Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 145
2.
Immunol Lett ; 267: 106864, 2024 Jun.
Article En | MEDLINE | ID: mdl-38705481

Tumor-associated myeloid cells, including macrophages and myeloid-derived suppressor cells, can be highly prevalent in solid tumors and play a significant role in the development of the tumor. Therefore, myeloid cells are being considered potential targets for cancer immunotherapies. In this review, we focused on strategies aimed at targeting tumor-associated macrophages (TAMs). Most strategies were studied preclinically but we also included a limited number of clinical studies based on these strategies. We describe possible underlying mechanisms and discuss future challenges and prospects.


Immunotherapy , Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Neoplasms/therapy , Neoplasms/immunology , Immunotherapy/methods , Animals , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor Microenvironment/immunology , Macrophages/immunology , Macrophages/metabolism , Cellular Reprogramming/immunology , Macrophage Activation/immunology
3.
Gene ; 925: 148601, 2024 Oct 20.
Article En | MEDLINE | ID: mdl-38788817

Tumor-derived exosomes (TDEs), as topologies of tumor cells, not only carry biological information from the mother, but also act as messengers for cellular communication. It has been demonstrated that TDEs play a key role in inducing an immunosuppressive tumor microenvironment (TME). They can reprogram immune cells indirectly or directly by delivering inhibitory proteins, cytokines, RNA and other substances. They not only inhibit the maturation and function of dendritic cells (DCs) and natural killer (NK) cells, but also remodel M2 macrophages and inhibit T cell infiltration to promote immunosuppression and create a favorable ecological niche for tumor growth, invasion and metastasis. Based on the specificity of TDEs, targeting TDEs has become a new strategy to monitor tumor progression and enhance treatment efficacy. This paper reviews the intricate molecular mechanisms underlying the immunosuppressive effects induced by TDEs to establish a theoretical foundation for cancer therapy. Additionally, the challenges of TDEs as a novel approach to tumor treatment are discussed.


Exosomes , Neoplasms , Tumor Microenvironment , Exosomes/immunology , Exosomes/metabolism , Humans , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/immunology , Animals , Dendritic Cells/immunology , Cellular Reprogramming/immunology , Killer Cells, Natural/immunology , Macrophages/immunology , Macrophages/metabolism , Cell Communication/immunology
4.
Immunopharmacol Immunotoxicol ; 46(3): 330-340, 2024 Jun.
Article En | MEDLINE | ID: mdl-38478467

Rheumatoid arthritis (RA) is a persistent autoimmune condition characterized by ongoing inflammation primarily affecting the synovial joint. This inflammation typically arises from an increase in immune cells such as neutrophils, macrophages, and T cells (TC). TC is recognized as a major player in RA pathogenesis. The involvement of HLA-DRB1 and PTPN-2 among RA patients confirms the TC involvement in RA. Metabolism of TC is maintained by various other factors like cytokines, mitochondrial proteins & other metabolites. Different TC subtypes utilize different metabolic pathways like glycolysis, oxidative phosphorylation and fatty acid oxidation for their activation from naive TC (T0). Although all subsets of TC are not deleterious for synovium, some subsets of TC are involved in joint repair using their anti-inflammatory properties. Hence artificially reprogramming of TC subset by interfering with their metabolic status poised a hope in future to design new molecules against RA.


Arthritis, Rheumatoid , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Animals , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/drug effects , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology
5.
Int Immunol ; 36(6): 261-278, 2024 Apr 27.
Article En | MEDLINE | ID: mdl-38364321

Adoptive cell therapy (ACT) is an immunotherapeutic approach that involves isolating T cells from a patient, culturing them ex vivo, then reinfusing the cells back into the patient. Although this strategy has shown remarkable efficacy in hematological malignancies, the solid-tumour microenvironment (TME) has presented serious challenges for therapy efficacy. Particularly, the TME has immunosuppressive signalling and presents a metabolically challenging environment that leads to T-cell suppression. T-cell metabolism is an expanding field of research with a focus on understanding its inherent link to T-cell function. Here, we review the current model of T-cell metabolism from naïve cells through effector and memory life stages, as well as updates to the model from recent literature. These models of metabolism have provided us with the tools and understanding to explore T-cell metabolic and mitochondrial insufficiency in the TME. We discuss manipulations that can be made to these mitochondrial and metabolic pathways to enhance the persistence of infused T cells, overcome the metabolically challenging TME and improve the efficacy of therapy in ACT models. Further understanding and investigation of the impact of metabolic pathways on T-cell performance could contribute to improving therapy efficacy for patients.


Immunotherapy, Adoptive , T-Lymphocytes , Humans , Immunotherapy, Adoptive/methods , Animals , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Cellular Reprogramming/immunology , Neoplasms/immunology , Neoplasms/therapy , Mitochondria/metabolism , Mitochondria/immunology
6.
Science ; 383(6679): eadf6493, 2024 01 12.
Article En | MEDLINE | ID: mdl-38207030

Neutrophils are increasingly recognized as key players in the tumor immune response and are associated with poor clinical outcomes. Despite recent advances characterizing the diversity of neutrophil states in cancer, common trajectories and mechanisms governing the ontogeny and relationship between these neutrophil states remain undefined. Here, we demonstrate that immature and mature neutrophils that enter tumors undergo irreversible epigenetic, transcriptional, and proteomic modifications to converge into a distinct, terminally differentiated dcTRAIL-R1+ state. Reprogrammed dcTRAIL-R1+ neutrophils predominantly localize to a glycolytic and hypoxic niche at the tumor core and exert pro-angiogenic function that favors tumor growth. We found similar trajectories in neutrophils across multiple tumor types and in humans, suggesting that targeting this program may provide a means of enhancing certain cancer immunotherapies.


Cellular Reprogramming , Neoplasms , Neovascularization, Pathologic , Neutrophils , Humans , Neoplasms/blood supply , Neoplasms/immunology , Neutrophils/immunology , Proteomics , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Epigenesis, Genetic , Hypoxia , Transcription, Genetic
7.
Clin Transl Med ; 11(12): e634, 2021 12.
Article En | MEDLINE | ID: mdl-34965016

BACKGROUND: Although microbioa-based therapies have shown putative effects on the treatment of non-alcoholic fatty liver disease (NAFLD), it is not clear how microbiota-derived metabolites contribute to the prevention of NAFLD. We explored the metabolomic signature of Lactobacillus lactis and Pediococcus pentosaceus in NAFLD mice and its association in NAFLD patients. METHODS: We used Western diet-induced NAFLD mice, and L. lactis and P. pentosaceus were administered to animals in the drinking water at a concentration of 109 CFU/g for 8 weeks. NAFLD severity was determined based on liver/body weight, pathology and biochemistry markers. Caecal samples were collected for the metagenomics by 16S rRNA sequencing. Metabolite profiles were obtained from caecum, liver and serum. Human stool samples (healthy control [n = 22] and NAFLD patients [n = 23]) were collected to investigate clinical reproducibility for microbiota-derived metabolites signature and metabolomics biomarker. RESULTS: L. lactis and P. pentosaceus supplementation effectively normalized weight ratio, NAFLD activity score, biochemical markers, cytokines and gut-tight junction. While faecal microbiota varied according to the different treatments, key metabolic features including short chain fatty acids (SCFAs), bile acids (BAs) and tryptophan metabolites were analogously restored by both probiotic supplementations. The protective effects of indole compounds were validated with in vitro and in vivo models, including anti-inflammatory effects. The metabolomic signatures were replicated in NAFLD patients, accompanied by the comparable levels of Firmicutes/Bacteroidetes ratio, which was significantly higher (4.3) compared with control (0.6). Besides, the consequent biomarker panel with six stool metabolites (indole, BAs, and SCFAs) showed 0.922 (area under the curve) in the diagnosis of NAFLD. CONCLUSIONS: NAFLD progression was robustly associated with metabolic dys-regulations in the SCFAs, bile acid and indole compounds, and NAFLD can be accurately diagnosed using the metabolites. L. lactis and P. pentosaceus ameliorate NAFLD progression by modulating gut metagenomic and metabolic environment, particularly tryptophan pathway, of the gut-liver axis.


Cellular Reprogramming/immunology , Gastrointestinal Microbiome/immunology , Lactobacillus/metabolism , Metabolome/immunology , Non-alcoholic Fatty Liver Disease/drug therapy , Pediococcus pentosaceus/metabolism , Animals , Benzofurans/metabolism , Cellular Reprogramming/physiology , Diet, Western/adverse effects , Disease Models, Animal , Feces/microbiology , Gastrointestinal Microbiome/physiology , Lactobacillus/pathogenicity , Metabolome/physiology , Mice , Non-alcoholic Fatty Liver Disease/physiopathology , Pediococcus pentosaceus/pathogenicity , Quinolines/metabolism
8.
Int J Mol Sci ; 22(19)2021 Sep 24.
Article En | MEDLINE | ID: mdl-34638609

Immune escape is one of the hallmarks of cancer. While metabolic reprogramming provides survival advantage to tumor cancer cells, accumulating data also suggest such metabolic rewiring directly affects the activation, differentiation and function of immune cells, particularly in the tumor microenvironment. Understanding how metabolic reprogramming affects both tumor and immune cells, as well as their interplay, is therefore critical to better modulate tumor immune microenvironment in the era of cancer immunotherapy. In this review, we discuss alterations in several essential metabolic pathways in both tumor and key immune cells, provide evidence on their dynamic interaction, and propose innovative strategies to improve cancer immunotherapy via the modulation of metabolic pathways.


Immunotherapy/methods , Neoplasms/therapy , Animals , Cellular Reprogramming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Progression , Humans , Immunotherapy/trends , Macrophages/immunology , Macrophages/metabolism , Metabolic Networks and Pathways , Neoplasms/immunology , Neoplasms/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology
9.
Mol Ther ; 29(11): 3192-3204, 2021 11 03.
Article En | MEDLINE | ID: mdl-34563675

Cell therapies based on reprogrammed adaptive immune cells have great potential as "living drugs." As first demonstrated clinically for engineered chimeric antigen receptor (CAR) T cells, the ability of such cells to undergo clonal expansion in response to an antigen promotes both self-renewal and self-regulation in vivo. B cells also have the potential to be developed as immune cell therapies, but engineering their specificity and functionality is more challenging than for T cells. In part, this is due to the complexity of the immunoglobulin (Ig) locus, as well as the requirement for regulated expression of both cell surface B cell receptor and secreted antibody isoforms, in order to fully recapitulate the features of natural antibody production. Recent advances in genome editing are now allowing reprogramming of B cells by site-specific engineering of the Ig locus with preformed antibodies. In this review, we discuss the potential of engineered B cells as a cell therapy, the challenges involved in editing the Ig locus and the advances that are making this possible, and envision future directions for this emerging field of immune cell engineering.


B-Lymphocytes/metabolism , CRISPR-Cas Systems , Cell- and Tissue-Based Therapy/methods , Gene Editing , Genetic Therapy/methods , Immunotherapy/methods , Animals , Antibodies/genetics , Antibodies/immunology , B-Lymphocytes/immunology , Cell Engineering , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Gene Expression Regulation , Genetic Engineering , Humans , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
10.
Immunity ; 54(9): 2024-2041.e8, 2021 09 14.
Article En | MEDLINE | ID: mdl-34473957

Sepsis results in elevated adenosine in circulation. Extracellular adenosine triggers immunosuppressive signaling via the A2a receptor (A2aR). Sepsis survivors develop persistent immunosuppression with increased risk of recurrent infections. We utilized the cecal ligation and puncture (CLP) model of sepsis and subsequent infection to assess the role of adenosine in post-sepsis immune suppression. A2aR-deficient mice showed improved resistance to post-sepsis infections. Sepsis expanded a subset of CD39hi B cells and elevated extracellular adenosine, which was absent in mice lacking CD39-expressing B cells. Sepsis-surviving B cell-deficient mice were more resistant to secondary infections. Mechanistically, metabolic reprogramming of septic B cells increased production of ATP, which was converted into adenosine by CD39 on plasmablasts. Adenosine signaling via A2aR impaired macrophage bactericidal activity and enhanced interleukin-10 production. Septic individuals exhibited expanded CD39hi plasmablasts and adenosine accumulation. Our study reveals CD39hi plasmablasts and adenosine as important drivers of sepsis-induced immunosuppression with relevance in human disease.


Adenosine/immunology , Antigens, CD/immunology , Apyrase/immunology , Immune Tolerance/immunology , Macrophages/immunology , Plasma Cells/immunology , Sepsis/immunology , Adenosine/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Cellular Reprogramming/immunology , Macrophages/metabolism , Mice , Plasma Cells/metabolism , Receptor, Adenosine A2A/immunology , Receptor, Adenosine A2A/metabolism , Sepsis/metabolism
11.
Front Immunol ; 12: 706583, 2021.
Article En | MEDLINE | ID: mdl-34489958

The burgeoning field of innate immune training, also called trained immunity, has given immunologists new insights into the role of innate responses in protection against infection and in modulating inflammation. Moreover, it has led to a paradigm shift in the way we think about immune memory and the interplay between innate and adaptive immune systems in conferring immunity against pathogens. Trained immunity is the term used to describe the medium-term epigenetic and metabolic reprogramming of innate immune cells in peripheral tissues or in the bone marrow stem cell niche. It is elicited by an initial challenge, followed by a significant period of rest that results in an altered response to a subsequent, unrelated challenge. Trained immunity can be associated with increased production of proinflammatory mediators, such as IL-1ß, TNF and IL-6, and increased expression of markers on innate immune cells associated with antigen presentation to T cells. The microenvironment created by trained innate immune cells during the secondary challenge may have profound effects on T cell responses, such as altering the differentiation, polarisation and function of T cell subtypes, including Th17 cells. In addition, the Th1 cytokine IFN-γ plays a critical role in establishing trained immunity. In this review, we discuss the evidence that trained immunity impacts on or can be impacted by T cells. Understanding the interplay between innate immune training and how it effects adaptive immunity will give insights into how this phenomenon may affect the development or progression of disease and how it could be exploited for therapeutic interventions or to enhance vaccine efficacy.


Adaptive Immunity/immunology , Immunity, Innate/immunology , Immunologic Memory/immunology , T-Lymphocytes/immunology , Animals , Cellular Reprogramming/immunology , Epigenesis, Genetic/immunology , Humans
12.
Front Immunol ; 12: 714822, 2021.
Article En | MEDLINE | ID: mdl-34367185

Advances in understanding how cancer cells interact with the immune system allowed the development of immunotherapeutic strategies, harnessing patients' immune system to fight cancer. Dendritic cell-based vaccines are being explored to reactivate anti-tumor adaptive immunity. Immune checkpoint inhibitors and chimeric antigen receptor T-cells (CAR T) were however the main approaches that catapulted the therapeutic success of immunotherapy. Despite their success across a broad range of human cancers, many challenges remain for basic understanding and clinical progress as only a minority of patients benefit from immunotherapy. In addition, cellular immunotherapies face important limitations imposed by the availability and quality of immune cells isolated from donors. Cell fate reprogramming is offering interesting alternatives to meet these challenges. Induced pluripotent stem cell (iPSC) technology not only enables studying immune cell specification but also serves as a platform for the differentiation of a myriad of clinically useful immune cells including T-cells, NK cells, or monocytes at scale. Moreover, the utilization of iPSCs allows introduction of genetic modifications and generation of T/NK cells with enhanced anti-tumor properties. Immune cells, such as macrophages and dendritic cells, can also be generated by direct cellular reprogramming employing lineage-specific master regulators bypassing the pluripotent stage. Thus, the cellular reprogramming toolbox is now providing the means to address the potential of patient-tailored immune cell types for cancer immunotherapy. In parallel, development of viral vectors for gene delivery has opened the door for in vivo reprogramming in regenerative medicine, an elegant strategy circumventing the current limitations of in vitro cell manipulation. An analogous paradigm has been recently developed in cancer immunotherapy by the generation of CAR T-cells in vivo. These new ideas on endogenous reprogramming, cross-fertilized from the fields of regenerative medicine and gene therapy, are opening exciting avenues for direct modulation of immune or tumor cells in situ, widening our strategies to remove cancer immunotherapy roadblocks. Here, we review current strategies for cancer immunotherapy, summarize technologies for generation of immune cells by cell fate reprogramming as well as highlight the future potential of inducing these unique cell identities in vivo, providing new and exciting tools for the fast-paced field of cancer immunotherapy.


Antineoplastic Agents, Immunological/therapeutic use , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Immunotherapy , Molecular Targeted Therapy , Neoplasms/etiology , Neoplasms/therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/therapeutic use , Cellular Reprogramming Techniques , Combined Modality Therapy , Disease Management , Genetic Engineering , Genetic Therapy , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy, Adoptive , Molecular Targeted Therapy/methods
13.
Front Immunol ; 12: 717421, 2021.
Article En | MEDLINE | ID: mdl-34394124

Regulatory T-cells (Tregs) are important for maintaining self-tolerance and tissue homeostasis. The functional plasticity of Tregs is a key feature of this lineage, as it allows them to adapt to different microenvironments, adopt transcriptional programs reflective of their environments and tailor their suppressive capacity in a context-dependent fashion. Tregs, particularly effector Tregs (eTregs), are abundant in many types of tumors. However, the functional and transcriptional plasticity of eTregs in tumors remain largely to be explored. Although depletion or inhibition of systemic Tregs can enhance anti-tumor responses, autoimmune sequelae have diminished the enthusiasm for such approaches. A more effective approach should specifically target intratumoral Tregs or subvert local Treg-mediated suppression. This mini-review will discuss the reported mechanisms by which the stability and suppressive function of tumoral Tregs are modulated, with the focus on eTregs and a subset of eTregs, follicular regulatory T (TFR) cells, and how to harness this knowledge for the future development of new effective cancer immunotherapies that selectively target the tumor local response while sparing the systemic side effects.


Cellular Reprogramming , Disease Susceptibility , Neoplasms/etiology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cell Lineage , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Humans , Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
15.
J Clin Invest ; 131(13)2021 07 01.
Article En | MEDLINE | ID: mdl-34043588

Peripheral T cell lymphomas (PTCLs) represent a significant unmet medical need with dismal clinical outcomes. The T cell receptor (TCR) is emerging as a key driver of T lymphocyte transformation. However, the role of chronic TCR activation in lymphomagenesis and in lymphoma cell survival is still poorly understood. Using a mouse model, we report that chronic TCR stimulation drove T cell lymphomagenesis, whereas TCR signaling did not contribute to PTCL survival. The combination of kinome, transcriptome, and epigenome analyses of mouse PTCLs revealed a NK cell-like reprogramming of PTCL cells with expression of NK receptors (NKRs) and downstream signaling molecules such as Tyrobp and SYK. Activating NKRs were functional in PTCLs and dependent on SYK activity. In vivo blockade of NKR signaling prolonged mouse survival, demonstrating the addiction of PTCLs to NKRs and downstream SYK/mTOR activity for their survival. We studied a large collection of human primary samples and identified several PTCLs recapitulating the phenotype described in this model by their expression of SYK and the NKR, suggesting a similar mechanism of lymphomagenesis and establishing a rationale for clinical studies targeting such molecules.


Lymphoma, T-Cell, Peripheral/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Natural Killer Cell/immunology , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, p53 , Humans , Killer Cells, Natural/immunology , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Natural Killer Cell/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Syk Kinase/metabolism , T-Lymphocytes/immunology
16.
Front Immunol ; 12: 609762, 2021.
Article En | MEDLINE | ID: mdl-33968014

Cancer is one of the leading causes of death and a major public health problem all over the world. Immunotherapy is becoming a revolutionary clinical management for various cancer types. Restoration of aberrant immune surveillance on cancers has achieved markable progress in the past years by either in vivo or ex vivo engineering of the immune cells. Here, we summarized the central roles of immune cells in tumor progression and regression, and the existing and emerging strategies for different immune cell-based immunotherapies. In addition, the current challenges and the potential solutions in translating the immunotherapies into the clinic are also discussed.


Cellular Reprogramming/immunology , Immunomodulation , Neoplasms/immunology , Neoplasms/therapy , Animals , Cellular Reprogramming/genetics , Genetic Engineering , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/diagnosis , Neoplasms/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
17.
Nat Commun ; 12(1): 2582, 2021 05 11.
Article En | MEDLINE | ID: mdl-33976133

Immune checkpoint blockers (ICBs) have failed in all phase III glioblastoma (GBM) trials. Here, we show that regulatory T (Treg) cells play a key role in GBM resistance to ICBs in experimental gliomas. Targeting glucocorticoid-induced TNFR-related receptor (GITR) in Treg cells using an agonistic antibody (αGITR) promotes CD4 Treg cell differentiation into CD4 effector T cells, alleviates Treg cell-mediated suppression of anti-tumor immune response, and induces potent anti-tumor effector cells in GBM. The reprogrammed GBM-infiltrating Treg cells express genes associated with a Th1 response signature, produce IFNγ, and acquire cytotoxic activity against GBM tumor cells while losing their suppressive function. αGITR and αPD1 antibodies increase survival benefit in three experimental GBM models, with a fraction of cohorts exhibiting complete tumor eradication and immune memory upon tumor re-challenge. Moreover, αGITR and αPD1 synergize with the standard of care treatment for newly-diagnosed GBM, enhancing the cure rates in these GBM models.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , Glucocorticoid-Induced TNFR-Related Protein/agonists , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/transplantation , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology , Disease Models, Animal , Female , Glioblastoma/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunologic Memory/drug effects , Male , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
18.
Leukemia ; 35(12): 3482-3496, 2021 12.
Article En | MEDLINE | ID: mdl-34021248

Immunotherapies have heralded a new era in the cancer treatment. In addition to checkpoint inhibitors, agonistic antibodies against co-stimulatory immune receptors hold the potential to invoke efficient antitumor immunity. Targeting CD137 has gained momentum based on its ability to drive NK- and T-cell-based responses. CD137-engaging mAbs have already entered clinical trials for different types of tumors showing promising results. Despite the efforts to translate CD137-mediated immunotherapy into clinical practice, little remains known regarding the role of CD137 in human monocytes/macrophages.We found CD137 being expressed on monocytes of healthy controls and at even higher levels in patients with multiple myeloma or CLL. CD137HI(GH) monocytes displayed a distinct phenotypic, transcriptomic, and metabolic profile. They possessed an increased phagocytic capacity enabling superior antibody-dependent phagocytosis (ADPC) of multiple myeloma and lymphoma cells that were treated with anti-CD38 or anti-CD20 mAbs. Triggering CD137 promoted both metabolic and tumoricidal activity in an extracellular signal-regulated kinase (ERK)-dependent fashion. In addition, we observed a phenotypic, transcriptomic, and functional skewing towards a M1-like phenotype.Overall, we introduce CD137 as a positive immune checkpoint on human monocytes/macrophages, which can have therapeutic implications especially in view of synergistic effects when combining CD137 agonists with tumor-targeting antibodies.


Immunotherapy/methods , Macrophages/immunology , Monocytes/immunology , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity , Cells, Cultured , Cellular Reprogramming/immunology , Humans , Killer Cells, Natural/immunology , Macrophages/metabolism , Monocytes/metabolism , Multiple Myeloma/blood , Multiple Myeloma/metabolism , Phagocytosis , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
19.
Nat Med ; 27(6): 1043-1054, 2021 06.
Article En | MEDLINE | ID: mdl-34017133

Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are prevalent liver conditions that underlie the development of life-threatening cirrhosis, liver failure and liver cancer. Chronic necro-inflammation is a critical factor in development of NASH, yet the cellular and molecular mechanisms of immune dysregulation in this disease are poorly understood. Here, using single-cell transcriptomic analysis, we comprehensively profiled the immune composition of the mouse liver during NASH. We identified a significant pathology-associated increase in hepatic conventional dendritic cells (cDCs) and further defined their source as NASH-induced boost in cycling of cDC progenitors in the bone marrow. Analysis of blood and liver from patients on the NAFLD/NASH spectrum showed that type 1 cDCs (cDC1) were more abundant and activated in disease. Sequencing of physically interacting cDC-T cell pairs from liver-draining lymph nodes revealed that cDCs in NASH promote inflammatory T cell reprogramming, previously associated with NASH worsening. Finally, depletion of cDC1 in XCR1DTA mice or using anti-XCL1-blocking antibody attenuated liver pathology in NASH mouse models. Overall, our study provides a comprehensive characterization of cDC biology in NASH and identifies XCR1+ cDC1 as an important driver of liver pathology.


Dendritic Cells/immunology , Fatty Liver/immunology , Non-alcoholic Fatty Liver Disease/immunology , Receptors, Chemokine/genetics , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Dendritic Cells/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/pathology , Female , Humans , Liver/immunology , Liver/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Chemokine/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
20.
Front Immunol ; 12: 631353, 2021.
Article En | MEDLINE | ID: mdl-34017325

Acute graft-vs.-host (GVHD) disease remains a common complication of allogeneic stem cell transplantation with very poor outcomes once the disease becomes steroid refractory. Mesenchymal stem cells (MSCs) represent a promising therapeutic approach for the treatment of GVHD, but so far this strategy has had equivocal clinical efficacy. Therapies using MSCs require optimization taking advantage of the plasticity of these cells in response to different microenvironments. In this study, we aimed to optimize cord blood tissue derived MSCs (CBti MSCs) by priming them using a regimen of inflammatory cytokines. This approach led to their metabolic reprogramming with enhancement of their glycolytic capacity. Metabolically reprogrammed CBti MSCs displayed a boosted immunosuppressive potential, with superior immunomodulatory and homing properties, even after cryopreservation and thawing. Mechanistically, primed CBti MSCs significantly interfered with glycolytic switching and mTOR signaling in T cells, suppressing T cell proliferation and ensuing polarizing toward T regulatory cells. Based on these data, we generated a Good Manufacturing Process (GMP) Laboratory protocol for the production and cryopreservation of primed CBti MSCs for clinical use. Following thawing, these cryopreserved GMP-compliant primed CBti MSCs significantly improved outcomes in a xenogenic mouse model of GVHD. Our data support the concept that metabolic profiling of MSCs can be used as a surrogate for their suppressive potential in conjunction with conventional functional methods to support their therapeutic use in GVHD or other autoimmune disorders.


Cellular Reprogramming Techniques/methods , Cellular Reprogramming/physiology , Fetal Blood/cytology , Graft vs Host Disease/prevention & control , Mesenchymal Stem Cells/metabolism , Animals , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology , Cytokines/pharmacology , Female , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred NOD , Quality Control
...