Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.088
Filtrer
2.
Theranostics ; 14(9): 3674-3692, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948057

RÉSUMÉ

Trophoblast cell surface antigen 2 (Trop2) is overexpressed in a range of solid tumors and participants in multiple oncogenic signaling pathways, making it an attractive therapeutic target. In the past decade, the rapid development of various Trop2-targeted therapies, notably marked by the advent of the antibody-drug conjugate (ADC), revolutionized the outcome for patients facing Trop2-positive tumors with limited treatment opinions, such as triple-negative breast cancer (TNBC). This review provides a comprehensive summary of advances in Trop2-targeted therapies, including ADCs, antibodies, multispecific agents, immunotherapy, cancer vaccines, and small molecular inhibitors, along with in-depth discussions on their designs, mechanisms of action (MOAs), and limitations. Additionally, we emphasize the clinical research progress of these emerging Trop2-targeted agents, focusing on their clinical application and therapeutic efficacy against tumors. Furthermore, we propose directions for future research, such as enhancing our understanding of Trop2's structure and biology, exploring the best combination strategies, and tailoring precision treatment based on Trop2 testing methodologies.


Sujet(s)
Antigènes néoplasiques , Molécules d'adhérence cellulaire , Immunoconjugués , Thérapie moléculaire ciblée , Tumeurs , Humains , Antigènes néoplasiques/immunologie , Molécules d'adhérence cellulaire/antagonistes et inhibiteurs , Molécules d'adhérence cellulaire/métabolisme , Immunoconjugués/usage thérapeutique , Immunoconjugués/pharmacologie , Thérapie moléculaire ciblée/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Immunothérapie/méthodes , Animaux , Vaccins anticancéreux/usage thérapeutique
3.
Anticancer Res ; 44(8): 3543-3550, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39060086

RÉSUMÉ

BACKGROUND: Current standard treatment for metastatic breast cancer (MBC) involves cyclin-dependent kinase 4/6 (CDK4/6) inhibitors with endocrine therapy, showing potential in enhancing anti-tumor immune responses. CASE REPORT: This report details a clinical case of MBC where palbociclib was co-administered with letrozole. The integration of allogeneic tumor vaccination to this treatment led to heightened interferon-γ production, expansion of CD8+ and NK cell populations, and positive delayed-type hypersensitivity reactions, indicating successful development of anti-tumor immunity. The induced production of interferon-γ by tumor vaccination was associated with manageable modulation of sensitivity to palbociclib-letrozole therapy. Administration of the BioNTech/Pfizer Covid-19 vaccine compromised the anti-tumor immune response by reducing cytotoxic cell populations and increasing immunosuppressive cytokine production. The patient undergoing combined treatment achieved a progressive-free survival of 42 months. CONCLUSION: Incorporating active tumor vaccination with CDK4/6 inhibitor therapy presents a feasible approach for metastatic breast cancer. The precise regulation of the microenvironment emerges as a crucial factor and warrants careful consideration.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Vaccins anticancéreux , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Pipérazines , Pyridines , Humains , Femelle , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Pipérazines/administration et posologie , Pipérazines/usage thérapeutique , Pyridines/administration et posologie , Pyridines/usage thérapeutique , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Létrozole/administration et posologie , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/immunologie , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/administration et posologie , Adulte d'âge moyen , Interféron gamma/métabolisme
4.
Cancer Immunol Immunother ; 73(9): 178, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954031

RÉSUMÉ

Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.


Sujet(s)
Tumeurs du cerveau , Vaccins anticancéreux , Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Vaccins à ADN , Animaux , Femelle , Humains , Souris , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Lignée cellulaire tumorale , Glioblastome/immunologie , Glioblastome/thérapie , Glioblastome/traitement médicamenteux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Intramolecular oxidoreductases , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Vaccins à ADN/immunologie , Vaccins à ADN/usage thérapeutique , Mâle , Enfant , Adulte d'âge moyen
5.
PLoS One ; 19(7): e0305413, 2024.
Article de Anglais | MEDLINE | ID: mdl-38976715

RÉSUMÉ

Pancreatic ductal adenocarcinoma is the most prevalent pancreatic cancer, which is considered a significant global health concern. Chemotherapy and surgery are the mainstays of current pancreatic cancer treatments; however, a few cases are suitable for surgery, and most of the cases will experience recurrent episodes. Compared to DNA or peptide vaccines, mRNA vaccines for pancreatic cancer have more promise because of their delivery, enhanced immune responses, and lower proneness to mutation. We constructed an mRNA vaccine by analyzing S100 family proteins, which are all major activators of receptors for advanced glycation end products. We applied immunoinformatic approaches, including physicochemical properties analysis, structural prediction and validation, molecular docking study, in silico cloning, and immune simulations. The designed mRNA vaccine was estimated to have a molecular weight of 165023.50 Da and was highly soluble (grand average of hydropathicity of -0.440). In the structural assessment, the vaccine seemed to be a well-stable and functioning protein (Z score of -8.94). Also, the docking analysis suggested that the vaccine had a high affinity for TLR-2 and TLR-4 receptors. Additionally, the molecular mechanics with generalized Born and surface area solvation analysis of the "Vaccine-TLR-2" (-141.07 kcal/mol) and "Vaccine-TLR-4" (-271.72 kcal/mol) complexes also suggests a strong binding affinity for the receptors. Codon optimization also provided a high expression level with a GC content of 47.04% and a codon adaptation index score 1.0. The appearance of memory B-cells and T-cells was also observed over a while, with an increased level of helper T-cells and immunoglobulins (IgM and IgG). Moreover, the minimum free energy of the mRNA vaccine was predicted at -1760.00 kcal/mol, indicating the stability of the vaccine following its entry, transcription, and expression. This hypothetical vaccine offers a groundbreaking tool for future research and therapeutic development of pancreatic cancer.


Sujet(s)
Vaccins anticancéreux , Simulation de docking moléculaire , Tumeurs du pancréas , Tumeurs du pancréas/immunologie , Humains , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Vaccins à ARNm/immunologie , Biologie informatique/méthodes , Récepteur de type Toll-4/immunologie , Récepteur de type Toll-4/métabolisme , Vaccinologie/méthodes , Récepteur de type Toll-2/immunologie , Simulation numérique , ARN messager/génétique , ARN messager/immunologie ,
6.
Egypt J Immunol ; 31(3): 123-130, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38995717

RÉSUMÉ

Cancer immunotherapy is a promising strategy in cancer management, including hepatocellular carcinoma (HCC). This experimental study aimed to evaluate interleukin-10 (IL-10) as a biomarker for monitoring the response of tumor-derived autophagosomes vaccine in inducing antitumor immunity in HCC induced mice. It was conducted on 56 BALB/c mice; divided into 20 normal and 36, cancer induced with human liver cancer cell line (HepG2) cells. The latter group was subdivided into a positive control group (n=6) and a treated group (n=30), that was subdivided into 3 subgroups: (A) treated with dendritic cells (DC) vaccine only, (B) treated with vaccine named Dribbles only, and (C) treated with DC plus Dribbles. Serum IL-10 was assessed after immunotherapy. The mean percentage of tumor volume reduction in mice vaccinated by DC plus Dribbles was significantly superior to DC and Dribbles groups (p= 0.013, and p= 0.043, respectively). There was a statistically significant difference in IL-10 levels between different immunotherapy groups (p= 0.0003). As the mean IL-10 level was 19.50 pg/ml for the positive control group, 13 pg/ml for Dribbles group, 10 pg/ml for DCs group and 3.50 pg/ml for DCs plus Dribbles group. We conclude that DC-Dribbles vaccine has a remarkable efficacy superior to either Dribbles alone or DC alone in the decline of HCC development and survival improvement. IL-10 is a predictive biomarker for response after immunotherapy.


Sujet(s)
Vaccins anticancéreux , Carcinome hépatocellulaire , Cellules dendritiques , Immunothérapie , Interleukine-10 , Tumeurs du foie , Souris de lignée BALB C , Animaux , Interleukine-10/sang , Interleukine-10/immunologie , Cellules dendritiques/immunologie , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/thérapie , Souris , Tumeurs du foie/immunologie , Tumeurs du foie/thérapie , Humains , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Immunothérapie/méthodes , Cellules HepG2 , Modèles animaux de maladie humaine , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/immunologie
8.
ACS Nano ; 18(29): 18801-18833, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38979917

RÉSUMÉ

Tumor vaccines, an important part of immunotherapy, prevent cancer or kill existing tumor cells by activating or restoring the body's own immune system. Currently, various formulations of tumor vaccines have been developed, including cell vaccines, tumor cell membrane vaccines, tumor DNA vaccines, tumor mRNA vaccines, tumor polypeptide vaccines, virus-vectored tumor vaccines, and tumor-in-situ vaccines. There are also multiple delivery systems for tumor vaccines, such as liposomes, cell membrane vesicles, viruses, exosomes, and emulsions. In addition, to decrease the risk of tumor immune escape and immune tolerance that may exist with a single tumor vaccine, combination therapy of tumor vaccines with radiotherapy, chemotherapy, immune checkpoint inhibitors, cytokines, CAR-T therapy, or photoimmunotherapy is an effective strategy. Given the critical role of tumor vaccines in immunotherapy, here, we look back to the history of tumor vaccines, and we discuss the antigens, adjuvants, formulations, delivery systems, mechanisms, combination therapy, and future directions of tumor vaccines.


Sujet(s)
Vaccins anticancéreux , Tumeurs , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/composition chimique , Humains , Tumeurs/immunologie , Tumeurs/thérapie , Immunothérapie , Systèmes de délivrance de médicaments , Animaux , Liposomes/composition chimique
9.
Front Immunol ; 15: 1387835, 2024.
Article de Anglais | MEDLINE | ID: mdl-39035008

RÉSUMÉ

Oral Squamous Cell Carcinoma (OSCC) is the most common malignant tumor of the oral cavity. Despite recent advances in the field of oral cancer therapy, including the introduction of immunotherapeutic approaches, the 5-year survival rate remains steadily assessed around 50%. Thus, there is an urgent need for new therapeutic strategies. After the characterization of the immune phenotype of three human OSCC cell lines (CAL-27, SCC-25, and SCC-4) and one mouse OSCC cell line (MOC2) showing their similarities to resected patient tumors, we explored for the first time an experimental preclinical model of therapeutic vaccination with mouse OSCC MOC2 cell line stably expressing MHC class II antigens after CIITA gene transfection (MOC2-CIITA). Mice injected with MOC2-CIITA reject or strongly retard tumor growth; more importantly, vaccinated animals that fully reject MOC2-CIITA tumors display anti-tumor immunological memory protective against challenge with parental MOC2 tumor cells. Further experiments of adoptive cell transfer or in vivo cell depletion show that both CD4+ and CD8+ T lymphocytes prove fundamental in tumor rejection. This unprecedented approach for oral cancer opens the way for possible future translation of novel immunotherapeutic strategies to the human setting for the treatment of this tumor.


Sujet(s)
Vaccins anticancéreux , Carcinome épidermoïde , Tumeurs de la bouche , Animaux , Tumeurs de la bouche/immunologie , Tumeurs de la bouche/thérapie , Souris , Humains , Lignée cellulaire tumorale , Vaccins anticancéreux/immunologie , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/thérapie , Lymphocytes T auxiliaires/immunologie , Vaccination , Transactivateurs/génétique , Transactivateurs/immunologie , Femelle , Mémoire immunologique , Lymphocytes T CD4+/immunologie , Protéines nucléaires
10.
ACS Nano ; 18(28): 18604-18621, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38952130

RÉSUMÉ

Chemo-immunotherapy has become a promising strategy for cancer treatment. However, the inability of the drugs to penetrate deeply into the tumor and form potent tumor vaccines in vivo severely restricts the antitumor effect of chemo-immunotherapy. In this work, an injectable sodium alginate platform is reported to promote penetration of the chemotherapeutic doxorubicin (DOX) and delivery of personalized tumor vaccines. The injectable multifunctional sodium alginate platform cross-links rapidly in the presence of physiological concentrations of Ca2+, forming a hydrogel that acts as a drug depot and releases loaded hyaluronidase (HAase), DOX, and micelles (IP-NPs) slowly and sustainedly. By degrading hyaluronic acid (HA) overexpressed in tumor tissue, HAase can make tumor tissue "loose" and favor other components to penetrate deeply. DOX induces potent immunogenic cell death (ICD) and produces tumor-associated antigens (TAAs), which could be effectively captured by polyethylenimine (PEI) coated IP-NPs micelles and form personalized tumor vaccines. The vaccines efficaciously facilitate the maturation of dendritic cells (DCs) and activation of T lymphocytes, thus producing long-term immune memory. Imiquimod (IMQ) loaded in the core could further activate the immune system and trigger a more robust antitumor immune effect. Hence, the research proposes a multifunctional drug delivery platform for the effective treatment of colorectal cancer.


Sujet(s)
Alginates , Doxorubicine , Hydrogels , Immunothérapie , Nanoparticules , Alginates/composition chimique , Hydrogels/composition chimique , Animaux , Nanoparticules/composition chimique , Souris , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Humains , Vaccins anticancéreux/composition chimique , Vaccins anticancéreux/administration et posologie , Hyaluronoglucosaminidase/métabolisme , Micelles , Lignée cellulaire tumorale
11.
ACS Nano ; 18(28): 18425-18443, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38975713

RÉSUMÉ

Tumor in situ vaccination (ISV) strategies have emerged in clinical trials as promising approaches, involving the release of tumor antigens through local radiotherapy and intratumorally adjuvant injections. However, the current fabrication strategy for achieving a sustainable immune response to ISV remains a pressing challenge. In this study, we present an empowered sustainable ISV method for antitumor therapy using 177Lu-labeled manganese-doped mesoporous hydroxyapatite (177Lu/Mn-HAP) microspheres. The ISV enables the sustained utilization of tumor antigens, leading to the activation of dendritic cells and polarization of macrophages toward the M1 subtype. Consequently, it facilitates the generation of potent CD8+ T-cell responses, enhancing the antitumor effects of internal radiation in both primary and distant tumors. Importantly, this approach achieves complete remission in all tumor-bearing mice and stimulates immune memory to prevent tumor recurrence. Our study highlights a universal and safe ISV strategy capable of inducing potent tumor-specific and sustainable immune response.


Sujet(s)
Vaccins anticancéreux , Durapatite , Microsphères , Durapatite/composition chimique , Animaux , Souris , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/composition chimique , Lymphocytes T CD8+/immunologie , Vaccination , Femelle , Souris de lignée C57BL , Radio-isotopes/composition chimique , Lignée cellulaire tumorale
12.
Front Biosci (Landmark Ed) ; 29(7): 268, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39082348

RÉSUMÉ

Liver cancer, primarily hepatocellular carcinoma (HCC), is the second leading cause of cancer-related deaths globally. It is typically characterized by rapid progression, poor prognosis, and high mortality rates. Given these challenges, the search for molecular targets aiding early diagnosis and targeted therapy remains imperative. Glypican 3 (GPC3), a cell-surface glycoprotein, emerges as a promising candidate for addressing HCC Overexpressed in HCC tissues; GPC3 is a credible immunohistochemical marker for liver cancer diagnosis and a potential marker for liquid biopsy through soluble GPC3 in serum. Various immunotherapies targeting GPC3 have been developed, including vaccines, anti-GPC3 immunotoxins, and chimeric antigen receptor-modified cells. This review comprehensively covers the structure, physicochemical properties, biological functions, and clinical applications of GPC3. It explores diagnostic and treatment strategies centered around GPC3, offering hope for improved early detection and targeted therapies in the challenging landscape of HCC.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome hépatocellulaire , Glypicanes , Immunothérapie , Tumeurs du foie , Glypicanes/immunologie , Glypicanes/métabolisme , Humains , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/diagnostic , Carcinome hépatocellulaire/thérapie , Tumeurs du foie/immunologie , Tumeurs du foie/thérapie , Tumeurs du foie/diagnostic , Tumeurs du foie/métabolisme , Immunothérapie/méthodes , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/immunologie , Marqueurs biologiques tumoraux/sang , Thérapie moléculaire ciblée/méthodes , Médecine de précision/méthodes , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique
13.
Biomed Pharmacother ; 177: 117083, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38968793

RÉSUMÉ

Cancer stem cells (CSCs) can self-renew and differentiate, contributing to tumor heterogeneity, metastasis, and recurrence. Their resistance to therapies, including immunotherapy, underscores the importance of targeting them for complete remission and relapse prevention. Olfactomedin 4 (OLFM4), a marker associated with various cancers such as colorectal cancer, is expressed on CSCs promoting immune evasion and tumorigenesis. However, its potential as a target for CSC-specific immunotherapy remains underexplored. The primary aim of this study is to evaluate the effectiveness of targeting OLFM4 with dendritic cell (DC)-based vaccines in inhibiting tumor growth and metastasis. To improve antigen delivery and immune response, OLFM4 was conjugated with a protein-transduction domain (PTD) from the antennapedia of Drosophila called penetratin, creating a fusion protein (P-OLFM4). The efficacy of DCs pulsed with P-OLFM4 (DCs [P-OLFM4]) was compared to DCs pulsed with OLFM4 (DCs [OLFM4]) and PBS (DCs [PBS]). DCs [P-OLFM4] inhibited tumor growth by 91.2 % and significantly reduced lung metastasis of OLFM4+ melanoma cells by 97 %, compared to the DCs [PBS]. DCs [OLFM4] also demonstrated a reduction in lung metastasis by 59.7 % compared to DCs [PBS]. Immunization with DCs [P-OLFM4] enhanced OLFM4-specific T-cell proliferation, interferon-γ production, and cytotoxic T cell activity in mice. The results indicate that OLFM4 is a viable target for CSC-focused immunotherapy. DC [P-OLFM4] vaccines can elicit robust immune responses, significantly inhibiting tumor growth and metastasis. This strategy holds promise for developing more effective cancer treatments that specifically target CSCs, potentially leading to better patient outcomes by reducing the likelihood of tumor relapse and metastasis.


Sujet(s)
Cellules dendritiques , Souris de lignée C57BL , Animaux , Cellules dendritiques/immunologie , Souris , Peptides de pénétration cellulaire , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Mélanome expérimental/anatomopathologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Femelle , Lignée cellulaire tumorale , Prolifération cellulaire , Immunothérapie/méthodes , Tumeurs du poumon/secondaire , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie , Tumeurs du poumon/anatomopathologie , Protéines de la matrice extracellulaire/métabolisme , Métastase tumorale
14.
Oncoimmunology ; 13(1): 2373526, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948931

RÉSUMÉ

Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.


Sujet(s)
Adjuvants immunologiques , Antigènes néoplasiques , Vaccins anticancéreux , Cellules dendritiques , Tumeurs de la prostate , ARN messager , Animaux , Mâle , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/thérapie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/traitement médicamenteux , Antigènes néoplasiques/immunologie , Souris , Cellules dendritiques/immunologie , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , ARN messager/génétique , ARN messager/métabolisme , ARN messager/administration et posologie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Humains , Souris de lignée C57BL , Lignée cellulaire tumorale , Vaccins à ARNm , Lymphocytes T CD8+/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Immunothérapie/méthodes , Activation des lymphocytes/effets des médicaments et des substances chimiques
15.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38955422

RÉSUMÉ

BACKGROUND: Despite advances in checkpoint inhibitor (CPI) therapy for cancer treatment, many cancers remain resistant. Tumors deemed "cold" based on lack of T cell infiltration show reduced potential for CPI therapy. Cancer vaccines may overcome the inadequacy of existing T cells by inducing the needed antitumor T cell response to synergize with CPIs and overcome resistance. METHODS: CT26 and TC1 tumor cells were injected subcutaneously into mice. Mice were treated with combinations of CPIs alone or a cancer vaccine specific to the tumor antigen E7 present in TC1 cells. CPIs for the TC1 model were selected because of immunophenotyping TC1 tumors. Antitumor and protumor immunity, tumor size and survival, sequence and timing of vaccine and CPI administration, and efficacy of treatment in young and aged mice were probed. RESULTS: While "hot" CT26 tumors are treatable with combinations of second-generation CPIs alone or with anti-TGFß, "cold" TC1 tumor reduction requires the synergy of a tumor-antigen-specific vaccine in combination with two CPIs, anti-TIGIT and anti-PD-L1, predicted by tumor microenvironment (TME) characterization. The synergistic triple combination delays tumor growth better than any pairwise combination and improves survival in a CD8+T cell-dependent manner. Depletion of CD4+T cells improved the treatment response, and depleting regulatory T cells (Treg) revealed Tregs to be inhibiting the response as also predicted from TME analysis. We found the sequence of CPI and vaccine administration dictates the success of the treatment, and the triple combination administered concurrently induces the highest E7-specific T cell response. Contrary to young mice, in aged mice, the cancer vaccine alone is ineffective, requiring the CPIs to delay tumor growth. CONCLUSIONS: These findings show how pre-existing or vaccine-mediated de novo T cell responses can both be amplified by and facilitate synergistic CPIs and Treg depletion that together lead to greater survival, and how analysis of the TME can help rationally design combination therapies and precision medicine to enhance clinical response to CPI and cancer vaccine therapy.


Sujet(s)
Vaccins anticancéreux , Inhibiteurs de points de contrôle immunitaires , Lymphocytes T régulateurs , Microenvironnement tumoral , Animaux , Vaccins anticancéreux/pharmacologie , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/immunologie , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Microenvironnement tumoral/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Femelle , Lignée cellulaire tumorale , Humains
16.
J Am Chem Soc ; 146(28): 19218-19228, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38955767

RÉSUMÉ

The messenger RNA (mRNA) vaccines hold great significance in contagion prevention and cancer immunotherapy. However, safely and effectively harnessing innate immunity to stimulate robust and durable adaptive immune protection is crucial, yet challenging. In this study, we synthesized a library of stimuli-responsive bivalent ionizable lipids (srBiv iLPs) with smart molecular blocks responsive to esterase, H2O2, cytochrome P450, alkaline phosphatase, nitroreductase, or glutathione (GSH), aiming to leverage physiological cues to trigger fast lipid degradation, promote mRNA translation, and induce robust antitumor immunity via reactive oxygen species (ROS)-mediated boosting. After subcutaneous immunization, esterase-responsive vaccine (eBiv-mVac) was rapidly internalized and transported into the draining lymph nodes. It then underwent fast decaging and self-immolative degradation in esterase-rich antigen-presenting cells, releasing sufficient mRNA for antigen translation and massive reactive quinone methides to elevate ROS levels. This resulted in broad activation of innate immunity to boost T cell response, prompting a large number of primed antigen-specific CD8+ T cells to circulate and infiltrate into tumors (>1000-fold versus unvaccinated control), thereby orchestrating innate and adaptive immunity to control tumor growth. Moreover, by further combining our vaccination strategy with immune checkpoint blockade, we demonstrated a synergism that significantly amplified the magnitude and function of antigen-specific CD8+ T cells. This, in turn, caused potent systemic antitumor efficacy and prolonged survival with high complete response rate in xenograft and metastasis models. Overall, our generalized stimuli-responsive mRNA delivery platform promises a paradigm shift in the design of potent vaccines for cancer immunotherapy, as well as effective and precise carriers for gene editing, protein replacement, and cell engineering.


Sujet(s)
Lymphocytes T CD8+ , Immunité innée , Espèces réactives de l'oxygène , Espèces réactives de l'oxygène/métabolisme , Immunité innée/effets des médicaments et des substances chimiques , Animaux , Lymphocytes T CD8+/immunologie , Souris , Vaccins à ARNm/composition chimique , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/composition chimique , ARN messager/immunologie , ARN messager/génétique , Humains , Souris de lignée C57BL
17.
PLoS One ; 19(7): e0307204, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008481

RÉSUMÉ

Malignant pleural mesothelioma (MPM) is an aggressive cancer with a very poor prognosis. Recently, immune checkpoint inhibition (ICI) has taken center stage in the currently ongoing revolution that is changing standard-of-care treatment for several malignancies, including MPM. As multiple arguments and accumulating lines of evidence are in support of the existence of a therapeutic synergism between chemotherapy and immunotherapy, as well as between different classes of immunotherapeutics, we designed a multicenter, single-arm, phase I/II trial in which both programmed-death-ligand 1 (PD-L1) inhibition and dendritic cell (DC) vaccination are integrated in the first-line conventional platinum/pemetrexed-based treatment scheme for epithelioid MPM patients (Immuno-MESODEC, ClinicalTrials.gov identifier NCT05765084). Fifteen treatment-naïve patients with unresectable epithelioid subtype MPM will be treated with four 3-weekly (±3 days) chemo-immunotherapy cycles. Standard-of-care chemotherapy consisting of cisplatinum (75mg/m2) and pemetrexed (500mg/m2) will be supplemented with the anti-PD-L1 antibody atezolizumab (1200 mg) and autologous Wilms' tumor 1 mRNA-electroporated dendritic cell (WT1/DC) vaccination (8-10 x 106 cells/vaccination). Additional atezolizumab (1680 mg) doses and/or WT1/DC vaccinations (8-10 x 106 cells/vaccination) can be administered optionally following completion of the chemo-immunotherapy scheme. Follow-up of patients will last for up to 90 days after final atezolizumab administration and/or WT1/DC vaccination or 24 months after diagnosis, whichever occurs later. The trial's primary endpoints are safety and feasibility, secondary endpoints are clinical efficacy and immunogenicity. This phase I/II trial will evaluate whether addition of atezolizumab and WT1/DC vaccination to frontline standard-of-care chemotherapy for the treatment of epithelioid MPM is feasible and safe. If so, this novel combination strategy should be further investigated as a promising advanced treatment option for this hard-to-treat cancer.


Sujet(s)
Anticorps monoclonaux humanisés , Antigène CD274 , Vaccins anticancéreux , Cellules dendritiques , Mésothéliome malin , Humains , Mésothéliome malin/traitement médicamenteux , Mésothéliome malin/immunologie , Cellules dendritiques/immunologie , Anticorps monoclonaux humanisés/usage thérapeutique , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Mâle , Femelle , Protéines WT1/immunologie , Tumeurs de la plèvre/immunologie , Tumeurs de la plèvre/traitement médicamenteux , Tumeurs de la plèvre/thérapie , Immunothérapie/méthodes , Adulte d'âge moyen , Adulte , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Sujet âgé , Vaccination , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Pémétrexed/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Mésothéliome/traitement médicamenteux , Mésothéliome/immunologie , Mésothéliome/thérapie , Cisplatine/usage thérapeutique , Cisplatine/pharmacologie
18.
J Immunother Cancer ; 12(7)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977329

RÉSUMÉ

The development of vaccines, especially RNA-based, directed against patient-specific tumor neoepitopes is an active and productive area of cancer immunotherapy. Promising clinical results in melanoma and other solid tumor types are emerging. As with all cancer therapy modalities, neoepitope vaccine development and delivery also has some drawbacks, including the level of effort to develop a patient-specific product, accuracy of algorithms to predict neoepitopes, and with the exception of melanoma and some other tumor types, biopsies of metastatic lesions of solid tumors are often not available. We hypothesize that in some circumstances the use of rationally designed combinations of "off-the-shelf" agents may prove an additional path to enable the patient to produce his/her own "neoepitope vaccine" in situ. These combination therapies may consist of agents to activate a tumor-associated T-cell response, potentiate that response, reduce or eliminate immunosuppressive entities in the tumor microenvironment, and/or alter the phenotype of tumor cells to render them more susceptible to immune-mediated lysis. Examples are provided in both preclinical and clinical studies in which combinations of "off-the-shelf" agents lead to the generation of T cells directed against tumor-derived neoepitopes with consequent antitumor activity.


Sujet(s)
Vaccins anticancéreux , Humains , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Immunothérapie/méthodes , Tumeurs/immunologie , Tumeurs/thérapie , Lymphocytes T/immunologie , Microenvironnement tumoral/immunologie
19.
AAPS PharmSciTech ; 25(6): 168, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39044047

RÉSUMÉ

Immunotherapy is one of the most promising therapeutic approaches in the field of cancer treatment. As a tumor progresses, tumor cells employ an array of immune-regulatory mechanisms to suppress immune responses within the tumor microenvironment. Using our understanding of these mechanisms, cancer immunotherapy has been developed to enhance the immune system's effectiveness in treating cancer. Numerous cancer immunotherapies are currently in clinical use, yet many others are either in different stages of development or undergoing clinical studies. In this paper, we briefly discuss the features and current status of cancer immunotherapies. This includes the application of monoclonal antibodies, immune checkpoint inhibitors, adoptive cell therapy, cytokine therapy, cancer vaccines, and gene therapy, all of which have gained significant recognition in clinical practice. Additionally, we discuss limitations that may hinder successful clinical utilization and promising strategies, such as combining immunotherapy with nanotechnology.


Sujet(s)
Vaccins anticancéreux , Immunothérapie , Tumeurs , Microenvironnement tumoral , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Immunothérapie/tendances , Microenvironnement tumoral/immunologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/administration et posologie , Animaux , Thérapie génétique/méthodes , Thérapie génétique/tendances , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Nanotechnologie/méthodes , Cytokines/métabolisme , Cytokines/immunologie
20.
Front Immunol ; 15: 1437774, 2024.
Article de Anglais | MEDLINE | ID: mdl-39055714

RÉSUMÉ

Alternative splicing (AS) functions as a crucial program in transcriptional modulation, leading to proteomic diversity and functional alterations of proteins. These splicing actions induce various neoantigens that hold prognostic significance and contribute to various aspects of cancer progression, including immune responses against cancer. The advent of immunotherapy has remarkably revolutionized tumor therapy. In this regard, AS-derived neoantigens are potent targets for cancer vaccines and chimeric antigen receptor (CAR) T cell therapies. In this review, we outline that AS-derived neoantigens serve as promising immunotherapeutic targets and guide immunotherapy strategies. This evidence contributes to a deeper comprehension of the complexity of proteomic diversity and provides novel perspectives and techniques for precision medicine in immunotherapy. Moreover, we underscore the obstacles that are awaited to be addressed for this novel approach to become clinically applicable.


Sujet(s)
Épissage alternatif , Antigènes néoplasiques , Tumeurs , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Antigènes néoplasiques/immunologie , Antigènes néoplasiques/génétique , Animaux , Immunothérapie/méthodes , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Immunothérapie adoptive/méthodes , Médecine de précision/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE