Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.558
Filtrer
1.
Biomaterials ; 312: 122740, 2025 Jan.
Article de Anglais | MEDLINE | ID: mdl-39096839

RÉSUMÉ

Metastasis stands as the primary contributor to mortality associated with tumors. Chemotherapy and immunotherapy are frequently utilized in the management of metastatic solid tumors. Nevertheless, these therapeutic modalities are linked to serious adverse effects and limited effectiveness in preventing metastasis. Here, we report a novel therapeutic strategy named starvation-immunotherapy, wherein an immune checkpoint inhibitor is combined with an ultra-long-acting L-asparaginase that is a fusion protein comprising L-asparaginase (ASNase) and an elastin-like polypeptide (ELP), termed ASNase-ELP. ASNase-ELP's thermosensitivity enables it to generate an in-situ depot following an intratumoral injection, yielding increased dose tolerance, improved pharmacokinetics, sustained release, optimized biodistribution, and augmented tumor retention compared to free ASNase. As a result, in murine models of oral cancer, melanoma, and cervical cancer, the antitumor efficacy of ASNase-ELP by selectively and sustainably depleting L-asparagine essential for tumor cell survival was substantially superior to that of ASNase or Cisplatin, a first-line anti-solid tumor medicine, without any observable adverse effects. Furthermore, the combination of ASNase-ELP and an immune checkpoint inhibitor was more effective than either therapy alone in impeding melanoma metastasis. Overall, the synergistic strategy of starvation-immunotherapy holds excellent promise in reshaping the therapeutic landscape of refractory metastatic tumors and offering a new alternative for next-generation oncology treatments.


Sujet(s)
Asparaginase , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Animaux , Asparaginase/usage thérapeutique , Asparaginase/pharmacologie , Asparaginase/composition chimique , Immunothérapie/méthodes , Femelle , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Humains , Lignée cellulaire tumorale , Synergie des médicaments , Élastine/composition chimique , Élastine/métabolisme , Métastase tumorale , Souris de lignée C57BL , Souris de lignée BALB C , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Distribution tissulaire
2.
Biomaterials ; 312: 122723, 2025 Jan.
Article de Anglais | MEDLINE | ID: mdl-39121732

RÉSUMÉ

The challenges generated by insufficient T cell activation and infiltration have constrained the application of immunotherapy. Making matters worse, the complex tumor microenvironment (TME), resistance to apoptosis collectively poses obstacles for cancer treatment. The carrier-free small molecular self-assembly strategy is a current research hotspot to overcome these challenges. This strategy can transform multiple functional agents into sustain-released hydrogel without the addition of any excipients. Herein, a coordination and hydrogen bond mediated tricomponent hydrogel (Cel hydrogel) composed of glycyrrhizic acid (GA), copper ions (Cu2+) and celastrol (Cel) was initially constructed. The hydrogel can regulate TME by chemo-dynamic therapy (CDT), which increases reactive oxygen species (ROS) in conjunction with GA and Cel, synergistically expediting cellular apoptosis. What's more, copper induced cuproptosis also contributes to the anti-tumor effect. In terms of regulating immunity, ROS generated by Cel hydrogel can polarize tumor-associated macrophages (TAMs) into M1-TAMs, Cel can induce T cell proliferation as well as activate DC mediated antigen presentation, which subsequently induce T cell proliferation, elevate T cell infiltration and enhance the specific killing of tumor cells, along with the upregulation of PD-L1 expression. Upon co-administration with aPD-L1, this synergy mitigated both primary and metastasis tumors, showing promising clinical translational value.


Sujet(s)
Cuivre , Hydrogels , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Activation des lymphocytes , Triterpènes pentacycliques , Espèces réactives de l'oxygène , Lymphocytes T , Microenvironnement tumoral , Triterpènes pentacycliques/pharmacologie , Hydrogels/composition chimique , Animaux , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Immunothérapie/méthodes , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Activation des lymphocytes/effets des médicaments et des substances chimiques , Cuivre/composition chimique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Humains , Souris de lignée C57BL , Acide glycyrrhizique/pharmacologie , Acide glycyrrhizique/composition chimique , Femelle , Triterpènes/pharmacologie , Triterpènes/composition chimique
3.
Front Immunol ; 15: 1444424, 2024.
Article de Anglais | MEDLINE | ID: mdl-39234253

RÉSUMÉ

Lymphocyte activation gene (Lag)-3 is an inhibitory co-receptor and target of immune checkpoint inhibitor (ICI) therapy for cancer. The dynamic behavior of Lag-3 was analyzed at the immune synapse upon T-cell activation to elucidate the Lag-3 inhibitory mechanism. Lag-3 formed clusters and co-localized with T-cell receptor microcluster (TCR-MC) upon T-cell activation similar to PD-1. Lag-3 blocking antibodies (Abs) inhibited the co-localization between Lag-3 and TCR-MC without inhibiting Lag-3 cluster formation. Lag-3 also inhibited MHC-II-independent stimulation and Lag-3 Ab, which did not block MHC-II binding could still block Lag-3's inhibitory function, suggesting that the Lag-3 Ab blocks the Lag-3 inhibitory signal by dissociating the co-assembly of TCR-MC and Lag-3 clusters. Consistent with the combination benefit of PD-1 and Lag-3 Abs to augment T-cell responses, bispecific Lag-3/PD-1 antagonists effectively inhibited both cluster formation and co-localization of PD-1 and Lag-3 with TCR-MC. Therefore, Lag-3 inhibits T-cell activation at TCR-MC, and the target of Lag-3 ICI is to dissociate the co-localization of Lag-3 with TCR-MC.


Sujet(s)
Antigènes CD , Inhibiteurs de points de contrôle immunitaires , Protéine LAG-3 , Activation des lymphocytes , Récepteurs aux antigènes des cellules T , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Antigènes CD/immunologie , Antigènes CD/métabolisme , Humains , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Animaux
4.
Technol Cancer Res Treat ; 23: 15330338241275947, 2024.
Article de Anglais | MEDLINE | ID: mdl-39228166

RÉSUMÉ

The programmed cell death protein 1 (PD-1, CD279) is an important therapeutic target in many oncological diseases. This checkpoint protein inhibits T lymphocytes from attacking other cells in the body and thus blocking it improves the clearance of tumor cells by the immune system. While there are already multiple FDA-approved anti-PD-1 antibodies, including nivolumab (Opdivo® from Bristol-Myers Squibb) and pembrolizumab (Keytruda® from Merck), there are ongoing efforts to discover new and improved checkpoint inhibitor therapeutics. In this study, we present multiple anti-PD-1 antibody fragments that were derived computationally using protein diffusion and evaluated through our scalable, in silico pipeline. Here we present nine synthetic Fv structures that are suitable for further empirical testing of their anti-PD-1 activity due to desirable predicted binding performance.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Humains , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Intelligence artificielle , Découverte de médicament , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Liaison aux protéines
6.
Expert Rev Anticancer Ther ; 24(10): 931-941, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39233324

RÉSUMÉ

INTRODUCTION: Although meningiomas are the most common primary brain tumor, there are limited treatment options for recurrent or aggressive lesions. Compared to other brain tumors, meningiomas may be uniquely amenable to immunotherapy by virtue of their location outside the blood-brain barrier. AREAS COVERED: This review describes our current understanding of the immunology of the meninges, as well as immune cell infiltration and immune signaling in meningioma. Current literature on meningioma immunology and immunotherapy was comprehensively reviewed and summarized by a comprehensive search of MEDLINE (1/1/1990-6/1/2024). Further, we describe the current state of immunotherapeutic approaches, as well as potential future targets. Potential immunotherapeutic approaches include immune checkpoint inhibition, CAR-T approaches, tumor vaccine therapy, and immunogenic molecular markers. EXPERT OPINION: Meningioma immunotherapy is in early stages, as no immunotherapies are currently included in treatment guidelines. There is substantial heterogeneity in immune cell infiltration, immunogenicity, and immune escape across tumors, even within tumor grade. Furthering our understanding of meningioma immunology and tumor classification will allow for careful selection of tumors and patient populations that may benefit from primary or adjunctive immunotherapy for meningioma.


Sujet(s)
Vaccins anticancéreux , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Tumeurs des méninges , Méningiome , Méningiome/thérapie , Méningiome/immunologie , Méningiome/anatomopathologie , Humains , Tumeurs des méninges/thérapie , Tumeurs des méninges/immunologie , Tumeurs des méninges/anatomopathologie , Immunothérapie/méthodes , Vaccins anticancéreux/administration et posologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Marqueurs biologiques tumoraux , Animaux , Échappement de la tumeur à la surveillance immunitaire , Sélection de patients , Récidive tumorale locale
7.
Oncol Rep ; 52(5)2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39301655

RÉSUMÉ

Lung cancer is increasingly recognized as a leading cause of cancer­related mortality. Immunotherapy has emerged as a promising therapeutic approach for lung cancer, particularly non­small cell lung cancer (NSCLC). Nonetheless, the response rate to programmed cell death 1 (PD­1) inhibitors remains less than optimal. It has been suggested that protein tyrosine phosphatase 1B (PTP1B) plays a crucial role in the development and progression of cancer by facilitating T cell expansion and cytotoxicity. Our previous research demonstrated that the combination of tumor necrosis factor receptor 2 (TNFR2) with immune activity treatments synergistically suppresses tumor growth. This insight led to exploring the efficacy of a combined treatment strategy involving PD­1 inhibitors, PTP1B inhibitors and TNFR2 antibodies (triple therapy) in NSCLC. In this context, the therapeutic effectiveness of these combination immunotherapies was validated in mouse models with NSCLC by analyzing the expansion and function of immune cells, thereby assessing their impact on tumor growth. The results indicated that inhibiting PTP1B decreases the expression of PD­L1 and TNFR2 on LLC cells, along with an increase in the proportion of CD4+T and CD8+T cells. Compared with other treatment groups, the triple therapy significantly reduced tumor volume in mice with NSCLC and extended their survival. Moreover, this combination therapy altered the distribution of myeloid­derived suppressor cells, dendritic cells, B cells and M1 macrophages, while increasing the proportion of CD8+T cells and reducing the proportion of Treg cells in the spleens, lymph nodes, and tumors of NSCLC models. The triple therapy also resulted in a decrease in PD­L1, PTP1B and TNFR2 expression within NSCLC tumor tissues in mice. Overall, the triple therapy effectively suppressed tumor growth and improved outcomes in mice with NSCLC by modulating immune cell distribution and reducing levels of target immune proteins.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Récepteur-1 de mort cellulaire programmée , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Récepteur au facteur de nécrose tumorale de type II , Animaux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Souris , Récepteur au facteur de nécrose tumorale de type II/antagonistes et inhibiteurs , Récepteur au facteur de nécrose tumorale de type II/immunologie , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Humains , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lignée cellulaire tumorale , Évolution de la maladie , Femelle , Immunothérapie/méthodes , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie
8.
Sci Adv ; 10(38): eado9746, 2024 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-39303028

RÉSUMÉ

While immune checkpoint inhibitors have revolutionized cancer therapy, many patients exhibit poor outcomes. Here, we show immunotherapy responses in bladder and non-small cell lung cancers are effectively predicted by factoring tumor mutation burden (TMB) into burdens on specific protein assemblies. This approach identifies 13 protein assemblies for which the assembly-level mutation burden (AMB) predicts treatment outcomes, which can be combined to powerfully separate responders from nonresponders in multiple cohorts (e.g., 76% versus 37% bladder cancer 1-year survival). These results are corroborated by (i) engineered disruptions in the predictive assemblies, which modulate immunotherapy response in mice, and (ii) histochemistry showing that predicted responders have elevated inflammation. The 13 assemblies have diverse roles in DNA damage checkpoints, oxidative stress, or Janus kinase/signal transducers and activators of transcription signaling and include unexpected genes (e.g., PIK3CG and FOXP1) for which mutation affects treatment response. This study provides a roadmap for using tumor cell biology to factor mutational effects on immune response.


Sujet(s)
Immunothérapie , Mutation , Humains , Immunothérapie/méthodes , Animaux , Souris , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Résultat thérapeutique , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/immunologie , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/thérapie , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Protéines tumorales/génétique , Protéines tumorales/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie
9.
J Immunother Cancer ; 12(9)2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39244215

RÉSUMÉ

BACKGROUND: Immune checkpoint blockade (ICB) has made remarkable achievements, but newly identified armored and cold tumors cannot respond to ICB therapy. The high prevalence of concomitant medications has huge impact on immunotherapeutic responses, but the clinical effects on the therapeutic outcome of armored and cold tumors are still unclear. METHODS: In this research, using large-scale transcriptomics datasets, the expression and potential biological functions of angiotensin II receptor 1 (AGTR1), the target of angiotensin receptor blocker (ARB), were investigated. Next, the roles of ARB in tumor cells and tumor microenvironment cells were defined by a series of in vitro and in vivo assays. In addition, the clinical impacts of ARB on ICB therapy were assessed by multicenter cohorts and meta-analysis. RESULTS: AGTR1 was overexpressed in armored and cold tumors and associated with poor response to ICB therapy. ARB, the inhibitor for AGTR1, only suppressed the aggressiveness of tumor cells with high AGTR1 expression, which accounted for a very small proportion. Further analysis revealed that AGTR1 was always highly expressed in cancer-associated fibroblasts (CAFs) and ARB inhibited type I collagen expression in CAFs by suppressing the RhoA-YAP axis. Moreover, ARB could also drastically reverse the phenotype of armored and cold to soft and hot in vivo, leading to a higher response to ICB therapy. In addition, both our in-house cohorts and meta-analysis further supported the idea that ARB can significantly enhance ICB efficacy. CONCLUSION: Overall, we identify AGTR1 as a novel target in armored and cold tumors and demonstrate the improved therapeutic efficacy of ICB in combination with ARB. These findings could provide novel clinical insight into how to treat patients with refractory armored and cold tumors.


Sujet(s)
Antagonistes des récepteurs aux angiotensines , Inhibiteurs de points de contrôle immunitaires , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Antagonistes des récepteurs aux angiotensines/usage thérapeutique , Antagonistes des récepteurs aux angiotensines/pharmacologie , Animaux , Souris , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Récepteur de type 1 à l'angiotensine-II/métabolisme , Récepteur de type 1 à l'angiotensine-II/génétique , Microenvironnement tumoral , Lignée cellulaire tumorale , Femelle
10.
Biomed Pharmacother ; 179: 117430, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39260322

RÉSUMÉ

Targeting checkpoints for immune cell activation has been acknowledged known as one of the most effective way to activate anti-tumor immune responses. Among them, drugs targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are approved for clinical treatment though several more are in advanced stages of development, which demonstrated durable response rates and manageable safety profile. However, its therapy efficacy is unsatisfactory in pancreatic cancer (PC), which can be limited by the overall condition of patients, the pathological type of PC, the expression level of tumor related genes, etc. To improve clinical efficiency, various researches have been conducted, and the efficacy of combination therapy showed significantly improvement compared to monotherapy. This review analyzed current strategies based on anti-CTLA-4 combination immunotherapy, providing totally new idea for future research.


Sujet(s)
Antigène CTLA-4 , Inhibiteurs de points de contrôle immunitaires , Tumeurs du pancréas , Humains , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/anatomopathologie , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Animaux , Immunothérapie/méthodes , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
11.
Sci Rep ; 14(1): 21625, 2024 09 16.
Article de Anglais | MEDLINE | ID: mdl-39285232

RÉSUMÉ

Lung cancer remains the leading cause of cancer death. Treatment with immune checkpoint inhibitor (ICI) alone or combination with chemotherapy served as first-line therapy in non-small cell lung cancer (NSCLC). However, only 20-50% of NSCLC patients respond to ICI. Necroptosis, an inflammatory form of cell death plays an important role in the regulation of tumor immune microenvironment which may affect prognosis and ICI response but its clinical significance in NSCLC patients has remained largely unknown. Therefore, we aimed to analyze the correlation between key necroptotic proteins and necroptosis and clinical outcomes, the status of tumor-infiltrating immune cells, and response to ICI in NSCLC patients. The expression of receptor-interacting protein kinase 3 (RIPK3), mixed lineage kinase domain-like protein (MLKL) and phosphorylated MLKL (pMLKL) were immunolocalized in 125 surgically resected NSCLC patients and 23 NSCLC patients administered with ICI therapy. CD8 + and FOXp3 + T cells and CD163 + M2 macrophages were also immunolocalized. High RIPK3 status was positively correlated with survival of the patients and RIPK3 turned out an independent favorable prognostic factor of the patients. RIPK3 was negatively correlated with CD8 + T cells, while MLKL positively correlated with CD163 + M2 macrophages, suggesting the possible involvement of RIPK3 and MLKL in formulating immunosuppressive microenvironment. In addition, high RIPK3 status tended to be associated with clinical resistance to ICI therapy (P-value = 0.057). Furthermore, NSCLC cells-expressing RIPK3 suppressed T cells response to ICI therapy in vitro. Therefore, RIPK3 and MLKL could induce an immunosuppressive microenvironment, resulting in low response to ICI therapy in NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Nécroptose , Protein kinases , Receptor-Interacting Protein Serine-Threonine Kinases , Microenvironnement tumoral , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/immunologie , Nécroptose/effets des médicaments et des substances chimiques , Pronostic , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme
12.
Nat Commun ; 15(1): 7875, 2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-39285180

RÉSUMÉ

Dysregulation of master regulator c-MYC (MYC) plays a central role in hepatocellular carcinoma (HCC) and other cancers but remains an elusive target for therapeutic intervention. MYC expression is epigenetically modulated within naturally occurring DNA loop structures, Insulated Genomic Domains (IGDs). We present a therapeutic approach using an epigenomic controller (EC), a programmable epigenomic mRNA medicine, to precisely modify MYC IGD sub-elements, leading to methylation of MYC regulatory elements and durable downregulation of MYC mRNA transcription. Significant antitumor activity is observed in preclinical models of HCC treated with the MYC-targeted EC, as monotherapy or in combination with tyrosine kinase or immune checkpoint inhibitors. These findings pave the way for clinical development of MYC-targeting epigenomic controllers in HCC patients and provide a framework for programmable epigenomic mRNA therapeutics for cancer and other diseases.


Sujet(s)
Carcinome hépatocellulaire , Méthylation de l'ADN , Régulation négative , Épigénomique , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Protéines proto-oncogènes c-myc , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Humains , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Animaux , Souris , Lignée cellulaire tumorale , Régulation négative/génétique , Épigénomique/méthodes , Épigenèse génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Transcription génétique , ARN messager/génétique , ARN messager/métabolisme
13.
Target Oncol ; 19(5): 769-778, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39222223

RÉSUMÉ

BACKGROUND: While guidelines recommend immune checkpoint inhibitor (ICI) rechallenge as second-line therapy for unresectable hepatocellular carcinoma (HCC), data supporting this remain limited, particularly regarding a standard regimen for first- and second-line treatments. Tremelimumab/durvalumab was recently approved but data on ICI rechallenge are lacking. OBJECTIVES: The purpose of this study was to evaluate the early efficacy and safety of tremelimumab/durvalumab for HCC as an ICI rechallenge following initial ICI therapy with atezolizumab/bevacizumab. PATIENTS AND METHODS: This multicenter retrospective study included patients with HCC who underwent treatment with tremelimumab/durvalumab, with relevant available clinical information. We evaluated the safety and efficacy of tremelimumab/durvalumab as ICI rechallenge following initial treatment with atezolizumab/bevacizumab. We analyzed the outcomes in patients who underwent tremelimumab/durvalumab as an ICI rechallenge and those who received tremelimumab/durvalumab as their initial ICI therapy RESULT: A total of 45 patients treated with tremelimumab/durvalumab were included, with 55.6% (25/45) undergoing ICI rechallenge. The objective-response and disease-control rates in patients who underwent ICI rechallenge were 14.3% (3/21) and 47.6% (10/21), respectively, similar to those in patients initially treated with tremelimumab/durvalumab. All patients (n = 3) who experienced the best response to progressive disease (PD) with initial atezolizumab/bevacizumab experienced PD during ICI rechallenge. The incidence rates of adverse events were similar between patient groups treated with tremelimumab/durvalumab as ICI rechallenge and initial ICI. Among patients experiencing immune-related adverse events (irAEs) with atezolizumab/bevacizumab, 75% (3/4) encountered similar irAEs during ICI rechallenge. CONCLUSION: Early safety and efficacy profiles of durvalumab/tremelimumab as ICI rechallenge are satisfactory.


Sujet(s)
Anticorps monoclonaux humanisés , Anticorps monoclonaux , Bévacizumab , Carcinome hépatocellulaire , Inhibiteurs de points de contrôle immunitaires , Tumeurs du foie , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/pharmacologie , Mâle , Femelle , Bévacizumab/usage thérapeutique , Bévacizumab/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Études rétrospectives , Sujet âgé , Adulte d'âge moyen , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/pharmacologie , Adulte , Sujet âgé de 80 ans ou plus , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie
14.
Mol Cancer ; 23(1): 197, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39272096

RÉSUMÉ

Anti-angiogenic agents elicit considerable immune modulatory effects within the tumor microenvironment, underscoring the rationale for synergistic clinical development of VEGF and immune checkpoint inhibitors in advanced gastric cancer (AGC). Early phase studies involving Asian patients demonstrated encouraging anti-tumor efficacies. We report the results of the REGOMUNE phase II study, in which Caucasian patients were administered regorafenib, a multi-tyrosine kinase inhibitor, in combination with avelumab, a PD-L1-targeting monoclonal antibody. This therapeutic regimen resulted in deep and durable responses in 19% of patients, with the median duration of response not yet reached. Notwithstanding, a significant proportion of AGC patients exhibited no therapeutic advantage, prompting investigations into mechanisms of inherent resistance. Comprehensive biomarker profiling elucidated that non-responders predominantly exhibited an augmented presence of M2 macrophages within the tumor microenvironment and a marked overexpression of S100A10 by neoplastic cells, a protein previously implicated in macrophage chemotaxis. Additionally, peripheral biomarker assessments identified elevated levels of cytokines, including CSF-1, IL-4, IL-8, and TWEAK, correlating with adverse clinical outcomes, thereby accentuating the role of macrophage infiltration in mediating resistance. These insights furnish an invaluable foundation for elucidating, and potentially circumventing, resistance mechanisms in current AGC therapeutic paradigms, emphasizing the integral role of tumor microenvironmental dynamics and immune modulation.


Sujet(s)
Antigène CD274 , Résistance aux médicaments antinéoplasiques , Protéomique , Tumeurs de l'estomac , Microenvironnement tumoral , Humains , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Protéomique/méthodes , Antigène CD274/métabolisme , Antigène CD274/génétique , Marqueurs biologiques tumoraux , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Femelle , Mâle , Transcriptome , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Adulte d'âge moyen , Analyse de profil d'expression de gènes
15.
J Nanobiotechnology ; 22(1): 563, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39272146

RÉSUMÉ

BACKGROUND: While small extracellular vesicles (sEVs)-derived circular RNAs (circRNAs) have been emerged as significant players in cancer, the function and underlying mechanism of sEVs-derived circRNAs in anti-cancer immunity remain unclear. METHODS: Gastric cancer (GC)-derived circRNAs were identified using RNA-seq data from GEO datasets and quantitative reverse transcription polymerase chain reaction (qRT-PCR), RNA immunoprecipitation, dual-luciferase assay, and bioinformatics analysis were performed to investigate the regulatory axis. Transwell assay, wound healing assay, cell counting kit-8 (CCK-8) assay, and xenograft models were used to evaluate its role in GC progression in vivo and in vitro. The delivery of specific circRNAs into sEVs were verified through electron microscopy, nanoparticle tracking analysis (NTA) and fuorescence in situ hybridization (FISH). Flow cytometric analysis and immunohistochemical staining were conducted to find out how specific circRNAs mediated CD8+ T cell exhaustion and resistant to anti-programmed cell death 1 (PD-1) therapy. RESULTS: We identified that circ_0001947, packaged by GC-derived sEVs, was obviously elevated in GC and was associated with poor clinical outcome. High circ0001947 level augmented the proliferation, migration, and invasion of GC cells. Mechanistically, circ0001947 sponged miR-661 and miR-671-5p to promote the expression of CD39, which further facilitated CD8+ T cell exhaustion and immune resistance. Conversely, blocking circ_0001947 attenuated CD8+ T cell exhaustion and increased the response to anti-PD-1 therapy. CONCLUSIONS: Our study manifested the therapeutic potential of targeting sEVs-transmitted circ_0001947 to prohibit CD8+ T cell exhaustion and immune resistance in GC.


Sujet(s)
Lymphocytes T CD8+ , Vésicules extracellulaires , ARN circulaire , Tumeurs de l'estomac , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/immunologie , ARN circulaire/génétique , Humains , Vésicules extracellulaires/métabolisme , Animaux , Lymphocytes T CD8+/immunologie , Souris , Lignée cellulaire tumorale , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Évolution de la maladie , Souris nude , Résistance aux médicaments antinéoplasiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Souris de lignée BALB C , Mâle , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Épuisement des cellules T
16.
Int J Mol Sci ; 25(17)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39273290

RÉSUMÉ

This study aimed to identify hub genes involved in regulatory T cell (Treg) function and migration, offering insights into potential therapeutic targets for cancer immunotherapy. We performed a comprehensive bioinformatics analysis using three gene expression microarray datasets from the GEO database. Differentially expressed genes (DEGs) were identified to pathway enrichment analysis to explore their functional roles and potential pathways. A protein-protein interaction network was constructed to identify hub genes critical for Treg activity. We further evaluated the co-expression of these hub genes with immune checkpoint proteins (PD-1, PD-L1, CTLA4) and assessed their prognostic significance. Through this comprehensive analysis, we identified CCR8 as a key player in Treg migration and explored its potential synergistic effects with ICIs. Our findings suggest that CCR8-targeted therapies could enhance cancer immunotherapy outcomes, with breast invasive carcinoma (BRCA) emerging as a promising indication for combination therapy. This study highlights the potential of CCR8 as a biomarker and therapeutic target, contributing to the development of targeted cancer treatment strategies.


Sujet(s)
Biologie informatique , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Cartes d'interactions protéiques , Lymphocytes T régulateurs , Humains , Biologie informatique/méthodes , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Tumeurs/génétique , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Analyse de profil d'expression de gènes , Pronostic , Réseaux de régulation génique , Récepteurs CCR8/génétique , Récepteurs CCR8/métabolisme , Marqueurs biologiques tumoraux/génétique
17.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39273605

RÉSUMÉ

Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), defective deoxyribonucleic acid (DNA) mismatch repair (dMMR), microsatellite instability (MSI), and the tumor microenvironment (TME). Over the past decade, several types of ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, anti-programmed cell death 1 (PD-1) antibodies, anti-programmed cell death ligand 1 (PD-L1) antibodies, and anti-lymphocyte activation gene-3 (LAG-3) antibodies have been studied and approved by the Food and Drug Administration (FDA), with ongoing research on others. Recent studies highlight the critical role of the gut microbiome in influencing a positive therapeutic response to ICIs, emphasizing the importance of modeling factors that can maintain a healthy microbiome. However, resistance mechanisms can emerge, such as increased expression of alternative immune checkpoints, T-cell immunoglobulin (Ig), mucin domain-containing protein 3 (TIM-3), LAG-3, impaired antigen presentation, and alterations in the TME. This review aims to synthesize the data regarding the interactions between microbiota and immunotherapy (IT). Understanding these mechanisms is essential for optimizing ICI therapy and developing effective combination strategies.


Sujet(s)
Marqueurs biologiques tumoraux , Résistance aux médicaments antinéoplasiques , Inhibiteurs de points de contrôle immunitaires , Tumeurs , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/immunologie , Marqueurs biologiques tumoraux/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
18.
Mol Biomed ; 5(1): 37, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39251538

RÉSUMÉ

The combination of anti-angiogenic drugs and immune checkpoint inhibitors (ICIs) in the treatment of tumors is emerging as a way to improve ICIs-resistant tumor therapy. In addition, gut microbes (GMs) are involved in angiogenesis in the tumor microenvironment and are also associated with the antitumor function of immune checkpoint inhibitors. However, it is unclear whether gut microbes have a role in anti-tumor function in the combination of anti-angiogenic drugs and immune checkpoint inhibitors for cancer treatment. Endostatin, an angiogenesis inhibitor, has been widely used as an antiangiogenic therapy for cancer. We showed that combined therapy with an adenovirus encoding human endostatin, named Ad-E, and PD-1 blockade dramatically abrogated MC38 tumor growth. The structure of intestinal microbes in mice was changed after combination treatment. We found that the antitumor function of combination therapy was inhibited after the elimination of intestinal microbes. In mice with depleted microbiota, oral gavage of Bacteroides fragilis salvaged the antitumor effects of combination Ad-E and αPD-1 monoclonal antibody (mAb) to a certain extent. Further, Bacteroides fragilis could improve CD3+T cells, NK cells, and IFNγ+CD8+ T cells in the tumor microenvironment to inhibit tumor growth. Besides, Bacteroides fragilis might restore antitumor function by down-regulating isobutyric acid (IBA). Our results suggested that GMs may be involved in the combination of Ad-E and αPD-1 mAb for cancer treatment, which has oncological implications for tumor growth dynamics and cancer immune surveillance.


Sujet(s)
Tumeurs colorectales , Endostatines , Microbiome gastro-intestinal , Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Endostatines/pharmacologie , Endostatines/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/immunologie , Tumeurs colorectales/microbiologie , Tumeurs colorectales/anatomopathologie , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Humains , Lignée cellulaire tumorale , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Souris de lignée C57BL , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Inhibiteurs de l'angiogenèse/administration et posologie , Femelle
19.
J Clin Invest ; 134(17)2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39225102

RÉSUMÉ

Tumor reliance on glycolysis is a hallmark of cancer. Immunotherapy is more effective in controlling glycolysis-low tumors lacking lactate dehydrogenase (LDH) due to reduced tumor lactate efflux and enhanced glucose availability within the tumor microenvironment (TME). LDH inhibitors (LDHi) reduce glucose uptake and tumor growth in preclinical models, but their impact on tumor-infiltrating T cells is not fully elucidated. Tumor cells have higher basal LDH expression and glycolysis levels compared with infiltrating T cells, creating a therapeutic opportunity for tumor-specific targeting of glycolysis. We demonstrate that LDHi treatment (a) decreases tumor cell glucose uptake, expression of the glucose transporter GLUT1, and tumor cell proliferation while (b) increasing glucose uptake, GLUT1 expression, and proliferation of tumor-infiltrating T cells. Accordingly, increasing glucose availability in the microenvironment via LDH inhibition leads to improved tumor-killing T cell function and impaired Treg immunosuppressive activity in vitro. Moreover, combining LDH inhibition with immune checkpoint blockade therapy effectively controls murine melanoma and colon cancer progression by promoting effector T cell infiltration and activation while destabilizing Tregs. Our results establish LDH inhibition as an effective strategy for rebalancing glucose availability for T cells within the TME, which can enhance T cell function and antitumor immunity.


Sujet(s)
Glucose , L-Lactate dehydrogenase , Microenvironnement tumoral , Animaux , Souris , Glucose/métabolisme , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , L-Lactate dehydrogenase/métabolisme , L-Lactate dehydrogenase/antagonistes et inhibiteurs , L-Lactate dehydrogenase/immunologie , Humains , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 1/antagonistes et inhibiteurs , Transporteur de glucose de type 1/immunologie , Transporteur de glucose de type 1/génétique , Lignée cellulaire tumorale , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Femelle , Lymphocytes TIL/immunologie , Lymphocytes TIL/effets des médicaments et des substances chimiques , Tumeurs du côlon/immunologie , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/métabolisme , Antienzymes/pharmacologie , Immunothérapie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique
20.
Mol Cancer ; 23(1): 183, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39223527

RÉSUMÉ

Programmed death receptor-1 (PD-1) and its ligand, programmed death ligand-1 (PD-L1) are essential molecules that are key in modulating immune responses. PD-L1 is constitutively expressed on various immune cells, epithelial cells, and cancer cells, where it functions as a co-stimulatory molecule capable of impairing T-cell mediated immune responses. Upon binding to PD-1 on activated T-cells, the PD-1/PD-L1 interaction triggers signaling pathways that can induce T-cell apoptosis or anergy, thereby facilitating the immune escape of tumors. In urological cancers, including bladder cancer (BCa), renal cell carcinoma (RCC), and prostate cancer (PCa), the upregulation of PD-L1 has been demonstrated. It is linked to poor prognosis and enhanced tumor immune evasion. Recent studies have highlighted the significant role of the PD-1/PD-L1 axis in the immune escape mechanisms of urological cancers. The interaction between PD-L1 and PD-1 on T-cells further contributes to immunosuppression by inhibiting T-cell activation and proliferation. Clinical applications of PD-1/PD-L1 checkpoint inhibitors have shown promising efficacy in treating advanced urological cancers, significantly improving patient outcomes. However, resistance to these therapies, either intrinsic or acquired, remains a significant challenge. This review aims to provide a comprehensive overview of the role of the PD-1/PD-L1 signaling pathway in urological cancers. We summarize the regulatory mechanism underlying PD-1 and PD-L1 expression and activity, including genetic, epigenetic, post-transcriptional, and post-translational modifications. Additionally, we discuss current clinical research on PD-1/PD-L1 inhibitors, their therapeutic potential, and the challenges associated with resistance. Understanding these mechanisms is crucial for developing new strategies to overcome therapeutic limitations and enhance the efficacy of cancer immunotherapy.


Sujet(s)
Antigène CD274 , Immunothérapie , Récepteur-1 de mort cellulaire programmée , Tumeurs urologiques , Humains , Antigène CD274/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Immunothérapie/méthodes , Tumeurs urologiques/thérapie , Tumeurs urologiques/métabolisme , Tumeurs urologiques/traitement médicamenteux , Tumeurs urologiques/immunologie , Tumeurs urologiques/étiologie , Tumeurs urologiques/anatomopathologie , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Échappement de la tumeur à la surveillance immunitaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE