Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 98
Filtrer
1.
Vaccine ; 42(9): 2394-2406, 2024 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-38448321

RÉSUMÉ

Malaria caused byPlasmodium vivaxis a pressing public health problem in tropical and subtropical areas.However, little progress has been made toward developing a P. vivaxvaccine, with only three candidates being tested in clinical studies. We previously reported that one chimeric recombinant protein (PvCSP-All epitopes) containing the conserved C-terminus of the P. vivax Circumsporozoite Protein (PvCSP), the three variant repeat domains, and aToll-like receptor-3 agonist,Poly(I:C), as an adjuvant (polyinosinic-polycytidylic acid, a dsRNA analog mimicking viral RNA), elicits strong antibody-mediated immune responses in mice to each of the three allelic forms of PvCSP. In the present study, a pre-clinical safety evaluation was performed to identify potential local and systemic toxic effects of the PvCSP-All epitopes combined with the Poly-ICLC (Poly I:C plus poly-L-lysine, Hiltonol®) or Poly-ICLC when subcutaneously injected into C57BL/6 mice and New Zealand White Rabbits followed by a 21-day recovery period. Overall, all observations were considered non-adverse and were consistent with the expected inflammatory response and immune stimulation following vaccine administration. High levels of vaccine-induced specific antibodies were detected both in mice and rabbits. Furthermore, mice that received the vaccine formulation were protected after the challenge with Plasmodium berghei sporozoites expressing CSP repeats from P. vivax sporozoites (Pb/Pv-VK210). In conclusion, in these non-clinical models, repeated dose administrations of the PvCSP-All epitopes vaccine adjuvanted with a Poly-ICLC were immunogenic, safe, and well tolerated.


Sujet(s)
Carboxyméthylcellulose de sodium/analogues et dérivés , Vaccins contre le paludisme , Paludisme à Plasmodium vivax , Polylysine/analogues et dérivés , Souris , Animaux , Lapins , Paludisme à Plasmodium vivax/prévention et contrôle , Poly I-C , Plasmodium vivax , Protéines de protozoaire/génétique , Souris de lignée C57BL , Adjuvants immunologiques , Protéines recombinantes , Épitopes , Anticorps antiprotozoaires
2.
Neuro Oncol ; 26(2): 335-347, 2024 02 02.
Article de Anglais | MEDLINE | ID: mdl-37758193

RÉSUMÉ

BACKGROUND: Central nervous system (CNS) WHO grade 2 low-grade glioma (LGG) patients are at high risk for recurrence and with unfavorable long-term prognosis due to the treatment resistance and malignant transformation to high-grade glioma. Considering the relatively intact systemic immunity and slow-growing nature, immunotherapy may offer an effective treatment option for LGG patients. METHODS: We conducted a prospective, randomized pilot study to evaluate the safety and immunological response of the multipeptide IMA950 vaccine with agonistic anti-CD27 antibody, varlilumab, in CNS WHO grade 2 LGG patients. Patients were randomized to receive combination therapy with IMA950 + poly-ICLC and varlilumab (Arm 1) or IMA950 + poly-ICLC (Arm 2) before surgery, followed by adjuvant vaccines. RESULTS: A total of 14 eligible patients were enrolled in the study. Four patients received pre-surgery vaccines but were excluded from postsurgery vaccines due to the high-grade diagnosis of the resected tumor. No regimen-limiting toxicity was observed. All patients demonstrated a significant increase of anti-IMA950 CD8+ T-cell response postvaccine in the peripheral blood, but no IMA950-reactive CD8+ T cells were detected in the resected tumor. Mass cytometry analyses revealed that adding varlilumab promoted T helper type 1 effector memory CD4+ and effector memory CD8+ T-cell differentiation in the PBMC but not in the tumor microenvironment. CONCLUSION: The combinational immunotherapy, including varlilumab, was well-tolerated and induced vaccine-reactive T-cell expansion in the peripheral blood but without a detectable response in the tumor. Further developments of strategies to overcome the blood-tumor barrier are warranted to improve the efficacy of immunotherapy for LGG patients.


Sujet(s)
Anticorps monoclonaux humanisés , Vaccins anticancéreux , Gliome , Peptides , Humains , Projets pilotes , Agranulocytes , Études prospectives , Gliome/traitement médicamenteux , Différenciation cellulaire , Microenvironnement tumoral
3.
NPJ Vaccines ; 8(1): 81, 2023 May 31.
Article de Anglais | MEDLINE | ID: mdl-37258518

RÉSUMÉ

Immunization with the Amastigote Surface Protein-2 (ASP-2) and Trans-sialidase (TS) antigens either in the form of recombinant protein, encoded in plasmids or human adenovirus 5 (hAd5) confers robust protection against various lineages of Trypanosoma cruzi. Herein we generated a chimeric protein containing the most immunogenic regions for T and B cells from TS and ASP-2 (TRASP) and evaluated its immunogenicity in comparison with our standard protocol of heterologous prime-boost using plasmids and hAd5. Mice immunized with TRASP protein associated to Poly-ICLC (Hiltonol) were highly resistant to challenge with T. cruzi, showing a large decrease in tissue parasitism, parasitemia and no lethality. This protection lasted for at least 3 months after the last boost of immunization, being equivalent to the protection induced by DNA/hAd5 protocol. TRASP induced high levels of T. cruzi-specific antibodies and IFNγ-producing T cells and protection was primarily mediated by CD8+ T cells and IFN-γ. We also evaluated the toxicity, immunogenicity, and efficacy of TRASP and DNA/hAd5 formulations in dogs. Mild collateral effects were detected at the site of vaccine inoculation. While the chimeric protein associated with Poly-ICLC induced high levels of antibodies and CD4+ T cell responses, the DNA/hAd5 induced no antibodies, but a strong CD8+ T cell response. Immunization with either vaccine protected dogs against challenge with T. cruzi. Despite the similar efficacy, we conclude that moving ahead with TRASP together with Hiltonol is advantageous over the DNA/hAd5 vaccine due to pre-existing immunity to the adenovirus vector, as well as the cost-benefit for development and large-scale production.

6.
J Clin Invest ; 132(3)2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-34882581

RÉSUMÉ

BACKGROUNDLong-term prognosis of WHO grade II low-grade gliomas (LGGs) is poor, with a high risk of recurrence and malignant transformation into high-grade gliomas. Given the relatively intact immune system of patients with LGGs and the slow tumor growth rate, vaccines are an attractive treatment strategy.METHODSWe conducted a pilot study to evaluate the safety and immunological effects of vaccination with GBM6-AD, lysate of an allogeneic glioblastoma stem cell line, with poly-ICLC in patients with LGGs. Patients were randomized to receive the vaccines before surgery (arm 1) or not (arm 2) and all patients received adjuvant vaccines. Coprimary outcomes were to evaluate safety and immune response in the tumor.RESULTSA total of 17 eligible patients were enrolled - 9 in arm 1 and 8 in arm 2. This regimen was well tolerated with no regimen-limiting toxicity. Neoadjuvant vaccination induced upregulation of type-1 cytokines and chemokines and increased activated CD8+ T cells in peripheral blood. Single-cell RNA/T cell receptor sequencing detected CD8+ T cell clones that expanded with effector phenotype and migrated into the tumor microenvironment (TME) in response to neoadjuvant vaccination. Mass cytometric analyses detected increased tissue resident-like CD8+ T cells with effector memory phenotype in the TME after the neoadjuvant vaccination.CONCLUSIONThe regimen induced effector CD8+ T cell response in peripheral blood and enabled vaccine-reactive CD8+ T cells to migrate into the TME. Further refinements of the regimen may have to be integrated into future strategies.TRIAL REGISTRATIONClinicalTrials.gov NCT02549833.FUNDINGNIH (1R35NS105068, 1R21CA233856), Dabbiere Foundation, Parker Institute for Cancer Immunotherapy, and Daiichi Sankyo Foundation of Life Science.


Sujet(s)
Lymphocytes T CD8+/immunologie , Vaccins anticancéreux , Carboxyméthylcellulose de sodium/analogues et dérivés , Gliome , Traitement néoadjuvant , Poly I-C/administration et posologie , Polylysine/analogues et dérivés , Microenvironnement tumoral/immunologie , Vaccination , Adulte , Sujet âgé , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Carboxyméthylcellulose de sodium/administration et posologie , Femelle , Gliome/immunologie , Gliome/thérapie , Humains , Mâle , Adulte d'âge moyen , Polylysine/administration et posologie
7.
J Control Release ; 337: 168-178, 2021 09 10.
Article de Anglais | MEDLINE | ID: mdl-34280415

RÉSUMÉ

Conventional cancer vaccines based on soluble vaccines and traditional adjuvants have produced suboptimal therapeutic efficacy in clinical trials. Thus, there is an urgent need for vaccine technologies that can generate potent T cell responses with strong anti-tumor efficacy. We have previously reported the development of synthetic high-density protein (sHDL) nanodiscs for efficient lymph node (LN)-targeted co-delivery of antigen peptides and CpG oligonucleotides (a Toll-like receptor-9 agonist). Here, we performed a comparative study in mice and non-human primates (NHPs) to identify an ideal vaccine platform for induction of CD8+ T cell responses. In particular, we compared the efficacy of CpG class B, CpG class C, and polyICLC (a synthetic double-stranded RNA analog, a TLR-3 agonist), each formulated with antigen-carrying sHDL nanodiscs. Here, we report that sHDL-Ag admixed with polyICLC elicited robust Ag-specific CD8+ T cell responses in mice, and when used in combination with α-PD-1 immune checkpoint inhibitor, sHDL-Ag + polyICLC eliminated large established (~100 mm3) MC-38 tumors in mice. Moreover, sHDL-Gag + polyICLC induced robust Simian immunodeficiency virus Gag-specific, polyfunctional CD8+ T cell responses in rhesus macaques and could further amplify the efficacy of recombinant adenovirus-based vaccine. Notably, while both sHDL-Ag-CpG-B and sHDL-Ag-CpG-C generated strong Ag-specific CD8+ T cell responses in mice, their results were mixed in NHPs. Overall, sHDL combined with polyICLC offers a strong platform to induce CD8+ T cells for vaccine applications.


Sujet(s)
Lymphocytes T CD8+ , Vaccins anticancéreux , Adjuvants immunologiques , Animaux , Macaca mulatta , Souris , Vaccins synthétiques
8.
J Immunother Cancer ; 9(6)2021 06.
Article de Anglais | MEDLINE | ID: mdl-34193567

RÉSUMÉ

BACKGROUND: While adoptive transfer of T-cells has been a major medical breakthrough for patients with B cell malignancies, the development of safe and effective T-cell-based immunotherapy for central nervous system (CNS) tumors, such as glioblastoma (GBM), still needs to overcome multiple challenges, including effective homing and persistence of T-cells. Based on previous observations that interleukin (IL)-17-producing T-cells can traffic to the CNS in autoimmune conditions, we evaluated CD8+ T-cells that produce IL-17 and interferon-γ (IFN-γ) (Tc17-1) cells in a preclinical GBM model. METHODS: We differentiated Pmel-1 CD8+ T-cells into Tc17-1 cells and compared their phenotypic and functional characteristics with those of IFN-γ-producing CD8+ T (Tc1) and IL-17-producing CD8+ T (Tc17) cells. We also evaluated the therapeutic efficacy, persistence, and tumor-homing of Tc17-1 cells in comparison to Tc1 cells using a mouse GL261 glioma model. RESULTS: In vitro, Tc17-1 cells demonstrated profiles of both Tc1 and Tc17 cells, including production of both IFN-γ and IL-17, although Tc17-1 cells demonstrated lesser degrees of antigen-specific cytotoxic activity compared with Tc1 cells. In mice-bearing intracranial GL261-Quad tumor and treated with temozolomide, Tc1 cells, but not Tc17-1, showed a significant prolongation of survival. However, when the T-cell transfer was combined with poly-ICLC and Pmel-1 peptide vaccine, both Tc1 and Tc17-1 cells exhibited significantly prolonged survival associated with upregulation of very late activation antigen-4 on Tc17-1 cells in vivo. Glioma cells that recurred following the therapy lost the susceptibility to Pmel-1-derived cytotoxic T-cells, indicating that immuno-editing was a mechanism of the acquired resistance. CONCLUSIONS: Tc17-1 cells were equally effective as Tc1 cells when combined with poly-ICLC and peptide vaccine treatment.


Sujet(s)
Lymphocytes T CD8+/métabolisme , Carboxyméthylcellulose de sodium/analogues et dérivés , Gliome/thérapie , Interféron gamma/métabolisme , Interleukine-17/métabolisme , Poly I-C/métabolisme , Polylysine/analogues et dérivés , Vaccins sous-unitaires/usage thérapeutique , Carboxyméthylcellulose de sodium/métabolisme , Humains , Polylysine/métabolisme
9.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-33562773

RÉSUMÉ

NSCLC (non-small cell lung cancer) is a leading cause of cancer-related deaths worldwide. Clinical trials showed that Hiltonol, a stable dsRNA representing an advanced form of polyI:C (polyinosinic-polycytidilic acid), is an adjuvant cancer-immunomodulator. However, its mechanisms of action and effect on lung cancer have not been explored pre-clinically. Here, we examined, for the first time, how a novel Hiltonol cocktail kills NSCLC cells. By retrospective analysis of NSCLC patient tissues obtained from the tumor biobank; pre-clinical studies with Hiltonol alone or Hiltonol+++ cocktail [Hiltonol+anti-IL6+AG490 (JAK2 inhibitor)+Stattic (STAT3 inhibitor)]; cytokine analysis; gene knockdown and gain/loss-of-function studies, we uncovered the mechanisms of action of Hiltonol+++. We demonstrated that Hiltonol+++ kills the cancer cells and suppresses the metastatic potential of NSCLC through: (i) upregulation of pro-apoptotic Caspase-9 and Caspase-3, (ii) induction of cytosolic cytochrome c, (iii) modulation of pro-inflammatory cytokines (GRO, MCP-1, IL-8, and IL-6) and anticancer IL-24 in NSCLC subtypes, and (iv) upregulation of tumor suppressors, PKR (protein kinase R) and OAS (2'5' oligoadenylate synthetase). In silico analysis showed that Lys296 of PKR and Lys66 of OAS interact with Hiltonol. These Lys residues are purportedly involved in the catalytic/signaling activity of the tumor suppressors. Furthermore, knockdown of PKR/OAS abrogated the anticancer action of Hiltonol, provoking survival of cancer cells. Ex vivo analysis of NSCLC patient tissues corroborated that loss of PKR and OAS is associated with cancer advancement. Altogether, our findings unraveled the significance of studying tumor biobank tissues, which suggests PKR and OAS as precision oncological suppressor candidates to be targeted by this novel Hiltonol+++ cocktail which represents a prospective drug for development into a potent and tailored therapy for NSCLC subtypes.


Sujet(s)
2',5'-Oligoadenylate synthetase/métabolisme , Antinéoplasiques immunologiques/pharmacologie , Carboxyméthylcellulose de sodium/analogues et dérivés , Carcinome pulmonaire non à petites cellules/métabolisme , S-Oxydes cycliques/pharmacologie , Tumeurs du poumon/métabolisme , Poly I-C/pharmacologie , Polylysine/analogues et dérivés , Tyrphostines/pharmacologie , eIF-2 Kinase/métabolisme , 2',5'-Oligoadenylate synthetase/composition chimique , 2',5'-Oligoadenylate synthetase/génétique , Cellules A549 , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Sites de fixation , Carboxyméthylcellulose de sodium/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Interleukine-6/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Modèles moléculaires , Polylysine/pharmacologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , eIF-2 Kinase/composition chimique , eIF-2 Kinase/génétique
10.
Semin Immunol ; 49: 101414, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-33011064

RÉSUMÉ

Immunotherapies have become the first line of treatment for many cancer types. Unfortunately, only a small fraction of patients benefits from these therapies. This low rate of success can be attributed to 3 main barriers: 1) low frequency of anti-tumor specific T cells; 2) lack of infiltration of the anti-tumor specific T cells into the tumor parenchyma and 3) accumulation of highly suppressive cells in the tumor mass that inhibit the effector function of the anti-tumor specific T cells. Thus, the identification of immunomodulators that can increase the frequency and/or the infiltration of antitumor specific T cells while reducing the suppressive capacity of the tumor microenvironment is necessary to ensure the effectiveness of T cell immunotherapies. In this review, we discuss the potential of poly-ICLC as a multi-functional immune modulator for treating cancer and its impact on the 3 above mentioned barriers. We describe the unique capacity of poly-ICLC in stimulating 2 separate pattern recognition receptors, TLR3 and cytosolic MDA5 and the consequences of these activations on cytokines and chemokines production. We emphasize the role of poly-ICLC as an adjuvant in the setting of peptide-based cancer vaccines and in situ tumor vaccination by mimicking natural immune responses to infections. Finally, we summarize the impact of poly-ICLC in enhancing T infiltration into the tumor parenchyma and address the implication of this finding in the clinic.


Sujet(s)
Antinéoplasiques/pharmacologie , Carboxyméthylcellulose de sodium/analogues et dérivés , Facteurs immunologiques/pharmacologie , Immunomodulation , Poly I-C/immunologie , Poly I-C/pharmacologie , Polylysine/analogues et dérivés , Animaux , Antinéoplasiques/usage thérapeutique , Carboxyméthylcellulose de sodium/pharmacologie , Carboxyméthylcellulose de sodium/usage thérapeutique , Cytokines/métabolisme , Humains , Immunité innée/effets des médicaments et des substances chimiques , Facteurs immunologiques/usage thérapeutique , Immunomodulation/effets des médicaments et des substances chimiques , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/étiologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Poly I-C/usage thérapeutique , Polylysine/immunologie , Polylysine/pharmacologie , Polylysine/usage thérapeutique , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , Récepteur de type Toll-3/métabolisme
11.
J Immunother Cancer ; 8(2)2020 09.
Article de Anglais | MEDLINE | ID: mdl-32958686

RÉSUMÉ

BACKGROUND: Immunotherapies, such as immune checkpoint inhibitors and adoptive cell therapies, have revolutionized cancer treatment and resulted in complete and durable responses in some patients. Unfortunately, most immunotherapy treated patients still fail to respond. Absence of T cell infiltration to the tumor site is one of the major obstacles limiting immunotherapy efficacy against solid tumors. Thus, the development of strategies that enhance T cell infiltration and broaden the antitumor efficacy of immunotherapies is greatly needed. METHODS: We used mouse tumor models, genetically deficient mice and vascular endothelial cells (VECs) to study the requirements for T cell infiltration into tumors. RESULTS: A specific formulation of poly-IC, containing poly-lysine and carboxymethylcellulose (PICLC) facilitated the traffic and infiltration of effector CD8 T cells into the tumors that reduced tumor growth. Surprisingly, intratumoral injection of PICLC was significantly less effective in inducing tumor T cell infiltration and controlling growth of tumors as compared with systemic (intravenous or intramuscular) administration. Systemically administered PICLC, but not poly-IC stimulated tumor VECs via the double-stranded RNA cytoplasmic sensor MDA5, resulting in enhanced adhesion molecule expression and the production of type I interferon (IFN-I) and T cell recruiting chemokines. Expression of IFNαß receptor in VECs was necessary to obtain the antitumor effects by PICLC and IFN-I was found to directly stimulate the secretion of T cell recruiting chemokines by VECs indicating that this cytokine-chemokine regulatory axis is crucial for recruiting effector T cells into the tumor parenchyma. Unexpectedly, these effects of PICLC were mostly observed in tumors and not in normal tissues. CONCLUSIONS: These findings have strong implications for the improvement of all types of T cell-based immunotherapies for solid cancers. We predict that systemic administration of PICLC will improve immune checkpoint inhibitor therapy, adoptive cell therapies and therapeutic cancer vaccines.


Sujet(s)
Immunothérapie/méthodes , Poly I-C/métabolisme , Lymphocytes T/métabolisme , Animaux , Modèles animaux de maladie humaine , Femelle , Humains , Souris
12.
J Clin Invest ; 130(12): 6325-6337, 2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-32817593

RÉSUMÉ

BACKGROUNDPatients with diffuse midline gliomas (DMGs), including diffuse intrinsic pontine glioma (DIPG), have dismal outcomes. We previously described the H3.3K27M mutation as a shared neoantigen in HLA-A*02.01+, H3.3K27M+ DMGs. Within the Pacific Pediatric Neuro-Oncology Consortium, we assessed the safety and efficacy of an H3.3K27M-targeted peptide vaccine.METHODSNewly diagnosed patients, aged 3-21 years, with HLA-A*02.01+ and H3.3K27M+ status were enrolled in stratum A (DIPG) or stratum B (nonpontine DMG). Vaccine was administered in combination with polyinosinic-polycytidylic acid-poly-I-lysine carboxymethylcellulose (poly-ICLC) every 3 weeks for 8 cycles, followed by once every 6 weeks. Immunomonitoring and imaging were performed every 3 months. Imaging was centrally reviewed. Immunological responses were assessed in PBMCs using mass cytometry.RESULTSA total of 19 patients were enrolled in stratum A (median age,11 years) and 10 in stratum B (median age, 13 years). There were no grade-4 treatment-related adverse events (TRAEs). Injection site reaction was the most commonly reported TRAE. Overall survival (OS) at 12 months was 40% (95% CI, 22%-73%) for patients in stratum A and 39% (95% CI, 16%-93%) for patients in stratum B. The median OS was 16.1 months for patients who had an expansion of H3.3K27M-reactive CD8+ T cells compared with 9.8 months for their counterparts (P = 0.05). Patients with DIPG with below-median baseline levels of myeloid-derived suppressor cells had prolonged OS compared with their counterparts (P < 0.01). Immediate pretreatment dexamethasone administration was inversely associated with H3.3K27M-reactive CD8+ T cell responses.CONCLUSIONAdministration of the H3.3K27M-specific vaccine was well tolerated. Patients with H3.3K27M-specific CD8+ immunological responses demonstrated prolonged OS compared with nonresponders.TRIAL REGISTRATIONClinicalTrials.gov NCT02960230.FUNDINGThe V Foundation, the Pacific Pediatric Neuro-Oncology Consortium Foundation, the Pediatric Brain Tumor Foundation, the Mithil Prasad Foundation, the MCJ Amelior Foundation, the Anne and Jason Farber Foundation, Will Power Research Fund Inc., the Isabella Kerr Molina Foundation, the Parker Institute for Cancer Immunotherapy, and the National Institute of Neurological Disorders and Stroke (NINDS), NIH (R35NS105068).


Sujet(s)
Tumeurs du tronc cérébral , Lymphocytes T CD8+/immunologie , Vaccins anticancéreux , Cytométrie en flux , Gliome , Histone , Immunité cellulaire/effets des médicaments et des substances chimiques , Mutation faux-sens , Protéines tumorales , Adolescent , Adulte , Substitution d'acide aminé , Tumeurs du tronc cérébral/génétique , Tumeurs du tronc cérébral/immunologie , Tumeurs du tronc cérébral/thérapie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/génétique , Vaccins anticancéreux/immunologie , Enfant , Enfant d'âge préscolaire , Femelle , Gliome/génétique , Gliome/immunologie , Gliome/thérapie , Histone/génétique , Histone/immunologie , Humains , Immunité cellulaire/génétique , Mâle , Protéines tumorales/génétique , Protéines tumorales/immunologie
13.
Nat Cancer ; 1(12): 1204-1217, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-35121932

RÉSUMÉ

Generating responses to tumor antigens poses a challenge for immunotherapy. This phase II trial (NCT02129075) tested fms-like tyrosine kinase 3 (Flt3) ligand pre-treatment enhancement of responses to dendritic cell (DC)-targeting vaccines. We evaluated a regimen of Flt3L (CDX-301) to increase DCs and other antigen-presenting cells, poly-ICLC (TLR3 agonist that activates DCs) and a vaccine comprising anti-DEC-205-NY-ESO-1, a fusion antibody targeting CD205, linked to NY-ESO-1. High-risk melanoma patients were randomized to vaccine, with and without CDX-301. The end point was immune response to NY-ESO-1. Flt3L increased peripheral monocytes and conventional DCs (cDCs), including cross-presenting cDC1 and cDC2 and plasmacytoid DCs. Significant increases in humoral and T-cell responses and activation of DCs, natural killer cells and T cells were elicited. Transcriptional analyses revealed gene signatures associated with CDX-301 induction of an early, durable immune response. This study reveals in vivo effects of Flt3L on innate immune cells in the setting of vaccination, leading to an immunogenic vaccine regimen.


Sujet(s)
Vaccins anticancéreux , Mélanome , Cellules dendritiques , Humains , Immunité , Protéines membranaires , Tyrosine kinase-3 de type fms
14.
Urol Clin North Am ; 47(4S): e1-e8, 2020 Nov.
Article de Anglais | MEDLINE | ID: mdl-33446322

RÉSUMÉ

Relatively simple, synthetic, double-stranded RNAs can be powerful viral pathogen-associated molecular pattern (PAMP) mimics, inducing a panoply of antiviral and antitumor responses that act at multiple stages of host defense. Their mechanisms of action and uses are beginning to be understood, alone, in combination with other therapeutics, or as novel PAMP-adjuvants providing the critical danger signal that has been missing from most cancer and other modern vaccines. Dose, timing, route of administration combinations, and other clinical variables can have a critical impact on immunogenicity. This article reviews advances in the use of polyinosinic-polycytidylic acid and derivatives, in particular poly-ICLC.


Sujet(s)
Adjuvants immunologiques/usage thérapeutique , Carboxyméthylcellulose de sodium/analogues et dérivés , Facteurs immunologiques/usage thérapeutique , Poly I-C/usage thérapeutique , Polylysine/analogues et dérivés , Tumeurs de la prostate/thérapie , ARN double brin/usage thérapeutique , Adjuvants immunologiques/physiologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Carboxyméthylcellulose de sodium/usage thérapeutique , Essais cliniques comme sujet , Humains , Facteurs immunologiques/immunologie , Mâle , Molécules contenant des motifs associés aux pathogènes/immunologie , Molécules contenant des motifs associés aux pathogènes/usage thérapeutique , Poly I-C/immunologie , Polylysine/immunologie , Polylysine/usage thérapeutique , Tumeurs de la prostate/immunologie , ARN double brin/immunologie
15.
Cancer Immunol Res ; 8(1): 70-80, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31699709

RÉSUMÉ

Given its ability to induce both humoral and cellular immune responses, NY-ESO-1 has been considered a suitable antigen for a cancer vaccine. Despite promising results from early-phase clinical studies in patients with melanoma, NY-ESO-1 vaccine immunotherapy has not been widely investigated in larger trials; consequently, many questions remain as to the optimal vaccine formulation, predictive biomarkers, and sequencing and timing of vaccines in melanoma treatment. We conducted an adjuvant phase I/II clinical trial in high-risk resected melanoma to optimize the delivery of poly-ICLC, a TLR-3/MDA-5 agonist, as a component of vaccine formulation. A phase I dose-escalation part was undertaken to identify the MTD of poly-ICLC administered in combination with NY-ESO-1 and montanide. This was followed by a randomized phase II part investigating the MTD of poly-ICLC with NY-ESO-1 with or without montanide. The vaccine regimens were generally well tolerated, with no treatment-related grade 3/4 adverse events. Both regimens induced integrated NY-ESO-1-specific CD4+ T-cell and humoral responses. CD8+ T-cell responses were mainly detected in patients receiving montanide. T-cell avidity toward NY-ESO-1 peptides was higher in patients vaccinated with montanide. In conclusion, NY-ESO-1 protein in combination with poly-ICLC is safe, well tolerated, and capable of inducing integrated antibody and CD4+ T-cell responses in most patients. Combination with montanide enhances antigen-specific T-cell avidity and CD8+ T-cell cross-priming in a fraction of patients, indicating that montanide contributes to the induction of specific CD8+ T-cell responses to NY-ESO-1.


Sujet(s)
Antigènes néoplasiques/administration et posologie , Vaccins anticancéreux/usage thérapeutique , Carboxyméthylcellulose de sodium/analogues et dérivés , Immunité cellulaire/immunologie , Immunité humorale/immunologie , Mannitol/analogues et dérivés , Mélanome/immunologie , Protéines membranaires/administration et posologie , Acides oléiques/administration et posologie , Poly I-C/administration et posologie , Polylysine/analogues et dérivés , Adjuvants immunologiques/administration et posologie , Sujet âgé , Antigènes néoplasiques/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/immunologie , Carboxyméthylcellulose de sodium/administration et posologie , Cross-priming/immunologie , Femelle , Humains , Inducteurs de l'interféron/administration et posologie , Mâle , Mannitol/administration et posologie , Mélanome/thérapie , Protéines membranaires/immunologie , Adulte d'âge moyen , Sécurité des patients , Polylysine/administration et posologie , Tumeurs cutanées/immunologie , Tumeurs cutanées/thérapie , Résultat thérapeutique
16.
Nat Med ; 25(5): 814-824, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30962585

RÉSUMÉ

Indolent non-Hodgkin's lymphomas (iNHLs) are incurable with standard therapy and are poorly responsive to checkpoint blockade. Although lymphoma cells are efficiently killed by primed T cells, in vivo priming of anti-lymphoma T cells has been elusive. Here, we demonstrate that lymphoma cells can directly prime T cells, but in vivo immunity still requires cross-presentation. To address this, we developed an in situ vaccine (ISV), combining Flt3L, radiotherapy, and a TLR3 agonist, which recruited, antigen-loaded and activated intratumoral, cross-presenting dendritic cells (DCs). ISV induced anti-tumor CD8+ T cell responses and systemic (abscopal) cancer remission in patients with advanced stage iNHL in an ongoing trial ( NCT01976585 ). Non-responding patients developed a population of PD1+CD8+ T cells after ISV, and murine tumors became newly responsive to PD1 blockade, prompting a follow-up trial of the combined therapy. Our data substantiate that recruiting and activating intratumoral, cross-priming DCs is achievable and critical to anti-tumor T cell responses and PD1-blockade efficacy.


Sujet(s)
Vaccins anticancéreux , Lymphome B/thérapie , Adulte , Sujet âgé , Animaux , Présentation d'antigène , Lymphocytes T CD8+/immunologie , Carboxyméthylcellulose de sodium/analogues et dérivés , Carboxyméthylcellulose de sodium/usage thérapeutique , Lignée cellulaire tumorale , Association thérapeutique , Cellules dendritiques/immunologie , Femelle , Humains , Immunothérapie adoptive , Lymphome B/immunologie , Lymphome B/anatomopathologie , Mâle , Protéines membranaires/immunologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , Adulte d'âge moyen , Poly I-C/usage thérapeutique , Polylysine/analogues et dérivés , Polylysine/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur de type Toll-3/agonistes , Vaccination
17.
Front Immunol ; 10: 725, 2019.
Article de Anglais | MEDLINE | ID: mdl-31024557

RÉSUMÉ

Objective: Toll-like receptor-3 agonist Poly-ICLC has been known to activate immune cells and induce HIV replication in pre-clinical experiments. In this study we investigated if Poly-ICLC could be used for disrupting HIV latency while simultaneously enhancing innate immune responses. Design: This was a randomized, placebo-controlled, double-blinded trial in aviremic, cART-treated HIV-infected subjects. Participants (n = 15) were randomized 3:1 to receive two consecutive daily doses of Poly-ICLC (1.4 mg subcutaneously) vs. placebo. Subjects were observed for adverse events, immune activation, and viral replication. Methods: Besides primary outcomes of safety and tolerability, several longitudinal immune parameters were evaluated including immune cell phenotype and function via flowcytometry, ELISA, and transcriptional profiling. PCR assays for plasma HIV-1 RNA, CD4+ T cell-associated HIV-1 RNA, and proviral DNA were performed to measure HIV reservoirs and latency. Results: Poly-ICLC was overall safe and well-tolerated. Poly-ICLC-related adverse events were Grade 1/2, with the exception of one Grade 3 neutropenia which was short-lived. Mild Injection site reactions were observed in nearly all participants in the Poly-ICLC arm. Transcriptional analyses revealed upregulation of innate immune pathways in PBMCs following Poly-ICLC treatment, including strong interferon signaling accompanied by transient increases in circulating IP-10 (CXCL10) levels. These responses generally peaked by 24-48 h after the first injection and returned to baseline by day 8. CD4+ T cell number and phenotype were unchanged, plasma viral control was maintained and no significant effect on HIV reservoirs was observed. Conclusions: These finding suggest that Poly-ICLC could be safely used for inducing transient innate immune responses in treated HIV+ subjects indicating promise as an adjuvant for HIV therapeutic vaccines. Trial Registration: www.ClinicalTrials.gov, identifier: NCT02071095.


Sujet(s)
Carboxyméthylcellulose de sodium/analogues et dérivés , Infections à VIH/traitement médicamenteux , Infections à VIH/immunologie , Immunité innée/effets des médicaments et des substances chimiques , Poly I-C/usage thérapeutique , Polylysine/analogues et dérivés , Récepteur de type Toll-3/métabolisme , Adulte , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Carboxyméthylcellulose de sodium/usage thérapeutique , Méthode en double aveugle , Femelle , VIH (Virus de l'Immunodéficience Humaine)/immunologie , Infections à VIH/métabolisme , Humains , Mâle , Adulte d'âge moyen , Polylysine/usage thérapeutique
18.
Cancer Immunol Immunother ; 68(3): 455-466, 2019 Mar.
Article de Anglais | MEDLINE | ID: mdl-30604041

RÉSUMÉ

Vaccines consisting of synthetic peptides representing cytotoxic T-lymphocyte (CTL) epitopes have long been considered as a simple and cost-effective approach to treat cancer. However, the efficacy of these vaccines in the clinic in patients with measurable disease remains questionable. We believe that the poor performance of peptide vaccines is due to their inability to generate sufficiently large CTL responses that are required to have a positive impact against established tumors. Peptide vaccines to elicit CTLs in the clinic have routinely been administered in the same manner as vaccines designed to induce antibody responses: injected subcutaneously and in many instances using Freund's adjuvant. We report here that peptide vaccines and poly-ICLC adjuvant administered via the unconventional intravenous route of immunization generate substantially higher CTL responses as compared to conventional subcutaneous injections, resulting in more successful antitumor effects in mice. Furthermore, amphiphilic antigen constructs such as palmitoylated peptides were shown to be better immunogens than long peptide constructs, which now are in vogue in the clinic. The present findings if translated into the clinical setting could help dissipate the wide-spread skepticism of whether peptide vaccines will ever work to treat cancer.


Sujet(s)
Vaccins anticancéreux/immunologie , Administration par voie intraveineuse , Animaux , Vaccins anticancéreux/administration et posologie , Injections sous-cutanées , Souris , Souris de lignée C57BL , Lymphocytes T cytotoxiques/immunologie , Vaccination , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/immunologie
19.
Antiviral Res ; 163: 179-184, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30611774

RÉSUMÉ

The potential protection of poly-ICLC (Hiltonol®) a double stranded RNA (dsRNA) against EBOV infection was assessed with prophylactic and therapeutic administration to wild type and TLR3-negative mice, and in non-human primates (NHPs) by measuring EBOL serum titers, survival extension, and serum liver and kidney function markers. Various doses of aqueous and liposomal poly-ICLC monotherapy provided robust protection in otherwise lethal murine EBOV challenge models, when treatment is started on the day 0 or one day after virus challenge. There was no advantage of liposomal vs. the aqueous poly-ICLC form. Protection appeared to be independent of TLR-3. NHPs treated with poly-ICLC and challenged with EBOV survived longer but eventually succumbed to Ebola infection. Nevertheless, the liver and kidney serum markers were markedly reduced in the infected and treated NHPs. In the two longest surviving poly-ICLC- treated NHPs, the day 10 serum EBOV titer was reduced 2.1 and 30 fold respectively.


Sujet(s)
Carboxyméthylcellulose de sodium/analogues et dérivés , Fièvre hémorragique à virus Ebola/traitement médicamenteux , Inducteurs de l'interféron/usage thérapeutique , Poly I-C/usage thérapeutique , Polylysine/analogues et dérivés , Animaux , Carboxyméthylcellulose de sodium/usage thérapeutique , République démocratique du Congo , Femelle , Macaca fascicularis , Souris , Souris de lignée BALB C , Polylysine/usage thérapeutique
20.
Nature ; 565(7738): 234-239, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30568305

RÉSUMÉ

Neoantigens, which are derived from tumour-specific protein-coding mutations, are exempt from central tolerance, can generate robust immune responses1,2 and can function as bona fide antigens that facilitate tumour rejection3. Here we demonstrate that a strategy that uses multi-epitope, personalized neoantigen vaccination, which has previously been tested in patients with high-risk melanoma4-6, is feasible for tumours such as glioblastoma, which typically have a relatively low mutation load1,7 and an immunologically 'cold' tumour microenvironment8. We used personalized neoantigen-targeting vaccines to immunize patients newly diagnosed with glioblastoma following surgical resection and conventional radiotherapy in a phase I/Ib study. Patients who did not receive dexamethasone-a highly potent corticosteroid that is frequently prescribed to treat cerebral oedema in patients with glioblastoma-generated circulating polyfunctional neoantigen-specific CD4+ and CD8+ T cell responses that were enriched in a memory phenotype and showed an increase in the number of tumour-infiltrating T cells. Using single-cell T cell receptor analysis, we provide evidence that neoantigen-specific T cells from the peripheral blood can migrate into an intracranial glioblastoma tumour. Neoantigen-targeting vaccines thus have the potential to favourably alter the immune milieu of glioblastoma.


Sujet(s)
Antigènes néoplasiques/immunologie , Vaccins anticancéreux/immunologie , Glioblastome/immunologie , Glioblastome/thérapie , Lymphocytes T/immunologie , Adulte , Sujet âgé , Méthylation de l'ADN , DNA modification methylases/génétique , Enzymes de réparation de l'ADN/génétique , Dexaméthasone/administration et posologie , Glioblastome/diagnostic , Glioblastome/génétique , Humains , Adulte d'âge moyen , Régions promotrices (génétique)/génétique , Récepteurs aux antigènes des cellules T/immunologie , Protéines suppresseurs de tumeurs/génétique , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...