Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 115
Filtrer
1.
Cell Rep Med ; 5(7): 101640, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38959885

RÉSUMÉ

CD8+ T cells must persist and function in diverse tumor microenvironments to exert their effects. Thus, understanding common underlying expression programs could better inform the next generation of immunotherapies. We apply a generalizable matrix factorization algorithm that recovers both shared and context-specific expression programs from diverse datasets to a single-cell RNA sequencing (scRNA-seq) compendium of 33,161 CD8+ T cells from 132 patients with seven human cancers. Our meta-single-cell analyses uncover a pan-cancer T cell dysfunction program that predicts clinical non-response to checkpoint blockade in melanoma and highlights CXCR6 as a pan-cancer marker of chronically activated T cells. Cxcr6 is trans-activated by AP-1 and repressed by TCF1. Using mouse models, we show that Cxcr6 deletion in CD8+ T cells increases apoptosis of PD1+TIM3+ cells, dampens CD28 signaling, and compromises tumor growth control. Our study uncovers a TCF1:CXCR6 axis that counterbalances PD1-mediated suppression of CD8+ cell responses and is essential for effective anti-tumor immunity.


Sujet(s)
Antigène CD28 , Lymphocytes T CD8+ , Facteur nucléaire hépatocytaire HNF-1 alpha , Récepteurs CXCR6 , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Animaux , Humains , Antigène CD28/métabolisme , Antigène CD28/génétique , Antigène CD28/immunologie , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Facteur nucléaire hépatocytaire HNF-1 alpha/génétique , Souris , Récepteurs CXCR6/métabolisme , Récepteurs CXCR6/génétique , Tumeurs/immunologie , Tumeurs/génétique , Tumeurs/anatomopathologie , Analyse sur cellule unique/méthodes , Transduction du signal , Microenvironnement tumoral/immunologie , Souris de lignée C57BL
2.
Cancer Biol Med ; 21(6)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38825813

RÉSUMÉ

In exploring persistent infections and malignancies, a distinctive subgroup of CD8+ T cells, progenitor exhausted CD8+ T (Tpex) cells, has been identified. These Tpex cells are notable for their remarkable self-renewal and rapid proliferation abilities. Recent strides in immunotherapy have demonstrated that Tpex cells expand and differentiate into responsive exhausted CD8+ T cells, thus underscoring their critical role in the immunotherapeutic retort. Clinical examinations have further clarified a robust positive correlation between the proportional abundance of Tpex cells and enhanced clinical prognosis. Tpex cells have found noteworthy applications in the formulation of inventive immunotherapeutic approaches against tumors. This review describes the functions of Tpex cells in the tumor milieu, particularly their potential utility in tumor immunotherapy. Precisely directing Tpex cells may be essential to achieving successful outcomes in immunotherapy against tumors.


Sujet(s)
Lymphocytes T CD8+ , Immunothérapie , Tumeurs , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Lymphocytes T CD8+/immunologie , Animaux , Microenvironnement tumoral/immunologie
3.
Cancer Sci ; 115(7): 2184-2195, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38590234

RÉSUMÉ

Recent studies have highlighted the pivotal roles of T cell transcription factors TCF-1 and TOX in modulating the immune response in cancer, with TCF-1 maintaining CD8+ T cell stemness and TOX promoting T cell exhaustion. The prognostic significance of these factors in lung adenocarcinoma (LUAD) remains a critical area of investigation. The retrospective study included 191 patients with LUAD who underwent surgery, of whom 83% were in stages II and III. These patients were divided into exploratory (n = 135) and validation (n = 56) groups based on the time of diagnosis. Multiplex fluorescence immunohistochemistry was used to examine the infiltration levels of CD8+ T cells, TCF1+ CD8+ T cells, and TOX+ CD8+ T cells. The percentage of CD8+ T cells in tumor was markedly lower than that in stroma (p < 0.05). In tumor-draining lymph nodes (TDLNs) invaded by tumor, the proportion of stem-like TCF1+ CD8+ T cells was significantly decreased (p < 0.01). Importantly, higher infiltration levels of CD8+ T cells and TCF1+ CD8+ T cells were associated with improved disease-free survival (DFS) (p = 0.009 and p = 0.006, respectively) and overall survival (OS) (p = 0.018 and p = 0.010, respectively). This study underscores the potential of TCF1+ CD8+ T cells as prognostic biomarkers in LUAD, providing insights into the tumor immune microenvironment and guiding future therapeutic strategies.


Sujet(s)
Adénocarcinome pulmonaire , Lymphocytes T CD8+ , Facteur nucléaire hépatocytaire HNF-1 alpha , Tumeurs du poumon , Lymphocytes TIL , Humains , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/mortalité , Femelle , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Pronostic , Mâle , Adulte d'âge moyen , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Facteur nucléaire hépatocytaire HNF-1 alpha/génétique , Sujet âgé , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Études rétrospectives , Microenvironnement tumoral/immunologie , Survie sans rechute , Protéines HMG/métabolisme , Transactivateurs
4.
Res Sq ; 2024 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-38496632

RÉSUMÉ

Radiotherapy (RT) and anti-PD-L1 synergize to enhance local and distant (abscopal) tumor control. However, clinical results in humans have been variable. With the goal of improving clinical outcomes, we investigated the underlying synergistic mechanism focusing on a CD8+ PD-1+ Tcf-1+ stem-like T cell subset in the tumor-draining lymph node (TdLN). Using murine melanoma models, we found that RT + anti-PD-L1 induces a novel differentiation program in the TdLN stem-like population which leads to their expansion and differentiation into effector cells within the tumor. Our data indicate that optimal synergy between RT + anti-PD-L1 is dependent on the TdLN stem-like T cell population as either blockade of TdLN egress or specific stem-like T cell depletion reduced tumor control. Together, these data demonstrate a multistep stimulation of stem-like T cells following combination therapy which is initiated in the TdLN and completed in the tumor.

5.
Cell Rep ; 43(3): 113898, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38451819

RÉSUMÉ

T cell exhaustion impairs tumor immunity and contributes to resistance against immune checkpoint inhibitors. The nuclear receptor subfamily 4 group A (NR4a) family of nuclear receptors plays a crucial role in driving T cell exhaustion. In this study, we observe that NR4a1 and NR4a2 deficiency in CD8+ tumor-infiltrating lymphocytes (TILs) results in potent tumor eradication and exhibits not only reduced exhaustion characteristics but also an increase in the precursors/progenitors of exhausted T (Pre-Tex) cell fraction. Serial transfers of NR4a1-/-NR4a2-/-CD8+ TILs into tumor-bearing mice result in the expansion of TCF1+ (Tcf7+) stem-like Pre-Tex cells, whereas wild-type TILs are depleted upon secondary transfer. NR4a1/2-deficient CD8+ T cells express higher levels of stemness/memory-related genes and illustrate potent mitochondrial oxidative phosphorylation. Collectively, these findings suggest that inhibiting NR4a in tumors represents a potent immuno-oncotherapy strategy by increasing stem-like Pre-Tex cells and reducing exhaustion of CD8+ T cells.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs , Animaux , Souris , Lymphocytes TIL , Tumeurs/génétique , Microenvironnement tumoral
6.
Cell ; 187(7): 1651-1665.e21, 2024 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-38490195

RÉSUMÉ

The immune checkpoint blockade (ICB) response in human cancers is closely linked to the gut microbiota. Here, we report that the abundance of commensal Lactobacillus johnsonii is positively correlated with the responsiveness of ICB. Supplementation with Lactobacillus johnsonii or tryptophan-derived metabolite indole-3-propionic acid (IPA) enhances the efficacy of CD8+ T cell-mediated αPD-1 immunotherapy. Mechanistically, Lactobacillus johnsonii collaborates with Clostridium sporogenes to produce IPA. IPA modulates the stemness program of CD8+ T cells and facilitates the generation of progenitor exhausted CD8+ T cells (Tpex) by increasing H3K27 acetylation at the super-enhancer region of Tcf7. IPA improves ICB responsiveness at the pan-cancer level, including melanoma, breast cancer, and colorectal cancer. Collectively, our findings identify a microbial metabolite-immune regulatory pathway and suggest a potential microbial-based adjuvant approach to improve the responsiveness of immunotherapy.


Sujet(s)
Lymphocytes T CD8+ , Immunothérapie , Lactobacillus , Tumeurs , Humains , Lactobacillus/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie , Indoles/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique
7.
Cell Rep ; 43(2): 113797, 2024 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-38363680

RÉSUMÉ

Immune checkpoint inhibitors exert clinical efficacy against various types of cancer through reinvigoration of exhausted CD8+ T cells that attack cancer cells directly in the tumor microenvironment (TME). Using single-cell sequencing and mouse models, we show that CXCL13, highly expressed in tumor-infiltrating exhausted CD8+ T cells, induces CD4+ follicular helper T (TFH) cell infiltration, contributing to anti-tumor immunity. Furthermore, a part of the TFH cells in the TME exhibits cytotoxicity and directly attacks major histocompatibility complex-II-expressing tumors. TFH-like cytotoxic CD4+ T cells have high LAG-3/BLIMP1 and low TCF1 expression without self-renewal ability, whereas non-cytotoxic TFH cells express low LAG-3/BLIMP1 and high TCF1 with self-renewal ability, closely resembling the relationship between terminally differentiated and stem-like progenitor exhaustion in CD8+ T cells, respectively. Our findings provide deep insights into TFH-like CD4+ T cell exhaustion with helper progenitor and cytotoxic differentiated functions, mediating anti-tumor immunity orchestrally with CD8+ T cells.


Sujet(s)
Épuisement des cellules T , Microenvironnement tumoral , Animaux , Souris , Lymphocytes T CD8+ , Différenciation cellulaire , Lymphocytes T CD4+
8.
Clin Epigenetics ; 16(1): 21, 2024 02 06.
Article de Anglais | MEDLINE | ID: mdl-38321550

RÉSUMÉ

BACKGROUND: Epigenetic remodeling at effector gene loci has been reported to be critical in regulating T cell differentiation and function. However, efforts to investigate underlying epigenetic mechanisms that control T cell behaviors have been largely hindered by very limited experimental tools, especially in humans. RESULTS: In this study, we employed a flow cytometric assay to analyze histone acetylation at single-cell level in human T cells. The data showed that histone acetylation was increased during T cell activation. Among T cell subsets, terminally differentiated effector memory T (TEMRA) cells robustly producing effector cytokines were hyper-acetylated. Conversely, these TEMRA cells had lower expression levels of TCF-1, a key transcription factor for maintaining stem cell features. Pharmaceutical inhibition of histone acetylation using a small molecule C646 restrained the production of effector molecules, but retained stem cell-like properties in T cells after expansion. CONCLUSIONS: Per-cell histone acetylation is associated with terminal differentiation and poor stemness in human T cells. These observations suggest a new approach to enhance the stem cell-like properties of T cells and improve the efficacy of immunotherapy.


Sujet(s)
Méthylation de l'ADN , Histone , Humains , Histone/métabolisme , Acétylation , Régulation de l'expression des gènes , Différenciation cellulaire/génétique
9.
Immunity ; 56(12): 2719-2735.e7, 2023 Dec 12.
Article de Anglais | MEDLINE | ID: mdl-38039966

RÉSUMÉ

Commensal microbes induce cytokine-producing effector tissue-resident CD4+ T cells, but the function of these T cells in mucosal homeostasis is not well understood. Here, we report that commensal-specific intestinal Th17 cells possess an anti-inflammatory phenotype marked by expression of interleukin (IL)-10 and co-inhibitory receptors. The anti-inflammatory phenotype of gut-resident commensal-specific Th17 cells was driven by the transcription factor c-MAF. IL-10-producing commensal-specific Th17 cells were heterogeneous and derived from a TCF1+ gut-resident progenitor Th17 cell population. Th17 cells acquired IL-10 expression and anti-inflammatory phenotype in the small-intestinal lamina propria. IL-10 production by CD4+ T cells and IL-10 signaling in intestinal macrophages drove IL-10 expression by commensal-specific Th17 cells. Intestinal commensal-specific Th17 cells possessed immunoregulatory functions and curbed effector T cell activity in vitro and in vivo in an IL-10-dependent and c-MAF-dependent manner. Our results suggest that tissue-resident commensal-specific Th17 cells perform regulatory functions in mucosal homeostasis.


Sujet(s)
Microbiome gastro-intestinal , Cellules Th17 , Interleukine-10/métabolisme , Muqueuse intestinale/métabolisme , Anti-inflammatoires
10.
Res Sq ; 2023 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-37790365

RÉSUMÉ

TCF1high progenitor CD8+ T cells mediate the efficacy of PD-1 blockade, however the mechanisms that govern their generation and maintenance are poorly understood. Here, we show that targeting glycolysis through deletion of pyruvate kinase muscle 2 (PKM2) results in elevated pentose phosphate pathway (PPP) activity, leading to enrichment of a TCF1high central memory-like phenotype and increased responsiveness to PD-1 blockade in vivo. PKM2KO CD8+ T cells showed reduced glycolytic flux, accumulation of glycolytic intermediates and PPP metabolites, and increased PPP cycling as determined by 1,2 13C glucose carbon tracing. Small molecule agonism of the PPP without acute glycolytic impairment skewed CD8+ T cells towards a TCF1high population, generated a unique transcriptional landscape, enhanced tumor control in mice in combination with PD-1 blockade, and promoted tumor killing in patient-derived tumor organoids. Our study demonstrates a new metabolic reprogramming that contributes to a progenitor-like T cell state amenable to checkpoint blockade.

11.
Cell Rep ; 42(10): 113298, 2023 10 31.
Article de Anglais | MEDLINE | ID: mdl-37862171

RÉSUMÉ

The miR-15/16 family targets a large network of genes in T cells to restrict their cell cycle, memory formation, and survival. Upon T cell activation, miR-15/16 are downregulated, allowing rapid expansion of differentiated effector T cells to mediate a sustained response. Here, we used conditional deletion of miR-15/16 in regulatory T cells (Tregs) to identify immune functions of the miR-15/16 family in T cells. miR-15/16 are indispensable to maintain peripheral tolerance by securing efficient suppression by a limited number of Tregs. miR-15/16 deficiency alters expression of critical Treg proteins and results in accumulation of functionally impaired FOXP3loCD25loCD127hi Tregs. Excessive proliferation in the absence of miR-15/16 shifts Treg fate and produces an effector Treg phenotype. These Tregs fail to control immune activation, leading to spontaneous multi-organ inflammation and increased allergic inflammation in a mouse model of asthma. Together, our results demonstrate that miR-15/16 expression in Tregs is essential to maintain immune tolerance.


Sujet(s)
microARN , Lymphocytes T régulateurs , Animaux , Souris , microARN/génétique , microARN/métabolisme , Division cellulaire , Phénotype , Inflammation/génétique , Inflammation/métabolisme , Facteurs de transcription Forkhead/métabolisme
12.
Cell Rep ; 42(10): 113155, 2023 10 31.
Article de Anglais | MEDLINE | ID: mdl-37756164

RÉSUMÉ

The ability of activated progenitor T cells to self-renew while producing differentiated effector cell descendants may underlie immunological memory and persistent responses to ongoing infection. The nature of stem-like T cells responding to cancer and during treatment with immunotherapy is not clear. The subcellular organization of dividing progenitor CD8+ T cells from mice challenged with syngeneic tumors is examined here. Three-dimensional microscopy reveals an activating hub composed of polarized CD3, CD28, and phosphatidylinositol 3-kinase (PI3K) activity at the putative immunological synapse with an inhibitory hub composed of polarized PD-1 and CD73 at the opposite pole of mitotic blasts. Progenitor T cells from untreated and inhibitory checkpoint blockade-treated mice yield a differentiated TCF1- daughter cell, which inherits the PI3K activation hub, alongside a discordantly fated, self-renewing TCF1+ sister cell. Dynamic organization of opposite activating and inhibitory signaling poles in mitotic lymphocytes may account for the enigmatic durability of specific immunity.


Sujet(s)
Lymphocytes T CD8+ , Phosphatidylinositol 3-kinases , Souris , Animaux , Différenciation cellulaire , Cellules souches , Transduction du signal
13.
Cancer Cell ; 41(9): 1662-1679.e7, 2023 09 11.
Article de Anglais | MEDLINE | ID: mdl-37625402

RÉSUMÉ

Stem-like CD8+ T cells are regulated by T cell factor 1 (TCF1) and are considered requisite for immune checkpoint blockade (ICB) response. However, recent findings indicate that reliance on TCF1+CD8+ T cells for ICB efficacy may differ across tumor contexts. We find that TCF1 is essential for optimal priming of tumor antigen-specific CD8+ T cells and ICB response in poorly immunogenic tumors that accumulate TOX+ dysfunctional T cells, but is dispensable for T cell priming and therapy response in highly immunogenic tumors that efficiently expand transitory effectors. Importantly, improving T cell priming by vaccination or by enhancing antigen presentation on tumors rescues the defective responses of TCF1-deficient CD8+ T cells upon ICB in poorly immunogenic tumors. Our study highlights TCF1's role during the early stages of anti-tumor CD8+ T cell responses with important implications for guiding optimal therapeutic interventions in cancers with low TCF1+CD8+ T cells and low-neo-antigen expression.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs , Facteur de transcription TCF-1 , Humains , Anticorps , Antigènes néoplasiques , Immunothérapie , Facteur de transcription TCF-1/génétique , Tumeurs/immunologie , Tumeurs/thérapie
14.
Cancer Lett ; 571: 216336, 2023 09 01.
Article de Anglais | MEDLINE | ID: mdl-37562671

RÉSUMÉ

The efficacy of HCC (hepatocellular carcinoma) immunotherapy is hindered by the limited reactivity and short duration of tumor-infiltrating T cells. These deficiencies may be ascribed to the proliferative ability of T cells. The primary objective of this study was to identify the key factor regulating tumor-infiltrating lymphocytes (TIL) proliferation within the HCC microenvironment. Through the utilization of tissue-infiltrated T cell proteomics and fraction proteomics, we analyzed the differential proteins in T cells among HCC, liver fibrosis, and hemangioma (serving as controls) groups. Additionally, we examined the differential regulatory TFs of T cells between the HCC and VH (volunteer healthy, as a control) groups. Using cyTOF and flow cytometry technologies, as well as generating CD8+ T-specific BMI1 knockout mice, we confirmed that BMI1 controls CD127+KLRG1+ memory cell differentiation. Through RNA-seq and MeRIP-seq, we verified that BMI1 regulates TCF1 expression independently of its classical function. Furthermore, by conducting Tyramide signal amplification (TSA) IHC analysis, employing a hydrodynamic mouse HCC model, and utilizing liver-specific nanoparticle targeting therapy, we demonstrated that BMI1 in HCC influences the proliferation of infiltrating CD8+T. BMI1 inhibition promotes effector T cell differentiation while suppressing memory T cell differentiation. Moreover, liver-specific BMI1 knockdown proves beneficial in ameliorating T cell dysfunction and decelerating HCC progression. Our research group has pioneered the exploration of the proteomics of HCC-infiltrated T cells, shedding light on the pivotal role of BMI1 in controlling CD127+KLRG1+ memory CD8+ T cell differentiation, which serves as the cornerstone for achieving immunotherapy efficacy in HCC.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Complexe répresseur Polycomb-1 , Protéines proto-oncogènes , Animaux , Souris , Carcinome hépatocellulaire/anatomopathologie , Lymphocytes T CD8+/métabolisme , Immunothérapie , Tumeurs du foie/anatomopathologie , Cellules T mémoire , Souris knockout , Microenvironnement tumoral , Complexe répresseur Polycomb-1/génétique , Protéines proto-oncogènes/génétique
15.
Cell Rep ; 42(8): 112924, 2023 08 29.
Article de Anglais | MEDLINE | ID: mdl-37540600

RÉSUMÉ

Lymphoid tissue inducer (LTi) cells, a subset of innate lymphoid cells (ILCs), play an essential role in the formation of secondary lymphoid tissues. However, the regulation of the development and functions of this ILC subset is still elusive. In this study, we report that the transcription factor T cell factor 1 (TCF-1), just as GATA3, is indispensable for the development of non-LTi ILC subsets. While LTi cells are still present in TCF-1-deficient mice, the organogenesis of Peyer's patches (PPs), but not of lymph nodes, is impaired in these mice. LTi cells from different tissues have distinct gene expression patterns, and TCF-1 regulates the expression of lymphotoxin specifically in PP LTi cells. Mechanistically, TCF-1 may directly and/or indirectly regulate Lta, including through promoting the expression of GATA3. Thus, the TCF-1-GATA3 axis, which plays an important role during T cell development, also critically regulates the development of non-LTi cells and tissue-specific functions of LTi cells.


Sujet(s)
Immunité innée , Facteur de transcription TCF-1 , Animaux , Souris , Lymphocytes , Tissu lymphoïde/métabolisme , Facteur de transcription TCF-1/métabolisme
16.
Thorac Cancer ; 14(27): 2745-2753, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37536668

RÉSUMÉ

BACKGROUND: T cell factor-1 (TCF-1) + stem-like tumor-infiltrating lymphocytes (stem-like TILs) are important memory cells in the tumor microenvironment. However, their relationship with clinicopathological features, CD8+ TIL densities, immune checkpoint inhibitors (ICs), and prognostic values remain unknown for lung adenocarcinomas (LUADs). In this study, we aimed to characterize TCF-1+ TILs and their prognostic significance in patients with surgically resected LUADs. METHODS: Expression of TCF-1, CD8, and ICs including programmed death-1 (PD-1), lymphocyte activating-3 (LAG-3), and T cell immunoglobulin and mucin-domain containing-3 (TIM-3) in TILs were estimated using immunohistochemistry of resected LUADs. The association between TCF-1 expressions and clinicopathological characteristics of patient prognoses were analyzed. RESULTS: Positive TCF-1 expression significantly correlated with advanced pathological stage, tumor grade, CD8+ TILs density, TIM-3 expression, LAG-3 expression, and PD-1 expression. TCF-1 positivity was significantly associated with a better recurrence-free survival (RFS), and overall survival (OS). Subgroup analysis revealed that the TCF-1+/CD8+ group had the best RFS and OS, while the TCF-1-/CD8- group had the worst RFS and OS. Similarly, patients with TCF-1 + PD-1- had the best prognoses and patients with TCF-1-PD-1+ had the worst prognoses. CONCLUSION: TCF-1 had relatively high positive expression and special clinicopathological features in patients with LUAD. TCF-1+ TILs were related to CD8 density, TIM-3 expression, LAG-3 expression, and PD-1 expression, and were associated with better prognoses in LUAD patients. A combination of TCF-1 and CD8 densities or PD-1 expression further stratified patients into different groups with distinct prognoses.


Sujet(s)
Adénocarcinome pulmonaire , Adénocarcinome , Tumeurs du poumon , Humains , Antigène CD274/métabolisme , Lymphocytes T CD8+/métabolisme , Récepteur cellulaire-2 du virus de l'hépatite A , Tumeurs du poumon/anatomopathologie , Pronostic , Récepteur-1 de mort cellulaire programmée/métabolisme , Microenvironnement tumoral
17.
Oncoimmunology ; 12(1): 2230666, 2023.
Article de Anglais | MEDLINE | ID: mdl-37435097

RÉSUMÉ

Human papilloma virus (HPV)-related oncogenesis in head and neck cancer establishes a local microenvironment rich with immune cells, however the composition of the microenvironment in recurrent disease following definitive treatment is poorly understood. Here, we investigate the composition and spatial relationships between tumor and immune cells in recurrent head and neck cancer following curative intent chemoradiotherapy. Multiplexed immunofluorescence with 12 unique markers, through two multiplex immunofluorescent panels, was performed to evaluate 27 tumor samples including 18 pre-treatment primary and 9 paired recurrent tumors. Tumor and immune cell populations were phenotyped and quantified using a previously validated semi-automated digital pathology platform for cell segmentation. Spatial analysis was conducted by evaluating immune cells within the tumor, peri-tumoral stroma, and distant stroma. Initial tumors in patients with subsequent recurrence were found to be enriched in tumor associated macrophages and displayed an immune excluded spatial distribution. Recurrent tumors after chemoradiation were hypo-inflamed, with a statistically significant reduction in the recently identified stem-like TCF1+ CD8 T-cells, which normally function to maintain HPV-specific immune responses in the setting of chronic antigen exposure. Our findings indicate that the tumor microenvironment of recurrent HPV-related head and neck cancers displays a reduction in stem-like T cells, consistent with an immune microenvironment with a reduced ability to mount T-cell-driven anti-tumor immune responses.


Sujet(s)
Infections à papillomavirus , Humains , Infections à papillomavirus/complications , Microenvironnement tumoral , Cellules souches , Carcinogenèse , Lymphocytes T CD8+ , Virus des Papillomavirus humains
18.
Semin Immunol ; 69: 101800, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37494738

RÉSUMÉ

The defense against infectious diseases, either through natural immunity or after vaccinations, relies on the generation and maintenance of protective T cell memory. Naïve T cells are at the center of memory T cell generation during primary responses. Upon activation, they undergo a complex, highly regulated differentiation process towards different functional states. Naïve T cells maintained into older age have undergone epigenetic adaptations that influence their fate decisions during differentiation. We review age-sensitive, molecular pathways and gene regulatory networks that bias naïve T cell differentiation towards effector cell generation at the expense of memory and Tfh cells. As a result, T cell differentiation in older adults is associated with release of bioactive waste products into the microenvironment, higher stress sensitivity as well as skewing towards pro-inflammatory signatures and shorter life spans. These maladaptations not only contribute to poor vaccine responses in older adults but also fuel a more inflammatory state.


Sujet(s)
Mémoire immunologique , Lymphocytes T , Humains , Sujet âgé , Différenciation cellulaire , Vieillissement , Activation des lymphocytes , Lymphocytes T CD8+
19.
Front Immunol ; 14: 1168125, 2023.
Article de Anglais | MEDLINE | ID: mdl-37122720

RÉSUMÉ

CD4+ T cells are typically considered as 'helper' or 'regulatory' populations that support and orchestrate the responses of other lymphocytes. However, they can also develop potent granzyme (Gzm)-mediated cytotoxic activity and CD4+ cytotoxic T cells (CTLs) have been amply documented both in humans and in mice, particularly in the context of human chronic infection and cancer. Despite the established description of CD4+ CTLs, as well as of the critical cytotoxic activity they exert against MHC class II-expressing targets, their developmental and memory maintenance requirements remain elusive. This is at least in part owing to the lack of a murine experimental system where CD4+ CTLs are stably induced. Here, we show that viral and bacterial vectors encoding the same epitope induce distinct CD4+ CTL responses in challenged mice, all of which are nevertheless transient in nature and lack recall properties. Consistent with prior reports, CD4+ CTL differentiation is accompanied by loss of TCF-1 expression, a transcription factor considered essential for memory T cell survival. Using genetic ablation of Tcf7, which encodes TCF-1, at the time of CD4+ T cell activation, we further show that, contrary to observations in CD8+ T cells, continued expression of TCF-1 is not required for CD4+ T cell memory survival. Whilst Tcf7-deficient CD4+ T cells persisted normally following retroviral infection, the CD4+ CTL subset still declined, precluding conclusive determination of the requirement for TCF-1 for murine CD4+ CTL survival. Using xenotransplantation of human CD4+ T cells into murine recipients, we demonstrate that human CD4+ CTLs develop and persist in the same experimental conditions where murine CD4+ CTLs fail to persist. These observations uncover a species-specific defect in murine CD4+ CTL persistence with implications for their use as a model system.


Sujet(s)
Lymphocytes T CD8+ , Cellules T mémoire , Animaux , Humains , Souris , Lymphocytes T CD4+ , Différenciation cellulaire , Lymphocytes T cytotoxiques/métabolisme
20.
Oncoimmunology ; 12(1): 2197360, 2023.
Article de Anglais | MEDLINE | ID: mdl-37025392

RÉSUMÉ

The immunocytokine PD1-IL2v was designed to overcome liabilities and improve efficacy of IL-2 therapies. PD1-IL2v preferentially targets PD-1+ T-cells and acts on antigen-specific stem-like PD-1+ TCF-1+ CD8+ T-cells expanding and differentiating them towards better effectors resulting in superior efficacy in LCMV chronic infection and tumor models compared to checkpoint inhibition.


Sujet(s)
Lymphocytes T CD8+ , Cytokines , Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Cytokines/pharmacologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Virus de la chorioméningite lymphocytaire , Lymphocytes T CD8+/immunologie , Récepteurs à l'interleukine-2 , Humains , Animaux , Souris , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Infections à Arenaviridae , Souris de lignée C57BL , Différenciation cellulaire , Immunothérapie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE