Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 11.439
Filtrer
1.
Biomaterials ; 313: 122776, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39236629

RÉSUMÉ

Surgical resection, the mainstay for melanoma treatment, faces challenges due to high tumor recurrence rates and complex postoperative wound healing. Chronic inflammation from residual disease and the risk of secondary infections impede healing. We introduce an innovative, injectable hydrogel system that integrates a multifaceted therapeutic approach. The hydrogel, crosslinked by calcium ions with sodium alginate, encapsulates a blood clot rich in dendritic cells (DCs) chemoattractants and melanoma cell-derived nanovesicles (NVs), functioning as a potent immunostimulant. This in situ recruitment strategy overcomes the limitations of subcutaneous tumor vaccine injections and more effectively achieves antitumor immunity. Additionally, the hydrogel incorporates Chlorella extracts, enhancing its antimicrobial properties to prevent wound infections and promote healing. One of the key findings of our research is the dual functionality of Chlorella extracts; they not only expedite the healing process of infected wounds but also increase the hydrogel's ability to stimulate an antitumor immune response. Given the patient-specific nature of the blood clot and NVs, our hydrogel system offers customizable solutions for individual postoperative requirements. This personalized approach is highlighted by our study, which demonstrates the synergistic impact of the composite hydrogel on preventing melanoma recurrence and hastening wound healing, potentially transforming postsurgical melanoma management.


Sujet(s)
Cellules dendritiques , Hydrogels , Mélanome , Cicatrisation de plaie , Hydrogels/composition chimique , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Mélanome/thérapie , Mélanome/anatomopathologie , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Humains , Récidive tumorale locale/prévention et contrôle , Souris de lignée C57BL , Anti-infectieux/usage thérapeutique , Anti-infectieux/pharmacologie , Souris , Lignée cellulaire tumorale , Femelle
2.
Front Immunol ; 15: 1451103, 2024.
Article de Anglais | MEDLINE | ID: mdl-39355255

RÉSUMÉ

Background: Immunotherapy has revolutionized skin cutaneous melanoma treatment, but response variability due to tumor heterogeneity necessitates robust biomarkers for predicting immunotherapy response. Methods: We used weighted gene co-expression network analysis (WGCNA), consensus clustering, and 10 machine learning algorithms to develop the immunotherapy-related gene model (ITRGM) signature. Multi-omics analyses included bulk and single-cell RNA sequencing of melanoma patients, mouse bulk RNA sequencing, and pathology sections of melanoma patients. Results: We identified 66 consensus immunotherapy prognostic genes (CITPGs) using WGCNA and differentially expressed genes (DEGs) from two melanoma cohorts. The CITPG-high group showed better prognosis and enriched immune activities. DEGs between CITPG-high and CITPG-low groups in the TCGA-SKCM cohort were analyzed in three additional melanoma cohorts using univariate Cox regression, resulting in 44 consensus genes. Using 101 machine learning algorithm combinations, we constructed the ITRGM signature based on seven model genes. The ITRGM outperformed 37 published signatures in predicting immunotherapy prognosis across the training cohort, three testing cohorts, and a meta-cohort. It effectively stratified patients into high-risk or low-risk groups for immunotherapy response. The low-risk group, with high levels of model genes, correlated with increased immune characteristics such as tumor mutation burden and immune cell infiltration, indicating immune-hot tumors with a better prognosis. The ITRGM's relationship with the tumor immune microenvironment was further validated in our experiments using pathology sections with GBP5, an important model gene, and CD8 IHC analysis. The ITRGM also predicted better immunotherapy response in eight cohorts, including urothelial carcinoma and stomach adenocarcinoma, indicating broad applicability. Conclusions: The ITRGM signature is a stable and robust predictor for stratifying melanoma patients into 'immune-hot' and 'immune-cold' tumors, enhancing prognosis and response to immunotherapy.


Sujet(s)
Marqueurs biologiques tumoraux , Immunothérapie , Apprentissage machine , Mélanome , Humains , Mélanome/thérapie , Mélanome/immunologie , Mélanome/génétique , Immunothérapie/méthodes , Marqueurs biologiques tumoraux/génétique , Pronostic , Tumeurs cutanées/immunologie , Tumeurs cutanées/thérapie , Tumeurs cutanées/génétique , Animaux , Analyse de profil d'expression de gènes , Transcriptome , Régulation de l'expression des gènes tumoraux , Souris , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Résultat thérapeutique , Réseaux de régulation génique
3.
Mol Biol (Mosk) ; 58(2): 189-203, 2024.
Article de Russe | MEDLINE | ID: mdl-39355878

RÉSUMÉ

Uveal melanoma (UM) is a neuroectodermal tumor that results from malignant transformation of melanocytes in the eye uvea, including the iris, the ciliary body, and the choroid. UM accounts for 5% of all melanoma cases and is extremely aggressive with half of the UM patients developing metastases within the first 1-2 years after tumor development. Molecular mechanisms of UM carcinogenesis are poorly understood, but are known to differ from those of skin melanoma. Activating mutations of the GNAQ and GNA11 genes, which code for the large G protein subunits Gq and G11, respectively, are found in 90% of UM patients. The Gaq/PKC/MAPK signaling pathway is a main signaling cascade that leads to the transformation of melanocytes of the uveal tract, and major regulators of the cascade provide targets for the development of drugs. Metastatic UM (MUM) is most often associated with mutations of BAP1, EIF1AX, GNA11, GNAQ, and SF3B1. A combination of a commercial expression test panel of 15 genes and a mutation panel of 7 genes, supplemented with data on the size of the primary tumor, is highly efficient in predicting the risk of metastasis. The risk of metastasis determines the choice of therapy and the patient follow-up regimen. However, no systemic therapy for MUM has been developed to date. New drugs undergoing clinical trials are mostly targeted drugs designed to inhibit the protein products of mutant genes or immunotherapeutic agents designed to stimulate the immune response against specific antigens. In addition to these approaches, potential therapeutic targets of epigenetic regulation of UM development are considered in the review.


Sujet(s)
Mélanome , Mutation , Tumeurs de l'uvée , Humains , Tumeurs de l'uvée/génétique , Tumeurs de l'uvée/anatomopathologie , Tumeurs de l'uvée/métabolisme , Tumeurs de l'uvée/traitement médicamenteux , Tumeurs de l'uvée/thérapie , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/traitement médicamenteux , Mélanome/métabolisme , Mélanome/thérapie , Sous-unités alpha des protéines G/génétique , Sous-unités alpha des protéines G/métabolisme , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme , Sous-unités alpha Gq-G11 des protéines G/génétique , Sous-unités alpha Gq-G11 des protéines G/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
4.
Science ; 386(6717): eabq7248, 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39361750

RÉSUMÉ

The premise of cancer immunotherapy is that cancers are specifically visible to an immune system tolerized to healthy self. The promise of cancer immunotherapy is that immune effector mechanisms and immunological memory can jointly eradicate cancers and inoperable metastases and de facto vaccinate against recurrence. For some patients with hitherto incurable diseases, including metastatic melanoma, this promise is being realized by game-changing immunotherapies based on αß T cells. Today's challenges are to bring benefit to greater numbers of patients of diverse ethnicities, target more cancer types, and achieve a cure while incurring fewer adverse events. In meeting those challenges, specific benefits may be offered by γδ T cells, which compose a second T cell lineage with distinct recognition capabilities and functional traits that bridge innate and adaptive immunity. γδ T cell-based clinical trials, including off-the-shelf adoptive cell therapy and agonist antibodies, are yielding promising results, although identifiable problems remain. In addressing those problems, we advocate that immunotherapies be guided by the distinctive biology of γδ T cells, as elucidated by ongoing research.


Sujet(s)
Immunothérapie adoptive , Lymphocytes intra-épithéliaux , Tumeurs , Récepteur lymphocytaire T antigène, gamma-delta , Animaux , Humains , Essais cliniques comme sujet , Immunothérapie adoptive/méthodes , Lymphocytes intra-épithéliaux/immunologie , Mélanome/thérapie , Mélanome/immunologie , Tumeurs/thérapie , Tumeurs/immunologie , Récepteur lymphocytaire T antigène, gamma-delta/immunologie
5.
Front Immunol ; 15: 1442573, 2024.
Article de Anglais | MEDLINE | ID: mdl-39391320

RÉSUMÉ

Melanoma, recognized as one of the most immunogenic malignancies in humans, holds paramount significance in the realm of immunotherapy. However, the emergence of drug resistance and the occurrence of adverse drug reactions underscore the pressing need to explore increasingly personalized immunotherapeutic modalities. Extracellular Vesicles (EVs), pivotal derivatives of immune cells, assume pivotal roles by encapsulating proteins, lipids, and nucleic acids within bilayer lipid structures, thereby facilitating targeted delivery to other immune cells. This orchestrated process orchestrates critical functions including antigen presentation, immune modulation, and the induction of apoptosis in tumor cells. A burgeoning body of evidence underscores the vast therapeutic potential of EVs in melanoma treatment. This comprehensive review aims to delineate the roles of EVs derived from immune cells such as dendritic cells, natural killer cells, macrophages, and T cells in the context of melanoma patients, thereby furnishing invaluable insights for the future direction of melanoma immunotherapy.


Sujet(s)
Vésicules extracellulaires , Mélanome , Humains , Mélanome/immunologie , Mélanome/thérapie , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Animaux , Immunomodulation , Immunothérapie/méthodes , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Microenvironnement tumoral/immunologie , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme
6.
Sci Adv ; 10(40): eado8366, 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39365866

RÉSUMÉ

Despite our increasing understanding of macrophage heterogeneity, drivers of macrophage phenotypic and functional polarization in the microenvironment are not fully elucidated. Here, our single-cell RNA sequencing data identify a subpopulation of macrophages expressing high levels of the phagocytic receptor MER proto-oncogene tyrosine kinase (MerTK+ macrophages), which is closely associated with melanoma progression and immunotherapy resistance. Adoptive transfer of the MerTK+ macrophages into recipient mice notably accelerated tumor growth regardless of macrophage depletion. Mechanistic studies further revealed that ALK And LTK Ligand 1 (ALKAL1), a target gene of aryl hydrocarbon receptor (AhR), facilitated MerTK phosphorylation, resulting in heightened phagocytic activity of MerTK+ macrophages and their subsequent polarization toward an immunosuppressive phenotype. Specifically targeted delivery of AhR antagonist to tumor-associated macrophages with mannosylated micelles could suppress MerTK expression and improved the therapeutic efficacy of anti-programmed cell death ligand 1 therapy. Our findings shed light on the regulatory mechanism of MerTK+ macrophages and provide strategies for improving the efficacy of melanoma immunotherapy.


Sujet(s)
Immunothérapie , Macrophages , Mélanome , Récepteurs à hydrocarbure aromatique , c-Mer Tyrosine kinase , Sujet âgé , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , c-Mer Tyrosine kinase/métabolisme , c-Mer Tyrosine kinase/génétique , Lignée cellulaire tumorale , Évolution de la maladie , Résistance aux médicaments antinéoplasiques , Immunothérapie/méthodes , Macrophages/métabolisme , Macrophages/immunologie , Mélanome/thérapie , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome/métabolisme , Mélanome expérimental/thérapie , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Souris de lignée C57BL , Phosphorylation , Proto-oncogène Mas , Récepteurs à hydrocarbure aromatique/métabolisme , Microenvironnement tumoral/immunologie , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme
7.
Clin Exp Med ; 24(1): 234, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39352553

RÉSUMÉ

Metastatic uveal melanoma (mUM) is associated with poor prognosis. Ipilimumab/nivolumab has shown antitumor efficacy in phase II studies. Tebentafusp resulted in longer overall survival (OS) compared to investigator`s choice in a phase III study. We sought to describe the radiological response patterns of mUM patients treated with immunotherapy. Patients with mUM treated with ipilimumab/nivolumab and tebentafusp between July 2018 and December 2022, with available radiological assessment per RECISTv1.1 and/or imPERCIST5, were retrospectively identified and included. Progression-free survival (PFS) and OS rates, liver-specific response and pathological assessment in available liver biopsies were evaluated. In the ipilimumab/nivolumab group, median PFS (mPFS) was 2.9 months (95% CI 2.2-28.6) and mOS 28.9 months (95% CI 12.7-NR). Complete (CMR) and partial (PMR) metabolic response per imPERCIST5, and partial response (PR) per RECISTv1.1 were associated with longer PFS and OS by trend, compared to morphologically and metabolically stable or progressive disease. In the tebentafusp group, mPFS was 2.7 months (95% CI 2.2-3) and mOS 18.6 months (95% CI 11.5-NR). PMR and PR were associated with longer PFS by trend. In both treatments, the overall treatment response was associated with the radiological response at the liver site. In available liver tumor biopsies, differences in pathological and radiological responses were noted. ImPERCIST5 and RECIST v1.1 are valuable tools in the radiological response assessment, but both methods display limitations. Accurate biomarkers to stratify patients at risk for disease progression and future translational studies to investigate mechanisms of response and resistance are required.


Sujet(s)
Immunothérapie , Ipilimumab , Mélanome , Nivolumab , Tumeurs de l'uvée , Humains , Tumeurs de l'uvée/anatomopathologie , Tumeurs de l'uvée/traitement médicamenteux , Tumeurs de l'uvée/mortalité , Tumeurs de l'uvée/thérapie , Tumeurs de l'uvée/immunologie , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Mélanome/thérapie , Mélanome/immunologie , Études rétrospectives , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Nivolumab/usage thérapeutique , Ipilimumab/usage thérapeutique , Immunothérapie/méthodes , Adulte , Résultat thérapeutique , Résistance aux médicaments antinéoplasiques , Sujet âgé de 80 ans ou plus , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Survie sans progression
8.
J Invest Surg ; 37(1): 2401125, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39370138

RÉSUMÉ

BACKGROUND: Malignant melanoma, a highly aggressive skin cancer, has remarkable incidence and mortality nowadays. This study aims to explore prognostic factors associated with nonmetastatic cutaneous melanoma of the limbs and to develop nomograms for predicting overall survival (OS) and cancer-specific survival (CSS). METHODS: The study cohort was derived from the Surveillance, Epidemiology, and End Results database. Univariate Cox regression, Lasso regression, and multivariate Cox regression analyses were conducted to identify prognostic factors and construct nomograms. The receiver operating characteristic (ROC) curve, time-dependent C-index, calibration curve, decision curve analysis (DCA) and Kaplan-Meier method were used to evaluate the accuracy and clinical applicability of the nomograms. RESULTS: A total of 15,606 patients were enrolled. Multivariate analysis identified several prognostic factors for OS and CSS including age, sex, histologic type, N stage, tumor thickness, depth of invasion, mitotic rate, ulceration, surgery of primary site, systemic therapy, race, and number of lymph nodes examined. A nomogram incorporating 12 independent predictors for OS was developed, with a C-index of 0.866 (95% confidence interval [CI]: 0.858-0.874) in the training cohort and 0.853 (95% CI: 0.839-0.867) in validation. For CSS, 10 independent predictors and one related factor were included, yielding a C-index of 0.913 (95% CI: 0.903-0.923) in the training cohort and 0.922 (95% CI: 0.908-0.936) in validation. The ROC curve, time-dependent C-index, calibration curve, DCA, and K-M plot demonstrated favorable discrimination, calibration, and clinical utility. CONCLUSION: The developed nomograms provide a precise and personalized predictive tool for risk management of patients with nonmetastatic limb melanoma.


Sujet(s)
Membres , Mélanome , Nomogrammes , Programme SEER , Tumeurs cutanées , Humains , Mélanome/mortalité , Mélanome/anatomopathologie , Mélanome/diagnostic , Mélanome/thérapie , Femelle , Mâle , Tumeurs cutanées/mortalité , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/diagnostic , Tumeurs cutanées/thérapie , Programme SEER/statistiques et données numériques , Adulte d'âge moyen , Sujet âgé , Membres/anatomopathologie , Pronostic , Adulte , Courbe ROC , Melanoma, Cutaneous Malignant , Estimation de Kaplan-Meier , Stadification tumorale , Sujet âgé de 80 ans ou plus , Études rétrospectives
9.
Front Immunol ; 15: 1328368, 2024.
Article de Anglais | MEDLINE | ID: mdl-39386211

RÉSUMÉ

Introduction: Point-of-care (POC) manufacturing of chimeric antigen receptor (CAR) modified T cell has expanded rapidly over the last decade. In addition to the use of CD19 CAR T cells for hematological diseases, there is a growing interest in targeting a variety of tumor-associated epitopes. Methods: Here, we report the manufacturing and characterization of autologous anti-CD20 CAR T cells from melanoma patients within phase I clinical trial (NCT03893019). Using a second-generation lentiviral vector for the production of the CD20 CAR T cells on the CliniMACS Prodigy®. Results: We demonstrated consistency in cell composition and functionality of the products manufactured at two different production sites. The T cell purity was >98.5%, a CD4/CD8 ratio between 2.5 and 5.5 and transduction rate between 34% and 61% on day 12 (harvest). Median expansion rate was 53-fold (range, 42-65-fold) with 1.7-3.8×109 CAR T cells at harvest, a sufficient number for the planned dose escalation steps (1×105/kg, 1×106/kg, 1×107/kg BW). Complementary research of some of the products pointed out that the CAR+ cells expressed mainly central memory T-cell phenotype. All tested CAR T cell products were capable to translate into T cell activation upon engagement of CAR target cells, indicated by the increase in pro-inflammatory cytokine release and by the increase in CAR T cell amplification. Notably, there were some interindividual, cell-intrinsic differences at the level of cytokine release and amplification. CAR-mediated T cell activation depended on the level of CAR cognate antigen. Discussion: In conclusion, the CliniMACS Prodigy® platform is well suited for decentralized POC manufacturing of anti-CD20 CAR T cells and may be likewise applicable for the rapid and automated manufacturing of CAR T cells directed against other targets. Clinical trial registration: https://clinicaltrials.gov/study/NCT03893019?cond=Melanoma&term=NCT03893019&rank=1, identifier NCT03893019.


Sujet(s)
Antigènes CD20 , Immunothérapie adoptive , Mélanome , Récepteurs chimériques pour l'antigène , Humains , Mélanome/thérapie , Mélanome/immunologie , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Antigènes CD20/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Stadification tumorale , Mâle
10.
J Exp Clin Cancer Res ; 43(1): 276, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39354629

RÉSUMÉ

BACKGROUND: In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms. METHODS: Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo. RESULTS: Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI. CONCLUSIONS: Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.


Sujet(s)
Immunothérapie , Facteur-2 de régulation d'interféron , Mélanome , Animaux , Humains , Souris , Facteur-2 de régulation d'interféron/génétique , Facteur-2 de régulation d'interféron/métabolisme , Mélanome/génétique , Mélanome/immunologie , Mélanome/traitement médicamenteux , Mélanome/thérapie , Immunothérapie/méthodes , Mélanome expérimental/immunologie , Mélanome expérimental/génétique , Mélanome expérimental/thérapie , Lignée cellulaire tumorale
11.
Nat Commun ; 15(1): 8354, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39333093

RÉSUMÉ

T cell inhibitory mechanisms prevent autoimmune reactions, while cancer immunotherapy aims to remove these inhibitory signals. Chronic ultraviolet (UV) exposure attenuates autoimmunity through promotion of poorly understood immune-suppressive mechanisms. Here we show that mice with subcutaneous melanoma are not responsive to anti-PD1 immunotherapy following chronic UV irradiation, given prior to tumor injection, due to the suppression of T cell killing ability in skin-draining lymph nodes. Using mass cytometry and single-cell RNA-sequencing analyzes, we discover that skin-specific, UV-induced suppression of T-cells killing activity is mediated by upregulation of a Ly6ahigh T-cell subpopulation. Independently of the UV effect, Ly6ahigh T cells are induced by chronic type-1 interferon in the tumor microenvironment. Treatment with an anti-Ly6a antibody enhances the anti-tumoral cytotoxic activity of T cells and reprograms their mitochondrial metabolism via the Erk/cMyc axis. Treatment with an anti-Ly6a antibody inhibits tumor growth in mice resistant to anti-PD1 therapy. Applying our findings in humans could lead to an immunotherapy treatment for patients with resistance to existing treatments.


Sujet(s)
Antigènes Ly , Lymphocytes T CD8+ , Immunothérapie , Microenvironnement tumoral , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Antigènes Ly/métabolisme , Antigènes Ly/immunologie , Souris , Immunothérapie/méthodes , Microenvironnement tumoral/immunologie , Souris de lignée C57BL , Lignée cellulaire tumorale , Humains , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Mélanome expérimental/anatomopathologie , Femelle , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Tumeurs cutanées/immunologie , Tumeurs cutanées/thérapie , Tumeurs cutanées/anatomopathologie , Mitochondries/métabolisme , Mélanome/immunologie , Mélanome/thérapie , Interféron de type I/métabolisme
12.
Int J Mol Sci ; 25(18)2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39337333

RÉSUMÉ

Malignant melanoma outcomes have drastically changed in recent years due to the introduction of immune checkpoint inhibitors (ICIs). However, many patients still experience intolerable side effects, therapy resistance, and disease progression on ICI therapy. Therefore, there remains a need for novel therapeutics that address this gap in treatment options. Cell-based therapies have gained wide attention as a therapeutic option that could address this gap in treatment options for advanced melanoma. These therapies work by extracting certain cell types produced in the human body such as T-cells, modifying them based on a specific target, and transfusing them back into the patient. In the realm of cancer therapy, cell-based therapies utilize immune cells to target tumor cells while sparing healthy cells. Recently, the Food and Drug Administration (FDA) has approved the usage of lifileucel, a tumor-infiltrating lymphocyte (TIL) therapy, in advanced melanoma. This came following recent results from the C-144-01 study (NCT02360579), which demonstrated the efficacy and safety of TILs in metastatic melanoma patients who otherwise failed on standard ICI/targeted therapy. Thus, the results of this trial as well as the recent FDA approval have proven the viability of utilizing cell-based therapies to fill the gap in treatment options for patients with advanced melanoma. This review aims to provide a comprehensive overview of major cell-based therapies that have been utilized in melanoma by delineating results of the most recent multi-center phase II/ III clinical trials that evaluate the efficacy and safety of major cell-based therapies in melanoma. Additionally, we provide a summary of current limitations in each cell-based therapeutic option as well as a future direction of how to further extrapolate these cell-based therapies in advanced melanoma.


Sujet(s)
Mélanome , Humains , Thérapie cellulaire et tissulaire/méthodes , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Mélanome/thérapie , Mélanome/immunologie , Mélanome/anatomopathologie , Essais cliniques comme sujet
14.
Hum Vaccin Immunother ; 20(1): 2394252, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39286868

RÉSUMÉ

Melanoma is a highly malignant tumor of melanocyte origin that is prone to early metastasis and has a very poor prognosis. Early melanoma treatment modalities are mainly surgical, and treatment strategies for advanced or metastatic melanoma contain chemotherapy, radiotherapy, targeted therapy and immunotherapy. The efficacy of chemotherapy and radiotherapy has been unsatisfactory due to low sensitivity and strong toxic side effects. And targeted therapy is prone to drug resistance, so its clinical application is limited. Melanoma has always been the leader of immunotherapy for solid tumors, and how to maximize the role of immunotherapy and how to implement immunotherapy more accurately are still urgent to be explored. This review summarizes the common immunotherapies and applications for melanoma, illustrates the current research status of melanoma immunotherapy delivery systems, and discusses the advantages and disadvantages of each delivery system and its prospects for clinical application.


Sujet(s)
Immunothérapie , Mélanome , Humains , Mélanome/thérapie , Mélanome/immunologie , Immunothérapie/méthodes , Systèmes de délivrance de médicaments/méthodes
15.
Cancer Cell ; 42(9): 1491-1493, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39255774

RÉSUMÉ

Combination immunotherapy improves outcomes in metastatic melanoma, but the underlying mechanisms remain unclear. In this issue of Cancer Cell, Wang et al.1 report dynamics and transcriptional states of CD8+ T cell clones over time in patients treated with anti-PD-1, anti-CTLA-4, or a combination of the two. These findings have important implications for understanding and monitoring combination immunotherapy.


Sujet(s)
Lymphocytes T CD8+ , Immunothérapie , Mélanome , Humains , Immunothérapie/méthodes , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Mélanome/immunologie , Mélanome/thérapie , Mélanome/traitement médicamenteux , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie
16.
Int J Nanomedicine ; 19: 9071-9090, 2024.
Article de Anglais | MEDLINE | ID: mdl-39253059

RÉSUMÉ

Purpose: Our study seeks to develop dual-modal organic-nanoagents for cancer therapy and real-time fluorescence imaging, followed by their pre-clinical evaluation on a murine model. Integrating NIR molecular imaging with nanotechnology, our aim is to improve outcomes for early-stage cutaneous melanoma by offering more effective and less invasive methods. This approach has the potential to enhance both photothermal therapy (PTT) and Sentinel Lymph Node Biopsy (SLNB) procedures for melanoma patients. Methods: NIR-797-isothiocyanate was encapsulated in poly(D,L-lactide-co-glycolide) acid (PLGA) nanoparticles (NPs) using a two-step protocol, followed by thorough characterization, including assessing loading efficiency, fluorescence stability, and photothermal conversion. Biocompatibility and cellular uptake were tested in vitro on melanoma cells, while PTT assay, with real-time thermal monitoring, was performed in vivo on tumor-bearing mice under irradiation with an 808 nm laser. Finally, ex vivo fluorescence microscopy, histopathological assay, and TEM imaging were performed. Results: Our PLGA NPs, with a diameter of 270 nm, negative charge, and 60% NIR-797 loading efficiency, demonstrated excellent stability and fluorescence properties, as well as efficient light-to-heat conversion. In vitro studies confirmed their biocompatibility and cellular internalization. In vivo experiments demonstrated their efficacy as photothermal agents, inducing mild hyperthermia with temperatures reaching up to 43.8 °C. Ex vivo microscopy of tumor tissue confirmed persistent NIR fluorescence and uniform distribution of the NPs. Histopathological and TEM assays revealed early apoptosis, immune cell response, ultrastructural damage, and intracellular material debris resulting from combined NP treatment and irradiation. Additionally, TEM analyses of irradiated zone margins showed attenuated cellular damage, highlighting the precision and effectiveness of our targeted treatment approach. Conclusion: Specifically tailored for dual-modal NIR functionality, our NPs offer a novel approach in cancer PTT and real-time fluorescence monitoring, signaling a promising avenue toward clinical translation.


Sujet(s)
Hyperthermie provoquée , Nanoparticules , Imagerie optique , Copolymère d'acide poly(lactique-co-glycolique) , Animaux , Nanoparticules/composition chimique , Souris , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Lignée cellulaire tumorale , Hyperthermie provoquée/méthodes , Humains , Thérapie photothermique/méthodes , Tumeurs cutanées/thérapie , Tumeurs cutanées/imagerie diagnostique , Tumeurs cutanées/anatomopathologie , Mélanome/thérapie , Mélanome/imagerie diagnostique , Photothérapie/méthodes
17.
Surg Oncol ; 56: 102127, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39236515

RÉSUMÉ

Modern systemic therapy has dramatically improved outcomes for many patients with advanced metastatic melanoma. The success of these therapies has attracted much scientific interest while these therapies have made their way into the treatment of earlier stages of disease. Randomized trials have led to the approval of adjuvant immunotherapy and targeted therapy for resected stage III melanoma. However, most recently, these therapies have gained traction in the neoadjuvant setting. Promising early results led to randomized controlled trials that have now established neoadjuvant therapy as standard of care in advanced melanoma patients. Questions remain regarding the optimal choice of therapy, duration and timing of neoadjuvant therapy, extent of surgery, and the need for additional adjuvant therapy for patients who received neoadjuvant therapy. Herein we provide an overview of neoadjuvant therapy for melanoma and dilemmas to its broader applications.


Sujet(s)
Mélanome , Traitement néoadjuvant , Humains , Mélanome/thérapie , Mélanome/anatomopathologie , Traitement néoadjuvant/méthodes , Tumeurs cutanées/thérapie , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/chirurgie , Immunothérapie/méthodes
18.
J Exp Clin Cancer Res ; 43(1): 263, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39272209

RÉSUMÉ

BACKGROUND: Macrophage-based cell therapies have shown modest success in clinical trials, which can be attributed to their phenotypic plasticity, where transplanted macrophages get reprogrammed towards a pro-tumor phenotype. In most tumor types, including melanoma, the balance between antitumor M1-like and tumor-promoting M2-like macrophages is critical in defining the local immune response with a higher M1/M2 ratio favoring antitumor immunity. Therefore, designing novel strategies to increase the M1/M2 ratio in the TME has high clinical significance and benefits macrophage-based cell therapies. METHODS: In this study, we reprogrammed antitumor and proinflammatory macrophages ex-vivo with HDAC6 inhibitors (HDAC6i). We administered the reprogrammed macrophages intratumorally as an adoptive cell therapy (ACT) in the syngeneic SM1 murine melanoma model and patient-derived xenograft bearing NSG-SGM3 humanized mouse models. We phenotyped the tumor-infiltrated immune cells by flow cytometry and histological analysis of tumor sections for macrophage markers. We performed bulk RNA-seq profiling of murine bone marrow-derived macrophages treated with vehicle or HDAC6i and single-cell RNA-seq profiling of SM1 tumor-infiltrated immune cells to determine the effect of intratumor macrophage ACT on the tumor microenvironment (TME). We further analyzed the single-cell data to identify key cell-cell interactions and trajectory analysis to determine the fate of tumor-associated macrophages post-ACT. RESULTS: Macrophage ACT resulted in diminished tumor growth in both mouse models. We also demonstrated that HDAC6 inhibition in macrophages suppressed the polarization toward tumor-promoting phenotype by attenuating STAT3-mediated M2 reprogramming. Two weeks post-transplantation, ACT macrophages were viable, and inhibition of HDAC6 rendered intratumor transplanted M1 macrophages resistant to repolarization towards protumor M2 phenotype in-vivo. Further characterization of tumors by flow cytometry, single-cell transcriptomics, and single-cell secretome analyses revealed a significant enrichment of antitumor M1-like macrophages, resulting in increased M1/M2 ratio and infiltration of CD8 effector T-cells. Computational analysis of single-cell RNA-seq data for cell-cell interactions and trajectory analyses indicated activation of monocytes and T-cells in the TME. CONCLUSIONS: In summary, for the first time, we demonstrated the potential of reprogramming macrophages ex-vivo with HDAC6 inhibitors as a viable macrophage cell therapy to treat solid tumors.


Sujet(s)
Macrophages , Mélanome , Animaux , Souris , Humains , Macrophages/immunologie , Macrophages/métabolisme , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome/thérapie , Thérapie cellulaire et tissulaire/méthodes , Lignée cellulaire tumorale , Microenvironnement tumoral , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Reprogrammation cellulaire , Modèles animaux de maladie humaine
19.
Cancer Res ; 84(17): 2792-2805, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39228255

RÉSUMÉ

Neoantigen-based immunotherapy is an attractive potential treatment for previously intractable tumors. To effectively broaden the application of this approach, stringent biomarkers are crucial to identify responsive patients. ARID1A, a frequently mutated subunit of SWI/SNF chromatin remodeling complex, has been reported to determine tumor immunogenicity in some cohorts; however, mutations and deletions of ARID1A are not always linked to clinical responses to immunotherapy. In this study, we investigated immunotherapeutic responses based on ARID1A status in targeted therapy-resistant cancers. Mouse and human BRAFV600E melanomas with or without ARID1A expression were transformed into resistant to vemurafenib, an FDA-approved specific BRAFV600E inhibitor. Anti-PD-1 antibody treatment enhanced antitumor immune responses in vemurafenib-resistant ARID1A-deficient tumors but not in ARID1A-intact tumors or vemurafenib-sensitive ARID1A-deficient tumors. Neoantigens derived from accumulated somatic mutations during vemurafenib resistance were highly expressed in ARID1A-deficient tumors and promoted tumor immunogenicity. Furthermore, the newly generated neoantigens could be utilized as immunotherapeutic targets by vaccines. Finally, targeted therapy resistance-specific neoantigen in experimental human melanoma cells lacking ARID1A were validated to elicit T-cell receptor responses. Collectively, the classification of ARID1A-mutated tumors based on vemurafenib resistance as an additional indicator of immunotherapy response will enable a more accurate prediction to guide cancer treatment. Furthermore, the neoantigens that emerge with therapy resistance can be promising therapeutic targets for refractory tumors. Significance: Chemotherapy resistance promotes the acquisition of immunogenic neoantigens in ARID1A-deficient tumors that confer sensitivity to immune checkpoint blockade and can be utilized for developing antitumor vaccines, providing strategies to improve immunotherapy efficacy.


Sujet(s)
Antigènes néoplasiques , Protéines de liaison à l'ADN , Résistance aux médicaments antinéoplasiques , Mélanome , Facteurs de transcription , Vémurafénib , Animaux , Humains , Facteurs de transcription/génétique , Facteurs de transcription/immunologie , Souris , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/immunologie , Résistance aux médicaments antinéoplasiques/immunologie , Antigènes néoplasiques/immunologie , Antigènes néoplasiques/génétique , Vémurafénib/pharmacologie , Vémurafénib/usage thérapeutique , Mélanome/immunologie , Mélanome/traitement médicamenteux , Mélanome/génétique , Mélanome/thérapie , Immunothérapie/méthodes , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/immunologie , Lignée cellulaire tumorale , Femelle , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mutation , Thérapie moléculaire ciblée/méthodes , Souris de lignée C57BL
20.
Front Immunol ; 15: 1441410, 2024.
Article de Anglais | MEDLINE | ID: mdl-39234260

RÉSUMÉ

Mucosal melanoma (MM) poses a significant clinical challenge due to its aggressive nature and limited treatment options. In recent years, immunotherapy has emerged as a promising strategy for MM, with a particular focus on immune checkpoint inhibitors such as PD-1 and CTLA-4 inhibitors. These inhibitors have demonstrated substantial efficacy by harnessing the body's immune response against tumors. Moreover, adoptive cell transfer (ACT), anti-angiogenic therapy, and combination therapies have garnered attention for their potential in MM treatment. ACT involves modifying T cells to target melanoma cells, showing promising antitumor activity. Anti-angiogenic therapy aims to impede tumor growth by inhibiting angiogenesis, while combination therapies, including immune checkpoint inhibitors and targeted therapies, offer a multifaceted approach to overcome treatment resistance. This comprehensive review explores the advancements in immunotherapy for MM, highlighting the role of diverse therapeutic modalities in enhancing treatment outcomes and addressing the challenges posed by this aggressive malignancy.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Mélanome , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mélanome/thérapie , Mélanome/immunologie , Mélanome/traitement médicamenteux , Immunothérapie/méthodes , Animaux , Résultat thérapeutique , Muqueuse/immunologie , Association thérapeutique , Immunothérapie adoptive/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE