Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.675
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(25): e2322264121, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38865265

RESUMO

Despite the tremendous clinical potential of nucleic acid-based vaccines, their efficacy to induce therapeutic immune response has been limited by the lack of efficient local gene delivery techniques in the human body. In this study, we develop a hydrogel-based organic electronic device (µEPO) for both transdermal delivery of nucleic acids and in vivo microarrayed cell electroporation, which is specifically oriented toward one-step transfection of DNAs in subcutaneous antigen-presenting cells (APCs) for cancer immunotherapy. The µEPO device contains an array of microneedle-shaped electrodes with pre-encapsulated dry DNAs. Upon a pressurized contact with skin tissue, the electrodes are rehydrated, electrically triggered to release DNAs, and then electroporate nearby cells, which can achieve in vivo transfection of more than 50% of the cells in the epidermal and upper dermal layer. As a proof-of-concept, the µEPO technique is employed to facilitate transdermal delivery of neoantigen genes to activate antigen-specific immune response for enhanced cancer immunotherapy based on a DNA vaccination strategy. In an ovalbumin (OVA) cancer vaccine model, we show that high-efficiency transdermal transfection of APCs with OVA-DNAs induces robust cellular and humoral immune responses, including antigen presentation and generation of IFN-γ+ cytotoxic T lymphocytes with a more than 10-fold dose sparing over existing intramuscular injection (IM) approach, and effectively inhibits tumor growth in rodent animals.


Assuntos
Eletroporação , Imunoterapia , Vacinas de DNA , Animais , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Eletroporação/métodos , Camundongos , Imunoterapia/métodos , Administração Cutânea , Neoplasias/terapia , Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Ovalbumina/imunologia , Ovalbumina/administração & dosagem , Células Apresentadoras de Antígenos/imunologia , Feminino , Camundongos Endogâmicos C57BL , Humanos , Vacinação/métodos
2.
IET Nanobiotechnol ; 2024: 5593879, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38863969

RESUMO

Cancer vaccines based on tumor cell components have shown promising results in animal and clinical studies. The vaccine system contains abundant tumor antigen components, which can activate the immune system by antigens. However, their efficacy has been limited by the inability of antigens delivery, which are the core components of vaccines, further fail to be presented and activation of effective cells. Nanotechnology offers a novel platform to enhance the immunogenicity of tumor-associated antigens and deliver them to antigen-presenting cells (APCs) more efficiently. In addition, nanotreatment of tumor cells derivate active ingredients could also help improve the effectiveness of cancer vaccines. In this review, we summarize recent advances in the development of cancer vaccines by the combination of nanotechnology and tumor-based ingredients, including liposomes, polymeric nanoparticles, metallic nanoparticles, virus-like particles and tumor cells membrane, tumor lysate, and specific tumor antigens. These nanovaccines have been designed to increase antigen uptake, prolong antigen presentation, and modulate immune responses through codelivery of immunostimulatory agents. We also further discuss challenges and opportunities in the clinical translation of these nanovaccines.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/química , Vacinas Anticâncer/administração & dosagem , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Animais , Nanopartículas/química , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/química
3.
Cell Rep Med ; 5(5): 101560, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38729159

RESUMO

Stimulator of IFN genes (STING) is a promising target for adjuvants utilized in in situ cancer vaccination approaches. However, key barriers remain for clinical translation, including low cellular uptake and accessibility, STING variability necessitating personalized STING agonists, and interferon (IFN)-independent signals that can promote tumor growth. Here, we identify C100, a highly deacetylated chitin-derived polymer (HDCP), as an attractive alternative to conventional STING agonists. C100 promotes potent anti-tumor immune responses, outperforming less deacetylated HDCPs, with therapeutic efficacy dependent on STING and IFN alpha/beta receptor (IFNAR) signaling and CD8+ T cell mediators. Additionally, C100 injection synergizes with systemic checkpoint blockade targeting PD-1. Mechanistically, C100 triggers mitochondrial stress and DNA damage to exclusively activate the IFN arm of the cGAS-STING signaling pathway and elicit sustained IFNAR signaling. Altogether, these results reveal an effective STING- and IFNAR-dependent adjuvant for in situ cancer vaccines with a defined mechanism and distinct properties that overcome common limitations of existing STING therapeutics.


Assuntos
Adjuvantes Imunológicos , Linfócitos T CD8-Positivos , Quitina , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta , Transdução de Sinais , Animais , Proteínas de Membrana/metabolismo , Proteínas de Membrana/imunologia , Proteínas de Membrana/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Receptor de Interferon alfa e beta/genética , Camundongos , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Humanos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Feminino , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Neoplasias/imunologia , Neoplasias/terapia
4.
J Immunother Cancer ; 12(5)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38782542

RESUMO

BACKGROUND: Neoantigens can serve as targets for T cell-mediated antitumor immunity via personalized neopeptide vaccines. Interim data from our clinical study NCT03715985 showed that the personalized peptide-based neoantigen vaccine EVX-01, formulated in the liposomal adjuvant, CAF09b, was safe and able to elicit EVX-01-specific T cell responses in patients with metastatic melanoma. Here, we present results from the dose-escalation part of the study, evaluating the feasibility, safety, efficacy, and immunogenicity of EVX-01 in addition to anti-PD-1 therapy. METHODS: Patients with metastatic melanoma on anti-PD-1 therapy were treated in three cohorts with increasing vaccine dosages (twofold and fourfold). Tumor-derived neoantigens were selected by the AI platform PIONEER and used in personalized therapeutic cancer peptide vaccines EVX-01. Vaccines were administered at 2-week intervals for a total of three intraperitoneal and three intramuscular injections. The study's primary endpoint was safety and tolerability. Additional endpoints were immunological responses, survival, and objective response rates. RESULTS: Compared with the base dose level previously reported, no new vaccine-related serious adverse events were observed during dose escalation of EVX-01 in combination with an anti-PD-1 agent given according to local guidelines. Two patients at the third dose level (fourfold dose) developed grade 3 toxicity, most likely related to pembrolizumab. Overall, 8 out of the 12 patients had objective clinical responses (6 partial response (PR) and 2 CR), with all 4 patients at the highest dose level having a CR (1 CR, 3 PR). EVX-01 induced peptide-specific CD4+ and/or CD8+T cell responses in all treated patients, with CD4+T cells as the dominating responses. The magnitude of immune responses measured by IFN-γ ELISpot assay correlated with individual peptide doses. A significant correlation between the PIONEER quality score and induced T cell immunogenicity was detected, while better CRs correlated with both the number of immunogenic EVX-01 peptides and the PIONEER quality score. CONCLUSION: Immunization with EVX-01-CAF09b in addition to anti-PD-1 therapy was shown to be safe and well tolerated and elicit vaccine neoantigen-specific CD4+and CD8+ T cell responses at all dose levels. In addition, objective tumor responses were observed in 67% of patients. The results encourage further assessment of the antitumor efficacy of EVX-01 in combination with anti-PD-1 therapy.


Assuntos
Antígenos de Neoplasias , Vacinas Anticâncer , Melanoma , Medicina de Precisão , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/uso terapêutico , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Melanoma/tratamento farmacológico , Melanoma/imunologia , Metástase Neoplásica , Medicina de Precisão/métodos , Vacinas de Subunidades Antigênicas/uso terapêutico , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem
5.
Nat Commun ; 15(1): 3882, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719809

RESUMO

In this randomized phase II clinical trial, we evaluated the effectiveness of adding the TLR agonists, poly-ICLC or resiquimod, to autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination in patients with newly-diagnosed or recurrent WHO Grade III-IV malignant gliomas. The primary endpoints were to assess the most effective combination of vaccine and adjuvant in order to enhance the immune potency, along with safety. The combination of ATL-DC vaccination and TLR agonist was safe and found to enhance systemic immune responses, as indicated by increased interferon gene expression and changes in immune cell activation. Specifically, PD-1 expression increases on CD4+ T-cells, while CD38 and CD39 expression are reduced on CD8+ T cells, alongside an increase in monocytes. Poly-ICLC treatment amplifies the induction of interferon-induced genes in monocytes and T lymphocytes. Patients that exhibit higher interferon response gene expression demonstrate prolonged survival and delayed disease progression. These findings suggest that combining ATL-DC with poly-ICLC can induce a polarized interferon response in circulating monocytes and CD8+ T cells, which may represent an important blood biomarker for immunotherapy in this patient population.Trial Registration: ClinicalTrials.gov Identifier: NCT01204684.


Assuntos
Linfócitos T CD8-Positivos , Vacinas Anticâncer , Carboximetilcelulose Sódica/análogos & derivados , Células Dendríticas , Glioma , Interferons , Poli I-C , Polilisina/análogos & derivados , Humanos , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Glioma/imunologia , Glioma/terapia , Feminino , Masculino , Pessoa de Meia-Idade , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Poli I-C/administração & dosagem , Poli I-C/farmacologia , Adulto , Receptores Toll-Like/agonistas , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Idoso , Vacinação , Monócitos/imunologia , Monócitos/efeitos dos fármacos , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/tratamento farmacológico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Imunoterapia/métodos , Agonistas do Receptor Semelhante a Toll
6.
J Control Release ; 370: 379-391, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38697317

RESUMO

Although various types of mRNA-based vaccines have been explored, the optimal conditions for induction of both humoral and cellular immunity remain rather unknown. In this study, mRNA vaccines of nucleoside-modified mRNA in lipoplexes (LPXs) or lipid nanoparticles (LNPs) were evaluated after administration in mice through different routes, assessing mRNA delivery, tolerability and immunogenicity. In addition, we investigated whether mRNA vaccines could benefit from the inclusion of the adjuvant alpha-galactosylceramide (αGC), an invariant Natural Killer T (iNKT) cell ligand. Intramuscular (IM) vaccination with ovalbumin (OVA)-encoding mRNA encapsulated in LNPs adjuvanted with αGC showed the highest antibody- and CD8+ T cell responses. Furthermore, we observed that addition of signal peptides and endocytic sorting signals of either LAMP1 or HLA-B7 in the OVA-encoding mRNA sequence further enhanced CD8+ T cell activation although reducing the induction of IgG antibody responses. Moreover, mRNA LNPs with the ionizable lipidoid C12-200 exhibited higher pro-inflammatory- and reactogenic activity compared to mRNA LNPs with SM-102, correlating with increased T cell activation and antitumor potential. We also observed that αGC could further enhance the cellular immunity of clinically relevant mRNA LNP vaccines, thereby promoting therapeutic antitumor potential. Finally, a Listeria monocytogenes mRNA LNP vaccine supplemented with αGC showed synergistic protective effects against listeriosis, highlighting a key advantage of co-activating iNKT cells in antibacterial mRNA vaccines. Taken together, our study offers multiple insights for optimizing the design of mRNA vaccines for disease applications, such as cancer and intracellular bacterial infections.


Assuntos
Vacinas Anticâncer , Galactosilceramidas , Camundongos Endogâmicos C57BL , Nanopartículas , Ovalbumina , Animais , Galactosilceramidas/administração & dosagem , Galactosilceramidas/química , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Feminino , Nanopartículas/química , Nanopartículas/administração & dosagem , Ovalbumina/imunologia , Ovalbumina/administração & dosagem , Vacinas de mRNA , Adjuvantes Imunológicos/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , RNA Mensageiro/administração & dosagem , Camundongos , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Lipídeos/química , Lipossomos
7.
J Control Release ; 370: 707-720, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38744346

RESUMO

Immunotherapy is gaining prominence as a promising strategy for treating triple-negative breast cancer (TNBC). Neoantigens (neoAgs) and cancer-testis antigens (CTAs) are tumor-specific targets originating from somatic mutations and epigenetic changes in cancer cells. These antigens hold great promise for personalized cancer vaccines, as supported by preclinical and early clinical evidence in TNBC. This review delves into the potential of neoAgs and CTAs as vaccine candidates, emphasizing diverse strategies and delivery approaches. It also highlights the current status of vaccination modalities undergoing clinical trials in TNBC therapy. A comprehensive understanding of neoAgs, CTAs, vaccination strategies, and innovative delivery methods is crucial for optimizing neoAg-based immunotherapies in clinical practice.


Assuntos
Antígenos de Neoplasias , Vacinas Anticâncer , Imunoterapia , Neoplasias de Mama Triplo Negativas , Humanos , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Antígenos de Neoplasias/imunologia , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/terapia , Feminino , Animais , Imunoterapia/métodos , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos/métodos
9.
Int J Nanomedicine ; 19: 4533-4568, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799699

RESUMO

Until now, there has been a lack of effective strategies for cancer treatment. Immunotherapy has high potential in treating several cancers but its efficacy is limited as a monotherapy. Chemoimmunotherapy (CIT) holds promise to be widely used in cancer treatment. Therefore, identifying their involvement and potential synergy in CIT approaches is decisive. Nano-based drug delivery systems (NDDSs) are ideal delivery systems because they can simultaneously target immune cells and cancer cells, promoting drug accumulation, and reducing the toxicity of the drug. In this review, we first introduce five current immunotherapies, including immune checkpoint blocking (ICB), adoptive cell transfer therapy (ACT), cancer vaccines, oncolytic virus therapy (OVT) and cytokine therapy. Subsequently, the immunomodulatory effects of chemotherapy by inducing immunogenic cell death (ICD), promoting tumor killer cell infiltration, down-regulating immunosuppressive cells, and inhibiting immune checkpoints have been described. Finally, the NDDSs-mediated collaborative drug delivery systems have been introduced in detail, and the development of NDDSs-mediated CIT nanoparticles has been prospected.


Assuntos
Imunoterapia , Neoplasias , Humanos , Imunoterapia/métodos , Neoplasias/terapia , Neoplasias/imunologia , Animais , Nanopartículas/química , Vacinas Anticâncer/administração & dosagem , Terapia Viral Oncolítica/métodos , Sistemas de Liberação de Fármacos por Nanopartículas/química , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Terapia Combinada/métodos
10.
Biomater Sci ; 12(12): 3175-3192, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38742916

RESUMO

The tumor immunosuppressive microenvironment (TIME) and uncontrollable release of antigens can lower the efficacy of nanovaccine-based immunotherapy (NBI). Therefore, it is necessary to develop a new strategy for TIME reshaping and controllable release of antigens to improve the NBI efficacy. Herein, an acidity-responsive Schiff base-conjugated polyphenol-coordinated nanovaccine was constructed for the first time to realize bidirectional TIME reshaping and controllable release of antigens for activating T cells. In particular, an acidity-responsive tannic acid-ovalbumin (TA-OVA) nanoconjugate was prepared via a Schiff base reaction. FeIII was coordinated with TA-OVA to produce a FeIII-TA-OVA nanosystem, and 1-methyltryptophan (1-MT) as an indoleamine 2,3-dioxygenase inhibitor was loaded to form a polyphenol-coordinated nanovaccine. The coordination between FeIII and TA could cause photothermal ablation of primary tumors, and the acidity-triggered Schiff base dissociation of TA-OVA could controllably release OVA to realize lysosome escape, initiating the body's immune response. More importantly, oxidative stress generated by a tumor-specific Fenton reaction of Fe ions could promote the polarization of tumor-associated macrophages from the M2 to M1 phenotype, resulting in the upregulation of cytotoxic T cells and helper T cells. Meanwhile, 1-MT could downregulate immunosuppressive regulatory T cells. Overall, such skillful combination of bidirectional TIME reshaping and controllable antigen release into one coordination nanosystem could effectively enhance the NBI efficacy of tumors.


Assuntos
Imunoterapia , Ovalbumina , Polifenóis , Bases de Schiff , Taninos , Microambiente Tumoral , Animais , Microambiente Tumoral/efeitos dos fármacos , Ovalbumina/imunologia , Ovalbumina/química , Ovalbumina/administração & dosagem , Polifenóis/química , Polifenóis/farmacologia , Camundongos , Taninos/química , Taninos/farmacologia , Bases de Schiff/química , Concentração de Íons de Hidrogênio , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Triptofano/química , Triptofano/análogos & derivados , Nanoconjugados/química , Camundongos Endogâmicos C57BL , Nanopartículas/química , Linhagem Celular Tumoral , Compostos Férricos/química , Nanovacinas
11.
Signal Transduct Target Ther ; 9(1): 118, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702343

RESUMO

Antitumor therapies based on adoptively transferred T cells or oncolytic viruses have made significant progress in recent years, but the limited efficiency of their infiltration into solid tumors makes it difficult to achieve desired antitumor effects when used alone. In this study, an oncolytic virus (rVSV-LCMVG) that is not prone to induce virus-neutralizing antibodies was designed and combined with adoptively transferred T cells. By transforming the immunosuppressive tumor microenvironment into an immunosensitive one, in B16 tumor-bearing mice, combination therapy showed superior antitumor effects than monotherapy. This occurred whether the OV was administered intratumorally or intravenously. Combination therapy significantly increased cytokine and chemokine levels within tumors and recruited CD8+ T cells to the TME to trigger antitumor immune responses. Pretreatment with adoptively transferred T cells and subsequent oncolytic virotherapy sensitizes refractory tumors by boosting T-cell recruitment, down-regulating the expression of PD-1, and restoring effector T-cell function. To offer a combination therapy with greater translational value, mRNA vaccines were introduced to induce tumor-specific T cells instead of adoptively transferred T cells. The combination of OVs and mRNA vaccine also displays a significant reduction in tumor burden and prolonged survival. This study proposed a rational combination therapy of OVs with adoptive T-cell transfer or mRNA vaccines encoding tumor-associated antigens, in terms of synergistic efficacy and mechanism.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Camundongos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Terapia Viral Oncolítica/métodos , Terapia Combinada , Vacinas de mRNA/imunologia , Melanoma Experimental/terapia , Melanoma Experimental/imunologia , Microambiente Tumoral/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T/imunologia , Humanos , Linhagem Celular Tumoral , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/administração & dosagem
12.
Rev Med Suisse ; 20(874): 984-989, 2024 May 15.
Artigo em Francês | MEDLINE | ID: mdl-38756036

RESUMO

For over a decade, immunotherapy has been transforming cancer treatment and prognosis. Tumor therapeutic vaccines trigger new immune responses and enhance existing immunity to more effectively combat cancer. These vaccines aim to curb the established disease or prevent recurrence, unlike conventional preventive vaccines. There are four categories of therapeutic vaccines: cellular, viral/bacterial, peptide, and nucleic acid, each with its own benefits and challenges. Advances in the understanding of anti-tumor immunity and advanced technologies such as mRNA vaccines support the development of this new treatment option. Currently in clinical trials, they could lead to promising and personalised anti-cancer therapies.


Depuis plus d'une décennie, l'immunothérapie améliore le traitement et le pronostic des patients atteints de cancer. Les vaccins thérapeutiques tumoraux activent de nouvelles réponses immunitaires et amplifient l'immunité existante pour combattre le cancer plus efficacement. Ces vaccins visent à freiner la maladie établie ou à éviter les récidives, à la différence des vaccins préventifs classiques. Il existe quatre catégories de vaccins thérapeutiques : cellulaire, viral/bactérien, peptidique et à acide nucléique, chacun avec des bénéfices et des défis spécifiques. Les avancées dans la compréhension de l'immunité antitumorale et dans les technologies de pointe, comme les vaccins à ARNm, favorisent le développement de cette nouvelle option de traitement. Actuellement en essais cliniques, ils pourraient aboutir à des thérapies anticancéreuses prometteuses et personnalisées.


Assuntos
Vacinas Anticâncer , Imunoterapia , Neoplasias , Humanos , Vacinas Anticâncer/administração & dosagem , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/prevenção & controle , Imunoterapia/métodos , Imunoterapia/tendências
13.
J Control Release ; 370: 287-301, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38679162

RESUMO

The success of lipid nanoparticles (LNPs) in treating COVID-19 promotes further research of mRNA vaccines for cancer vaccination. Aiming at overcoming the constraints of currently available mRNA carriers, various alternative nano-vectors have been developed for delivering tumor antigen encoding mRNA and showed versatility to induce potent anti-tumor immunity. The rationally designed nano-vaccines increase the immune activation capacity of the mRNA vaccines by promoting crucial aspects including mRNA stability, cellular uptake, endosomal escape and targeting of immune cells or organs. Herein, we summarized the research progress of various mRNA based nano-vaccines that have been reported for cancer vaccination, including LNPs, lipid enveloped hybrid nanoparticles, polymeric nanoparticles etc. Several strategies that have been reported for further enhancing the immune stimulation efficacy of mRNA nano-vaccines, including developing nano-vaccines for co-delivering adjuvants, combination of immune checkpoint inhibitors, and optimizing the injection routes for boosting immune responses, have been reviewed. The progress of mRNA nano-vaccines in clinical trials and the prospect of the mRNA vaccines for cancer vaccination are also discussed.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Vacinas de mRNA , Humanos , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Neoplasias/terapia , Neoplasias/imunologia , Nanopartículas/administração & dosagem , Animais , Vacinas de mRNA/administração & dosagem , RNA Mensageiro/administração & dosagem , RNA Mensageiro/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Sistemas de Liberação de Medicamentos/métodos , Lipídeos/química , Lipossomos
14.
Mol Ther ; 32(6): 1917-1933, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38637990

RESUMO

Cancer immunotherapy has greatly improved the prognosis of tumor-bearing patients. Nevertheless, cancer patients exhibit low response rates to current immunotherapy drugs, such as PD1 and PDL1 antibodies. Cyclic dinucleotide analogs are a promising class of immunotherapeutic agents. In this study, in situ autologous tumor vaccines, composed of bis-2'-F-cGSASMP phosphonothioate isomers (FGA-di-pS-2 or FGA-di-pS-4) and cytidinyl/cationic lipids (Mix), were constructed. Intravenous and intratumoral injection of FGA-di-pS-2/Mix or FGA-di-pS-4/Mix enhanced the immunogenic cell death of tumor cells in vivo, leading to the exposure and presentation of whole tumor antigens, inhibiting tumor growth in both LLC and EO771 tumor in situ murine models and increasing their survival rates to 50% and 23%, respectively. Furthermore, the tumor-bearing mice after treatment showed potent immune memory efficacy and exhibited 100% protection against tumor rechallenge. Intravenous administration of FGA-di-pS-2/Mix potently promoted DC maturation, M1 macrophage polarization and CD8+ T cell activation and decreased the proportion of Treg cells in the tumor microenvironment. Notably, two doses of ICD-debris (generated by FGA-di-pS-2 or 4/Mix-treated LLC cells) protected 100% of mice from tumor growth. These tumor vaccines showed promising results and may serve as personalized cancer vaccinations in the future.


Assuntos
Vacinas Anticâncer , Imunoterapia , Animais , Camundongos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Imunoterapia/métodos , Linhagem Celular Tumoral , Humanos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Células Dendríticas/imunologia , Feminino , Antígenos de Neoplasias/imunologia
15.
J Control Release ; 369: 687-695, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38575073

RESUMO

Extracts of the Chilean soapbark tree, Quillaja Saponaria (QS) are the source of potent immune-stimulatory saponin compounds. This study compared the adjuvanticity and toxicity of QS-18 and QS-21, assessing the potential to substitute QS-18 in place of QS-21 for vaccine development. QS-18, the most abundant QS saponin fraction, has been largely overlooked due to safety concerns. We found that QS-18 spontaneously inserted into liposomes, thereby neutralizing hemolytic activity, and following administration did not induce local reactogenicity in a footpad swelling test in mice. With high-dose intramuscular administration, transient weight loss was minor, and QS-18 did not induce significantly more weight loss compared to a liposome vaccine adjuvant system lacking it. Two days after administration, no elevation of inflammatory cytokines was detected in murine serum. In a formulation including cobalt-porphyrin-phospholipid (CoPoP) for short peptide sequestration, QS-18 did not impact the formation of peptide nanoparticles. With immunization, QS-18 peptide particles induced higher levels of cancer neoepitope-specific and tumor-associated antigen-specific CD8+ T cells compared to QS-21 particles, without indication of greater toxicity based on mouse body weight. T cell receptor sequencing of antigen-specific CD8+ T cells showed that QS-18 induced significantly more T cell transcripts. In two murine cancer models, vaccination with QS-18 peptide particles induced a similar therapeutic effect as QS-21 particles, without indication of increased toxicity. Antigen-specific CD8+ T cells in the tumor microenvironment were found to express the exhaustion marker PD-1, pointing to the rationale for exploring combination therapy. Taken together, these data demonstrate that QS-18, when formulated in liposomes, can be a safe and effective adjuvant to induce tumor-inhibiting cellular responses in murine models with potential to facilitate or diminish costs of production for vaccine adjuvant systems. Further studies are warranted to assess liposomal QS-18 immunogic, reactogenic and toxicological profiles in mice and other animal species.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer , Lipossomos , Quillaja , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Quillaja/química , Adjuvantes Imunológicos/administração & dosagem , Feminino , Camundongos Endogâmicos C57BL , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Camundongos , Saponinas de Quilaia , Citocinas , Saponinas/administração & dosagem , Saponinas/farmacologia , Linhagem Celular Tumoral , Vacinas de Subunidades Proteicas
16.
ACS Nano ; 18(18): 11910-11920, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38680054

RESUMO

Personalized antitumor immunotherapy utilizing neoantigen vaccines holds great promise. However, the limited immunogenicity of existing recognized neoantigens and the inadequate stimulation of antitumor immune responses by conventional adjuvants pose significant challenges. To address these limitations, we developed a nanovaccine that combines a BCG bacterial cell wall skeleton (BCG-CWS) based nanoscale adjuvant (BCNA) with peptide neoantigens (M27 and M30). This integrated approach provides an efficient translational strategy for cancer immunotherapy. The BCNA nanovaccine, formulated with PLGA as an emulsifier, exhibits excellent biocompatibility and superior antigen presentation compared with conventional BCG-CWS adjuvants. Subcutaneous immunization with the BCNA-based nanovaccine effectively targets lymph nodes, eliciting robust innate and tumor-specific immune responses. Importantly, our findings demonstrate that BCNAs significantly enhance neoantigen immunogenicity while minimizing acute systemic toxicity. Furthermore, when combined with a mouse PD-L1 antibody, our strategy achieves complete tumor elimination in 60% of cases and prevents 25% of tumor growth in a melanoma mouse model. In conclusion, our BCNA-based nanovaccine represents a promising avenue for advancing personalized therapeutic neoantigen vaccines and holds significant implications for enhancing personalized immunotherapy and improving patient outcomes in the field of cancer treatment.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer , Imunoterapia , Animais , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Antígenos de Neoplasias/imunologia , Feminino , Humanos , Parede Celular/imunologia , Parede Celular/química , Mycobacterium bovis/imunologia , Nanopartículas/química , Vacina BCG/imunologia , Linhagem Celular Tumoral
17.
Theranostics ; 14(6): 2290-2303, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646651

RESUMO

Background: Neoantigen nanovaccine has been recognized as a promising treatment modality for personalized cancer immunotherapy. However, most current nanovaccines are carrier-dependent and the manufacturing process is complicated, resulting in potential safety concerns and suboptimal codelivery of neoantigens and adjuvants to antigen-presenting cells (APCs). Methods: Here we report a facile and general methodology for nanoassembly of peptide and oligonucleotide by programming neoantigen peptide with a short cationic module at N-terminus to prepare nanovaccine. The programmed peptide can co-assemble with CpG oligonucleotide (TLR9 agonist) into monodispersed nanostructures without the introduction of artificial carrier. Results: We demonstrate that the engineered nanovaccine promoted the codelivery of neoantigen peptides and adjuvants to lymph node-residing APCs and instigated potent neoantigen-specific T-cell responses, eliciting neoantigen-specific antitumor immune responses with negligible systemic toxicity. Furthermore, the antitumor T-cell immunity is profoundly potentiated when combined with anti-PD-1 therapy, leading to significant inhibition or even complete regression of established melanoma and MC-38 colon tumors. Conclusions: Collectively, this work demonstrates the feasibility and effectiveness of personalized cancer nanovaccine preparation with high immunogenicity and good biosafety by programming neoantigen peptide for nanoassembly with oligonucleotides without the aid of artificial carrier.


Assuntos
Antígenos de Neoplasias , Vacinas Anticâncer , Peptídeos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Animais , Camundongos , Antígenos de Neoplasias/imunologia , Peptídeos/imunologia , Peptídeos/química , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Oligodesoxirribonucleotídeos/química , Células Apresentadoras de Antígenos/imunologia , Linhagem Celular Tumoral , Imunoterapia/métodos , Humanos , Feminino , Linfócitos T/imunologia , Nanoestruturas/química , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Neoplasias do Colo/tratamento farmacológico
18.
Adv Sci (Weinh) ; 11(22): e2309770, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38528670

RESUMO

Messenger RNA (mRNA) cancer vaccines are a new class of immunotherapies that can activate the immune system to recognize and destroy cancer cells. However, their effectiveness in treating colorectal cancer located on the mucosal surface of the gut is limited due to the insufficient activation of mucosal immune response and inadequate infiltration of cytotoxic T cells into tumors. To address this issue, a new mRNA cancer vaccine is developed that can stimulate mucosal immune responses in the gut by co-delivering all-trans-retinoic acid (ATRA) and mRNA using lipid nanoparticle (LNP). The incorporation of ATRA has not only improved the mRNA transfection efficiency of LNP but also induced high expression of gut-homing receptors on vaccine-activated T cells. Additionally, the use of LNP improves the aqueous solubility of ATRA, eliminating the need for toxic solvents to administer ATRA. Upon intramuscular injections, ATRA-adjuvanted mRNA-LNP significantly increase the infiltration of antigen-specific, cytotoxic T cells in the lamina propria of the intestine, mesenteric lymph nodes, and orthotopic colorectal tumors, resulting in significantly improved tumor inhibition and prolonged animal survival compared to conventional mRNA-LNP without ATRA. Overall, this study provides a promising approach for improving the therapeutic efficacy of mRNA cancer vaccines against colorectal cancer.


Assuntos
Vacinas Anticâncer , Neoplasias Colorretais , Tretinoína , Tretinoína/farmacologia , Tretinoína/administração & dosagem , Animais , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Neoplasias Colorretais/tratamento farmacológico , Camundongos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Imunidade nas Mucosas/efeitos dos fármacos , Imunidade nas Mucosas/imunologia , Modelos Animais de Doenças , Nanopartículas , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Feminino , Humanos , Camundongos Endogâmicos BALB C , Vacinas de mRNA , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/administração & dosagem
19.
Clin Cancer Res ; 30(11): 2412-2423, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38506710

RESUMO

PURPOSE: Personalized vaccines targeting multiple neoantigens (nAgs) are a promising strategy for eliciting a diversified antitumor T-cell response to overcome tumor heterogeneity. NOUS-PEV is a vector-based personalized vaccine, expressing 60 nAgs and consists of priming with a nonhuman Great Ape Adenoviral vector (GAd20) followed by boosts with Modified Vaccinia Ankara. Here, we report data of a phase Ib trial of NOUS-PEV in combination with pembrolizumab in treatment-naïve patients with metastatic melanoma (NCT04990479). PATIENTS AND METHODS: The feasibility of this approach was demonstrated by producing, releasing, and administering to 6 patients 11 of 12 vaccines within 8 weeks from biopsy collection to GAd20 administration. RESULTS: The regimen was safe, with no treatment-related serious adverse events observed and mild vaccine-related reactions. Vaccine immunogenicity was demonstrated in all evaluable patients receiving the prime/boost regimen, with detection of robust neoantigen-specific immune responses to multiple neoantigens comprising both CD4 and CD8 T cells. Expansion and diversification of vaccine-induced T-cell receptor (TCR) clonotypes was observed in the posttreatment biopsies of patients with clinical response, providing evidence of tumor infiltration by vaccine-induced neoantigen-specific T cells. CONCLUSIONS: These findings indicate the ability of NOUS-PEV to amplify and broaden the repertoire of tumor-reactive T cells to empower a diverse, potent, and durable antitumor immune response. Finally, a gene signature indicative of the reduced presence of activated T cells together with very poor expression of the antigen-processing machinery genes has been identified in pretreatment biopsies as a potential biomarker of resistance to the treatment.


Assuntos
Adenoviridae , Antígenos de Neoplasias , Vacinas Anticâncer , Vetores Genéticos , Medicina de Precisão , Humanos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/genética , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , Feminino , Pessoa de Meia-Idade , Masculino , Medicina de Precisão/métodos , Adenoviridae/genética , Adenoviridae/imunologia , Melanoma/terapia , Melanoma/imunologia , Idoso , Vacinação/métodos , Linfócitos T/imunologia , Adulto , Linfócitos T CD8-Positivos/imunologia
20.
Adv Healthc Mater ; 13(13): e2304384, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38301259

RESUMO

Complete surgical resection of tumor is difficult as the invasiveness of cancer, making the residual tumor a lethal threat to patients. The situation is deteriorated by the immune suppression state after surgery, which further nourishes tumor recurrence and metastasis. Immunotherapy is promising to combat tumor metastasis, but is limited by severe toxicity of traditional immunostimulants and complexity of multiple functional units. Here, it is reported that the simple "trans-surgical bed" delivery of Cu2- xSe nanozyme (CSN) by a microneedle-patch can turn the threat to therapy by efficient in situ vaccination. The biocompatible CSN exhibits both peroxidase and glutathione oxidase-like activities, efficiently exhausting glutathione, boosting free radical generation, and inducing immunogenic cell death. The once-for-all inserting of the patch on surgical bed facilitates sustained catalytic action, leading to drastic decrease of recurrence rate and complete suppression of tumor-rechallenge in cured mice. In vivo mechanism interrogation reveals elevated cytotoxic T cell infiltration, re-educated macrophages, increased dendritic cell maturation, and memory T cells formation. Importantly, preliminary metabolism and safety evaluation validated that the metal accumulation is marginable, and the important biochemical indexes are in normal range during therapy. This study has provided a simple, safe, and robust tumor vaccination approach for postsurgical metastasis control.


Assuntos
Vacinas Anticâncer , Animais , Camundongos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/química , Cobre/química , Linhagem Celular Tumoral , Catálise , Imunoterapia/métodos , Selênio/química , Selênio/farmacologia , Vacinação , Feminino , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/patologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...