Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.522
Filtrer
1.
Oncoimmunology ; 13(1): 2372118, 2024.
Article de Anglais | MEDLINE | ID: mdl-38939518

RÉSUMÉ

The need for reliable biomarkers to predict clinical benefit from anti-PD1 treatment in metastatic melanoma (MM) patients remains unmet. Several parameters have been considered in the tumor environment or the blood, but none has yet achieved sufficient accuracy for routine clinical practice. Whole blood samples from MM patients receiving second-line anti-PD1 treatment (NCT02626065), collected longitudinally, were analyzed by flow cytometry to assess the immune cell subsets absolute numbers, the expression of immune checkpoints or ligands on T cells and the functionality of innate immune cells and T cells. Clinical response was assessed according to Progression-Free Survival (PFS) status at one-year following initiation of anti-PD1 (responders: PFS > 1 year; non-responders: PFS ≤ 1 year). At baseline, several phenotypic and functional alterations in blood immune cells were observed in MM patients compared to healthy donors, but only the proportion of polyfunctional memory CD4+ T cells was associated with response to anti-PD1. Under treatment, a decreased frequency of HVEM on CD4+ and CD8+ T cells after 3 months of treatment identified responding patients, whereas its receptor BTLA was not modulated. Both reduced proportion of CD69-expressing CD4+ and CD8+ T cells and increased number of polyfunctional blood memory T cells after 3 months of treatment were associated with response to anti-PD1. Of upmost importance, the combination of changes of all these markers accurately discriminated between responding and non-responding patients. These results suggest that drugs targeting HVEM/BTLA pathway may be of interest to improve anti-PD1 efficacy.


Sujet(s)
Mélanome , Récepteur-1 de mort cellulaire programmée , Récepteurs immunologiques , Membre-14 de la superfamille des récepteurs au TNF , Humains , Mélanome/traitement médicamenteux , Mélanome/immunologie , Mélanome/anatomopathologie , Mâle , Récepteurs immunologiques/métabolisme , Femelle , Adulte d'âge moyen , Sujet âgé , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Membre-14 de la superfamille des récepteurs au TNF/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Adulte , Résultat thérapeutique , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme
2.
Commun Biol ; 7(1): 773, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937521

RÉSUMÉ

Distinct Natural Killer (NK)-like CD57+ and PD-1+ CD8+ exhausted-like T cell populations (Tex) have both been linked to beneficial immunotherapy response in autoimmune type 1 diabetes (T1D) patients. The origins and relationships between these cell types are poorly understood. Here we show that while PD-1+ and CD57+ Tex populations are epigenetically similar, CD57+ Tex cells display unique increased chromatin accessibility of inhibitory Killer Cell Immunoglobulin-like Receptor (iKIR) and other NK cell genes. PD-1+ and CD57+ Tex also show reciprocal expression of Inhibitory Receptors (IRs) and iKIRs accompanied by chromatin accessibility of Tcf1 and Tbet transcription factor target sites, respectively. CD57+ Tex show unappreciated gene expression heterogeneity and share clonal relationships with PD-1+ Tex, with these cells differentiating along four interconnected lineage trajectories: Tex-PD-1+, Tex-CD57+, Tex-Branching, and Tex-Fluid. Our findings demonstrate new relationships between Tex-like populations in human autoimmune disease and suggest that modulating common precursor populations may enhance response to autoimmune disease treatment.


Sujet(s)
Lymphocytes T CD8+ , Diabète de type 1 , Cellules tueuses naturelles , Diabète de type 1/immunologie , Diabète de type 1/génétique , Humains , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Antigènes CD57/métabolisme , Lignage cellulaire/génétique , Facteur nucléaire hépatocytaire HNF-1 alpha/génétique , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Femelle , Mâle , Adulte
3.
World J Surg Oncol ; 22(1): 172, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937736

RÉSUMÉ

BACKGROUND: The alteration of the immune microenvironment in the axillary metastatic lymph nodes of luminal A breast cancer patients is still unclear. METHODS: Postsurgical tissues from the enrolled luminal A BCs were divided into five categories: primary BC lesion at stage N0 (PL1), primary BC lesion at stage N1 (PL2), negative axillary lymph node at stage N0 BC (LN1), negative axillary lymph node at stage N1 BC (LN2), and positive axillary lymph node at stage N1 BC (LN3). The frequencies of positive immune markers (CD4, CD8, PD1, PD-L1, T-cell immunoglobulin and mucin domain 3 (TIM3), and forkhead box protein 3 (Foxp3)) in the above tissues were quantified by AKOYA Opal Polaris 7 Color Manual IHC Detection Kit. RESULTS: A total of 50 female patients with luminal A BC were enrolled in this study. Among these patients, 23 had stage N1 disease, and 27 had stage N0 disease. Compared with that in the PL2 subgroup, the frequency of PD-1-positive cells was significantly greater in the PL1 subgroup, whether at the stromal or intratumoral level (P value < 0.05). Both the frequency of CD8 + T cells in LN1 and that in LN2 were significantly greater than that in LN3 (P value < 0.05). The frequency of TIM3 + T cells in LN1 was significantly greater than that in PL1 (P value < 0.05). The frequency of CD8 + TIM3 + T cells was significantly greater in both the LN2 and LN3 groups than in the PL2 group (P value < 0.05). The frequency of CD4 + Foxp3 + T cells was significantly greater in LN1 than in PL1 (P value < 0.05), which was the same for both LN3 and PL2 (P value < 0.05). CONCLUSION: Increased frequencies of CD8 + PD1+, CD8 + TIM3 + and CD4 + Foxp3 + T cells might inhibit the immune microenvironment of axillary metastatic lymph nodes in luminal A breast cancer patients and subsequently promote lymph node metastasis.


Sujet(s)
Aisselle , Tumeurs du sein , Noeuds lymphatiques , Métastase lymphatique , Microenvironnement tumoral , Humains , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Tumeurs du sein/chirurgie , Microenvironnement tumoral/immunologie , Adulte d'âge moyen , Noeuds lymphatiques/anatomopathologie , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/chirurgie , Adulte , Pronostic , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/immunologie , Sujet âgé , Études de suivi , Stadification tumorale , Lymphocytes TIL/immunologie , Lymphocytes TIL/anatomopathologie , Antigène CD274/métabolisme , Antigène CD274/immunologie , Récepteur-1 de mort cellulaire programmée/métabolisme
4.
Cancer Med ; 13(12): e7346, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923758

RÉSUMÉ

OBJECTIVES: Previous studies have demonstrated that obesity may impact the efficacy of anti-PD1 therapy, but the underlying mechanism remains unclear. In this study, our objective was to determine the prognostic value of obesity in patients with oral tongue squamous cell carcinoma (OTSCC) treated with pembrolizumab and establish a subtype based on fatty acid metabolism-related genes (FAMRGs) for immunotherapy. MATERIALS AND METHODS: We enrolled a total of 56 patients with OTSCC who underwent neoadjuvant anti-PD1 therapy. Univariate and multivariate Cox regression analyses, Kaplan-Meier survival analysis, and immunohistochemistry staining were performed. Additionally, we acquired the gene expression profiles of pan-cancer samples and conducted GSEA and KEGG pathway analysis. Moreover, data from TCGA, MSigDB, UALCAN, GEPIA and TIMER were utilized to construct the FAMRGs subtype. RESULTS: Our findings indicate that high Body Mass Index (BMI) was significantly associated with improved PFS (HR = 0.015; 95% CI, 0.001 to 0.477; p = 0.015), potentially attributed to increased infiltration of PD1 + T cells. A total of 91 differentially expressed FAMRGs were identified between the response and non-response groups in pan-cancer patients treated with immunotherapy. Of these, 6 hub FAMRGs (ACSL5, PLA2G2D, PROCA1, IL4I1, UBE2L6 and PSME1) were found to affect PD-1 expression and T cell infiltration in HNSCC, which may impact the efficacy of anti-PD1 therapy. CONCLUSION: This study demonstrates that obesity serves as a robust prognostic predictor for patients with OTSCC undergoing neoadjuvant anti-PD1 therapy. Furthermore, the expression of 6 hub FAMRGs (ACSL5, PLA2G2D, PROCA1, IL4I1, UBE2L6 and PSME1) plays a pivotal role in the context of anti-PD1 therapy and deserves further investigation.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Traitement néoadjuvant , Obésité , Tumeurs de la langue , Humains , Tumeurs de la langue/traitement médicamenteux , Tumeurs de la langue/métabolisme , Tumeurs de la langue/immunologie , Tumeurs de la langue/anatomopathologie , Tumeurs de la langue/mortalité , Tumeurs de la langue/génétique , Femelle , Mâle , Traitement néoadjuvant/méthodes , Obésité/métabolisme , Obésité/complications , Adulte d'âge moyen , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Pronostic , Sujet âgé , 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/mortalité , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/thérapie , Anticorps monoclonaux humanisés/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Adulte , Indice de masse corporelle , Marqueurs biologiques tumoraux/métabolisme
5.
J Immunother Cancer ; 12(6)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38925680

RÉSUMÉ

BACKGROUND: The majority of anti-programmed cell-death 1 (PD-1) monoclonal antibodies (mAbs) use S228P mutation IgG4 as the structural basis to avoid the activation of immune cells or complement. However, little attention has been paid to the Fc-Fc interactions between IgG4 and other IgG Fc fragments that could result in adverse effects. Fc-null IgG1 framework is a potential safer alternative to avoid the undesirable Fc-Fc interactions and Fc receptor binding derived effects observed with IgG4. This study provides a comprehensive evaluation of anti-PD-1 mAbs of these two frameworks. METHODS: Trastuzumab and rituximab (both IgG1), wildtype IgG1 and IgG4 were immobilized on nitrocellulose membranes, coated to microplates and biosensor chips, and bound to tumor cells as targets for Fc-Fc interactions. Wildtype IgG1 and IgG4, anti-PD-1 mAb nivolumab (IgG4 S228P), penpulimab (Fc-null IgG1), and tislelizumab (Fc-null IgG4 S228P-R409K) were assessed for their binding reactions to the immobilized IgG proteins and quantitative kinetic data were obtained. To evaluate the effects of the two anti-PD-1 mAbs on immune responses mediated by trastuzumab and rituximab in the context of combination therapy, we employed classic immune models for antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement dependent cytotoxicity. Tumor-bearing mouse models, both wildtype and humanized, were used for in vivo investigation. Furthermore, we also examined the effects of IgG1 and IgG4 on diverse immune cell populations RESULTS: Experiments demonstrated that wildtype IgG4 and nivolumab bound to immobilized IgG through Fc-Fc interactions, diminishing antibody-dependent cell-mediated cytotoxicity and phagocytosis reactions. Quantitative analysis of kinetic parameters suggests that nivolumab and wildtype IgG4 exhibit comparable binding affinities to immobilized IgG1 in both non-denatured and denatured states. IgG4 exerted inhibitory effects on various immune cell types. Wildtype IgG4 and nivolumab both promoted tumor growth in wildtype mouse models. Conversely, wildtype IgG1, penpulimab, and tislelizumab did not show similar adverse effects. CONCLUSIONS: Fc-null IgG1 represents a safer choice for anti-PD-1 immunotherapies by avoiding both the adverse Fc-Fc interactions and Fc-related immune inhibitory effects of IgG4. Fc-null IgG4 S228P-R409K and Fc-null IgG1 displayed similar structural properties and benefits. This study contributes to the understanding of immunotherapy resistance and the advancement of safer immune therapies for cancer.


Sujet(s)
Immunoglobuline G , Immunothérapie , Immunoglobuline G/immunologie , Animaux , Souris , Humains , Immunothérapie/méthodes , Fragments Fc des immunoglobulines/pharmacologie , Femelle , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme
6.
Nature ; 630(8018): 968-975, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38867043

RÉSUMÉ

Obesity is a leading risk factor for progression and metastasis of many cancers1,2, yet can in some cases enhance survival3-5 and responses to immune checkpoint blockade therapies, including anti-PD-1, which targets PD-1 (encoded by PDCD1), an inhibitory receptor expressed on immune cells6-8. Although obesity promotes chronic inflammation, the role of the immune system in the obesity-cancer connection and immunotherapy remains unclear. It has been shown that in addition to T cells, macrophages can express PD-19-12. Here we found that obesity selectively induced PD-1 expression on tumour-associated macrophages (TAMs). Type I inflammatory cytokines and molecules linked to obesity, including interferon-γ, tumour necrosis factor, leptin, insulin and palmitate, induced macrophage PD-1 expression in an mTORC1- and glycolysis-dependent manner. PD-1 then provided negative feedback to TAMs that suppressed glycolysis, phagocytosis and T cell stimulatory potential. Conversely, PD-1 blockade increased the level of macrophage glycolysis, which was essential for PD-1 inhibition to augment TAM expression of CD86 and major histocompatibility complex I and II molecules and ability to activate T cells. Myeloid-specific PD-1 deficiency slowed tumour growth, enhanced TAM glycolysis and antigen-presentation capability, and led to increased CD8+ T cell activity with a reduced level of markers of exhaustion. These findings show that obesity-associated metabolic signalling and inflammatory cues cause TAMs to induce PD-1 expression, which then drives a TAM-specific feedback mechanism that impairs tumour immune surveillance. This may contribute to increased cancer risk yet improved response to PD-1 immunotherapy in obesity.


Sujet(s)
Glycolyse , Obésité , Récepteur-1 de mort cellulaire programmée , Macrophages associés aux tumeurs , Animaux , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Obésité/immunologie , Souris , Glycolyse/effets des médicaments et des substances chimiques , Mâle , Femelle , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Souris de lignée C57BL , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Tumeurs/immunologie , Tumeurs/anatomopathologie , Humains , Lignée cellulaire tumorale , Phagocytose/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Lymphocytes T/métabolisme
7.
Oncoimmunology ; 13(1): 2371051, 2024.
Article de Anglais | MEDLINE | ID: mdl-38915783

RÉSUMÉ

Improving cancer immunotherapy efficacy hinges on identifying key T-cell populations critical for tumor control and response to Immune Checkpoint Blockade (ICB). We have recently reported that while the co-expression of PD-1 and CD28 is associated with impaired functionality in peripheral blood, it significantly enhances T-cell fitness in the tumor site of non-small cell lung cancer (NSCLC) patients. To uncover the underlying mechanisms, we explored the role of CD26, a key player in T-cell activation through its interaction with adenosine deaminase (ADA), a crucial intra/extracellular enzyme able to neutralize local adenosine (ADO). We found that an autocrine ADA/CD26 axis enhances CD8+PD-1+CD28+ T-cell function, particularly within an immunosuppressive environment marked by CD39 expression. Then, we interrogated the TCGA and OAK datasets to gain insight into the prognostic/predictive potential of our findings. We identified a signature predicting overall survival (OS) in LUAD patients and response to atezolizumab in advanced LUAD cases. These findings suggest promising avenues for therapeutic intervention targeting the ADA/CD26 axis.


Sujet(s)
Adenosine deaminase , Antigène CD28 , Lymphocytes T CD8+ , Carcinome pulmonaire non à petites cellules , Dipeptidyl peptidase 4 , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Récepteur-1 de mort cellulaire programmée , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Antigène CD28/métabolisme , Pronostic , Récepteur-1 de mort cellulaire programmée/métabolisme , Dipeptidyl peptidase 4/métabolisme , Dipeptidyl peptidase 4/génétique , Adenosine deaminase/métabolisme , Adenosine deaminase/génétique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/administration et posologie , Femelle , Mâle , Apyrase/métabolisme
8.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38928437

RÉSUMÉ

Multiple sclerosis (MS) onset at an advanced age is associated with a higher risk of developing progressive forms and a greater accumulation of disability for which there are currently no effective disease-modifying treatments. Immunosenescence is associated with the production of the senescence-associated secretory phenotype (SASP), with IL-6 being one of the most prominent cytokines. IL-6 is a determinant for the development of autoimmunity and neuroinflammation and is involved in the pathogenesis of MS. Herein, we aimed to preclinically test the therapeutic inhibition of IL-6 signaling in experimental autoimmune encephalomyelitis (EAE) as a potential age-specific treatment for elderly MS patients. Young and aged mice were immunized with myelin oligodendrocyte protein (MOG)35-55 and examined daily for neurological signs. Mice were randomized and treated with anti-IL-6 antibody. Inflammatory infiltration was evaluated in the spinal cord and the peripheral immune response was studied. The blockade of IL-6 signaling did not improve the clinical course of EAE in an aging context. However, IL-6 inhibition was associated with an increase in the peripheral immunosuppressive response as follows: a higher frequency of CD4 T cells producing IL-10, and increased frequency of inhibitory immune check points PD-1 and Tim-3 on CD4+ T cells and Lag-3 and Tim-3 on CD8+ T cells. Our results open the window to further studies aimed to adjust the anti-IL-6 treatment conditions to tailor an effective age-specific therapy for elderly MS patients.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Interleukine-6 , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/métabolisme , Animaux , Souris , Interleukine-6/métabolisme , Interleukine-6/antagonistes et inhibiteurs , Femelle , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Souris de lignée C57BL , Glycoprotéine MOG/immunologie , Sclérose en plaques/traitement médicamenteux , Sclérose en plaques/immunologie , Sclérose en plaques/métabolisme , Vieillissement/immunologie , Interleukine-10/métabolisme , Moelle spinale/métabolisme , Moelle spinale/anatomopathologie , Moelle spinale/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/immunologie , Transduction du signal/effets des médicaments et des substances chimiques
9.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38928456

RÉSUMÉ

The management of advanced bladder carcinoma involves a multidisciplinary approach, but the prognosis remains poor for many patients. The immune system plays a crucial role in this disease, influencing both tumor development and response to treatment, and exploiting the immune system against the tumor can be a valuable strategy to destroy neoplastic cells. This is the biological principle underlying Bacillus Calmette-Guérin (BCG) use and, more recently, immune checkpoint inhibitors (ICIs), like PD-1 (programmed death-1)/PD-L1 (programmed death-ligand 1) inhibitors. In fact, one of the best studied immune checkpoints is represented by the PD-1/PD-L1 axis, which is a well-known immune escape system adopted by neoplastic bladder cells. PD-L1 expression has been associated with a higher pathologic stage and has shown prognostic value in bladder carcinoma. Interestingly, high-grade bladder cancers tend to express higher levels of PD-1 and PD-L1, suggesting a potential role of such an axis in mediating disease progression. Immunotherapy with PD-1 and PD-L1 inhibitors has therefore emerged as a valuable treatment option and has shown efficacy in advanced bladder cancer patients, with high PD-L1 expression levels associated with better treatment responses. Our review aims to provide a comprehensive overview of the role of PD-L1 in advanced bladder cancer, focusing on its implications for treatment decisions and the prediction of treatment response. Overall, our work aims to contribute to the understanding of PD-L1 as a predictive biomarker and highlight its role in shaping therapeutic approaches for advanced bladder cancer.


Sujet(s)
Antigène CD274 , Tumeurs de la vessie urinaire , Humains , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Antigène CD274/métabolisme , Antigène CD274/génétique , Pronostic , Marqueurs biologiques tumoraux/métabolisme , Immunohistochimie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs
10.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928479

RÉSUMÉ

Endometriosis is a chronic inflammatory disease characterized by the presence of endometrial-like tissue outside the uterine cavity, causing pain and infertility. Despite the rather unclear etiopathogenesis, recent studies suggest the involvement of the immune system in the development and progression of endometriosis. The role of the PD-1/PD-L1 axis in the modulation of the immune response in this disease seems to be particularly interesting. This preliminary study aimed to investigate the expression of PD-1 and PD-L1 on T and B lymphocytes in peripheral blood in patients with endometriosis to assess their potential impact on disease progression. Our study involved peripheral blood samples from 80 patients diagnosed with endometriosis and 20 healthy women as a control group were analyzed. Flow cytometry was used to assess the expression of PD-1 and PD-L1 on T and B lymphocytes, and enzyme-linked immunosorbent assays were used to assess their soluble forms in serum and peritoneal fluid.in our research we observe significantly higher expression of PD-1 and PD-L1 on T and B lymphocytes was found in patients with endometriosis compared to the control group. Higher expression of both tested molecules correlated with the stage of endometriosis. The results of our preliminary studies indicate a potential role of the PD-1/PD-L1 axis in the modulation of the immune response in endometriosis. Modified expression of these proteins may contribute to immune evasion by ectopic tissues, supporting their survival and proliferation. These findings suggest that targeting PD-1/PD-L1 could be explored as a therapeutic option for the treatment of endometriosis, though further research with larger sample sizes is necessary to confirm these results and clarify the role of PD-1/PD-L1 in the pathogenesis of the disease.


Sujet(s)
Lymphocytes B , Antigène CD274 , Endométriose , Récepteur-1 de mort cellulaire programmée , Humains , Endométriose/métabolisme , Endométriose/immunologie , Endométriose/anatomopathologie , Femelle , Antigène CD274/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Adulte , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme
11.
Nanoscale ; 16(25): 12149-12162, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38833269

RÉSUMÉ

Together, tumor and virus-specific tissue-resident CD8+ memory T cells (TRMs) of hepatocellular carcinoma (HCC) patients with Hepatitis B virus (HBV) infection can provide rapid frontline immune surveillance. The quantity and activity of CD8+ TRMs were correlated with the relapse-free survival of patients with improved health. However, HBV-specific CD8+ TRMs have a more exhausted phenotype and respond more actively under anti-PDL1 or PD1 treatment of HBV+HCC patients. Vaccination strategies that induce a strong and sustained CD8+ TRMs response are quite promising. Herein, a biodegradable poly(D,L-lactide-co-glycolide) microsphere and nanosphere particle (PLGA N.M.P) delivery system co-assembled by anti-PD1 antibodies (aPD1) and loaded with ovalbumin (OVA-aPD1 N.M.P) was fabricated and characterized for size (200 nm and 1 µm diameter), charge (-15 mV), and loading efficiencies of OVA (238 µg mg-1 particles) and aPD1 (40 µg mg-1 particles). OVA-aPD1 N.M.P could stimulate the maturation of BMDCs and enhance the antigen uptake and presentation by 2-fold compared to free OVA. The nanoparticles also induced the activation of macrophages (RAW 264.7) to produce a high level of cytokines, including TNF-α, IL-6 and IL-10. In vivo stimulation of mice using OVA-aPD1 N.M.P robustly enhanced IFN-γ-producing-CD8+ T cell infiltration in tumor tissues and the secretion of IgG and IgG2a/IgG1 antibodies. OVA-aPD1 N.M.P delivered OVA to increase the activation and proliferation of OVA-specific CD8+ TRMs, and its combination with anti-PD1 antibodies promoted complete tumor rejection by the reversal of tumor-infiltrating CD8+ T cell exhaustion. Thus, PLGA N.M.P could induce a strong CD8+ TRMs response, further highlighting its therapeutic potential in enhancing an antitumor immune response.


Sujet(s)
Lymphocytes T CD8+ , Carcinome hépatocellulaire , Tumeurs du foie , Copolymère d'acide poly(lactique-co-glycolique) , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/thérapie , Souris , Tumeurs du foie/immunologie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/thérapie , Souris de lignée C57BL , Ovalbumine/immunologie , Ovalbumine/composition chimique , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Nanoparticules/composition chimique , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/composition chimique , Cellules T mémoire/immunologie , Vaccination , Humains , Cellules RAW 264.7 , Mémoire immunologique
12.
Cancer Immunol Immunother ; 73(8): 147, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38833156

RÉSUMÉ

BACKGROUND: This study aimed to investigate the relationship between signal regulatory protein gamma (SIRPG) and tumor immune microenvironment phenotypes or T cell mediated-adaptive antitumor immunity, and its predictive value for response to PD-1 blockade in cancers. METHODS: Pan-cancer analysis of SIRPG expression and immune deconvolution was performed using transcriptomic data across 33 tumor types. Transcriptomic and clinical data from 157 patients with non-small-cell lung cancer (NSCLC) and melanoma received PD-1 blockade were analyzed. Expression characteristics of SIRPG were investigated using single-cell RNA sequencing (scRNA-seq) data of 103,599 cells. The effect of SIRPG expression was evaluated via SIRPG knockdown or overexpression in Jurkat T cells. RESULTS: The results showed that most cancers with high SIRPG expression had significantly higher abundance of T cells, B cells, NK cells, M1 macrophages and cytotoxic lymphocytes and increased expression level of immunomodulatory factors regulating immune cell recruitment, antigen presentation, T cell activation and cytotoxicity, but markedly lower abundance of neutrophils, M2 macrophages, and myeloid-derived suppressor cells. High SIRPG expression was associated with favorable response to PD-1 blockade in both NSCLC and melanoma. scRNA-seq data suggested SIRPG was mainly expressed in CD8+ exhausted T and CD4+ regulatory T cells, and positively associated with immune checkpoint expression including PDCD1 and CTLA4. In vitro test showed SIRPG expression in T cells could facilitate expression of PDCD1 and CTLA4. CONCLUSION: High SIRPG expression is associated with an inflamed immune phenotype in cancers and favorable response to PD-1 blockade, suggesting it would be a promising predictive biomarker for PD-1 blockade and novel immunotherapeutic target.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Marqueurs biologiques tumoraux/métabolisme , Mélanome/immunologie , Mélanome/métabolisme , Mélanome/génétique
13.
Molecules ; 29(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38893521

RÉSUMÉ

The PD-1/PD-L1 complex is an immune checkpoint responsible for regulating the natural immune response, but also allows tumors to escape immune surveillance. Inhibition of the PD-1/PD-L1 axis positively contributes to the efficacy of cancer treatment. The only available therapeutics targeting PD-1/PD-L1 are monoclonal antibody-based drugs, which have several limitations. Therefore, small molecule compounds are emerging as an attractive alternative that can potentially overcome the drawbacks of mAb-based therapy. In this article, we present a novel class of small molecule compounds based on the terphenyl scaffold that bind to PD-L1. The general architecture of the presented structures is characterized by axial symmetry and consists of three elements: an m-terphenyl core, an additional aromatic ring, and a solubilizing agent. Using molecular docking, we designed a series of final compounds, which were subsequently synthesized and tested in HTRF assay and NMR binding assay to evaluate their activity. In addition, we performed an in-depth analysis of the mutual arrangement of the phenyl rings of the terphenyl core within the binding pocket of PD-L1 and found several correlations between the plane angle values and the affinity of the compounds towards the protein.


Sujet(s)
Antigène CD274 , Simulation de docking moléculaire , Récepteur-1 de mort cellulaire programmée , Liaison aux protéines , Dérivés du terphényle , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Antigène CD274/composition chimique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/composition chimique , Humains , Dérivés du terphényle/composition chimique , Dérivés du terphényle/pharmacologie , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Inhibiteurs de points de contrôle immunitaires/composition chimique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Structure moléculaire , Relation structure-activité , Sites de fixation
14.
J Cell Mol Med ; 28(12): e18404, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38888489

RÉSUMÉ

In patients with nasopharyngeal carcinoma (NPC), the alteration of immune responses in peripheral blood remains unclear. In this study, we established an immune cell profile for patients with NPC and used flow cytometry and machine learning (ML) to identify the characteristics of this profile. After isolation of circulating leukocytes, the proportions of 104 immune cell subsets were compared between NPC group and the healthy control group (HC). Data obtained from the immune cell profile were subjected to ML training to differentiate between the immune cell profiles of the NPC and HC groups. We observed that subjects in the NPC group presented higher proportions of T cells, memory B cells, short-lived plasma cells, IgG-positive B cells, regulatory T cells, MHC II+ T cells, CTLA4+ T cells and PD-1+ T cells than subjects in the HC group, indicating weaker and compromised cellular and humoral immune responses. ML revealed that monocytes, PD-1+ CD4 T cells, memory B cells, CTLA4+ CD4 Treg cells and PD-1+ CD8 T cells were strongly contributed to the difference in immune cell profiles between the NPC and HC groups. This alteration can be fundamental in developing novel immunotherapies for NPC.


Sujet(s)
Cytométrie en flux , Apprentissage machine , Cancer du nasopharynx , Tumeurs du rhinopharynx , Humains , Cancer du nasopharynx/immunologie , Cancer du nasopharynx/anatomopathologie , Cytométrie en flux/méthodes , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du rhinopharynx/immunologie , Tumeurs du rhinopharynx/anatomopathologie , Adulte , Récepteur-1 de mort cellulaire programmée/métabolisme , Lymphocytes T CD8+/immunologie , Études cas-témoins , Sujet âgé
15.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892153

RÉSUMÉ

The role of programmed death cell protein 1 (PD-1) has already been described in a range of various diseases, including COVID-19. This study provides new, innovative data, related to the expression of PD-1 and the risk of Paediatric Inflammatory Multisystem Syndrome, temporally associated with SARS-CoV-2 infection (PIMS-TS)-a rare, but potentially life-threatening complication of COVID-19. In this study, we evaluated the expression of PD-1 protein in patients with PIMS. Blood samples were taken from patients at the time of diagnosis (n = 33), after 6 weeks (n = 33), 3 months (n = 24), 6 months (n = 24) and 12 months (n = 8). The immunophenotypes were evaluated in flow cytometry. The control group consisted of 35 healthy children with negative SARS-CoV-2 antigen/PCR test, who were asymptomatic and had no history of allergic, autoimmune or oncological diseases. The associations between immunophenotypes, biochemical findings and clinical data were analysed. Significant increases in the expression of PD-1 for CD4+ and CD8+ T cells, compared to the control group, were observed in the day of admission, with a gradual decrease during the first weeks from initiation of treatment. This study sheds new light on the pathogenesis of PIMS-TS, emphasizing the role of PD-1 protein. Future research is essential for early risk prediction in SARS-CoV-2 patients and for devising effective clinical prevention and management strategies.


Sujet(s)
COVID-19 , Récepteur-1 de mort cellulaire programmée , SARS-CoV-2 , Syndrome de réponse inflammatoire généralisée , Humains , COVID-19/complications , COVID-19/immunologie , COVID-19/sang , COVID-19/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Syndrome de réponse inflammatoire généralisée/sang , Syndrome de réponse inflammatoire généralisée/diagnostic , Mâle , Enfant , Femelle , Enfant d'âge préscolaire , Études prospectives , Adolescent , Nourrisson , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Immunophénotypage
16.
Immunobiology ; 229(4): 152824, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38875763

RÉSUMÉ

Recent evidence has shown that T cell exhaustion is implicated in Allergen-specific Immunotherapy (AIT). However, how T cell exhaustion plays a role in AIT is far from clear. Our study aimed to investigate T cell exhaustion associated with allergen exposure during AIT in mice. Ovalbumin (OVA) - sensitized C57BL/6J asthma mouse and AIT mouse models were constructed. Quantitative real-time PCR (qRTPCR) and flow cytometry were used to monitor the occurrence of local and systemic CD4+ T cells and Th2+T cells exhaustion in OVA-sensitized mice. The inhibitory surface marker programmed cell death protein 1 (PD-1) on CD4+ T cells and Th2+T cells was significantly upregulated in AIT mice compared with asthmatic and control mice. The level of PD-1 on the surface of CD4+T cells of asthma mice was significantly higher than that of control mice. The inhibitory surface marker cytotoxic T lymphocyte-associated protein 4 (CTLA-4) on CD4+ T cells and Th2+T cells showed no significant difference between the AIT, asthma and control mice. Collectively, our study indicated that the expression of PD-1 on CD4+ T cells and Th2+T cells was increased in AIT. Allergen exposure promotes the expression of PD-1 on the surface of CD4+ T cells. T cell exhaustion plays an important role in AIT.


Sujet(s)
Allergènes , Asthme , Lymphocytes T CD4+ , Désensibilisation immunologique , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée , Lymphocytes auxiliaires Th2 , Animaux , Récepteur-1 de mort cellulaire programmée/métabolisme , Souris , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Asthme/immunologie , Asthme/thérapie , Allergènes/immunologie , Désensibilisation immunologique/méthodes , Lymphocytes auxiliaires Th2/immunologie , Modèles animaux de maladie humaine , Femelle , Marqueurs biologiques , Ovalbumine/immunologie
17.
J Chem Inf Model ; 64(12): 4877-4896, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38856697

RÉSUMÉ

Programmed death-ligand 1 (PD-L1) has emerged as a promising therapeutic target for various cancers due to its crucial role in promoting tumor immune evasion. Here, we report a novel class of chroman-like small-molecule PD-L1 inhibitors exhibiting significant activity in inhibiting the PD-1/PD-L1 interaction. Employing a "ring-close" strategy for conformational restriction, we have achieved compound C27, which demonstrates superior PD-1/PD-L1 inhibitory activity compared to the positive control. Molecular dynamics simulation and binding free energy calculation predict that (R)-C27 with inhibitory activity surpassed (S)-C27. The experimental results from bioassay and X-ray structural analysis corroborate these findings. All these results collectively indicate that (R)-C27 is a promising lead compound deserving further exploration.


Sujet(s)
Antigène CD274 , Chromanes , Conception de médicament , Récepteur-1 de mort cellulaire programmée , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Humains , Chromanes/composition chimique , Chromanes/pharmacologie , Simulation de dynamique moléculaire
18.
Cancer Cell ; 42(6): 1051-1066.e7, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38861924

RÉSUMÉ

PD-1 blockade unleashes potent antitumor activity in CD8+ T cells but can also promote immunosuppressive T regulatory (Treg) cells, which may worsen the response to immunotherapy. Tumor-Treg inhibition is a promising strategy to improve the efficacy of checkpoint blockade immunotherapy; however, our understanding of the mechanisms supporting tumor-Tregs during PD-1 immunotherapy is incomplete. Here, we show that PD-1 blockade increases tumor-Tregs in mouse models of melanoma and metastatic melanoma patients. Mechanistically, Treg accumulation is not caused by Treg-intrinsic inhibition of PD-1 signaling but depends on an indirect effect of activated CD8+ T cells. CD8+ T cells produce IL-2 and colocalize with Tregs in mouse and human melanomas. IL-2 upregulates the anti-apoptotic protein ICOS on tumor-Tregs, promoting their accumulation. Inhibition of ICOS signaling before PD-1 immunotherapy improves control over immunogenic melanoma. Thus, interrupting the intratumor CD8+ T cell:Treg crosstalk represents a strategy to enhance the therapeutic efficacy of PD-1 immunotherapy.


Sujet(s)
Lymphocytes T CD8+ , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Protéine inductible de costimulation du lymphocyte T , Interleukine-2 , Mélanome , Récepteur-1 de mort cellulaire programmée , Lymphocytes T régulateurs , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T régulateurs/immunologie , Humains , Souris , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Mélanome/immunologie , Mélanome/thérapie , Mélanome/traitement médicamenteux , Protéine inductible de costimulation du lymphocyte T/métabolisme , Immunothérapie/méthodes , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Interleukine-2/immunologie , Souris de lignée C57BL , Transduction du signal , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Lignée cellulaire tumorale
19.
Nat Commun ; 15(1): 5280, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902261

RÉSUMÉ

The regulatory circuits dictating CD8+ T cell responsiveness versus exhaustion during anti-tumor immunity are incompletely understood. Here we report that tumor-infiltrating antigen-specific PD-1+ TCF-1- CD8+ T cells express the immunosuppressive cytokine Fgl2. Conditional deletion of Fgl2 specifically in mouse antigen-specific CD8+ T cells prolongs CD8+ T cell persistence, suppresses phenotypic and transcriptomic signatures of T cell exhaustion, and improves control of the tumor. In a mouse model of chronic viral infection, PD-1+ CD8+ T cell-derived Fgl2 also negatively regulates virus-specific T cell responses. In humans, CD8+ T cell-derived Fgl2 is associated with poorer survival in patients with melanoma. Mechanistically, the dampened responsiveness of WT Fgl2-expressing CD8+ T cells, when compared to Fgl2-deficient CD8+ T cells, is underpinned by the cell-intrinsic interaction of Fgl2 with CD8+ T cell-expressed FcγRIIB and concomitant caspase 3/7-mediated apoptosis. Our results thus illuminate a cell-autonomous regulatory axis by which PD-1+ CD8+ T cells both express the receptor and secrete its ligand in order to mediate suppression of anti-tumor and anti-viral immunity.


Sujet(s)
Lymphocytes T CD8+ , Mélanome , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée , Récepteurs du fragment Fc des IgG , Animaux , Lymphocytes T CD8+/immunologie , Souris , Humains , Récepteurs du fragment Fc des IgG/métabolisme , Récepteurs du fragment Fc des IgG/génétique , Récepteurs du fragment Fc des IgG/immunologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Mélanome/immunologie , Mélanome/génétique , Souris knockout , Apoptose , Femelle
20.
Sci Rep ; 14(1): 14248, 2024 06 20.
Article de Anglais | MEDLINE | ID: mdl-38902343

RÉSUMÉ

Treatment of advanced triple-negative breast cancer (TNBC) is a great challenge in clinical practice. The immune checkpoints are a category of immunosuppressive molecules that cancer could hijack and impede anti-tumor immunity. Targeting immune checkpoints, such as anti-programmed cell death 1 (PD-1) therapy, is a promising therapeutic strategy in TNBC. The efficacy and safety of PD-1 monoclonal antibody (mAb) with chemotherapy have been validated in TNBC patients. However, the precise mechanisms underlying the synergistic effect of chemotherapy and anti-PD-1 therapy have not been elucidated, causing the TNBC patients that might benefit from this combination regimen not to be well selected. In the present work, we found that IL-23, an immunological cytokine, is significantly upregulated after chemotherapy in TNBC cells and plays a vital role in enhancing the anti-tumor immune response of cytotoxic T cells (CTLs), especially in combination with PD-1 mAb. In addition, the combination of IL-23 and PD-1 mAb could synergistically inhibit the expression of Phosphoinositide-3-Kinase Regulatory Subunit 1 (PIK3R1), which is a regulatory subunit of PI3K and inhibit p110 activity, and promote phosphorylation of AKT in TNBC-specific CTLs. Our findings might provide a molecular marker that could be used to predict the effects of combination chemotherapy therapy and PD-1 mAb in TNBC.


Sujet(s)
Sous-unité p19 de l'interleukine-23 , Phosphatidylinositol 3-kinases , Récepteur-1 de mort cellulaire programmée , Protéines proto-oncogènes c-akt , Transduction du signal , Lymphocytes T cytotoxiques , Tumeurs du sein triple-négatives , Humains , Protéines proto-oncogènes c-akt/métabolisme , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Lignée cellulaire tumorale , Femelle , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/métabolisme , Sous-unité p19 de l'interleukine-23/métabolisme , Animaux , Souris , Anticorps monoclonaux/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...