Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.203
Filter
1.
Mol Biol Rep ; 51(1): 571, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662155

ABSTRACT

Leukemia is a malignancy of the bone marrow and blood originating from self-renewing cancerous immature blast cells or transformed leukocytes. Despite improvements in treatments, leukemia remains still a serious disease with poor prognosis because of disease heterogeneity, drug resistance and relapse. There is emerging evidence that differentially expression of co-signaling molecules play a critical role in tumor immune evasion. Galectin-9 (Gal-9) is one of the key proteins that leukemic cells express, secrete, and use to proliferate, self-renew, and survive. It also suppresses host immune responses controlled by T and NK cells, enabling leukemic cells to evade immune surveillance. The present review provides the molecular mechanisms of Gal-9-induced immune evasion in leukemia. Understanding the complex immune evasion machinery driven by Gal-9 expressing leukemic cells will enable the identification of novel therapeutic strategies for efficient immunotherapy in leukemic patients. Combined treatment approaches targeting T-cell immunoglobulin and mucin domain-3 (Tim-3)/Gal-9 and other immune checkpoint pathways can be considered, which may enhance the efficacy of host effector cells to attack leukemic cells.


Subject(s)
Cell Transformation, Neoplastic , Galectins , Hepatitis A Virus Cellular Receptor 2 , Leukemia , Humans , Galectins/metabolism , Leukemia/immunology , Hepatitis A Virus Cellular Receptor 2/metabolism , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/genetics , Animals , Immune Tolerance , Signal Transduction , Tumor Escape , Cell Proliferation , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism
2.
Front Immunol ; 15: 1382099, 2024.
Article in English | MEDLINE | ID: mdl-38665912

ABSTRACT

Introduction: Chimerism is closely correlated with disease relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, chimerism rate is dynamic changes, and the sensitivity of different chimerism requires further research. Methods: To investigate the predictive value of distinct chimerism for relapse, we measured bone marrow (BM), peripheral blood (PB), and T-cell (isolated from BM) chimerism in 178 patients after allo-HSCT. Results: Receiver operating characteristic (ROC) curve showed that T-cell chimerism was more suitable to predict relapse after allo-HSCT compared with PB and BM chimerism. The cutoff value of T-cell chimerism for predicting relapse was 99.45%. Leukemia and myelodysplastic syndrome (MDS) relapse patients' T-cell chimerism was a gradual decline from 2 months to 9 months after allo-HSCT. Higher risk of relapse and death within 1 year after allo-HSCT. The T-cell chimerism rates in remission and relapse patients were 99.43% and 94.28% at 3 months after allo-HSCT (P = 0.009), 99.31% and 95.27% at 6 months after allo-HSCT (P = 0.013), and 99.26% and 91.32% at 9 months after allo-HSCT (P = 0.024), respectively. There was a significant difference (P = 0.036) for T-cell chimerism between early relapse (relapse within 9 months after allo-HSCT) and late relapse (relapse after 9 months after allo-HSCT) at 2 months after allo-HSCT. Every 1% increase in T-cell chimerism, the hazard ratio for disease relapse was 0.967 (95% CI: 0.948-0.987, P<0.001). Discussion: We recommend constant monitoring T-cell chimerism at 2, 3, 6, and 9 months after allo-HSCT to predict relapse.


Subject(s)
Hematopoietic Stem Cell Transplantation , Recurrence , T-Lymphocytes , Transplantation Chimera , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Female , Adult , Middle Aged , T-Lymphocytes/immunology , Transplantation Chimera/immunology , Adolescent , Young Adult , Child , Child, Preschool , Chimerism , Myelodysplastic Syndromes/therapy , Myelodysplastic Syndromes/immunology , Leukemia/therapy , Leukemia/immunology , Leukemia/mortality , Predictive Value of Tests , Graft vs Host Disease/immunology , Graft vs Host Disease/etiology
3.
Exp Hematol ; 133: 104211, 2024 May.
Article in English | MEDLINE | ID: mdl-38527589

ABSTRACT

Leukemias are a set of clonal hematopoietic malignant diseases that develop in the bone marrow. Several factors influence leukemia development and progression. Among these, the gut microbiota is a major factor influencing a wide array of its processes. The gut microbial composition is linked to the risk of tumor development and the host's ability to respond to treatment, mostly due to the immune-modulatory effects of their metabolites. Despite such strong evidence, its role in the development of hematologic malignancies still requires attention of investigators worldwide. In this review, we make an effort to discuss the role of host gut microbiota-immune crosstalk in leukemia development and progression. Additionally, we highlight certain recently developed strategies to modify the gut microbial composition that may help to overcome dysbiosis in leukemia patients in the near future.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Leukemia , Humans , Dysbiosis/immunology , Dysbiosis/complications , Gastrointestinal Microbiome/immunology , Leukemia/immunology , Leukemia/microbiology , Leukemia/etiology , Animals
4.
Nature ; 615(7951): 315-322, 2023 03.
Article in English | MEDLINE | ID: mdl-36755094

ABSTRACT

Further advances in cell engineering are needed to increase the efficacy of chimeric antigen receptor (CAR) and other T cell-based therapies1-5. As T cell differentiation and functional states are associated with distinct epigenetic profiles6,7, we hypothesized that epigenetic programming may provide a means to improve CAR T cell performance. Targeting the gene that encodes the epigenetic regulator ten-eleven translocation 2 (TET2)8 presents an interesting opportunity as its loss may enhance T cell memory9,10, albeit not cause malignancy9,11,12. Here we show that disruption of TET2 enhances T cell-mediated tumour rejection in leukaemia and prostate cancer models. However, loss of TET2 also enables antigen-independent CAR T cell clonal expansions that may eventually result in prominent systemic tissue infiltration. These clonal proliferations require biallelic TET2 disruption and sustained expression of the AP-1 factor BATF3 to drive a MYC-dependent proliferative program. This proliferative state is associated with reduced effector function that differs from both canonical T cell memory13,14 and exhaustion15,16 states, and is prone to the acquisition of secondary somatic mutations, establishing TET2 as a guardian against BATF3-induced CAR T cell proliferation and ensuing genomic instability. Our findings illustrate the potential of epigenetic programming to enhance T cell immunity but highlight the risk of unleashing unchecked proliferative responses.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Cell Proliferation , DNA-Binding Proteins , Dioxygenases , Immunotherapy, Adoptive , Lymphocyte Activation , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Male , Cell Differentiation/genetics , Dioxygenases/metabolism , DNA-Binding Proteins/metabolism , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Leukemia/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Epigenesis, Genetic , Immunologic Memory , Basic-Leucine Zipper Transcription Factors/metabolism
5.
Nature ; 609(7925): 174-182, 2022 09.
Article in English | MEDLINE | ID: mdl-36002574

ABSTRACT

The efficacy of adoptive T cell therapies for cancer treatment can be limited by suppressive signals from both extrinsic factors and intrinsic inhibitory checkpoints1,2. Targeted gene editing has the potential to overcome these limitations and enhance T cell therapeutic function3-10. Here we performed multiple genome-wide CRISPR knock-out screens under different immunosuppressive conditions to identify genes that can be targeted to prevent T cell dysfunction. These screens converged on RASA2, a RAS GTPase-activating protein (RasGAP) that we identify as a signalling checkpoint in human T cells, which is downregulated upon acute T cell receptor stimulation and can increase gradually with chronic antigen exposure. RASA2 ablation enhanced MAPK signalling and chimeric antigen receptor (CAR) T cell cytolytic activity in response to target antigen. Repeated tumour antigen stimulations in vitro revealed that RASA2-deficient T cells show increased activation, cytokine production and metabolic activity compared with control cells, and show a marked advantage in persistent cancer cell killing. RASA2-knockout CAR T cells had a competitive fitness advantage over control cells in the bone marrow in a mouse model of leukaemia. Ablation of RASA2 in multiple preclinical models of T cell receptor and CAR T cell therapies prolonged survival in mice xenografted with either liquid or solid tumours. Together, our findings highlight RASA2 as a promising target to enhance both persistence and effector function in T cell therapies for cancer treatment.


Subject(s)
Antigens, Neoplasm , Neoplasms , T-Lymphocytes , ras GTPase-Activating Proteins , Animals , Antigens, Neoplasm/immunology , Bone Marrow , CRISPR-Cas Systems , Disease Models, Animal , Gene Knockdown Techniques , Humans , Immunotherapy, Adoptive , Leukemia/immunology , Leukemia/pathology , Leukemia/therapy , Mice , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Xenograft Model Antitumor Assays , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
6.
Sci Rep ; 12(1): 3234, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35217717

ABSTRACT

Leukemic cells proliferate faster than non-transformed counterparts. This requires them to change their metabolism to adapt to their high growth. This change can stress cells and facilitate recognition by immune cells such as cytotoxic lymphocytes, which express the activating receptor Natural Killer G2-D (NKG2D). The tumor suppressor gene p53 regulates cell metabolism, but its role in the expression of metabolism-induced ligands, and subsequent recognition by cytotoxic lymphocytes, is unknown. We show here that dichloroacetate (DCA), which induces oxidative phosphorylation (OXPHOS) in tumor cells, induces the expression of such ligands, e.g. MICA/B, ULBP1 and ICAM-I, by a wtp53-dependent mechanism. Mutant or null p53 have the opposite effect. Conversely, DCA sensitizes only wtp53-expressing cells to cytotoxic lymphocytes, i.e. cytotoxic T lymphocytes and NK cells. In xenograft in vivo models, DCA slows down the growth of tumors with low proliferation. Treatment with DCA, monoclonal antibodies and NK cells also decreased tumors with high proliferation. Treatment of patients with DCA, or a biosimilar drug, could be a clinical option to increase the effectiveness of CAR T cell or allogeneic NK cell therapies.


Subject(s)
Antineoplastic Agents , Leukemia , Tumor Suppressor Protein p53 , Antineoplastic Agents/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia/immunology , Leukemia/metabolism , Ligands , NK Cell Lectin-Like Receptor Subfamily K/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
7.
Nature ; 602(7897): 503-509, 2022 02.
Article in English | MEDLINE | ID: mdl-35110735

ABSTRACT

The adoptive transfer of T lymphocytes reprogrammed to target tumour cells has demonstrated potential for treatment of various cancers1-7. However, little is known about the long-term potential and clonal stability of the infused cells. Here we studied long-lasting CD19-redirected chimeric antigen receptor (CAR) T cells in two patients with chronic lymphocytic leukaemia1-4 who achieved a complete remission in 2010. CAR T cells remained detectable more than ten years after infusion, with sustained remission in both patients. Notably, a highly activated CD4+ population emerged in both patients, dominating the CAR T cell population at the later time points. This transition was reflected in the stabilization of the clonal make-up of CAR T cells with a repertoire dominated by a small number of clones. Single-cell profiling demonstrated that these long-persisting CD4+ CAR T cells exhibited cytotoxic characteristics along with ongoing functional activation and proliferation. In addition, longitudinal profiling revealed a population of gamma delta CAR T cells that prominently expanded in one patient concomitant with CD8+ CAR T cells during the initial response phase. Our identification and characterization of these unexpected CAR T cell populations provide novel insight into the CAR T cell characteristics associated with anti-cancer response and long-term remission in leukaemia.


Subject(s)
CD4-Positive T-Lymphocytes , Immunotherapy, Adoptive , Leukemia , Receptors, Chimeric Antigen , Antigens, CD19/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Humans , Leukemia/immunology , Leukemia/therapy , Receptors, Chimeric Antigen/immunology , Time Factors
8.
Int J Hematol ; 115(3): 371-381, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35037229

ABSTRACT

Genomic deletion of donor-patient-mismatched HLA alleles in leukemic cells is a major cause of relapse after allogeneic hematopoietic stem cell transplantation (HSCT). Mismatched HLA is frequently lost as an individual allele or a whole region in HLA-class I, however, it is downregulated in HLA-class II. We hypothesized that there might be a difference in T cell recognition capacity against epitopes associated with HLA-class I and HLA-class II and consequently such allogeneic immune pressure induced HLA alterations in leukemic cells. To investigate this, we conducted in vitro experiments with T cell receptor-transduced T (TCR-T) cells. The cytotoxic activity of NY-ESO-1-specific TCR-T cells exhibited similarly against K562 cells with low HLA-A*02:01 expression. However, we demonstrated that the cytokine production against low HLA-DPB1*05:01 expression line decreased gradually from the HLA expression level approximately 2-log lower than normal expressors. Using sort-purified leukemia cells before and after HSCT, we applied the next-generation sequencing, and revealed that there were several marked downregulations of HLA-class II alleles which demonstrated consistently low expression from pre-transplantation. The marked downregulation of HLA-class II may lead to decreased antigen recognition ability of antigen-specific T cells and may be one of immune evasion mechanism associated with HLA-class II downregulation.


Subject(s)
Down-Regulation , Epitopes/immunology , Hematopoietic Stem Cell Transplantation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Leukemia/genetics , Leukemia/immunology , T-Lymphocytes/immunology , Transplantation, Homologous , Alleles , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Humans , K562 Cells , Leukemia/therapy , Recurrence
9.
Cells ; 11(1)2022 01 05.
Article in English | MEDLINE | ID: mdl-35011737

ABSTRACT

David Mason started his research career at a time when lymphoma diagnosis was based primarily on cellular morphology, clinical symptoms and special cytochemical stains using formalin fixed tissue sections. There were occasions, however, where the morphology was unhelpful, such as in the case of anaplastic or poorly differentiated tumours, where a distinction between lymphoma and a non-haematopoietic tumour was often problematical. Accurate diagnosis became even more important with the developments in the clinical staging of lymphoma and the availability of more effective treatments. One way forward to improve diagnosis was to use immunohistochemistry to study the antigens expressed by the tumor cells.


Subject(s)
Carcinoma/pathology , Cooperative Behavior , Lymphoma/pathology , Carcinoma/diagnosis , Carcinoma/immunology , Humans , Leukemia/diagnosis , Leukemia/immunology , Lymphoma/diagnosis , Lymphoma/immunology
10.
Leukemia ; 36(2): 525-531, 2022 02.
Article in English | MEDLINE | ID: mdl-34545183

ABSTRACT

Chronic eosinophilic leukemia-not otherwise specified (CEL-NOS) is a rare, aggressive, fatal disease characterized by blood eosinophilia and dysfunction of organs infiltrated with eosinophils. Clinically, the disease manifests with weight loss, cough, weakness, diarrhea, and multi-organ dysfunction that is unresponsive to therapy. We developed a one-time gene therapy for CEL-NOS using an adeno-associated virus (AAV) expressing an anti-eosinophil monoclonal antibody (AAVrh.10mAnti-Eos) to provide sustained suppression of eosinophil numbers in blood, thus reducing eosinophil tissue invasion and organ dysfunction. A novel CEL-NOS model was developed in NOD-scid IL2rγnull (NSG) mice by administration of AAV expressing the cytokine IL5 (AAVrh.10mIL5), resulting in marked peripheral and tissue eosinophilia of the heart, lung, liver, and spleen, and eventually death. Mice were administered AAVrh.10mAnti-Eos (1011 genome copies) 4 wk after administration of AAVrh.10mIL5 and evaluated for anti-eosinophil antibody expression, blood eosinophil counts, organ eosinophil invasion, and survival. AAVrh.10mAnti-Eos expressed persistent levels of the anti-eosinophil antibody for >24 wk. Strikingly, CEL-NOS treated mice had markedly lower blood eosinophil levels and reduced mortality when compared with control treated mice. These results suggest that a single treatment with AAVrh.10mAnti-Eos has the potential to provide substantial therapeutic benefit to patients with CEL-NOS, a fatal malignant disorder.


Subject(s)
Antibodies, Monoclonal/pharmacology , Dependovirus/genetics , Disease Models, Animal , Eosinophils/immunology , Genetic Therapy , Hypereosinophilic Syndrome/therapy , Interleukin-5/genetics , Leukemia/therapy , Animals , Eosinophils/drug effects , Female , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/immunology , Leukemia/genetics , Leukemia/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID
12.
Br J Haematol ; 196(4): 892-901, 2022 02.
Article in English | MEDLINE | ID: mdl-34761389

ABSTRACT

Patients with haematological malignancies have a high risk of severe infection and death from SARS-CoV-2. In this prospective observational study, we investigated the impact of cancer type, disease activity, and treatment in 877 unvaccinated UK patients with SARS-CoV-2 infection and active haematological cancer. The primary end-point was all-cause mortality. In a multivariate analysis adjusted for age, sex and comorbidities, the highest mortality was in patients with acute leukaemia [odds ratio (OR) = 1·73, 95% confidence interval (CI) 1·1-2·72, P = 0·017] and myeloma (OR 1·3, 95% CI 0·96-1·76, P = 0·08). Having uncontrolled cancer (newly diagnosed awaiting treatment as well as relapsed or progressive disease) was associated with increased mortality risk (OR = 2·45, 95% CI 1·09-5·5, P = 0·03), as was receiving second or beyond line of treatment (OR = 1·7, 95% CI 1·08-2·67, P = 0·023). We found no association between recent cytotoxic chemotherapy or anti-CD19/anti-CD20 treatment and increased risk of death within the limitations of the cohort size. Therefore, disease control is an important factor predicting mortality in the context of SARS-CoV-2 infection alongside the possible risks of therapies such as cytotoxic treatment or anti-CD19/anti-CD20 treatments.


Subject(s)
Antigens, CD20/immunology , Antineoplastic Agents, Immunological/therapeutic use , COVID-19/complications , Hematologic Neoplasms/complications , Hematologic Neoplasms/drug therapy , Adult , Antineoplastic Agents, Immunological/adverse effects , COVID-19/etiology , COVID-19/immunology , Female , Hematologic Neoplasms/immunology , Humans , Leukemia/complications , Leukemia/drug therapy , Leukemia/immunology , Male , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Prospective Studies , Risk Factors
13.
Bull Cancer ; 108(10S): S181-S194, 2021 Oct.
Article in French | MEDLINE | ID: mdl-34920802

ABSTRACT

Bispecific therapies targeting CD3, so-called T-cell engagers (TCE), belong to the new spectrum of anti-tumor immunotherapies stimulating T-lymphocytes. TCE are unique constructs targeting the MHC-independent CD3 epsilon subunit (CD3e) and a tumor antigen. To date, only blinatumomab have reached market agreements in lymphoid malignancies with constructs targeting CD3exCD19. Other TCE are in advances development, with promising results targeting CD20 and BSMA in lymphoma and myeloma. These successes have relaunched the development of TCE in solid tumors, bringing mixed results so far (notably in terms of tolerance). Still, TCE pave the way to new immunotherapy in tumors considered to be refractory to inhibitors of immune checkpoints such as prostate cancer or colorectal cancer.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , T-Lymphocytes/immunology , Antibodies, Bispecific/therapeutic use , Antigens, CD19/immunology , Antigens, CD20/immunology , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Digestive System Neoplasms/immunology , Digestive System Neoplasms/therapy , Female , Humans , Immune Tolerance , Leukemia/immunology , Leukemia/therapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lymphoma/immunology , Lymphoma/therapy , Male , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Neoplasms/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/therapy
15.
J Hematol Oncol ; 14(1): 199, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34809678

ABSTRACT

Although chimeric antigen receptor T cells demonstrated remarkable efficacy in patients with chemo-resistant hematologic malignancies, a significant portion still resist or relapse. This immune evasion may be due to CAR T cells dysfunction, a hostile tumor microenvironment, or resistant cancer cells. Here, we review the intrinsic resistance mechanisms of cancer cells to CAR T cell therapy and potential strategies to circumvent them.


Subject(s)
Hematologic Neoplasms/therapy , Immunotherapy, Adoptive , Animals , Hematologic Neoplasms/immunology , Humans , Immunotherapy, Adoptive/methods , Leukemia/immunology , Leukemia/therapy , Lymphoma/immunology , Lymphoma/therapy , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/therapy , Tumor Escape
16.
Nat Immunol ; 22(12): 1577-1589, 2021 12.
Article in English | MEDLINE | ID: mdl-34811546

ABSTRACT

Single-cell genomics technology has transformed our understanding of complex cellular systems. However, excessive cost and a lack of strategies for the purification of newly identified cell types impede their functional characterization and large-scale profiling. Here, we have generated high-content single-cell proteo-genomic reference maps of human blood and bone marrow that quantitatively link the expression of up to 197 surface markers to cellular identities and biological processes across all main hematopoietic cell types in healthy aging and leukemia. These reference maps enable the automatic design of cost-effective high-throughput cytometry schemes that outperform state-of-the-art approaches, accurately reflect complex topologies of cellular systems and permit the purification of precisely defined cell states. The systematic integration of cytometry and proteo-genomic data enables the functional capacities of precisely mapped cell states to be measured at the single-cell level. Our study serves as an accessible resource and paves the way for a data-driven era in cytometry.


Subject(s)
Blood Cells/metabolism , Bone Marrow Cells/metabolism , Cell Separation , Flow Cytometry , Gene Expression Profiling , Proteome , Proteomics , Single-Cell Analysis , Transcriptome , Age Factors , Blood Cells/immunology , Blood Cells/pathology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cells, Cultured , Databases, Genetic , Healthy Aging/genetics , Healthy Aging/immunology , Healthy Aging/metabolism , Humans , Leukemia/genetics , Leukemia/immunology , Leukemia/metabolism , Leukemia/pathology , RNA-Seq , Systems Biology
17.
Cell Rep ; 37(6): 109992, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758319

ABSTRACT

To elucidate mechanisms by which T cells eliminate leukemia, we study donor lymphocyte infusion (DLI), an established immunotherapy for relapsed leukemia. We model T cell dynamics by integrating longitudinal, multimodal data from 94,517 bone marrow-derived single T cell transcriptomes in addition to chromatin accessibility and single T cell receptor sequencing from patients undergoing DLI. We find that responsive tumors are defined by enrichment of late-differentiated T cells before DLI and rapid, durable expansion of early differentiated T cells after treatment, highly similar to "terminal" and "precursor" exhausted subsets, respectively. Resistance, in contrast, is defined by heterogeneous T cell dysfunction. Surprisingly, early differentiated T cells in responders mainly originate from pre-existing and novel clonotypes recruited to the leukemic microenvironment, rather than the infusion. Our work provides a paradigm for analyzing longitudinal single-cell profiling of scenarios beyond adoptive cell therapy and introduces Symphony, a Bayesian approach to infer regulatory circuitry underlying T cell subsets, with broad relevance to exhaustion antagonists across cancers.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia/immunology , Lymphocyte Activation/immunology , Lymphocyte Transfusion/methods , Neoplasm Recurrence, Local/immunology , Stem Cell Transplantation/methods , T-Lymphocytes/immunology , Clonal Evolution , Humans , Leukemia/pathology , Leukemia/therapy , Longitudinal Studies , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Tissue Donors , Transplantation, Homologous
18.
Nat Commun ; 12(1): 5773, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599159

ABSTRACT

Protein localisation and translocation between intracellular compartments underlie almost all physiological processes. The hyperLOPIT proteomics platform combines mass spectrometry with state-of-the-art machine learning to map the subcellular location of thousands of proteins simultaneously. We combine global proteome analysis with hyperLOPIT in a fully Bayesian framework to elucidate spatiotemporal proteomic changes during a lipopolysaccharide (LPS)-induced inflammatory response. We report a highly dynamic proteome in terms of both protein abundance and subcellular localisation, with alterations in the interferon response, endo-lysosomal system, plasma membrane reorganisation and cell migration. Proteins not previously associated with an LPS response were found to relocalise upon stimulation, the functional consequences of which are still unclear. By quantifying proteome-wide uncertainty through Bayesian modelling, a necessary role for protein relocalisation and the importance of taking a holistic overview of the LPS-driven immune response has been revealed. The data are showcased as an interactive application freely available for the scientific community.


Subject(s)
Inflammation/metabolism , Leukemia/metabolism , Leukemia/pathology , Lipopolysaccharides/pharmacology , Proteomics , Algorithms , Anti-Infective Agents/metabolism , Anti-Inflammatory Agents/metabolism , Antigen Presentation , Autophagosomes/metabolism , Bayes Theorem , Cell Cycle Checkpoints , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Shape , Humans , Immunity , Inflammation/pathology , Leukemia/immunology , Lymphocyte Activation/immunology , Lysosomes/metabolism , Neoplasm Proteins/metabolism , Protein Transport , Proteome/metabolism , Signal Transduction , T-Lymphocytes/immunology , THP-1 Cells , Time Factors , Transport Vesicles/metabolism , Up-Regulation , rho GTP-Binding Proteins/metabolism
19.
Front Immunol ; 12: 730507, 2021.
Article in English | MEDLINE | ID: mdl-34671352

ABSTRACT

To avoid graft rejection, the hematopoietic stem cells with matched classical human leukocyte antigen (HLA) alleles are the primary choice for clinical allogeneic transplantation. However, even if the fully HLA-matched hematopoietic stem cells are used for transplantation, some patients still have poor prognosis after hematopoietic stem cell transplantation (HSCT), suggesting that the HLA system was not the only determinant of the outcomes of HSCT. In this study, we investigated whether the single-nucleotide polymorphisms (SNPs) of the co-stimulatory genes within non-HLA regions were related to the outcomes of HSCT. The genomic DNAs of 163 patients who had acute leukemia and received HSCT and their respective donors were collected for analysis. Thirty-four SNPs located in the four co-stimulatory genes including cytotoxic T-lymphocyte associated protein 4 (CTLA4), CD28, tumor necrosis factor ligand superfamily 4 (TNFSF4), and programmed cell death protein 1 (PDCD1) were selected to explore their relationship with the adverse outcomes after transplantation, including mortality, cytomegalovirus infection, graft-versus-host disease, and relapse. Our results revealed that nine SNPs in the CTLA4 gene, five SNPs in the PDCD1 gene, two SNPs in the TNFSF4 gene, and four SNPs in the CD28 gene were significantly associated with the occurrence of adverse outcomes post-HSCT. These SNPs may play important roles in immune response to allografts post-HSCT and can be the targets for developing strategy to identify appropriate donors.


Subject(s)
CD28 Antigens/genetics , CTLA-4 Antigen/genetics , HLA Antigens/immunology , Hematopoietic Stem Cell Transplantation , Leukemia/surgery , OX40 Ligand/genetics , Polymorphism, Single Nucleotide , Programmed Cell Death 1 Receptor/genetics , Adolescent , Adult , Aged , CD28 Antigens/immunology , CTLA-4 Antigen/immunology , Child , Child, Preschool , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Donor Selection , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Humans , Infant , Leukemia/genetics , Leukemia/immunology , Leukemia/mortality , Male , Middle Aged , OX40 Ligand/immunology , Programmed Cell Death 1 Receptor/immunology , Recurrence , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , Young Adult
20.
Front Immunol ; 12: 729085, 2021.
Article in English | MEDLINE | ID: mdl-34630403

ABSTRACT

Recently, many discoveries have elucidated the cellular and molecular diversity in the leukemic microenvironment and improved our knowledge regarding their complex nature. This has allowed the development of new therapeutic strategies against leukemia. Advances in biotechnology and the current understanding of T cell-engineering have led to new approaches in this fight, thus improving cell-mediated immune response against cancer. However, most of the investigations focus only on conventional cytotoxic cells, while ignoring the potential of unconventional T cells that until now have been little studied. γδ T cells are a unique lymphocyte subpopulation that has an extensive repertoire of tumor sensing and may have new immunotherapeutic applications in a wide range of tumors. The ability to respond regardless of human leukocyte antigen (HLA) expression, the secretion of antitumor mediators and high functional plasticity are hallmarks of γδ T cells, and are ones that make them a promising alternative in the field of cell therapy. Despite this situation, in particular cases, the leukemic microenvironment can adopt strategies to circumvent the antitumor response of these lymphocytes, causing their exhaustion or polarization to a tumor-promoting phenotype. Intervening in this crosstalk can improve their capabilities and clinical applications and can make them key components in new therapeutic antileukemic approaches. In this review, we highlight several characteristics of γδ T cells and their interactions in leukemia. Furthermore, we explore strategies for maximizing their antitumor functions, aiming to illustrate the findings destined for a better mobilization of γδ T cells against the tumor. Finally, we outline our perspectives on their therapeutic applicability and indicate outstanding issues for future basic and clinical leukemia research, in the hope of contributing to the advancement of studies on γδ T cells in cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive , Intraepithelial Lymphocytes/drug effects , Intraepithelial Lymphocytes/transplantation , Leukemia/therapy , Animals , Antineoplastic Agents, Immunological/adverse effects , Cell Proliferation/drug effects , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy, Adoptive/adverse effects , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Leukemia/genetics , Leukemia/immunology , Leukemia/metabolism , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...