observed that CD8(+) T cells that had been precultured with DiMix-DCs or TriMix-DCs were partially protected against subsequent Treg suppression. Finally, we showed that Tregs cocultured in the presence of TriMix-DCs, but not DiMix-DCs, partially lost their suppressive capacity. This was accompanied by a decrease in CD27 and CD25 expression on Tregs, as well as an increase in the expression of T-bet and secretion of IFN-γ, TNF-α, and IL-10, suggesting a shift of the Treg phenotype toward a Th1 phenotype. In conclusion, these data suggest that TriMix-DCs are not only able to suppress Treg functions, but moreover could be able to reprogram Tregs to Th1 cells under certain circumstances.


Sujet(s)
Antigènes CD70/physiologie , Ligand de CD40/physiologie , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Tolérance immunitaire/immunologie , Lymphopoïèse/physiologie , Lymphocytes T régulateurs/immunologie , Récepteur de type Toll-4/physiologie , Antigènes CD70/génétique , Lymphocytes T CD4+/cytologie , Ligand de CD40/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Division cellulaire , Cellules cultivées , Techniques de coculture , Cytokines/pharmacologie , Cellules dendritiques/métabolisme , Électroporation , Humains , Immunophénotypage , Activation des lymphocytes , MAP Kinase Kinase 1/génétique , MAP Kinase Kinase 1/physiologie , Monocytes/cytologie , Monocytes/effets des médicaments et des substances chimiques , ARN messager/administration et posologie , ARN messager/génétique , Protéines de fusion recombinantes/métabolisme , Lymphocytes T régulateurs/cytologie , Lymphocytes auxiliaires Th1/immunologie , Récepteur de type Toll-4/génétique
9.
Biomed Res Int ; 2013: 976383, 2013.
Article de Anglais | MEDLINE | ID: mdl-23509826

RÉSUMÉ

Treatment of melanoma patients with mRNA electroporated dendritic cells (TriMixDC-MEL) stimulates T-cell responses against the presented tumor-associated antigens (TAAs). In the current clinical trials, melanoma patients with systemic metastases are treated, requiring priming and/or expansion of preexisting TAA-specific T cells that are able to migrate to both the skin and internal organs. We monitored the presence of TAA-specific CD8(+) T cells infiltrating the skin at sites of intradermal TriMixDC-MEL injection (SKILs) and within the circulation of melanoma patients treated in two clinical trials. In 10 out of fourteen (71%) patients screened, CD8(+) T cells recognizing any of the four TAA presented by TriMixDC-MEL cellular vaccine were found in both compartments. In total, 30 TAA-specific T-cell responses were detected among the SKILs and 29 among peripheral blood T cells, of which 24 in common. A detailed characterization of the antigen specificity of CD8(+) T-cell populations in four patients indicates that the majority of the epitopes detected were only recognized by CD8(+) T cells derived from either skin biopsies or peripheral blood, indicating that some compartmentalization occurs after TriMix-DC therapy. To conclude, functional TAA-specific CD8(+) T cells distribute both to the skin and peripheral blood of patients after TriMixDC-MEL therapy.


Sujet(s)
Antigènes néoplasiques/immunologie , Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/usage thérapeutique , Cellules dendritiques/transplantation , Mélanome/thérapie , Tumeurs cutanées/thérapie , Biopsie , Mouvement cellulaire , Électroporation , Épitopes/immunologie , Humains , Métastase tumorale , Projets pilotes , Récidive , Peau/métabolisme
10.
Oncoimmunology ; 1(8): 1392-1394, 2012 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-23243604

RÉSUMÉ

A decade of collective work by tumor immunologists has led to improved large scale generation, maturation, antigen loading and administration of dendritic cells (DCs) to cancer patients, promoting enhanced antitumor activity. We alleviated the HLA-restriction in DC therapy and demonstrated that it is meaningful to treat patients with DCs irrespective of their HLA type.

11.
Mol Ther ; 20(5): 1063-74, 2012 May.
Article de Anglais | MEDLINE | ID: mdl-22371843

RÉSUMÉ

It is generally thought that dendritic cells (DCs) loaded with full-length tumor antigen could improve immunotherapy by stimulating broad T-cell responses and by allowing treatment irrespective of the patient's human leukocyte antigen (HLA) type. To investigate this, we determined the specificity of T cells from melanoma patients treated with DCs loaded with mRNA encoding a full-length tumor antigen fused to a signal peptide and an HLA class II sorting signal, allowing presentation in HLA class I and II. In delayed-type hypersensitive (DTH)-biopsies and blood, we found functional CD8(+) and CD4(+) T cells recognizing novel treatment-antigen-derived epitopes, presented by several HLA types. Additionally, we identified a CD8(+) response specific for the signal peptide incorporated to elicit presentation by HLA class II and a CD4(+) response specific for the fusion region of the signal peptide and one of the antigens. This demonstrates that the fusion proteins contain newly created immunogenic sequences and provides evidence that ex vivo-generated mRNA-modified DCs can induce effector CD8(+) and CD4(+) T cells from the naive T-cell repertoire of melanoma patients. Thus, this work provides definitive proof that DCs presenting the full antigenic spectrum of tumor antigens can induce T cells specific for novel epitopes and can be administered to patients irrespective of their HLA type.


Sujet(s)
Antigènes néoplasiques/immunologie , Cellules dendritiques/transplantation , Antigènes HLA-D/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Immunothérapie adoptive/méthodes , Mélanome/thérapie , ARN messager/immunologie , Tumeurs cutanées/thérapie , Séquence d'acides aminés , Présentation d'antigène , Antigènes néoplasiques/génétique , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Cytotoxicité immunologique , Cellules dendritiques/immunologie , Électroporation , Antigènes HLA-D/génétique , Antigènes d'histocompatibilité de classe I/génétique , Humains , Activation des lymphocytes , Mélanome/immunologie , Mélanome/anatomopathologie , Données de séquences moléculaires , Signaux de triage des protéines , ARN messager/génétique , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie , Transfection
12.
J Immunol Methods ; 377(1-2): 23-36, 2012 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-22269772

RÉSUMÉ

The efficacy of cancer immunotherapy can be improved by treatment with full-length tumor antigen and by combining several antigens. This approach allows the induction of a broad immune response irrespective of the patient's HLA type which at the same time challenges immune monitoring. Also, the number of available lymphocytes is most often limited and minimal in vitro restimulations of the lymphocytes should maintain information about the actual in vivo situation. To overcome these hurdles, we developed a method to measure the CD8(+) and CD4(+) T-cell responses directly ex vivo. Skin biopsies taken from dendritic cell (DC)-induced DTH reactions from melanoma patients participating in a DC-clinical trial served as lymphocyte source. Antigen-specificity of skin infiltrating lymphocytes was investigated by coculture with antigen-presenting autologous B cells and assessed for CD137 upregulation and enhanced cytokine secretion. Using this approach we could detect treatment-specific CD8(+) T-cells without restimulation in vitro. Upregulation of the activation marker CD137 correlated with the upregulation of the lytic marker CD107a. CD137 upregulation by treatment-specific CD4(+) lymphocytes however was more pronounced after antigen-specific in vitro restimulation. Both CD8(+) and CD4(+) lymphocytes could be further expanded using the same B cells as for screening allowing characterization of the recognized antigenic region. In addition, this technique can be extended to detect a broader array of T-cell functions and to monitor a large cohort of patients. We believe that this approach of direct ex vivo monitoring, irrespective of the patient's HLA-type or the recognized peptide, and using a limited number of lymphocytes is a valuable tool in the immune monitoring of current cellular immunotherapies.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Immunothérapie adoptive/méthodes , Mélanome/immunologie , Mélanome/thérapie , Antigènes néoplasiques/immunologie , Biopsie , Cytométrie en flux , Humains , Hypersensibilité retardée/immunologie , Immunophénotypage , Interféron gamma/analyse , Interféron gamma/immunologie , Protéine de membrane-1 associée au lysosome/analyse , Protéine de membrane-1 associée au lysosome/immunologie , Peau/cytologie , Peau/immunologie , Antigènes CD137/analyse , Antigènes CD137/immunologie , Facteur de nécrose tumorale alpha/analyse , Facteur de nécrose tumorale alpha/immunologie
13.
Cancer Immunol Immunother ; 61(7): 1033-43, 2012 Jul.
Article de Anglais | MEDLINE | ID: mdl-22159452

RÉSUMÉ

Dendritic cells (DCs) electroporated with mRNA encoding CD70, CD40L and a constitutively active toll-like receptor 4 (TriMix-DC) have an increased T-cell stimulatory capacity. In a prospective phase IB clinical trial, we treated melanoma patients with intradermal and intravenous injections of autologous TriMix-DC co-electroporated with mRNA encoding full-length MAGE-A3, MAGE-C2, tyrosinase and gp100. We report here the immunological and clinical results obtained in one patient with a particularly favorable outcome. This patient had stage IV-M1c melanoma with documented progression during dacarbazine chemotherapy and received 5 TriMix-DC injections. Following DC therapy, a broad CD8(+) T-cell response against multiple epitopes derived from all four treatment antigens was found in the blood and among T cells derived from DTH biopsy. In addition, CD4(+) T cells recognizing different MAGE-A3-derived epitopes were detected in DTH-derived cells. A spontaneous anti-MAGE-C2 CD8(+) T-cell response was present prior to TriMix-DC therapy and increased during treatment. The tumor response was assessed with 18-fluorodeoxyglucose-positron emission/computed tomography. We documented a partial tumor response according to RECIST criteria with a marked reduction in (18)F-FDG-uptake by lung, lymph node and bone metastases. The patient remains free from progression after 12 months of follow-up. This case report indicates that administration of autologous TriMix-DC by the combined intradermal and intravenous route can mediate a durable objective tumor response accompanied by a broad T-cell response in a chemorefractory stage IV-M1c melanoma patient.


Sujet(s)
Cellules dendritiques/immunologie , Hélium/administration et posologie , Immunothérapie adoptive/méthodes , Mélanome/immunologie , Mélanome/thérapie , Azote/administration et posologie , Oxygène/administration et posologie , Lymphocytes T/immunologie , Séquence d'acides aminés , Antigènes CD70/biosynthèse , Antigènes CD70/génétique , Antigènes CD70/immunologie , Ligand de CD40/biosynthèse , Ligand de CD40/génétique , Ligand de CD40/immunologie , Cellules dendritiques/anatomopathologie , Électroporation/méthodes , Humains , Hypersensibilité retardée/immunologie , Mâle , Mélanome/anatomopathologie , Adulte d'âge moyen , Données de séquences moléculaires , Stadification tumorale , Études prospectives , ARN messager/administration et posologie , ARN messager/génétique , Récepteur de type Toll-4/biosynthèse , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/immunologie
14.
J Immunother ; 34(5): 448-56, 2011 Jun.
Article de Anglais | MEDLINE | ID: mdl-21577140

RÉSUMÉ

The immunostimulatory capacity of dendritic cells is improved by co-electroporation with mRNA encoding CD40 ligand, constitutively active toll-like receptor 4, and CD70 (TriMix-DC). This pilot clinical trial evaluated the feasibility, safety, and immunogenicity of a therapeutic vaccination containing autologous TriMix-DC co-electroporated with mRNA encoding a human leukocyte antigen class II-targeting signal linked to 1 of 4 melanoma-associated antigens (MAGE-A3, MAGE-C2, tyrosinase, and gp100) in patients with advanced melanoma. Thirty-five American Joint Committee on Cancer stage III/IV melanoma patients received autologous TriMix-DC (4 administrations 2 weeks apart). Immune monitoring was performed by evaluating skin biopsies of delayed type IV hypersensitivity (DTH) reactions for presence of vaccinal antigen-specific DTH-infiltrating lymphocytes (DIL). Thereafter, patients could receive interferon-alpha-2b (IFN-α-2b) 5 MU subcutaneously 3 times weekly and additional TriMix-DC every 8 weeks. TriMix-DC-related adverse events comprised grade 2 local injection site reactions (all patients), and grade 2 fever and lethargy (2 patients). Vaccinal antigen-specific DIL were found in 0/6 patients tested at vaccine initiation and in 12/21 (57.1%) assessed after the fourth vaccine. A positive postvaccination DTH test correlated with IL-12p70 secretion capacity of TriMix-DC. No objective responses to TriMix-DC alone were seen according to RECIST. Twenty-nine patients received IFN-α-2b after the fourth vaccine without unexpected adverse events. During TriMix-DC/IFN-α-2b combination therapy, 1 partial response and 5 stable disease (disease control of >6 months with regression of metastases) were observed in 17 patients with evaluable disease at baseline. In conclusion, this study demonstrated that therapeutic vaccination with autologous TriMix-DC is feasible, safe, and immunogenic and can be combined with sequential IFN-α-2b.


Sujet(s)
Antigènes néoplasiques/immunologie , Vaccins anticancéreux/immunologie , Cellules dendritiques/immunologie , Association de médicaments/méthodes , Interféron alpha/administration et posologie , Mélanome , ARN messager/immunologie , Tumeurs cutanées/traitement médicamenteux , Adulte , Sujet âgé , Antigènes CD70/immunologie , Antigènes CD70/métabolisme , Antigènes CD40/immunologie , Antigènes CD40/métabolisme , Vaccins anticancéreux/administration et posologie , Cellules dendritiques/cytologie , Cellules dendritiques/métabolisme , Électroporation , Femelle , Antigènes d'histocompatibilité de classe II/immunologie , Antigènes d'histocompatibilité de classe II/métabolisme , Humains , Hypersensibilité retardée/immunologie , Interféron alpha-2 , Mâle , Adulte d'âge moyen , Stadification tumorale , ARN messager/métabolisme , Protéines recombinantes , Tumeurs cutanées/immunologie , Tumeurs cutanées/mortalité , Tumeurs cutanées/anatomopathologie , Analyse de survie , Récepteur de type Toll-4/immunologie , Récepteur de type Toll-4/métabolisme , Vaccination
15.
Melanoma Res ; 21(2): 152-9, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-21317818

RÉSUMÉ

Metastatic melanoma runs a predictable detrimental course in the vast majority of patients. New modalities of immunotherapy, such as melanoma antigen-specific therapeutic vaccination and cytotoxic T-lymphocyte antigen 4 (CTLA-4) receptor blockade by monoclonal antibodies (mAbs), have been associated with atypical kinetics of tumor response that differ from those observed during cytotoxic treatment. Recently, new tumor response criteria have been proposed based on the tumor response characteristics observed in clinical studies with ipilimumab (the so-called 'immune-related response criteria'). We report three illustrative cases of the American Joint Committee on Cancer stage IV-M1c melanoma patients who experienced atypical kinetics of tumor response to the treatment with the CTLA-4-blocking mAb, ipilimumab (case 1), or an autologous dendritic cell vaccine in combination with interferon α-2b (cases 2 and 3). These cases show that atypical response patterns not only relate to the outcome of CTLA-4-blocking mAb therapy but also to the treatment with therapeutic vaccines and interferon α-2b.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Immunothérapie/méthodes , Mélanome/immunologie , Mélanome/thérapie , Adulte , Sujet âgé , Femelle , Humains , Ipilimumab , Mâle , Mélanome/anatomopathologie , Mélanome/secondaire , Adulte d'âge moyen , Métastase tumorale
16.
Methods Mol Biol ; 629: 405-52, 2010.
Article de Anglais | MEDLINE | ID: mdl-20387165

RÉSUMÉ

Since decades, the main goal of tumor immunologists has been to increase the capacity of the immune system to mediate tumor regression. Considerable progress has been made in enhancing the efficacy of therapeutic anticancer vaccines. First, dendritic cells (DCs) have been identified as the key players in orchestrating primary immune responses. A better understanding of their biology and the development of procedures to generate vast amounts of DCs in vitro have accelerated the development of potent immunotherapeutic strategies for cancer. Second, tumor-associated antigens have been identified which are either selectively or preferentially expressed by tumor cells and can be recognized by the immune system. Finally, several studies have been performed on the genetic modification of DCs with tumor antigens. In this regard, loading the DCs with mRNA, which enables them to produce/process and present the tumor antigens themselves, has emerged as a promising strategy. Here, we will first overview the different aspects that must be taken into account when generating an mRNA-based DC vaccine and the published clinical studies exploiting mRNA-loaded DCs. Second, we will give a detailed description of a novel procedure to generate a vaccine consisting of tumor antigen-expressing dendritic cells with an in vitro superior capacity to induce anti-tumor immune responses. Here, immature DCs are electroporated with mRNAs encoding a tumor antigen, CD40 ligand (CD40L), CD70, and constitutively active (caTLR4) to generate mature antigen-presenting DCs.


Sujet(s)
Antigènes néoplasiques/immunologie , Cellules dendritiques/immunologie , Électroporation/méthodes , Immunothérapie/méthodes , Mélanome/immunologie , Mélanome/thérapie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Différenciation cellulaire , Cellules dendritiques/cytologie , Humains , Monitorage immunologique , Coiffes des ARN/biosynthèse , ARN messager/génétique , ARN messager/métabolisme , Vaccination
17.
Melanoma Res ; 19(6): 385-90, 2009 Dec.
Article de Anglais | MEDLINE | ID: mdl-19707165

RÉSUMÉ

Melanoma metastases are characterized by pronounced neo-angiogenesis and spontaneous bleeding frequently occurring within central nervous system metastases. Clinically apparent spontaneous hemorrhage within subcutaneous melanoma metastases, however, is a rare event that coincides with progression of such metastases. We report, to our knowledge the first observation, on regression of subcutaneous metastases with hemorrhage of the overlying skin in three patients with stage IV melanoma who participated in clinical trials on therapeutic vaccination. In two patients, loss of arterial flow on Doppler ultrasound imaging was documented in the metastasis at the time of hematoma formation. One patient suffered from an intracranial hemorrhage in a subcentimetric brain metastasis coincident with the hemorrhagic regression of some of his skin metastases.


Sujet(s)
Vaccins anticancéreux/usage thérapeutique , Tumeurs du système nerveux central/secondaire , Hémorragie/complications , Mélanome/secondaire , Adulte , Tumeurs du système nerveux central/vascularisation , Tumeurs du système nerveux central/anatomopathologie , Évolution de la maladie , Femelle , Humains , Hémorragies intracrâniennes/étiologie , Mâle , Mélanome/vascularisation , Mélanome/thérapie , Adulte d'âge moyen , Néovascularisation pathologique , Tumeurs cutanées/vascularisation , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/thérapie , Vaccination
18.
Clin Cancer Res ; 15(10): 3366-75, 2009 May 15.
Article de Anglais | MEDLINE | ID: mdl-19417017

RÉSUMÉ

PURPOSE: A critical factor determining the effectiveness of currently used dendritic cell (DC)-based vaccines is the DC activation or maturation status. We have recently shown that the T-cell stimulatory capacity of DCs pulsed with tumor-antigen-derived peptides can be considerably increased by activating the DCs through electroporation with mRNA encoding CD40 ligand, CD70, and a constitutively active Toll-like receptor 4 (TriMix DCs). Here, we investigate whether TriMix DCs can be coelectroporated with whole tumor-antigen-encoding mRNA. EXPERIMENTAL DESIGN: The T-cell stimulatory capacity of TriMix DCs pulsed with the immunodominant MelanA-A2 peptide and that of TriMix DCs coelectroporated with MelanA mRNA were compared in vitro. TriMix DCs were also coelectroporated with mRNA encoding Mage-A3, Mage-C2, tyrosinase, or gp100. The capacity of these DCs to stimulate tumor-antigen-specific T cells in melanoma patients was investigated both in vitro before vaccination and after DC vaccination. RESULTS: Like peptide-pulsed TriMix DCs, TriMix DCs coelectroporated with MelanA mRNA are very potent in inducing MelanA-specific CD8(+) T cells in vitro. These T cells have an activated phenotype, show cytolytic capacity, and produce inflammatory cytokines in response to specific stimulation. TriMix DCs coelectroporated with tyrosinase are able to stimulate tyrosinase-specific CD8(+) T cells in vitro from the blood of nonvaccinated melanoma patients. Furthermore, TriMix DCs coelectroporated with Mage-A3, Mage-C2, or tyrosinase are able to induce antigen-specific CD8(+) T cells through therapeutic DC vaccination. CONCLUSIONS: TriMix DCs coelectroporated with whole tumor-antigen mRNA stimulate antigen-specific T cells in vitro and induce antigen-specific T-cell responses in melanoma patients through vaccination. Therefore, they represent a promising new approach for antitumor immunotherapy.


Sujet(s)
Antigènes néoplasiques/immunologie , Cellules dendritiques/immunologie , Électroporation/méthodes , Mélanome/immunologie , ARN messager/génétique , Antigènes néoplasiques/génétique , Antigènes CD70/génétique , Antigènes CD70/immunologie , Vaccins anticancéreux/génétique , Vaccins anticancéreux/immunologie , Cytokines/métabolisme , Cellules dendritiques/cytologie , Cellules dendritiques/métabolisme , Femelle , Cytométrie en flux , Humains , Protéine de membrane-1 associée au lysosome/métabolisme , Antigène MART-1 , Mâle , Mélanome/anatomopathologie , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/immunologie , Monophenol monooxygenase/génétique , Monophenol monooxygenase/immunologie , Protéines tumorales/génétique , Protéines tumorales/immunologie , Stadification tumorale , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/immunologie , Antigènes CD137/métabolisme , Vaccination/méthodes , Antigène gp100 du mélanome
19.
Mol Ther ; 16(6): 1170-80, 2008 Jun.
Article de Anglais | MEDLINE | ID: mdl-18431362

RÉSUMÉ

The effectiveness of the dendritic cell (DC) vaccination protocols that are currently in use could be improved by providing the DCs with a more potent maturation signal. We therefore investigated whether the T-cell stimulatory capacity of human monocyte-derived DCs could be increased by co-electroporation with different combinations of CD40L, CD70, and constitutively active toll-like receptor 4 (caTLR4) encoding mRNA. We show that immature DCs electroporated with CD40L and/or caTLR4 mRNA, but not those electroporated with CD70 mRNA, acquire a mature phenotype along with an enhanced secretion of several cytokines/chemokines. Moreover, these DCs are very potent in inducing naive CD4(+) T cells to differentiate into interferon-gamma (IFN-gamma)-secreting type 1 T helper (Th1) cells. Further, we assessed the capacity of the electroporated DCs to activate naive HLA-A2-restricted MelanA-specific CD8(+) T cells without the addition of any exogenous cytokines. When all three molecules were combined, a >500-fold increase in MelanA-specific CD8(+) T cells was observed when compared with immature DCs, and a >200-fold increase when compared with cytokine cocktail-matured DCs. In correlation, we found a marked increase in cytolytic and IFN-gamma/tumor necrosis factor-alpha (TNF-alpha) secreting CD8(+) T cells. Our data indicate that immature DCs genetically modified to express stimulating molecules can induce tumor antigen-specific T cells in vitro and could prove to be a significant improvement over DCs matured with the methods currently in use.


Sujet(s)
Antigènes CD70/métabolisme , Ligand de CD40/métabolisme , Cellules dendritiques/cytologie , Électroporation/méthodes , Lymphocytes T/cytologie , Récepteur de type Toll-4/métabolisme , Lymphocytes T CD8+/métabolisme , Différenciation cellulaire , Antigène HLA-A2/métabolisme , Humains , Interféron gamma/métabolisme , Cellules K562 , Modèles biologiques , Lymphocytes auxiliaires Th1/cytologie , Facteur de nécrose tumorale alpha/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE