Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 165
Filtrer
1.
Nat Biotechnol ; 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39079964

RÉSUMÉ

Although chimeric antigen receptor (CAR) T cell therapies have demonstrated promising clinical outcomes, durable remissions remain limited. To extend the efficacy of CAR T cells, we develop a CAR enhancer (CAR-E), comprising a CAR T cell antigen fused to an immunomodulatory molecule. Here we demonstrate this strategy using B cell maturation antigen (BCMA) CAR T cells for the treatment of multiple myeloma, with a CAR-E consisting of the BCMA fused to a low-affinity interleukin 2 (IL-2). This selectively induces IL-2 signaling in CAR T cells upon antigen-CAR binding, enhancing T cell activation and antitumor activity while reducing IL-2-associated toxicities. We show that the BCMA CAR-E selectively binds CAR T cells and increases CAR T cell proliferation, clearance of tumor cells and development of memory CAR T cells. The memory cells retain the ability to re-expand upon restimulation, effectively controlling tumor growth upon rechallenge. Mechanistic studies reveal the involvement of both CAR and IL-2 receptor endodomains in the CAR-E mechanism of action. The CAR-E approach avoids the need for specific engineering and enables CAR T cell therapy with lower cell doses.

2.
Curr Opin Oncol ; 36(3): 136-142, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38573202

RÉSUMÉ

PURPOSE OF REVIEW: Neoadjuvant (presurgical) immune checkpoint blockade (ICB) has shown promising clinical activity in head and neck cancer and other cancers, including FDA approvals for neoadjuvant approaches for triple-negative breast cancer and nonsmall cell lung cancer. Here we will review recent data from clinical trials in head and neck squamous cell carcinoma (HNSCC), including mechanistic studies highlighting local and systemic effects on T cell-mediated immunity. RECENT FINDINGS: A series of clinical trials of neoadjuvant ICB have documented evidence of clinical activity, including clinical to pathologic downstaging and pathologic response in a subset of patients. Also, emerging data suggest improved survival outcomes for patients with tumors responsive to neoadjuvant ICB. In depth mechanistic studies have documented intra-tumoral expansion of CD8 T cell populations characterized by tissue residency and cytotoxicity programs. Treatment also leads to expansion of activated CD8 T cells in the blood, many of which share TCR sequences with tumor-infiltrating T cells. The frequency of activated circulating CD8 T cell populations is correlated with the degree of pathologic response within tumors. SUMMARY: Even a short duration of neoadjuvant immunotherapy can enhance local and systemic tumor-reactive T cell populations. Downstaging induced by neoadjuvant ICB can reduce the extent of surgical resection in this anatomically sensitive location.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs de la tête et du cou , Tumeurs du poumon , Humains , Traitement néoadjuvant , Inhibiteurs de points de contrôle immunitaires , Tumeurs de la tête et du cou/traitement médicamenteux
4.
Front Immunol ; 15: 1368586, 2024.
Article de Anglais | MEDLINE | ID: mdl-38550583

RÉSUMÉ

MICA and MICB are Class I MHC-related glycoproteins that are upregulated on the surface of cells in response to stress, for instance due to infection or malignant transformation. MICA/B are ligands for NKG2D, an activating receptor on NK cells, CD8+ T cells, and γδ T cells. Upon engagement of MICA/B with NKG2D, these cytotoxic cells eradicate MICA/B-positive targets. MICA is frequently overexpressed on the surface of cancer cells of epithelial and hematopoietic origin. Here, we created nanobodies that recognize MICA. Nanobodies, or VHHs, are the recombinantly expressed variable regions of camelid heavy chain-only immunoglobulins. They retain the capacity of antigen recognition but are characterized by their stability and ease of production. The nanobodies described here detect surface-disposed MICA on cancer cells in vitro by flow cytometry and can be used therapeutically as nanobody-drug conjugates when fused to the Maytansine derivative DM1. The nanobody-DM1 conjugate selectively kills MICA positive tumor cells in vitro.


Sujet(s)
Tumeurs , Anticorps à domaine unique , Humains , Lymphocytes T CD8+ , Anticorps à domaine unique/usage thérapeutique , Antigènes d'histocompatibilité de classe I , Sous-famille K des récepteurs de cellules NK de type lectine , Tumeurs/diagnostic , Tumeurs/thérapie , Immunothérapie
5.
Blood ; 143(12): 1124-1138, 2024 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-38153903

RÉSUMÉ

ABSTRACT: The CD161 inhibitory receptor is highly upregulated by tumor-infiltrating T cells in multiple human solid tumor types, and its ligand, CLEC2D, is expressed by both tumor cells and infiltrating myeloid cells. Here, we assessed the role of the CD161 receptor in hematological malignancies. Systematic analysis of CLEC2D expression using the Cancer Cell Line Encyclopedia revealed that CLEC2D messenger RNA was most abundant in hematological malignancies, including B-cell and T-cell lymphomas as well as lymphocytic and myelogenous leukemias. CLEC2D protein was detected by flow cytometry on a panel of cell lines representing a diverse set of hematological malignancies. We, therefore, used yeast display to generate a panel of high-affinity, fully human CD161 monoclonal antibodies (mAbs) that blocked CLEC2D binding. These mAbs were specific for CD161 and had a similar affinity for human and nonhuman primate CD161, a property relevant for clinical translation. A high-affinity CD161 mAb enhanced key aspects of T-cell function, including cytotoxicity, cytokine production, and proliferation, against B-cell lines originating from patients with acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and Burkitt lymphoma. In humanized mouse models, this CD161 mAb enhanced T-cell-mediated immunity, resulting in a significant survival benefit. Single cell RNA-seq data demonstrated that CD161 mAb treatment enhanced expression of cytotoxicity genes by CD4 T cells as well as a tissue-residency program by CD4 and CD8 T cells that is associated with favorable survival outcomes in multiple human cancer types. These fully human mAbs, thus, represent potential immunotherapy agents for hematological malignancies.


Sujet(s)
Tumeurs hématologiques , Tumeurs , Animaux , Souris , Humains , Lymphocytes T CD4+ , Immunité cellulaire , Lymphocytes T CD8+ , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Tumeurs hématologiques/génétique , Tumeurs hématologiques/thérapie , Sous-famille B des récepteurs de cellules NK de type lectine/génétique
6.
Cell Rep ; 42(12): 113564, 2023 12 26.
Article de Anglais | MEDLINE | ID: mdl-38100350

RÉSUMÉ

Triple-negative breast cancer (TNBC) is a heterogeneous disease with limited treatment options. To characterize TNBC heterogeneity, we defined transcriptional, epigenetic, and metabolic subtypes and subtype-driving super-enhancers and transcription factors by combining functional and molecular profiling with computational analyses. Single-cell RNA sequencing revealed relative homogeneity of the major transcriptional subtypes (luminal, basal, and mesenchymal) within samples. We found that mesenchymal TNBCs share features with mesenchymal neuroblastoma and rhabdoid tumors and that the PRRX1 transcription factor is a key driver of these tumors. PRRX1 is sufficient for inducing mesenchymal features in basal but not in luminal TNBC cells via reprogramming super-enhancer landscapes, but it is not required for mesenchymal state maintenance or for cellular viability. Our comprehensive, large-scale, multiplatform, multiomics study of both experimental and clinical TNBC is an important resource for the scientific and clinical research communities and opens venues for future investigation.


Sujet(s)
Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/anatomopathologie , Facteurs de transcription/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines à homéodomaine/métabolisme
7.
Med ; 4(11): 755-760, 2023 11 10.
Article de Anglais | MEDLINE | ID: mdl-37951209

RÉSUMÉ

Frontline treatment and resultant cure rates in patients with advanced ovarian cancer have changed little over the past several decades. Here, we outline a multidisciplinary approach aimed at gaining novel therapeutic insights by focusing on the poorly understood minimal residual disease phase of ovarian cancer that leads to eventual incurable recurrences.


Sujet(s)
Tumeurs de l'ovaire , Humains , Femelle , Maladie résiduelle , Tumeurs de l'ovaire/traitement médicamenteux , Carcinome épithélial de l'ovaire/thérapie
8.
bioRxiv ; 2023 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-37873273

RÉSUMÉ

Targeting of specific metabolic pathways in tumor cells has the potential to sensitize them to immune-mediated attack. Here we provide evidence for a specific means of mitochondrial respiratory Complex I (CI) inhibition that improves tumor immunogenicity and sensitivity to immune checkpoint blockade (ICB). Targeted genetic deletion of the CI subunits Ndufs4 and Ndufs6 , but not other subunits, induces an immune-dependent tumor growth attenuation in mouse melanoma models. We show that deletion of Ndufs4 induces expression of the transcription factor Nlrc5 and genes in the MHC class I antigen presentation and processing pathway. This induction of MHC-related genes is driven by an accumulation of pyruvate dehydrogenase-dependent mitochondrial acetyl-CoA downstream of CI subunit deletion. This work provides a novel functional modality by which selective CI inhibition restricts tumor growth, suggesting that specific targeting of Ndufs4 , or related CI subunits, increases T-cell mediated immunity and sensitivity to ICB.

9.
Cancer Cell ; 41(7): 1207-1221.e12, 2023 07 10.
Article de Anglais | MEDLINE | ID: mdl-37327789

RÉSUMÉ

The cell-autonomous balance of immune-inhibitory and -stimulatory signals is a critical process in cancer immune evasion. Using patient-derived co-cultures, humanized mouse models, and single-cell RNA-sequencing of patient melanomas biopsied before and on immune checkpoint blockade, we find that intact cancer cell-intrinsic expression of CD58 and ligation to CD2 is required for anti-tumor immunity and is predictive of treatment response. Defects in this axis promote immune evasion through diminished T cell activation, impaired intratumoral T cell infiltration and proliferation, and concurrently increased PD-L1 protein stabilization. Through CRISPR-Cas9 and proteomics screens, we identify and validate CMTM6 as critical for CD58 stability and upregulation of PD-L1 upon CD58 loss. Competition between CD58 and PD-L1 for CMTM6 binding determines their rate of endosomal recycling over lysosomal degradation. Overall, we describe an underappreciated yet critical axis of cancer immunity and provide a molecular basis for how cancer cells balance immune inhibitory and stimulatory cues.


Sujet(s)
Antigène CD274 , Mélanome , Souris , Animaux , Antigène CD274/génétique , Lymphocytes T , Antigènes CD58/composition chimique , Antigènes CD58/métabolisme , Mélanome/génétique , Mélanome/métabolisme , Activation des lymphocytes
10.
Int J Radiat Oncol Biol Phys ; 116(4): 906-915, 2023 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-37201759

RÉSUMÉ

PURPOSE: The aim of this work was to determine whether intratumoral injections of a liquid oxygen solution are effective at boosting radiation-induced abscopal effects. METHODS AND MATERIALS: A liquid oxygen solution, comprising slow-release polymer-shelled oxygen microparticles, was fabricated and injected intratumorally to locally elevate tumor oxygen levels before and after treatment with radiation therapy. Changes in tumor volume were monitored. In a subset of studies, CD8-positive cells were depleted and the experiments were repeated. Histologic analyses of the tumor tissues were performed to quantify the concentration of infiltrating immune cells. RESULTS: Daily intratumoral injections of oxygen-filled microparticles significantly retarded primary and secondary tumor growth, boosted infiltration of cytotoxic T cells, and improved overall survival when used as an adjuvant to radiation therapy. The findings also demonstrated that efficacy requires both radiation and oxygen, suggesting that they act synergistically to enhance in situ vaccination and systemic antitumor immune responses. CONCLUSIONS: This study demonstrated the potential advantages of intratumoral injections of a liquid oxygen solution as a strategy to boost radiation-induced abscopal effects, and the findings warrant future efforts toward clinical translation of the injectable liquid oxygen solution.


Sujet(s)
Tumeurs , Oxygène , Humains , Tumeurs/anatomopathologie , Lymphocytes T CD8+ , Vaccination , Immunité
11.
Med ; 4(7): 457-477.e8, 2023 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-37172578

RÉSUMÉ

BACKGROUND: The advent of chimeric antigen receptor (CAR) T cell therapies has transformed the treatment of hematological malignancies; however, broader therapeutic success of CAR T cells has been limited in solid tumors because of their frequently heterogeneous composition. Stress proteins in the MICA and MICB (MICA/B) family are broadly expressed by tumor cells following DNA damage but are rapidly shed to evade immune detection. METHODS: We have developed a novel CAR targeting the conserved α3 domain of MICA/B (3MICA/B CAR) and incorporated it into a multiplexed-engineered induced pluripotent stem cell (iPSC)-derived natural killer (NK) cell (3MICA/B CAR iNK) that expressed a shedding-resistant form of the CD16 Fc receptor to enable tumor recognition through two major targeting receptors. FINDINGS: We demonstrated that 3MICA/B CAR mitigates MICA/B shedding and inhibition via soluble MICA/B while simultaneously exhibiting antigen-specific anti-tumor reactivity across an expansive library of human cancer cell lines. Pre-clinical assessment of 3MICA/B CAR iNK cells demonstrated potent antigen-specific in vivo cytolytic activity against both solid and hematological xenograft models, which was further enhanced in combination with tumor-targeted therapeutic antibodies that activate the CD16 Fc receptor. CONCLUSIONS: Our work demonstrated 3MICA/B CAR iNK cells to be a promising multi-antigen-targeting cancer immunotherapy approach intended for solid tumors. FUNDING: Funded by Fate Therapeutics and NIH (R01CA238039).


Sujet(s)
Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Lignée cellulaire tumorale , Immunothérapie adoptive , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/transplantation , Récepteur Fc/métabolisme
12.
Cancer Discov ; 13(5): 1186-1209, 2023 05 04.
Article de Anglais | MEDLINE | ID: mdl-36811466

RÉSUMÉ

Tumor heterogeneity is a major barrier to cancer therapy, including immunotherapy. Activated T cells can efficiently kill tumor cells following recognition of MHC class I (MHC-I)-bound peptides, but this selection pressure favors outgrowth of MHC-I-deficient tumor cells. We performed a genome-scale screen to discover alternative pathways for T cell-mediated killing of MHC-I-deficient tumor cells. Autophagy and TNF signaling emerged as top pathways, and inactivation of Rnf31 (TNF signaling) and Atg5 (autophagy) sensitized MHC-I-deficient tumor cells to apoptosis by T cell-derived cytokines. Mechanistic studies demonstrated that inhibition of autophagy amplified proapoptotic effects of cytokines in tumor cells. Antigens from apoptotic MHC-I-deficient tumor cells were efficiently cross-presented by dendritic cells, resulting in heightened tumor infiltration by IFNγ-and TNFα-producing T cells. Tumors with a substantial population of MHC-I-deficient cancer cells could be controlled by T cells when both pathways were targeted using genetic or pharmacologic approaches. SIGNIFICANCE: Tumor heterogeneity is a major barrier to immunotherapy. We show that MHC-I-deficient tumor cells are forced into apoptosis by T cell-derived cytokines when TNF signaling and autophagy pathways are targeted. This approach enables T cell-mediated elimination of tumors with a substantial population of resistant, MHC-I-deficient tumor cells. This article is highlighted in the In This Issue feature, p. 1027.


Sujet(s)
Tumeurs , Lymphocytes T , Humains , Cytokines , Antigènes d'histocompatibilité de classe I/métabolisme , Tumeurs/génétique , Tumeurs/thérapie , Gènes MHC de classe I
13.
Proc Natl Acad Sci U S A ; 120(1): e2213222120, 2023 01 03.
Article de Anglais | MEDLINE | ID: mdl-36577059

RÉSUMÉ

Adoptive T cell transfer (ACT) therapies suffer from a number of limitations (e.g., poor control of solid tumors), and while combining ACT with cytokine therapy can enhance effectiveness, this also results in significant side effects. Here, we describe a nanotechnology approach to improve the efficacy of ACT therapies by metabolically labeling T cells with unnatural sugar nanoparticles, allowing direct conjugation of antitumor cytokines onto the T cell surface during the manufacturing process. This allows local, concentrated activity of otherwise toxic cytokines. This approach increases T cell infiltration into solid tumors, activates the host immune system toward a Type 1 response, encourages antigen spreading, and improves control of aggressive solid tumors and achieves complete blood cancer regression with otherwise noncurative doses of CAR-T cells. Overall, this method provides an effective and easily integrated approach to the current ACT manufacturing process to increase efficacy in various settings.


Sujet(s)
Cytokines , Tumeurs , Humains , Cytokines/métabolisme , Immunothérapie adoptive/méthodes , Récepteurs aux antigènes des cellules T , Lymphocytes T , Tumeurs/anatomopathologie , Thérapie cellulaire et tissulaire
14.
Annu Rev Immunol ; 41: 17-38, 2023 04 26.
Article de Anglais | MEDLINE | ID: mdl-36446137

RÉSUMÉ

T cells and natural killer (NK) cells have complementary roles in tumor immunity, and dual T cell and NK cell attack thus offers opportunities to deepen the impact of immunotherapy. Recent work has also shown that NK cells play an important role in recruiting dendritic cells to tumors and thus enhance induction of CD8 T cell responses, while IL-2 secreted by T cells activates NK cells. Targeting of immune evasion mechanisms from the activating NKG2D receptor and its MICA and MICB ligands on tumor cells offers opportunities for therapeutic intervention. Interestingly, T cells and NK cells share several important inhibitory and activating receptors that can be targeted to enhance T cell- and NK cell-mediated immunity. These inhibitory receptor-ligand systems include CD161-CLEC2D, TIGIT-CD155, and NKG2A/CD94-HLA-E. We also discuss emerging therapeutic strategies based on inhibitory and activating cytokines that profoundly impact the function of both lymphocyte populations within tumors.


Sujet(s)
Cellules tueuses naturelles , Tumeurs , Humains , Animaux , Antigènes d'histocompatibilité de classe I , Lymphocytes T CD8+ , Immunothérapie , Immunité cellulaire
15.
Cancer Res ; 83(4): 613-625, 2023 02 15.
Article de Anglais | MEDLINE | ID: mdl-36548402

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cell therapy can lead to dramatic clinical responses in B-cell malignancies. However, early clinical trials with CAR T-cell therapy in non-B-cell malignancies have been disappointing to date, suggesting that tumor-intrinsic features contribute to resistance. To investigate tumor-intrinsic modes of resistance, we performed genome scale CRISPR-Cas9 screens in mesothelin (MSLN)-expressing pancreatic cancer cells. Co-culture with MSLN-targeting CAR T cells identified both antigen-dependent and antigen-independent modes of resistance. In particular, loss of the majority of the genes involved in the pathway responsible for GPI-anchor biosynthesis and attachment abrogated the ability of CAR T cells to target pancreatic cancer cells, suggesting that disruption of this pathway may permit MSLN CAR T-cell evasion in the clinic. Antigen-independent mediators of CAR T-cell response included members of the death receptor pathway as well as genes that regulate tumor transcriptional responses, including TFAP4 and INTS12. TFAP4-mediated CAR T resistance depended on the NFκB transcription factor p65, indicating that tumor resistance to CAR T-cell therapy likely involves alterations in tumor-intrinsic states. Overall, this study uncovers multiple antigen-dependent and -independent mechanisms of CAR T-cell evasion by pancreatic cancer, paving the way for overcoming resistance in this disease that is notoriously refractory to immunotherapy. SIGNIFICANCE: The identification and validation of key determinants of CAR T-cell response in pancreatic cancer provide insights into the landscape of tumor cell intrinsic resistance mechanisms and into approaches to improve therapeutic efficacy.


Sujet(s)
Immunothérapie adoptive , Tumeurs du pancréas , Récepteurs chimériques pour l'antigène , Humains , Lignée cellulaire tumorale , Thérapie cellulaire et tissulaire , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/thérapie , Récepteurs aux antigènes des cellules T , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/usage thérapeutique , Tumeurs du pancréas
16.
Nat Commun ; 13(1): 7558, 2022 12 07.
Article de Anglais | MEDLINE | ID: mdl-36476730

RÉSUMÉ

Cancer prevention has a profound impact on cancer-associated mortality and morbidity. We previously identified TGFß signaling as a candidate regulator of mammary epithelial cells associated with breast cancer risk. Here, we show that short-term TGFBR inhibitor (TGFBRi) treatment of peripubertal ACI inbred and Sprague Dawley outbred rats induces lasting changes and prevents estrogen- and carcinogen-induced mammary tumors, respectively. We identify TGFBRi-responsive cell populations by single cell RNA-sequencing, including a unique epithelial subpopulation designated secretory basal cells (SBCs) with progenitor features. We detect SBCs in normal human breast tissues and find them to be associated with breast cancer risk. Interactome analysis identifies SBCs as the most interactive cell population and the main source of insulin-IGF signaling. Accordingly, inhibition of TGFBR and IGF1R decrease proliferation of organoid cultures. Our results reveal a critical role for TGFß in regulating mammary epithelial cells relevant to breast cancer and serve as a proof-of-principle cancer prevention strategy.


Sujet(s)
Tumeurs , Rats , Humains , Animaux , Rats de lignée ACI , Rat Sprague-Dawley
17.
Science ; 377(6614): 1519-1529, 2022 09 30.
Article de Anglais | MEDLINE | ID: mdl-36173860

RÉSUMÉ

Gain-of-function mutations in isocitrate dehydrogenase (IDH) in human cancers result in the production of d-2-hydroxyglutarate (d-2HG), an oncometabolite that promotes tumorigenesis through epigenetic alterations. The cancer cell-intrinsic effects of d-2HG are well understood, but its tumor cell-nonautonomous roles remain poorly explored. We compared the oncometabolite d-2HG with its enantiomer, l-2HG, and found that tumor-derived d-2HG was taken up by CD8+ T cells and altered their metabolism and antitumor functions in an acute and reversible fashion. We identified the glycolytic enzyme lactate dehydrogenase (LDH) as a molecular target of d-2HG. d-2HG and inhibition of LDH drive a metabolic program and immune CD8+ T cell signature marked by decreased cytotoxicity and impaired interferon-γ signaling that was recapitulated in clinical samples from human patients with IDH1 mutant gliomas.


Sujet(s)
Lymphocytes T CD8+ , Carcinogenèse , Glutarates , Isocitrate dehydrogenases , Tumeurs , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Mutation gain de fonction , Glutarates/métabolisme , Humains , Interféron gamma/métabolisme , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme , L-Lactate dehydrogenase/antagonistes et inhibiteurs , L-Lactate dehydrogenase/métabolisme , Souris , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/métabolisme
18.
Nat Mater ; 21(8): 939-950, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35817965

RÉSUMÉ

Myelofibrosis is a progressive bone marrow malignancy associated with monocytosis, and is believed to promote the pathological remodelling of the extracellular matrix. Here we show that the mechanical properties of myelofibrosis, namely the liquid-to-solid properties (viscoelasticity) of the bone marrow, contribute to aberrant differentiation of monocytes. Human monocytes cultured in stiff, elastic hydrogels show proinflammatory polarization and differentiation towards dendritic cells, as opposed to those cultured in a viscoelastic matrix. This mechanically induced cell differentiation is blocked by inhibiting a myeloid-specific isoform of phosphoinositide 3-kinase, PI3K-γ. We further show that murine bone marrow with myelofibrosis has a significantly increased stiffness and unveil a positive correlation between myelofibrosis grading and viscoelasticity. Treatment with a PI3K-γ inhibitor in vivo reduced frequencies of monocyte and dendritic cell populations in murine bone marrow with myelofibrosis. Moreover, transcriptional changes driven by viscoelasticity are consistent with transcriptional profiles of myeloid cells in other human fibrotic diseases. These results demonstrate that a fibrotic bone marrow niche can physically promote a proinflammatory microenvironment.


Sujet(s)
Myélofibrose primitive , Animaux , Moelle osseuse/anatomopathologie , Différenciation cellulaire , Fibrose , Humains , Souris , Monocytes , Phosphatidylinositol 3-kinases , Myélofibrose primitive/anatomopathologie
19.
Cell ; 185(16): 2918-2935.e29, 2022 08 04.
Article de Anglais | MEDLINE | ID: mdl-35803260

RÉSUMÉ

Neoadjuvant immune checkpoint blockade has shown promising clinical activity. Here, we characterized early kinetics in tumor-infiltrating and circulating immune cells in oral cancer patients treated with neoadjuvant anti-PD-1 or anti-PD-1/CTLA-4 in a clinical trial (NCT02919683). Tumor-infiltrating CD8 T cells that clonally expanded during immunotherapy expressed elevated tissue-resident memory and cytotoxicity programs, which were already active prior to therapy, supporting the capacity for rapid response. Systematic target discovery revealed that treatment-expanded tumor T cell clones in responding patients recognized several self-antigens, including the cancer-specific antigen MAGEA1. Treatment also induced a systemic immune response characterized by expansion of activated T cells enriched for tumor-infiltrating T cell clonotypes, including both pre-existing and emergent clonotypes undetectable prior to therapy. The frequency of activated blood CD8 T cells, notably pre-treatment PD-1-positive KLRG1-negative T cells, was strongly associated with intra-tumoral pathological response. These results demonstrate how neoadjuvant checkpoint blockade induces local and systemic tumor immunity.


Sujet(s)
Tumeurs , Récepteur-1 de mort cellulaire programmée , Lymphocytes T CD8+ , Humains , Immunothérapie , Lymphocytes TIL , Traitement néoadjuvant , Tumeurs/thérapie , Microenvironnement tumoral
20.
Nature ; 606(7916): 992-998, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35614223

RÉSUMÉ

Most cancer vaccines target peptide antigens, necessitating personalization owing to the vast inter-individual diversity in major histocompatibility complex (MHC) molecules that present peptides to T cells. Furthermore, tumours frequently escape T cell-mediated immunity through mechanisms that interfere with peptide presentation1. Here we report a cancer vaccine that induces a coordinated attack by diverse T cell and natural killer (NK) cell populations. The vaccine targets the MICA and MICB (MICA/B) stress proteins expressed by many human cancers as a result of DNA damage2. MICA/B serve as ligands for the activating NKG2D receptor on T cells and NK cells, but tumours evade immune recognition by proteolytic MICA/B cleavage3,4. Vaccine-induced antibodies increase the density of MICA/B proteins on the surface of tumour cells by inhibiting proteolytic shedding, enhance presentation of tumour antigens by dendritic cells to T cells and augment the cytotoxic function of NK cells. Notably, this vaccine maintains efficacy against MHC class I-deficient tumours resistant to cytotoxic T cells through the coordinated action of NK cells and CD4+ T cells. The vaccine is also efficacious in a clinically important setting: immunization following surgical removal of primary, highly metastatic tumours inhibits the later outgrowth of metastases. This vaccine design enables protective immunity even against tumours with common escape mutations.


Sujet(s)
Syndromes myélodysplasiques , Tumeurs , Maladies génétiques de la peau , Vaccins , Antigènes d'histocompatibilité de classe I , Humains , Cellules tueuses naturelles , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Tumeurs/prévention et contrôle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE