Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 684
Filtrer
1.
Sci Rep ; 14(1): 21751, 2024 09 18.
Article de Anglais | MEDLINE | ID: mdl-39294296

RÉSUMÉ

Gastric cancer (GC) is a prevalent malignancy with high mortality rates. Immunogenic cell death (ICD) is a unique form of programmed cell death that is closely linked to antitumor immunity and plays a critical role in modulating the tumor microenvironment (TME). Nevertheless, elucidating the precise effect of ICD on GC remains a challenging endeavour. ICD-related genes were identified in single-cell sequencing datasets and bulk transcriptome sequencing datasets via the AddModuleScore function, weighted gene co-expression network (WGCNA), and differential expression analysis. A robust signature associated with ICD was constructed using a machine learning computational framework incorporating 101 algorithms. Furthermore, multiomics analysis, including single-cell sequencing analysis, bulk transcriptomic analysis, and proteomics analysis, was conducted to verify the correlation of these hub genes with the immune microenvironment features of GC and with GC invasion and metastasis. We screened 59 genes associated with ICD and developed a robust ICD-related gene signature (ICDRS) via a machine learning computational framework that integrates 101 different algorithms. Furthermore, we identified five key hub genes (SMAP2, TNFAIP8, LBH, TXNIP, and PIK3IP1) from the ICDRS. Through single-cell analysis of GC tumor s, we confirmed the strong correlations of the hub genes with immune microenvironment features. Among these five genes, LBH exhibited the most significant associations with a poor prognosis and with the invasion and metastasis of GC. Finally, our findings were validated through immunohistochemical staining of a large clinical sample set, and the results further supported that LBH promotes GC cell invasion by activating the epithelial-mesenchymal transition (EMT) pathway.


Sujet(s)
Mort cellulaire immunogène , Apprentissage machine , Analyse sur cellule unique , Tumeurs de l'estomac , Microenvironnement tumoral , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/mortalité , Humains , Analyse sur cellule unique/méthodes , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Régulation de l'expression des gènes tumoraux , Analyse de profil d'expression de gènes , Protéomique/méthodes , Transcriptome , Biologie informatique/méthodes , Réseaux de régulation génique , Multi-omique
2.
Nat Commun ; 15(1): 8187, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39294133

RÉSUMÉ

Cancer is a significant cause of death around the world, and for many varieties, treatment is not successful. Therefore, there is a need for the development of innovative, efficacious, and precisely targeted treatments. Here, we develop a series of Au(I) complexes (1-4) through rational manipulation of ligand structures, thereby achieving tumor cell specific targeting and orchestrated tumor eradication via chemo-phototherapy and induced immunogenic cell death. A comprehensive exploration based on in vitro and in vivo female mice experimentation shows that complex 4 exhibits proficiency in specific tumor imaging, endoplasmic reticulum targeting, and has robust therapeutic capabilities. Mechanistic elucidation indicates that the anticancer effect derives from the synergistic actions of thioredoxin reductase inhibition, highly efficient reactive oxygen species production and immunogenic cell death. This work presents a report on a robust Au(I) complex integrating three therapeutic modalities within a singular system. The strategy presented in this work provides a valuable reference for the development of high-performance therapeutic agents.


Sujet(s)
Or , Mort cellulaire immunogène , Espèces réactives de l'oxygène , Animaux , Or/composition chimique , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Femelle , Souris , Humains , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Thioredoxin-disulfide reductase/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Tumeurs/thérapie , Tumeurs/immunologie , Photothérapie/méthodes , Complexes de coordination/composition chimique , Complexes de coordination/pharmacologie , Complexes de coordination/usage thérapeutique
3.
PLoS One ; 19(9): e0307024, 2024.
Article de Anglais | MEDLINE | ID: mdl-39231199

RÉSUMÉ

Radiotherapy is a commonly used method in the treatment of bladder cancers (BC). Radiation-induced immunogenic cell death (ICD) is related to the immune response against cancers and their prognoses. Even though dendritic cells (DC) act as powerful antigen-presenting cells in the body, their precise role in this ICD process remains unclear. Accordingly, an in vitro study was undertaken to ascertain whether high-dose radiation-induced ICD of BC cells could regulate the immune response of DC. The results indicated that high-dose radiation treatments of BC cells significantly increased their levels of apoptosis, blocked their cell cycle in the G2/M phase, increased their expression of ICD-related proteins, and upregulated their secretion of CCL5 and CCL21 which control the directed migration of DC. It was also noted that expression of CD80, CD86, CCR5, and CCR7 on DC was upregulated in the medium containing the irradiated cells. In conclusion, the present findings illustrate that high-dose radiation can induce the occurrence of ICD within BC cells, concomitantly resulting in the activation of DC. Such findings could be of great significance in increasing the understanding how radiotherapy of BC may work to bring about reductions in cell activity and how these processes in turn lead to immunoregulation of the function of DC.


Sujet(s)
Apoptose , Cellules dendritiques , Mort cellulaire immunogène , Tumeurs de la vessie urinaire , Cellules dendritiques/immunologie , Cellules dendritiques/effets des radiations , Tumeurs de la vessie urinaire/immunologie , Tumeurs de la vessie urinaire/radiothérapie , Tumeurs de la vessie urinaire/anatomopathologie , Humains , Lignée cellulaire tumorale , Apoptose/effets des radiations , Mort cellulaire immunogène/effets des radiations , Chimiokine CCL21/métabolisme , Récepteurs CCR7/métabolisme , Chimiokine CCL5/métabolisme , Récepteurs CCR5/métabolisme , Antigène CD86/métabolisme , Mouvement cellulaire/effets des radiations , Antigène CD80/métabolisme , Relation dose-effet des rayonnements
4.
Nat Commun ; 15(1): 7664, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39227567

RÉSUMÉ

The immunosuppressive tumor microenvironment (TME) remains a major obstacle to tumor control and causes suboptimal responses to immune checkpoint blockade (ICB) therapy. Thus, developing feasible therapeutic strategies that trigger inflammatory responses in the TME could improve the ICB efficacy. Mitochondria play an essential role in inflammation regulation and tumor immunogenicity induction. Herein, we report the discovery and characterization of a class of small molecules that can recapitulate aqueous self-assembly behavior, specifically target cellular organelles (e.g., mitochondria), and invigorate tumor cell immunogenicity. Mechanistically, this nanoassembly platform dynamically rewires mitochondria, induces endoplasmic reticulum stress, and causes apoptosis/paraptosis-associated immunogenic cell death. After treatment, stressed and dying tumor cells can act as prophylactic or therapeutic cancer vaccines. In preclinical mouse models of cancers with intrinsic or acquired resistance to PD-1 blockade, the local administration of nanoassemblies inflames the immunologically silent TME and synergizes with ICB therapy, generating potent antitumor immunity. This chemically programmed small-molecule immune enhancer acts distinctly from regular cytotoxic therapeutics and offers a promising strategy for synchronous and dynamic tailoring of innate immunity to achieve traceless cancer therapy and overcome immunosuppression in cancers.


Sujet(s)
Mitochondries , Tumeurs , Microenvironnement tumoral , Animaux , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Souris , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Humains , Lignée cellulaire tumorale , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/immunologie , Apoptose/effets des médicaments et des substances chimiques , Femelle , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Nanoparticules/composition chimique , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Immunothérapie/méthodes
5.
Theranostics ; 14(12): 4667-4682, 2024.
Article de Anglais | MEDLINE | ID: mdl-39239517

RÉSUMÉ

Background: Effective innate immunity activation could dramatically improve the anti-tumor efficacy and increase the beneficiary population of immunotherapy. However, the anti-tumor effect of unimodal immunotherapy is still not satisfactory. Methods: Herein, a novel relay-type innate immunity activation strategy based on photo-immunotherapy mediated by a water-soluble aggregation-induced emission luminogen, PEG420-TQ, with the assistant of toll-like receptor 7 (TLR-7) agonist, imiquimod (R837), was developed and constructed. Results: The strategy could promote tumor cells to undergo immunogenic cell death (ICD) induced by the well-designed PEG420-TQ@R837 (PTQ@R) nanoplatform under light irradiation, which in turn enhanced the infiltration of immune cells and the activation of innate immune cells to achieve the first innate immunity activation. The second innate immunity activation was subsequently achieved by drug delivery of R837 via apoptotic bodies (ApoBDs), further enhancing the anti-tumor activity of infiltrated immune cells. Conclusion: The strategy ultimately demonstrated robust innate immunity activation and achieved excellent performance against tumor growth and metastasis. The construction of the relay-type innate immunity activation strategy could provide a new idea for the application of immunotherapy in clinical trials.


Sujet(s)
Imiquimod , Immunité innée , Immunothérapie , Immunité innée/effets des médicaments et des substances chimiques , Animaux , Immunothérapie/méthodes , Souris , Imiquimod/usage thérapeutique , Imiquimod/pharmacologie , Lignée cellulaire tumorale , Humains , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Eau/composition chimique , Récepteur de type Toll-7/agonistes , Femelle , Photothérapie/méthodes , Nanoparticules/composition chimique , Souris de lignée BALB C , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Rayons infrarouges
6.
J Med Chem ; 67(17): 15098-15117, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39145486

RÉSUMÉ

Ferroptosis is a unique type of cell death, characterized by its reliance on iron dependency and lipid peroxidation (LPO). Consequently, small-molecule ferroptosis modulators have garnered substantial interest as a promising avenue for cancer therapy. Herein, we explored the ferroptosis sensitivity of epigenetic modulators and found that the antiproliferative effects of class I histone deacetylase (HDAC) inhibitors are significantly reliant on ferroptosis. Subsequently, we developed a novel series of HDAC inhibitors, identifying HL-5s with robust inhibitory activity against class I HDACs, particularly HDAC1. Notably, HL-5s induces ferroptosis by augmenting LPO production. Mechanistically, HL-5s increased the YB-1 acetylation and inhibited the Nrf2/HO-1 signaling pathway. Furthermore, HL-5s not only significantly suppresses tumor growth in the PC-9 xenograft model but also remodels the tumor microenvironment in the LLC allograft model. Our study has unveiled that class I HDAC inhibitors can exert antitumor effects by triggering ferroptosis, and HL-5s may serve as a promising candidate for future cancer treatment.


Sujet(s)
Antinéoplasiques , Benzimidazoles , Ferroptose , Inhibiteurs de désacétylase d'histone , Mort cellulaire immunogène , Ferroptose/effets des médicaments et des substances chimiques , Humains , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/synthèse chimique , Animaux , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Benzimidazoles/pharmacologie , Benzimidazoles/composition chimique , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Histone deacetylases/métabolisme , Lignée cellulaire tumorale , Relation structure-activité , Découverte de médicament , Prolifération cellulaire/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Chem Biol Interact ; 400: 111177, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39097071

RÉSUMÉ

Tartrolon D (TRL) is produced by Teredinibacter turnerae, a symbiotic cellulose-degrading bacteria in shipworm gills. Immunogenic cell death (ICD) induction contributes to a better and longer-lasting response to anticancer treatment. Tumor cells undergoing ICD trigger activation of the immune system, as a vaccine. AIMS: This study aimed to evaluate ICD induction by TRL. MAIN METHODS: Cell viability was evaluated by SRB assay. Cell stress, cell death, ICD features and antigen-presenting molecules were evaluated by flow cytometry and immunoblot. KEY FINDINGS: TRL showed antiproliferative activity on 7 tumor cell lines (L929, HCT 116, B16-F10, WM293A, SK-MEL-28, PC-3M, and MCF-7) and a non-tumor cell (HEK293A), with an inhibition concentration mean (IC50) ranging from 0.03 µM to 13 µM. Metastatic melanomas, SK-MEL-28, B16-F10, and WM293A, were more sensitive cell lines, with IC50 ranging from 0.07 to 1.2 µM. TRL induced apoptosis along with autophagy and endoplasmic reticulum stress and release of typical damage-associated molecular patterns (DAMPs) of ICD such calreticulin, ERp57, and HSP70 exposure, and HMGB1 release. Additionally, melanoma B16-F10 exposed to TRL increased expression of antigen-presenting molecules MHC II and CD1d and induced activation of splenocytes of C57BL/6 mice. SIGNIFICANCE: In spite of recent advances provided by target therapy and immunotherapy, advanced metastatic melanoma is incurable for more than half of patients. ICD inducers yield better and long-lasting responses to anticancer treatment. Our findings shed light on an anticancer candidate of marine origin that induces ICD in melanoma.


Sujet(s)
Mort cellulaire immunogène , Mélanome , Humains , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome/traitement médicamenteux , Animaux , Apoptose/effets des médicaments et des substances chimiques , Souris , Autophagie/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/immunologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules HEK293 , Calréticuline/métabolisme
8.
ACS Appl Mater Interfaces ; 16(33): 43257-43271, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39119624

RÉSUMÉ

Prostate cancer presents as a challenging disease, as it is often characterized as an immunologically "cold" tumor, leading to suboptimal outcomes with current immunotherapeutic approaches in clinical settings. Photodynamic therapy (PDT) harnesses reactive oxygen species generated by photosensitizers (PSs) to disrupt the intracellular redox equilibrium. This process induces DNA damage in both the mitochondria and nucleus, activating the process of immunogenic cell death (ICD) and the cGAS-STING pathway. Ultimately, this cascade of events leads to the initiation of antitumor immune responses. Nevertheless, existing PSs face challenges, including suboptimal tumor targeting, aggregation-induced quenching, and insufficient oxygen levels in the tumor regions. To this end, a versatile bionic nanoplatform has been designed for the simultaneous delivery of the aggregation-induced emission PS TPAQ-Py-PF6 and paclitaxel (PTX). The cell membrane camouflage of the nanoplatform leads to its remarkable abilities in tumor targeting and cellular internalization. Upon laser irradiation, the utilization of TPAQ-Py-PF6 in conjunction with PTX showcases a notable and enhanced synergistic antitumor impact. Additionally, the nanoplatform has the capability of initiating the cGAS-STING pathway, leading to the generation of cytokines. The presence of damage-associated molecular patterns induced by ICD collaborates with these aforementioned cytokines lead to the recruitment and facilitation of dendritic cell maturation. Consequently, this elicits a systemic immune response against tumors. In summary, this promising strategy highlights the use of a multifunctional biomimetic nanoplatform, combining chemotherapy, PDT, and immunotherapy to enhance the effectiveness of antitumor treatment.


Sujet(s)
Mort cellulaire immunogène , Immunothérapie , Protéines membranaires , Nucleotidyltransferases , Photothérapie dynamique , Photosensibilisants , Tumeurs de la prostate , Humains , Nucleotidyltransferases/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/thérapie , Tumeurs de la prostate/anatomopathologie , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Mort cellulaire immunogène/effets des radiations , Protéines membranaires/métabolisme , Mâle , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/usage thérapeutique , Animaux , Souris , Paclitaxel/composition chimique , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments , Porphyrines/composition chimique , Porphyrines/pharmacologie
9.
Nat Commun ; 15(1): 7037, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39147750

RÉSUMÉ

The quest for targeted therapies is critical in the battle against cancer. The RAS/MAP kinase pathway is frequently implicated in neoplasia, with ERK playing a crucial role as the most distal kinase in the RAS signaling cascade. Our previous research demonstrated that the interaction between ERK and MYD88, an adaptor protein in innate immunity, is crucial for RAS-dependent transformation and cancer cell survival. In this study, we examine the biological consequences of disrupting the ERK-MYD88 interaction through the ERK D-recruitment site (DRS), while preserving ERK's kinase activity. Our results indicate that EI-52, a small-molecule benzimidazole targeting ERK-MYD88 interaction induces an HRI-mediated integrated stress response (ISR), resulting in immunogenic apoptosis specific to cancer cells. Additionally, EI-52 exhibits anti-tumor efficacy in patient-derived tumors and induces an anti-tumor T cell response in mice in vivo. These findings suggest that inhibiting the ERK-MYD88 interaction may be a promising therapeutic approach in cancer treatment.


Sujet(s)
Benzimidazoles , Extracellular Signal-Regulated MAP Kinases , Facteur de différenciation myéloïde-88 , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Humains , Animaux , Souris , Extracellular Signal-Regulated MAP Kinases/métabolisme , Lignée cellulaire tumorale , Benzimidazoles/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Femelle , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
10.
Nano Lett ; 24(34): 10664-10673, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39140448

RÉSUMÉ

Here we report a brand-new bioactive polymer featuring sulfonium moieties that exhibits the capability of inducing immunogenic cell death (ICD) for anticancer therapy. The optimized polysulfonium presents a wide spectrum of potent anticancer activity and remarkable selectivity. In-depth mechanistic studies reveal that the polymer exerts its cytotoxic effects on cancer cells through a membrane-disrupting mechanism. This further initiates the release of a plethora of damage-associated molecular patterns, effectively triggering ICD and resulting in systemic anticancer immune responses. Notably, the compound demonstrated significant efficacy in suppressing tumor growth in the B16-F10 melanoma tumor model. Furthermore, it exhibits robust immune memory effects, effectively suppressing tumor recurrence and metastasis in both the rechallenge model and the lung metastatic tumor model. To the best of our knowledge, the study represents the pioneering exportation of cationic polysulfoniums, showcasing not only their remarkable safety and efficacy against primary tumors but also their unique ability in activating long-term immune memory.


Sujet(s)
Antinéoplasiques , Mort cellulaire immunogène , Polymères , Animaux , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Souris , Humains , Lignée cellulaire tumorale , Polymères/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Composés de sulfonium/composition chimique , Composés de sulfonium/pharmacologie , Composés de sulfonium/usage thérapeutique , Mélanome expérimental/immunologie , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/anatomopathologie
11.
Front Immunol ; 15: 1447817, 2024.
Article de Anglais | MEDLINE | ID: mdl-39185425

RÉSUMÉ

The field of oncology has transformed in recent years, with treatments shifting from traditional surgical resection and radiation therapy to more diverse and customized approaches, one of which is immunotherapy. ICD (immunogenic cell death) belongs to a class of regulatory cell death modalities that reactivate the immune response by facilitating the interaction between apoptotic cells and immune cells and releasing specific signaling molecules, and DAMPs (damage-associated molecular patterns). The inducers of ICD can elevate the expression of specific proteins to optimize the TME (tumor microenvironment). The use of nanotechnology has shown its unique potential. Nanomaterials, due to their tunability, targeting, and biocompatibility, have become powerful tools for drug delivery, immunomodulators, etc., and have shown significant efficacy in clinical trials. In particular, these nanomaterials can effectively activate the ICD, trigger a potent anti-tumor immune response, and maintain long-term tumor suppression. Different types of nanomaterials, such as biological cell membrane-modified nanoparticles, self-assembled nanostructures, metallic nanoparticles, mesoporous materials, and hydrogels, play their respective roles in ICD induction due to their unique structures and mechanisms of action. Therefore, this review will explore the latest advances in the application of these common nanomaterials in tumor ICD induction and discuss how they can provide new strategies and tools for cancer therapy. By gaining a deeper understanding of the mechanism of action of these nanomaterials, researchers can develop more precise and effective therapeutic approaches to improve the prognosis and quality of life of cancer patients. Moreover, these strategies hold the promise to overcome resistance to conventional therapies, minimize side effects, and lead to more personalized treatment regimens, ultimately benefiting cancer treatment.


Sujet(s)
Mort cellulaire immunogène , Immunothérapie , Nanostructures , Tumeurs , Microenvironnement tumoral , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Nanostructures/usage thérapeutique , Nanostructures/composition chimique , Animaux , Immunothérapie/méthodes , Microenvironnement tumoral/immunologie
12.
J Immunother Cancer ; 12(7)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39089738

RÉSUMÉ

BACKGROUND: Enhanced glucose metabolism has been reported in many cancers. Glucose-6-phosphate dehydrogenase (G6PD) is a rate-limiting enzyme involved in the pentose phosphate pathway, which maintains NADPH levels and protects cells from oxidative damage. We recently found that low G6PD expression correlates with active tumor immunity. However, the mechanism involving G6PD and tumor immunity remained unclear. METHODS: We conducted in vitro studies using G6PD-knocked down malignant melanoma cells, pathway analysis using the GEO dataset, in vivo studies in combination with immune checkpoint inhibitors (ICIs) using a mouse melanoma model, and prognostic analysis in 42 melanoma patients and 30 lung cancer patients who were treated with ICIs. RESULTS: Inhibition of G6PD, both chemically and genetically, has been shown to decrease the production of NADPH and reduce their oxidative stress tolerance. This leads to cell death, which is accompanied by the release of high mobility group box 1 and the translocation of calreticulin to the plasma membrane. These findings suggested that inhibiting G6PD can induce immunogenic cell death. In experiments with C57BL/6 mice transplanted with G6PD-knockdown B16 melanoma cells and treated with anti-PD-L1 antibody, a significant reduction in tumor size was observed. Interestingly, inhibiting G6PD in only a part of the lesions increased the sensitivity of other lesions to ICI. Additionally, out of 42 melanoma patients and 30 lung cancer patients treated with ICIs, those with low G6PD expression had a better prognosis than those with high G6PD expression (p=0.0473; melanoma, p=0.0287; lung cancer). CONCLUSION: G6PD inhibition is a potent therapeutic strategy that triggers immunogenic cell death in tumors, significantly augmenting the efficacy of immunotherapies.


Sujet(s)
Glucose 6-phosphate dehydrogenase , Mort cellulaire immunogène , Immunothérapie , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/antagonistes et inhibiteurs , Animaux , Humains , Souris , Immunothérapie/méthodes , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/immunologie , Mélanome expérimental/métabolisme , Femelle , Souris de lignée C57BL , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Lignée cellulaire tumorale , Mâle , Mélanome/traitement médicamenteux , Mélanome/immunologie , Mélanome/anatomopathologie
13.
Expert Rev Vaccines ; 23(1): 830-844, 2024.
Article de Anglais | MEDLINE | ID: mdl-39193620

RÉSUMÉ

BACKGROUND: In this study, effective antigens of mRNA vaccine were excavated from the perspective of ICD, and ICD subtypes of PRAD were further distinguished to establish an ICD landscape, thereby determining suitable vaccine recipients. RESEARCH DESIGN AND METHODS: TCGA and MSKCC databases were applied to acquire RNA-seq data and corresponding clinical data of 554 and 131 patients, respectively. GEPIA was employed to measure prognostic indices. Then, a comparison of genetic alterations was performed utilizing cBioPortal, and correlation of identified ICD antigens with immune infiltrating cells was analyzed employing TIMER. Moreover, ICD subtypes were identified by means of consensus cluster, and ICD landscape of PRAD was depicted utilizing graph learning-based dimensional reduction. RESULTS: In total, 4 PRAD antigens were identified in PRAD, including FUS, LMNB2, RNPC3, and ZNF700, which had association with adverse prognosis and infiltration of APCs. PRAD patients were classified as two ICD subtypes based on their differences in molecular, cellular, and clinical features. Furthermore, ICD modulators and immune checkpoints were also differentially expressed between two ICD subtype tumors. Finally, the ICD landscape of PRAD showed substantial heterogeneity among individual patients. CONCLUSIONS: In summary, the research may provide a theoretical foundation for developing mRNA vaccine against PRAD as well as determining appropriate vaccine recipients.


Sujet(s)
Adénocarcinome , Antigènes néoplasiques , Vaccins anticancéreux , Mort cellulaire immunogène , Tumeurs de la prostate , Vaccins à ARNm , Humains , Tumeurs de la prostate/immunologie , Mâle , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Antigènes néoplasiques/immunologie , Adénocarcinome/immunologie , Pronostic
14.
Eur J Pharmacol ; 981: 176913, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39154830

RÉSUMÉ

Tumor immunotherapy, especially immune checkpoint inhibitors (ICIs), has been applied in clinical practice, but low response to immune therapies remains a thorny issue. Oncolytic viruses (OVs) are considered promising for cancer treatment because they can selectively target and destroy tumor cells followed by spreading to nearby tumor tissues for a new round of infection. Immunogenic cell death (ICD), which is the major mechanism of OVs' anticancer effects, is induced by endoplasmic reticulum stress and reactive oxygen species overload after virus infection. Subsequent release of specific damage-associated molecular patterns (DAMPs) from different types of tumor cells can transform the tumor microenvironment from "cold" to "hot". In this paper, we broadly define ICD as those types of cell death that is immunogenic, and describe their signaling pathways respectively. Focusing on ICD, we also elucidate the advantages and disadvantages of recent combination therapies and their future prospects.


Sujet(s)
Mort cellulaire immunogène , Immunothérapie , Tumeurs , Thérapie virale de cancers , Virus oncolytiques , Thérapie virale de cancers/méthodes , Humains , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Animaux , Virus oncolytiques/physiologie , Virus oncolytiques/immunologie , Microenvironnement tumoral/immunologie , Stress du réticulum endoplasmique/immunologie , Transduction du signal
15.
Adv Mater ; 36(36): e2405673, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39022876

RÉSUMÉ

Immunogenic cell death (ICD) often results in the production and accumulation of adenosine (ADO), a byproduct that negatively impacts the therapeutic effect as well as facilitates tumor development and metastasis. Here, an innovative strategy is elaborately developed to effectively activate ICD while avoiding the generation of immunosuppressive adenosine. Specifically, ZIF-90, an ATP-responsive consumer, is synthesized as the core carrier to encapsulate AB680 (CD73 inhibitor) and then coated with an iron-polyphenol layer to prepare the ICD inducer (AZTF), which is further grafted onto prebiotic bacteria via the esterification reaction to obtain the engineered biohybrid (Bc@AZTF). Particularly, the designed Bc@AZTF can actively enrich in tumor sites and respond to the acidic tumor microenvironment to offload AZTF nanoparticles, which can consume intracellular ATP (iATP) content and simultaneously inhibit the ATP-adenosine axis to reduce the accumulation of adenosine, thereby alleviating adenosine-mediated immunosuppression and strikingly amplifying ICD effect. Importantly, the synergy of anti-PD-1 (αPD-1) with Bc@AZTF not only establishes a collaborative antitumor immune network to potentiate effective tumoricidal immunity but also activates long-lasting immune memory effects to manage tumor recurrence and rechallenge, presenting a new paradigm for ICD treatment combined with adenosine metabolism.


Sujet(s)
Adénosine triphosphate , Adénosine , Mort cellulaire immunogène , Immunothérapie , Adénosine/composition chimique , Adénosine triphosphate/métabolisme , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Souris , Animaux , Lignée cellulaire tumorale , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/anatomopathologie , Nanoparticules/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
16.
Theranostics ; 14(10): 3777-3792, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994018

RÉSUMÉ

Rationale: Growing evidence has demonstrated that miRNA-21 (miR-21) upregulation is closely associated with tumor pathogenesis. However, the mechanisms by which miR-21 inhibition modulates the immunosuppressive tumor microenvironment (TME) and improves tumor sensitivity to immune checkpoint blockade therapies remain largely unexplored. In this study, we demonstrate the precise delivery of anti-miR-21 using a PD-L1-targeting peptide conjugate (P21) to the PD-L1high TME. Methods: Investigating miR-21 inhibition mechanisms involved conducting quantitative real-time PCR, western blot, flow cytometry, and confocal microscopy analyses. The antitumor efficacy and immune profile of P21 monotherapy, or combined with anti-PD-L1 immune checkpoint inhibitors, were assessed in mouse models bearing CT26.CL25 tumors and 4T1 breast cancer. Results Inhibition of oncogenic miR-21 in cancer cells by P21 efficiently activates tumor suppressor genes, inducing autophagy and endoplasmic reticulum stress. Subsequent cell-death-associated immune activation (immunogenic cell death) is initiated via the release of damage-associated molecular patterns. The in vivo results also illustrated that the immunogenic cell death triggered by P21 could effectively sensitize the immunosuppressive TME. That is, P21 enhances CD8+ T cell infiltration in tumor tissues by conferring immunogenicity to dying cancer cells and promoting dendritic cell maturation. Meanwhile, combining P21 with an anti-PD-L1 immune checkpoint inhibitor elicits a highly potent antitumor effect in a CT26.CL25 tumor-bearing mouse model and 4T1 metastatic tumor model. Conclusions: Collectively, we have clarified a miR-21-related immunogenic cell death mechanism through the precise delivery of anti-miR-21 to the PD-L1high TME. These findings highlight the potential of miR-21 as a target for immunotherapeutic interventions.


Sujet(s)
Antigène CD274 , Mort cellulaire immunogène , Immunothérapie , microARN , Microenvironnement tumoral , microARN/génétique , microARN/métabolisme , Animaux , Souris , Antigène CD274/métabolisme , Antigène CD274/antagonistes et inhibiteurs , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Immunothérapie/méthodes , Femelle , Souris de lignée BALB C , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Autophagie/effets des médicaments et des substances chimiques , Tumeurs du sein/immunologie , Tumeurs du sein/thérapie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique
17.
Biomed Pharmacother ; 177: 117099, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38981240

RÉSUMÉ

Immunogenic cell death (ICD) can activate adaptive immune response in the host with normal immune system. Some synthetic chemotherapeutic drugs and natural compounds have shown promising results in cancer treatment by triggering the release of damage-associated molecules (DAMPs) to trigger ICD. However, most chemotherapeutic drugs exhibit non-selective cytotoxicity and may also induce and promote metastasis, thereby significantly reducing their clinical efficacy. Among the natural compounds that can induce ICD, plant-derived compounds account for the largest proportion, which are of increasing value in the treatment of cancer. Understanding which plant-derived natural compounds can induce ICD and how they induce ICD is crucial for developing strategies to improve chemotherapy outcomes. In this review, we focus on the recent findings regarding plant-derived natural compounds that induce ICD according to the classification of flavonoids, alkaloids, glycosides, terpenoids and discuss the potential mechanisms including endoplasmic reticulum (ER) stress, DNA damage, apoptosis, necroptosis autophagy, ferroptosis. In addition, plant-derived natural compounds that can enhance the ICD induction ability of conventional therapies for cancer treatment is also elaborated. The rational use of plant-derived natural compounds to induce ICD is helpful for the development of new cancer treatment methods.


Sujet(s)
Mort cellulaire immunogène , Tumeurs , Humains , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/anatomopathologie , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Animaux , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Produits biologiques/pharmacologie , Produits biologiques/usage thérapeutique , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques
18.
Cell Commun Signal ; 22(1): 352, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970078

RÉSUMÉ

BACKGROUND: In triple-negative breast cancer (TNBC) therapy, insufficient tumor infiltration by lymphocytes significantly hinders the efficacy of immune checkpoint inhibitors. We have previously demonstrated that Hainanenin-1 (HN-1), a host defense peptide (HDP) identified from Hainan frog skin, induces breast cancer apoptosis and boots anti-tumor immunity via unknown mechanism. METHODS: We used in vitro experiments to observe immunogenic cell death (ICD) indicators in HN-1-treated TNBC cell lines, a mouse tumor model to verify HN-1 promotion of mice anti-tumor immune response, and an in vitro drug sensitivity test of patient-derived breast cancer cells to verify the inhibitory effect of HN-1. RESULTS: HN-1 induced ICD in TNBC in a process during which damage-associated molecular patterns (DAMPs) were released that could further increase the anti-tumor immune response. The secretion level of interleukin 2 (IL-2), IL-12, and interferon γ in the co-culture supernatant was increased, and dendritic cells (DCs) were activated via a co-culture with HN-1-pretreated TNBC cells. As a result, HN-1 increased the infiltration of anti-tumor immune cells (DCs and T lymphocytes) in the mouse model bearing both 4T1 and EMT6 tumors. Meanwhile, regulatory T cells and myeloid-derived suppressor cells were suppressed. In addition, HN-1 induced DNA damage, and double-strand DNA release in the cytosol was significantly enhanced, indicating that HN-1 might stimulate ICD via activation of STING pathway. The knockdown of STING inhibited HN-1-induced ICD. Of note, HN-1 exhibited inhibitory effects on patient-derived breast cancer cells under three-dimensional culture conditions. CONCLUSIONS: Collectively, our study demonstrated that HN-1 could be utilized as a potential compound that might augment immunotherapy effects in patients with TNBC.


Sujet(s)
Mort cellulaire immunogène , Protéines membranaires , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/immunologie , Animaux , Humains , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Femelle , Souris , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Lignée cellulaire tumorale , Souris de lignée BALB C , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/métabolisme
19.
Front Immunol ; 15: 1396349, 2024.
Article de Anglais | MEDLINE | ID: mdl-39011040

RÉSUMÉ

Introduction: Immunogenic cell death (ICD) has emerged as a novel option for cancer immunotherapy. The key determinants of ICD encompass antigenicity (the presence of antigens) and adjuvanticity, which involves the release of damage-associated molecular patterns (DAMPs) and various cytokines and chemokines. CX3CL1, also known as neurotactin or fractalkine, is a chemokine involved in cellular signalling and immune cell interactions. CX3CL1 has been denoted as a "find me" signal that stimulates chemotaxis of immune cells towards dying cells, facilitating efferocytosis and antigen presentation. However, in the context of ICD, it is uncertain whether CX3CL1 is an important mediator of the effects of ICD. Methods: In this study, we investigated the intricate role of CX3CL1 in immunogenic apoptosis induced by mitoxantrone (MTX) in cancer cells. The Luminex xMAP technology was used to quantify murine cytokines, chemokines and growth factors to identify pivotal regulatory cytokines released by murine fibrosarcoma MCA205 and melanoma B16-F10 cells undergoing ICD. Moreover, a murine tumour prophylactic vaccination model was employed to analyse the effect of CX3CL1 on the activation of an adaptive immune response against MCA205 cells undergoing ICD. Furthermore, thorough analysis of the TCGA-SKCM public dataset from 98 melanoma patients revealed the role of CX3CL1 and its receptor CX3CR1 in melanoma patients. Results: Our findings demonstrate enhanced CX3CL1 release from apoptotic MCA205 and B16-F10 cells (regardless of the cell type) but not if they are undergoing ferroptosis or accidental necrosis. Moreover, the addition of recombinant CX3CL1 to non-immunogenic doses of MTX-treated, apoptotically dying cancer cells in the murine prophylactic tumour vaccination model induced a robust immunogenic response, effectively increasing the survival of the mice. Furthermore, analysis of melanoma patient data revealed enhanced survival rates in individuals exhibiting elevated levels of CD8+ T cells expressing CX3CR1. Conclusion: These data collectively underscore the importance of the release of CX3CL1 in eliciting an immunogenic response against dying cancer cells and suggest that CX3CL1 may serve as a key switch in conferring immunogenicity to apoptosis.


Sujet(s)
Apoptose , Chimiokine CX3CL1 , Animaux , Chimiokine CX3CL1/métabolisme , Souris , Humains , Lignée cellulaire tumorale , Souris de lignée C57BL , Mélanome expérimental/immunologie , Femelle , Mort cellulaire immunogène , Cytokines/métabolisme
20.
J Nanobiotechnology ; 22(1): 408, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992664

RÉSUMÉ

BACKGROUND: Ovarian cancer (OC) has the highest fatality rate among all gynecological malignancies, necessitating the exploration of novel, efficient, and low-toxicity therapeutic strategies. Ferroptosis is a type of programmed cell death induced by iron-dependent lipid peroxidation and can potentially activate antitumor immunity. Developing highly effective ferroptosis inducers may improve OC prognosis. RESULTS: In this study, we developed an ultrasonically controllable two-dimensional (2D) piezoelectric nanoagonist (Bi2MoO6-MXene) to induce ferroptosis. A Schottky heterojunction between Bi2MoO6 (BMO) and MXene reduced the bandgap width by 0.44 eV, increased the carrier-separation efficiency, and decreased the recombination rate of electron-hole pairs under ultrasound stimulation. Therefore, the reactive oxygen species yield was enhanced. Under spatiotemporal ultrasound excitation, BMO-MXene effectively inhibited OC proliferation by more than 90%, induced lipid peroxidation, decreased mitochondrial-membrane potential, and inactivated the glutathione peroxidase and cystathionine transporter protein system, thereby causing ferroptosis in tumor cells. Ferroptosis in OC cells further activated immunogenic cell death, facilitating dendritic cell maturation and stimulating antitumor immunity. CONCLUSION: We have succeeded in developing a highly potent ferroptosis inducer (BMO-MXene), capable of inhibiting OC progression through the sonodynamic-ferroptosis-immunogenic cell death pathway.


Sujet(s)
Ferroptose , Mort cellulaire immunogène , Tumeurs de l'ovaire , Ferroptose/effets des médicaments et des substances chimiques , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Humains , Animaux , Lignée cellulaire tumorale , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Souris , Espèces réactives de l'oxygène/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Bismuth/pharmacologie , Bismuth/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE