Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 570
Filtrer
1.
Cancer Immunol Immunother ; 73(11): 234, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39271499

RÉSUMÉ

The clinical response to immune checkpoint inhibitors may vary by tumor type and many tumors present with either primary or acquired resistance to immunotherapy. Improved understanding of the molecular and immunologic mechanisms underlying immunotherapy resistance is essential for developing biomarkers and for guiding the optimum approach to selecting treatment regimens and sequencing. This is increasingly important for tumors with primary resistance as effective biomarkers in this setting can guide clinicians about appropriate treatment regimen selection in the first-line setting. Multiple potential biological mechanisms of primary resistance have been proposed but most are yet to be validated in prospective clinical cohorts. Individual biomarkers have poor specificity and sensitivity, and the development of validated and integrated predictive models may guide which patient will benefit from monotherapy versus combination therapy. In this review, we discuss the emerging data identifying the molecular mechanisms of primary resistance to immunotherapy and explore potential therapeutic strategies to target these.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Inhibiteurs de points de contrôle immunitaires , Tumeurs , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/thérapie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/immunologie , Génomique/méthodes , Marqueurs biologiques tumoraux/génétique , Immunothérapie/méthodes , Animaux
2.
Int J Oncol ; 65(4)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39219258

RÉSUMÉ

The use of antitumor drugs represents a reliable strategy for cancer therapy. Unfortunately, drug resistance has become increasingly common and contributes to tumor metastasis and local recurrence. The tumor immune microenvironment (TME) consists of immune cells, cytokines and immunomodulators, and collectively they influence the response to treatment. Epigenetic changes including DNA methylation and histone modification, as well as increased drug exportation have been reported to contribute to the development of drug resistance in cancers. In the past few years, the majority of studies on tumors have only focused on the development and progression of a tumor from a mechanistic standpoint; few studies have examined whether the changes in the TME can also affect tumor growth and drug resistance. Recently, emerging evidence have raised more concerns regarding the role of TME in the development of drug resistance. In the present review, it was discussed how the suppressive TME adapts to drug resistance characterized by the cooperation of immune cells, cytokines, immunomodulators, stromal cells and extracellular matrix. Furthermore, it was reviewed how these immunological or metabolic changes alter immuno­surveillance and thus facilitate tumor drug resistance. In addition, potential targets present in the TME for developing novel therapeutic strategies to improve individualized therapy for cancer treatment were revealed.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Tumeurs , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/immunologie , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/anatomopathologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Épigenèse génétique/effets des médicaments et des substances chimiques , Animaux , Cytokines/métabolisme
3.
Cancer Res ; 84(17): 2792-2805, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39228255

RÉSUMÉ

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


Sujet(s)
Antigènes néoplasiques , Protéines de liaison à l'ADN , Résistance aux médicaments antinéoplasiques , Mélanome , Facteurs de transcription , Vémurafénib , Animaux , Humains , Facteurs de transcription/génétique , Facteurs de transcription/immunologie , Souris , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/immunologie , Résistance aux médicaments antinéoplasiques/immunologie , Antigènes néoplasiques/immunologie , Antigènes néoplasiques/génétique , Vémurafénib/pharmacologie , Vémurafénib/usage thérapeutique , Mélanome/immunologie , Mélanome/traitement médicamenteux , Mélanome/génétique , Mélanome/thérapie , Immunothérapie/méthodes , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/immunologie , Lignée cellulaire tumorale , Femelle , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mutation , Thérapie moléculaire ciblée/méthodes , Souris de lignée C57BL
4.
Biochem Biophys Res Commun ; 733: 150686, 2024 Nov 12.
Article de Anglais | MEDLINE | ID: mdl-39278093

RÉSUMÉ

Our study aims to explore the effects of neoadjuvant chemotherapy (NACT) on tumour cells and immune cells in the immune microenvironment of patients with high-grade serous ovarian cancer (HGSOC). Single-cell RNA sequencing data of paired ovarian cancer tissues were analysed before and after NACT in 11 patients with HGSOC. The effect of NACT on two major cell components of the tumour microenvironment, epithelial cells and CD8+T cells, was investigated. The mechanisms of epithelial cell evasion by NACT and immune killing were explored from the perspectives of gene expression, functional characteristics, transcriptional regulation, and cell communication. Key targets for reversing NACT resistance were identified and possible therapeutic strategies proposed. While NACT improved the de novo differentiation of anti-tumour CD8+T cells, enhancing their anti-tumour function, it increased the proportion of cancer cells with high HSP90B1 expression. Thus, the potential reasons for NACT resistance were identified as: 1) high levels of endoplasmic reticulum stress (ERS) characteristics, 2) high expression of the MDK-NCL ligand-receptor pair between them and exhausted CD8+T cells before NACT, and 3) high expression of the NECTIN2-TIGIT immune ligand-receptor pair between them and exhausted CD8+T cells after NACT. Thus, our study reveals the mechanisms underlying NACT resistance in patients with HGSOC from the perspective of the independent and interactive roles of cancer cells and CD8+T cells. We propose therapeutic strategies targeting the ERS marker HSP90B1 and the immune escape marker MDK before or during NACT, while targeting NECTIN2 blockade after NACT. This approach may offer new insights into combination treatments for patients with HGSOC displaying NACT resistance.


Sujet(s)
Lymphocytes T CD8+ , Résistance aux médicaments antinéoplasiques , Stress du réticulum endoplasmique , Traitement néoadjuvant , Tumeurs de l'ovaire , Humains , Femelle , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Résistance aux médicaments antinéoplasiques/immunologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/immunologie , Traitement néoadjuvant/méthodes , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Adulte d'âge moyen
5.
Trends Cancer ; 10(10): 893-919, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39214782

RÉSUMÉ

Bispecific antibodies (bsAbs) are engineered molecules designed to target two different epitopes or antigens. The mechanism of action is determined by the bsAb molecular targets and structure (or format), which can be manipulated to create variable and novel functionalities, including linking immune cells with tumor cells, or dual signaling pathway blockade. Several bsAbs have already changed the treatment landscape of hematological malignancies and select solid cancers. However, the mechanisms of resistance to these agents are understudied and the management of toxicities remains challenging. Herein, we review the principles in bsAb engineering, current understanding of mechanisms of action and resistance, data for clinical application, and provide a perspective on ongoing challenges and future developments in this field.


Sujet(s)
Anticorps bispécifiques , Tumeurs , Médecine de précision , Anticorps bispécifiques/usage thérapeutique , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/immunologie , Humains , Médecine de précision/méthodes , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/immunologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques immunologiques/usage thérapeutique , Antinéoplasiques immunologiques/pharmacologie , Thérapie moléculaire ciblée/méthodes
6.
World J Gastroenterol ; 30(31): 3654-3667, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39193002

RÉSUMÉ

Despite advances in cancer treatment, pancreatic cancer (PC) remains a disease with high mortality rates and poor survival outcomes. The B7 homolog 3 (B7-H3) checkpoint molecule is overexpressed among many malignant tumors, including PC, with low or absent expression in healthy tissues. By modulating various immunological and nonimmunological molecular mechanisms, B7-H3 may influence the progression of PC. However, the impact of B7-H3 on the survival of patients with PC remains a subject of debate. Still, most available scientific data recognize this molecule as a suppressive factor to antitumor immunity in PC. Furthermore, it has been demonstrated that B7-H3 stimulates the migration, invasion, and metastasis of PC cells, and enhances resistance to chemotherapy. In preclinical models of PC, B7-H3-targeting monoclonal antibodies have exerted profound antitumor effects by increasing natural killer cell-mediated antibody-dependent cellular cytotoxicity and delivering radioisotopes and cytotoxic drugs to the tumor site. Finally, PC treatment with B7-H3-targeting antibody-drug conjugates and chimeric antigen receptor T cells is being tested in clinical studies. This review provides a comprehensive analysis of all PC-related studies in the context of B7-H3 and points to deficiencies in the current data that should be overcome by future research.


Sujet(s)
Antigènes B7 , Tumeurs du pancréas , Humains , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/mortalité , Antigènes B7/métabolisme , Antigènes B7/antagonistes et inhibiteurs , Antigènes B7/immunologie , Animaux , Résistance aux médicaments antinéoplasiques/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Antinéoplasiques immunologiques/usage thérapeutique , Anticorps monoclonaux/usage thérapeutique
7.
Nat Commun ; 15(1): 7077, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39152118

RÉSUMÉ

Enoblituzumab, an immunotherapeutic agent targeting CD276, shows both safety and efficacy in activating T cells and oligodendrocyte-like cells against various cancers. Preclinical studies and mouse models suggest that therapies targeting CD276 may outperform PD1/PD-L1 blockade. However, data from mouse models indicate a significant non-responsive population to anti-CD276 treatment, with the mechanisms of resistance still unclear. In this study, we evaluate the activity of anti-CD276 antibodies in a chemically-induced murine model of head and neck squamous cell carcinoma. Using models of induced and orthotopic carcinogenesis, we identify ITGB6 as a key gene mediating differential responses to anti-CD276 treatment. Through single-cell RNA sequencing and gene-knockout mouse models, we find that ITGB6 regulates the expression of the tumor-associated chemokine CX3CL1, which recruits and activates PF4+ macrophages that express high levels of CX3CR1. Inhibition of the CX3CL1-CX3CR1 axis suppresses the infiltration and secretion of CXCL16 by PF4+ macrophages, thereby reinvigorating cytotoxic CXCR6+ CD8+ T cells and enhancing sensitivity to anti-CD276 treatment. Further investigations demonstrate that inhibiting ITGB6 restores sensitivity to PD1 antibodies in mice resistant to anti-PD1 treatment. In summary, our research reveals a resistance mechanism associated with immune checkpoint inhibitor therapy and identifies potential targets to overcome resistance in cancer treatment.


Sujet(s)
Antigènes B7 , Tumeurs de la tête et du cou , Souris knockout , Animaux , Souris , Antigènes B7/métabolisme , Antigènes B7/génétique , Antigènes B7/antagonistes et inhibiteurs , Humains , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Lignée cellulaire tumorale , Souris de lignée C57BL , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme , Modèles animaux de maladie humaine , Femelle , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
8.
Crit Rev Oncol Hematol ; 202: 104457, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39038527

RÉSUMÉ

Tumor resistance poses a significant challenge to effective cancer treatment, making it imperative to explore new therapeutic strategies. Recent studies have highlighted the profound involvement of immune cells in the development of tumor resistance. Within the tumor microenvironment, macrophages undergo polarization into the M2 phenotype, thus promoting the emergence of drug-resistant tumors. Neutrophils contribute to tumor resistance by forming extracellular traps. While T cells and natural killer (NK) cells exert their impact through direct cytotoxicity against tumor cells. Additionally, dendritic cells (DCs) have been implicated in preventing tumor drug resistance by stimulating T cell activation. In this review, we provide a comprehensive summary of the current knowledge regarding immune cell-mediated modulation of tumor resistance at the molecular level, with a particular focus on macrophages, neutrophils, DCs, T cells, and NK cells. The targeting of immune cell modulation exhibits considerable potential for addressing drug resistance, and an in-depth understanding of the molecular interactions between immune cells and tumor cells holds promise for the development of innovative therapies. Furthermore, we explore the clinical implications of these immune cells in the treatment of drug-resistant tumors. This review emphasizes the exploration of novel approaches that harness the functional capabilities of immune cells to effectively overcome drug-resistant tumors.


Sujet(s)
Cellules dendritiques , Résistance aux médicaments antinéoplasiques , Cellules tueuses naturelles , Tumeurs , Microenvironnement tumoral , Humains , Tumeurs/immunologie , Tumeurs/thérapie , Résistance aux médicaments antinéoplasiques/immunologie , Microenvironnement tumoral/immunologie , Cellules tueuses naturelles/immunologie , Cellules dendritiques/immunologie , Lymphocytes T/immunologie , Animaux , Macrophages/immunologie , Granulocytes neutrophiles/immunologie
9.
Nat Cancer ; 5(8): 1206-1226, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38844817

RÉSUMÉ

Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y6. Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Immunothérapie , Uridine diphosphate , Humains , Uridine diphosphate/métabolisme , Immunothérapie/méthodes , Résistance aux médicaments antinéoplasiques/immunologie , Animaux , Souris , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/traitement médicamenteux , Cytidine deaminase/métabolisme , Cytidine deaminase/génétique , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Lignée cellulaire tumorale , Récepteurs purinergiques P2/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/traitement médicamenteux , Nucléotides/métabolisme , Tolérance immunitaire , Récepteur-1 de mort cellulaire programmée
10.
Cell Stem Cell ; 31(8): 1101-1112, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38925125

RÉSUMÉ

Cancer stem cells (CSCs) are heterogeneous, possess self-renewal attributes, and orchestrate important crosstalk in tumors. We propose that the CSC state represents "mimicry" by cancer cells that leads to phenotypic plasticity. CSC mimicry is suggested as CSCs can impersonate immune cells, vasculo-endothelia, or lymphangiogenic cells to support cancer growth. CSCs facilitate both paracrine and juxtracrine signaling to prime tumor-associated immune and stromal cells to adopt pro-tumoral phenotypes, driving therapeutic resistance. Here, we outline the ingenuity of CSCs' mimicry in their quest to evade immune detection, which leads to immunotherapeutic resistance, and highlight CSC-mimicry-targeted therapeutic strategies for robust immunotherapy.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Échappement immunitaire , Tumeurs , Cellules souches tumorales , Humains , Cellules souches tumorales/immunologie , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/anatomopathologie , Résistance aux médicaments antinéoplasiques/immunologie , Animaux , Immunothérapie/méthodes
11.
Front Immunol ; 15: 1384121, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903504

RÉSUMÉ

The past decade has witnessed a revolution in cancer treatment, shifting from conventional drugs (chemotherapies) towards targeted molecular therapies and immune-based therapies, in particular immune-checkpoint inhibitors (ICIs). These immunotherapies release the host's immune system against the tumor and have shown unprecedented durable remission for patients with cancers that were thought incurable, such as metastatic melanoma, metastatic renal cell carcinoma (RCC), microsatellite instability (MSI) high colorectal cancer and late stages of non-small cell lung cancer (NSCLC). However, about 80% of the patients fail to respond to these immunotherapies and are therefore left with other less effective and potentially toxic treatments. Identifying and understanding the mechanisms that enable cancerous cells to adapt to and eventually overcome therapy can help circumvent resistance and improve treatment. In this review, we describe the recent discoveries on the onco-immunological processes which govern the tumor microenvironment and their impact on the resistance to PD-1/PD-L1 checkpoint blockade.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Inhibiteurs de points de contrôle immunitaires , Tumeurs , Microenvironnement tumoral , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Résistance aux médicaments antinéoplasiques/immunologie , Microenvironnement tumoral/immunologie , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Animaux , Immunothérapie/méthodes , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie
12.
Trends Immunol ; 45(7): 486-494, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876831

RÉSUMÉ

Immunotherapies have revolutionized the treatment of certain cancers, but challenges remain in overcoming immunotherapy resistance. Research shows that metabolic modulation of the tumor microenvironment can enhance antitumor immunity. Here, we discuss recent preclinical and clinical evidence for the efficacy of combining metabolic modifiers with immunotherapies. While this combination holds great promise, a few key areas must be addressed, which include identifying the effects of metabolic modifiers on immune cell metabolism, the putative biomarkers of therapeutic efficacy, the efficacy of modifiers on tumors harboring metabolic heterogeneity, and the potential development of resistance due to tumor reliance on alternative metabolic pathways. We propose solutions to these problems and posit that assessing these parameters is crucial for considering the potential of metabolic modifiers in sensitizing tumors to immunotherapies.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Immunothérapie , Voies et réseaux métaboliques , Tumeurs , Microenvironnement tumoral , Humains , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/métabolisme , Immunothérapie/méthodes , Microenvironnement tumoral/immunologie , Animaux , Résistance aux médicaments antinéoplasiques/immunologie
13.
Math Biosci ; 374: 109239, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38906526

RÉSUMÉ

Recent studies have utilized evolutionary mechanisms to impede the emergence of drug-resistant populations. In this paper, we develop a mathematical model that integrates hormonal treatment, immunotherapy, and the interactions among three cell types: drug-sensitive cancer cells, drug-resistant cancer cells and immune effector cells. Dynamical analysis is performed, examining the existence and stability of equilibria, thereby confirming the model's interpretability. Model parameters are calibrated using available prostate cancer data and literature. Through bifurcation analysis for drug sensitivity under different immune effector cells recruitment responses, we find that resistant cancer cells grow rapidly under weak recruitment response, maintain at a low level under strong recruitment response, and both may occur under moderate recruitment response. To quantify the competitiveness of sensitive and resistant cells, we introduce the comprehensive measures R1 and R2, respectively, which determine the outcome of competition. Additionally, we introduce the quantitative indicators CIE1 and CIE2 as comprehensive measures of the immune effects on sensitive and resistant cancer cells, respectively. These two indicators determine whether the corresponding cancer cells can maintain at a low level. Our work shows that the immune system is an important factor affecting the evolution of drug resistance and provides insights into how to enhance immune response to control resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Humains , Résistance aux médicaments antinéoplasiques/immunologie , Mâle , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/traitement médicamenteux , Modèles biologiques , Immunothérapie/méthodes , Dynamique non linéaire , Concepts mathématiques
14.
Front Biosci (Landmark Ed) ; 29(6): 204, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38940035

RÉSUMÉ

BACKGROUND: Lung cancer is the primary cause of cancer-related deaths, with one of the highest incidence and mortality rates of all malignant tumors. Dysregulated expression of DEPDC1B has been reported to occur in various tumor types. However, the functional implications of this alteration in lung adenocarcinoma (LUAD) and the underlying molecular mechanism remains unclear. In this study, we investigated the role and clinical significance of DEPDC1B in LUAD. METHODS: The expression of DEPDC1B in LUAD and its relationship with prognosis were systematically evaluated in several publically available datasets. The effects of DEPDC1B knockdown on the proliferation and motility of LUAD cells were assessed using the JULI Stage Real-time Cell History Recorder, while the effect of knockdown on the cell cycle was studied by flow cytometry. Furthermore, RNA-Sequencing (RNA-Seq) analysis was conducted to identify the downstream target genes and pathways regulated by DEPDC1B. Correlations between the expression of DEPDC1B and immune cell infiltration, immunotherapy resistance, and chemoresistance were also examined. Additionally, molecular biological methods were used to explore the regulatory mechanism of B-Myb on DEPDC1B expression. RESULTS: DEPDC1B was found to be upregulated in LUAD patients and this was associated with poor clinical outcomes. Knockdown of DEPDC1B inhibited cell growth, migration and motility, as well as cell cycle progression. Knockdown also resulted in the down-regulation of several downstream genes, including NID1, FN1, and EGFR, as well as the inactivation of multiple critical pathways, such as the ERK and PI3K-AKT pathways. Analysis of the tumor immuno-environment in LUAD revealed that high DEPDC1B expression was associated with an abundance of activated CD4+ memory T cells, M0 macrophages, M1 macrophages, and CD8+ T cells. Moreover, these tumors responded poorly to immunotherapy. Analysis of chemo-drug sensitivity showed that LUADs with high DEPDC1B expression were more responsive to frontline chemotherapeutic drugs such as Vinorelbine, Cisplatin, and Etoposide. Additionally, mechanistic investigations revealed that DEPDC1B is a direct target gene of B-Myb, and that its knockdown attenuated the proliferation and motility effects of B-Myb. CONCLUSIONS: In summary, our findings indicate that DEPDC1B is a critical regulator during the malignant progression of LUAD. DEPDC1B could therefore be a promising prognostic marker and therapeutic target in LUAD diagnosis and treatment.


Sujet(s)
Adénocarcinome pulmonaire , Mouvement cellulaire , Prolifération cellulaire , Protéines d'activation de la GTPase , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , Humains , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Protéines d'activation de la GTPase/génétique , Protéines d'activation de la GTPase/métabolisme , Tumeurs du poumon/immunologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Évolution de la maladie , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Pronostic , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/immunologie , Mâle , Techniques de knock-down de gènes , Transduction du signal , Protéines tumorales , Transactivateurs
15.
Immunol Res ; 72(4): 592-604, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38816670

RÉSUMÉ

Cervical cancer affects thousands of women globally with recurring high-risk HPV infections being at the centre of cervical pathology. Oncological treatment strategies are continually challenged by both chemoresistance and metastasis within patients. Although both work hand-in-hand, targeting their individual mechanisms could prove highly beneficial for treatment outcomes. Such targets include the metastatic-promoting stem cell marker, CD44, which is abundant in cervical cancer cells and is common to both chemoresistance and metastatic mechanisms. Seeing that many existing advanced-stage cervical cancer treatment regimes, such as platinum-based chemotherapy regimens, remain limited and are rarely curative, alternative treatment options within the field of immunology are being considered. The use of immune checkpoint inhibition therapy, which targets immune checkpoints, CTLA-4 and PD-1/PD-L1, has shown promise as an alternate standard of care for patients suffering from advanced-stage cervical cancer. Therefore, this review aims to assess whether immune checkpoint inhibition can mitigate the pathological effects of CD44-induced EMT, metastasis, and chemoresistance in cervical cancer patients.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Antigènes CD44 , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Tumeurs du col de l'utérus , Humains , Tumeurs du col de l'utérus/immunologie , Tumeurs du col de l'utérus/thérapie , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/anatomopathologie , Antigènes CD44/métabolisme , Femelle , Résistance aux médicaments antinéoplasiques/immunologie , Immunothérapie/méthodes , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Métastase tumorale , Évolution de la maladie , Animaux , Transition épithélio-mésenchymateuse/immunologie
16.
Front Immunol ; 15: 1341079, 2024.
Article de Anglais | MEDLINE | ID: mdl-38817612

RÉSUMÉ

Despite the efforts, pancreatic ductal adenocarcinoma (PDAC) is still highly lethal. Therapeutic challenges reside in late diagnosis and establishment of peculiar tumor microenvironment (TME) supporting tumor outgrowth. This stromal landscape is highly heterogeneous between patients and even in the same patient. The organization of functional sub-TME with different cellular compositions provides evolutive advantages and sustains therapeutic resistance. Tumor progressively establishes a TME that can suit its own needs, including proliferation, stemness and invasion. Cancer-associated fibroblasts and immune cells, the main non-neoplastic cellular TME components, follow soluble factors-mediated neoplastic instructions and synergize to promote chemoresistance and immune surveillance destruction. Unveiling heterotypic stromal-neoplastic interactions is thus pivotal to breaking this synergism and promoting the reprogramming of the TME toward an anti-tumor milieu, improving thus the efficacy of conventional and immune-based therapies. We underscore recent advances in the characterization of immune and fibroblast stromal components supporting or dampening pancreatic cancer progression, as well as novel multi-omic technologies improving the current knowledge of PDAC biology. Finally, we put into context how the clinic will translate the acquired knowledge to design new-generation clinical trials with the final aim of improving the outcome of PDAC patients.


Sujet(s)
Carcinome du canal pancréatique , Résistance aux médicaments antinéoplasiques , Tumeurs du pancréas , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/thérapie , Résistance aux médicaments antinéoplasiques/immunologie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Animaux , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/métabolisme , Tolérance immunitaire
17.
Hum Cell ; 37(4): 931-943, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38814516

RÉSUMÉ

Immunosuppressive regulatory cells (IRCs) play important roles in negatively regulating immune response, and are mainly divided into myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). Large numbers of preclinical and clinical studies have shown that inhibition or reduction of IRCs could effectively elevate antitumor immune responses. However, several studies also reported that excessive inhibition of IRCs function is one of the main reasons causing the side effects of cancer immunotherapy. Therefore, the reasonable regulation of IRCs is crucial for improving the safety and efficiency of cancer immunotherapy. In this review, we summarised the recent research advances in the cancer immunotherapy by regulating the proportion of IRCs, and discussed the roles of IRCs in regulating tumour immune evasion and drug resistance to immunotherapies. Furthermore, we also discussed how to balance the potential opportunities and challenges of using IRCs to improve the safety of cancer immunotherapies.


Sujet(s)
Immunothérapie , Cellules myéloïdes suppressives , Tumeurs , Lymphocytes T régulateurs , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Lymphocytes T régulateurs/immunologie , Cellules myéloïdes suppressives/immunologie , Échappement de la tumeur à la surveillance immunitaire/immunologie , Résistance aux médicaments antinéoplasiques/immunologie , Animaux , Immunosuppression thérapeutique
19.
Nat Cancer ; 5(9): 1318-1333, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38641734

RÉSUMÉ

Markers that predict response and resistance to chimeric antigen receptor (CAR) T cells in relapsed/refractory multiple myeloma are currently missing. We subjected mononuclear cells isolated from peripheral blood and bone marrow before and after the application of approved B cell maturation antigen-directed CAR T cells to single-cell multiomic analyses to identify markers associated with resistance and early relapse. Differences between responders and nonresponders were identified at the time of leukapheresis. Nonresponders showed an immunosuppressive microenvironment characterized by increased numbers of monocytes expressing the immune checkpoint molecule CD39 and suppressed CD8+ T cell and natural killer cell function. Analysis of CAR T cells showed cytotoxic and exhausted phenotypes in hyperexpanded clones compared to low/intermediate expanded clones. We identified potential immunotherapy targets on CAR T cells, like PD1, to improve their functionality and durability. Our work provides evidence that an immunosuppressive microenvironment causes resistance to CAR T cell therapies in multiple myeloma.


Sujet(s)
Antigène de maturation des cellules B , Immunothérapie adoptive , Myélome multiple , Récepteurs chimériques pour l'antigène , Myélome multiple/immunologie , Myélome multiple/thérapie , Humains , Récepteurs chimériques pour l'antigène/immunologie , Antigène de maturation des cellules B/immunologie , Immunothérapie adoptive/méthodes , Microenvironnement tumoral/immunologie , Analyse sur cellule unique/méthodes , Mâle , Lymphocytes T CD8+/immunologie , Récidive tumorale locale/immunologie , Femelle , Résistance aux médicaments antinéoplasiques/immunologie , Adulte d'âge moyen , Cellules tueuses naturelles/immunologie , Sujet âgé
20.
Clin Cancer Res ; 30(14): 3006-3022, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38687588

RÉSUMÉ

PURPOSE: Bispecific antibodies (BsAb) directed against B-cell maturation antigen (teclistamab) or the orphan G protein-coupled receptor GPRC5D (talquetamab) induce deep and durable responses in heavily pretreated patients with multiple myeloma. However, mechanisms underlying primary and acquired resistance remain poorly understood. EXPERIMENTAL DESIGN: The anti-multiple myeloma activity of teclistamab and talquetamab was evaluated in bone marrow (BM) samples from patients with multiple myeloma. T-cell phenotype and function were assessed in BM/peripheral blood samples obtained from patients with multiple myeloma who were treated with these BsAb. RESULTS: In ex vivo killing assays with 41 BM samples from BsAb-naive patients with multiple myeloma, teclistamab- and talquetamab-mediated multiple myeloma lysis was strongly correlated (r = 0.73, P < 0.0001). Both BsAb exhibited poor activity in samples with high regulatory T-cell (Treg) numbers and a low T-cell/multiple myeloma cell ratio. Furthermore, comprehensive phenotyping of BM samples derived from patients treated with teclistamab or talquetamab revealed that high frequencies of PD-1+ CD4+ T cells, CTLA4+ CD4+ T cells, and CD38+ CD4+ T cells were associated with primary resistance. Although this lack of response was linked to a modest increase in the expression of inhibitory receptors, increasing T-cell/multiple myeloma cell ratios by adding extra T cells enhanced sensitivity to BsAb. Further, treatment with BsAb resulted in an increased proportion of T cells expressing exhaustion markers (PD-1, TIGIT, and TIM-3), which was accompanied by reduced T-cell proliferative potential and cytokine secretion, as well as impaired antitumor efficacy in ex vivo experiments. CONCLUSIONS: Primary resistance is characterized by a low T-cell/multiple myeloma cell ratio and Treg-driven immunosuppression, whereas reduced T-cell fitness due to continuous BsAb-mediated T-cell activation may contribute to the development of acquired resistance.


Sujet(s)
Anticorps bispécifiques , Résistance aux médicaments antinéoplasiques , Myélome multiple , Myélome multiple/immunologie , Myélome multiple/traitement médicamenteux , Myélome multiple/anatomopathologie , Humains , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/usage thérapeutique , Résistance aux médicaments antinéoplasiques/immunologie , Antigène de maturation des cellules B/immunologie , Lymphocytes T régulateurs/immunologie , Femelle , Mâle , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Adulte d'âge moyen , Sujet âgé , Récepteurs couplés aux protéines G
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE