Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 139(4): 608-623, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34657151

ABSTRACT

The key immunologic signatures associated with clinical outcomes after posttransplant cyclophosphamide (PTCy)-based HLA-haploidentical (haplo) and HLA-matched bone marrow transplantation (BMT) are largely unknown. To address this gap in knowledge, we used machine learning to decipher clinically relevant signatures from immunophenotypic, proteomic, and clinical data and then examined transcriptome changes in the lymphocyte subsets that predicted major posttransplant outcomes. Kinetics of immune subset reconstitution after day 28 were similar for 70 patients undergoing haplo and 75 patients undergoing HLA-matched BMT. Machine learning based on 35 candidate factors (10 clinical, 18 cellular, and 7 proteomic) revealed that combined elevations in effector CD4+ conventional T cells (Tconv) and CXCL9 at day 28 predicted acute graft-versus-host disease (aGVHD). Furthermore, higher NK cell counts predicted improved overall survival (OS) due to a reduction in both nonrelapse mortality and relapse. Transcriptional and flow-cytometric analyses of recovering lymphocytes in patients with aGVHD identified preserved hallmarks of functional CD4+ regulatory T cells (Tregs) while highlighting a Tconv-driven inflammatory and metabolic axis distinct from that seen with conventional GVHD prophylaxis. Patients developing early relapse displayed a loss of inflammatory gene signatures in NK cells and a transcriptional exhaustion phenotype in CD8+ T cells. Using a multimodality approach, we highlight the utility of systems biology in BMT biomarker discovery and offer a novel understanding of how PTCy influences alloimmune responses. Our work charts future directions for novel therapeutic interventions after these increasingly used GVHD prophylaxis platforms. Specimens collected on NCT0079656226 and NCT0080927627 https://clinicaltrials.gov/.


Subject(s)
Bone Marrow Transplantation , Cyclophosphamide/therapeutic use , Graft vs Host Disease/diagnosis , Immunosuppressive Agents/therapeutic use , Adult , Bone Marrow Transplantation/adverse effects , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Humans , Immune Reconstitution , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Machine Learning , Male , Middle Aged , Proteomics , Transcriptome , Young Adult
2.
Blood ; 137(18): 2544-2557, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33534893

ABSTRACT

Patients with chronic graft-versus-host disease (cGVHD) have increased B cell-activating factor (BAFF) levels, but whether BAFF promotes disease after allogeneic bone marrow transplantation (allo-BMT) remains unknown. In a major histocompatibility complex-mismatched model with cGVHD-like manifestations, we first examined B-lymphopenic µMT allo-BMT recipients and found that increased BAFF levels in cGVHD mice were not merely a reflection of B-cell number. Mice that later developed cGVHD had significantly increased numbers of recipient fibroblastic reticular cells with higher BAFF transcript levels. Increased BAFF production by donor cells also likely contributed to cGVHD, because BAFF transcript in CD4+ T cells from diseased mice and patients was increased. cGVHD manifestations in mice were associated with high BAFF/B-cell ratios and persistence of B-cell receptor (BCR)-activated B cells in peripheral blood and lesional tissue. By employing BAFF transgenic (Tg) mice donor cells, we addressed whether high BAFF contributed to BCR activation in cGVHD. BAFF increased NOTCH2 expression on B cells, augmenting BCR responsiveness to surrogate antigen and NOTCH ligand. BAFF Tg B cells had significantly increased protein levels of the proximal BCR signaling molecule SYK, and high SYK protein was maintained by BAFF after in vitro BCR activation or when alloantigen was present in vivo. Using T cell-depleted (BM only) BAFF Tg donors, we found that BAFF promoted cGVHD manifestations, circulating GL7+ B cells, and alloantibody production. We demonstrate that pathologic production of BAFF promotes an altered B-cell compartment and augments BCR responsiveness. Our findings compel studies of therapeutic targeting of BAFF and BCR pathways in patients with cGVHD.


Subject(s)
B-Cell Activating Factor/metabolism , Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/pathology , Proto-Oncogene Proteins c-bcr/metabolism , Receptor, Notch2/metabolism , Syk Kinase/metabolism , T-Lymphocytes/immunology , Animals , B-Cell Activating Factor/genetics , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Isoantibodies/immunology , Isoantigens/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcr/genetics , Receptor, Notch2/genetics , Syk Kinase/genetics , Transplantation, Homologous
3.
Immunology ; 166(3): 310-326, 2022 07.
Article in English | MEDLINE | ID: mdl-35322421

ABSTRACT

Ability of IL-17-producing CD8+ T cells (Tc17) to transform into cytotoxic anti-tumour effectors makes them a promising candidate for immune effector cell (IEC) therapy. However, key factors regulating Tc17 reprogramming remain poorly defined, hindering translation of Tc17-based IEC use from bench to bedside. We probed the effects of multiple cytokines and underlying signalling pathways on Tc17 cells and identified pivotal role for IL-4 and PI3K/AKT in promoting Tc17 transformation into cytotoxic IFN-γ-producing IECs, an effect dependent on Eomes expression. IL-4 not only triggered Tc17 cytotoxicity, but also induced cell expansion, which significantly improved the antitumour potential of Tc17 cells compared to that of IFN-γ-producing CD8+ T cells (Tc1) in a murine model. Furthermore, IL-4/AKT signalling drove the upregulation of the T-cell receptor-associated transmembrane adaptor 1 (Trat1) in Tc17 cells to promote IL-4-induced T-cell receptor stabilization and Tc17 cytotoxicity. Finally, we proposed a possible procedure to expand human Tc17 from peripheral blood of cancer patients, and confirmed the function of IL-4 in Tc17 reprogramming. Collectively, these results document a novel IL-4/AKT/Eomes/Trat1 axis that promotes expansion and transformation of Tc17 cells into cytotoxic effectors with a therapeutic potential. IL-4 priming of Tc17 cells should be further explored as a cell therapy engineering strategy to generate IECs to augment anti-tumour responses.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-4 , Adoptive Transfer , Animals , Humans , Interleukin-17/metabolism , Interleukin-4/metabolism , Mice , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt/metabolism
4.
J Immunol ; 203(2): 557-568, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31182480

ABSTRACT

Graft-versus-host disease (GVHD) is the most serious complication of allogeneic hematopoietic cell transplantation. Notch signals delivered during the first 48 h after transplantation drive proinflammatory cytokine production in conventional T cells (Tconv) and inhibit the expansion of regulatory T cells (Tregs). Short-term Notch inhibition induces long-term GVHD protection. However, it remains unknown whether Notch blockade blunts GVHD through its effects on Tconv, Tregs, or both and what early Notch-regulated molecular events occur in alloantigen-specific T cells. To address these questions, we engineered T cell grafts to achieve selective Notch blockade in Tconv versus Tregs and evaluated their capacity to trigger GVHD in mice. Notch blockade in Tconv was essential for GVHD protection as GVHD severity was similar in the recipients of wild-type Tconv combined with Notch-deprived versus wild-type Tregs. To identify the impact of Notch signaling on the earliest steps of T cell activation in vivo, we established a new acute GVHD model mediated by clonal alloantigen-specific 4C CD4+ Tconv. Notch-deprived 4C T cells had preserved early steps of activation, IL-2 production, proliferation, and Th cell polarization. In contrast, Notch inhibition dampened IFN-γ and IL-17 production, diminished mTORC1 and ERK1/2 activation, and impaired transcription of a subset of Myc-regulated genes. The distinct Notch-regulated signature had minimal overlap with known Notch targets in T cell leukemia and developing T cells, highlighting the specific impact of Notch signaling in mature T cells. Our findings uncover a unique molecular program associated with the pathogenic effects of Notch in T cells at the earliest stages of GVHD.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Isoantigens/immunology , Receptors, Notch/immunology , Animals , Bone Marrow Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous/adverse effects
5.
Blood ; 132(20): 2188-2200, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30181175

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is a major complication of allogeneic hematopoietic cell transplantation (allo-HCT) and remains an area of unmet clinical need with few treatment options available. Notch blockade prevents acute GVHD in multiple mouse models, but the impact of Notch signaling on cGVHD remains unknown. Using genetic and antibody-mediated strategies of Notch inhibition, we investigated the role of Notch signaling in complementary mouse cGVHD models that mimic several aspects of human cGVHD in search of candidate therapeutics. In the B10.D2→BALB/c model of sclerodermatous cGVHD, Delta-like ligand 4 (Dll4)-driven Notch signaling was essential for disease development. Antibody-mediated Dll4 inhibition conferred maximum benefits when pursued early in a preventative fashion, with anti-Dll1 enhancing early protection. Notch-deficient alloantigen-specific T cells showed no early defects in proliferation or helper polarization in vivo but subsequently exhibited markedly decreased cytokine secretion and enhanced accumulation of FoxP3+ regulatory T cells. In the B6→B10.BR major histocompatibility complex-mismatched model with multi-organ system cGVHD and prominent bronchiolitis obliterans (BO), but not skin manifestations, absence of Notch signaling in T cells provided long-lasting disease protection that was replicated by systemic targeting of Dll1, Dll4, or both Notch ligands, even during established disease. Notch inhibition decreased target organ damage and germinal center formation. Moreover, decreased BO-cGVHD was observed upon inactivation of Notch1 and/or Notch2 in T cells. Systemic targeting of Notch2 alone was safe and conferred therapeutic benefits. Altogether, Notch ligands and receptors regulate key pathogenic steps in cGVHD and emerge as novel druggable targets to prevent or treat different forms of cGVHD.


Subject(s)
Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Intercellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , Receptors, Notch/immunology , Adaptor Proteins, Signal Transducing , Animals , Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/pathology , Calcium-Binding Proteins , Chronic Disease , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Isoantigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transplantation, Homologous/adverse effects
6.
J Immunol ; 194(6): 2899-908, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25687759

ABSTRACT

Rejection remains a major clinical challenge limiting allograft survival after solid organ transplantation. Both cellular and humoral immunity contribute to this complication, with increased recognition of Ab-mediated damage during acute and chronic rejection. Using a mouse model of MHC-mismatched heart transplantation, we report markedly protective effects of Notch inhibition, dampening both T cell and Ab-driven rejection. T cell-specific pan-Notch blockade prolonged heart allograft survival and decreased IFN-γ and IL-4 production by alloreactive T cells, especially when combined with depletion of recipient CD8(+) T cells. These effects were associated with decreased infiltration by conventional T cells and an increased proportion of regulatory T cells in the graft. Transient administration of neutralizing Abs specific for delta-like (Dll)1/4 Notch ligands in the peritransplant period led to prolonged acceptance of allogeneic hearts, with superior outcome over Notch inhibition only in T cells. Systemic Dll1/4 inhibition decreased T cell cytokines and graft infiltration, germinal center B cell and plasmablast numbers, as well as production of donor-specific alloantibodies and complement deposition in the transplanted hearts. Dll1 or Dll4 inhibition alone provided partial protection. Thus, pathogenic signals delivered by Dll1/4 Notch ligands early after transplantation promote organ rejection through several complementary mechanisms. Transient interruption of these signals represents an attractive new therapeutic strategy to enhance long-term allograft survival.


Subject(s)
Antibodies, Neutralizing/immunology , Graft Rejection/immunology , Heart Transplantation/methods , Immunity/immunology , Intercellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Neutralizing/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Calcium-Binding Proteins , Flow Cytometry , Graft Rejection/prevention & control , Graft Survival/immunology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/immunology , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transplantation, Homologous
7.
Curr Opin Hematol ; 23(4): 362-70, 2016 07.
Article in English | MEDLINE | ID: mdl-27135981

ABSTRACT

PURPOSE OF REVIEW: This article highlights recent discoveries about Notch activation and its oncogenic functions in lymphoid malignancies, and discusses the therapeutic potential of Notch inhibition. RECENT FINDINGS: NOTCH mutations arise in a broad spectrum of lymphoid malignancies and are increasingly scrutinized as putative therapeutic targets. In T-cell acute lymphoblastic leukemia (T-ALL), NOTCH1 mutations affect the extracellular negative regulatory region and lead to constitutive Notch activation, although mutated receptors remain sensitive to Notch ligands. Other NOTCH1 mutations in T-ALL and NOTCH1/2 mutations in multiple B-cell malignancies truncate the C-terminal proline (P), glutamic acid (E), serine (S), threonine (T)-rich (PEST) domain, leading to decreased Notch degradation after ligand-mediated activation. Thus, targeting Notch ligand-receptor interactions could provide therapeutic benefits. In addition, we discuss recent reports on clinical testing of Notch inhibitors in T-ALL that influenced contemporary thinking on the challenges of targeting Notch in cancer. We review advances in the laboratory to address these challenges in regards to drug targets, the Notch-driven metabolome, and the sophisticated protein-protein interactions at Notch-dependent superenhancers that underlie oncogenic Notch functions. SUMMARY: Notch signaling is a recurrent oncogenic pathway in multiple T- and B-cell lymphoproliferative disorders. Understanding the complexity and consequences of Notch activation is critical to define optimal therapeutic strategies targeting the Notch pathway.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Lymphoproliferative Disorders/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Biomarkers , Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology , Molecular Targeted Therapy , Protein Binding , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
9.
Blood ; 134(11): 848-849, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31515227
10.
Transfus Apher Sci ; 52(2): 157-61, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25716169

ABSTRACT

Chronic graft-versus-host disease (GVHD) is the most important cause of late morbidity and mortality in recipients of allogeneic blood and marrow transplantation. Despite increased understanding of biology of chronic GVHD, treatment options remain limited and ineffective. While corticosteroids represent the backbone of initial chronic GVHD treatment, they have significant long-term toxicity and more than half of the patients require second-line therapy. Among the second-line treatments for chronic GVHD, extracorporeal photopheresis (ECP) is one of the most extensively studied modalities. While high quality studies establishing true value of ECP in chronic GVHD patients are lacking, its benefits in chronic GVHD are well documented. Its putative immunomodulatory, but not immunosuppressive, properties represent an attractive alternative to the other strategies leading to global immunosuppression and the resulting risks of opportunistic infections or malignancy relapse.


Subject(s)
Graft vs Host Disease/prevention & control , Graft vs Host Disease/therapy , Photopheresis/methods , Adrenal Cortex Hormones/adverse effects , Adult , Animals , Bone Marrow Transplantation/adverse effects , Child , Chronic Disease , Humans , Immunologic Factors , Immunosuppression Therapy/adverse effects , Immunosuppressive Agents , Recurrence , Transfusion Reaction
11.
Blood ; 130(19): 2053-2054, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29122773
13.
Blood ; 117(1): 299-308, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20870902

ABSTRACT

Graft-versus-host disease (GVHD) remains the major barrier to the success of allogeneic hematopoietic stem cell transplantation (HSCT). GVHD is caused by donor T cells that mediate host tissue injury through multiple inflammatory mechanisms. Blockade of individual effector molecules has limited efficacy in controlling GVHD. Here, we report that Notch signaling is a potent regulator of T-cell activation, differentiation, and function during acute GVHD. Inhibition of canonical Notch signaling in donor T cells markedly reduced GVHD severity and mortality in mouse models of allogeneic HSCT. Although Notch-deprived T cells proliferated and expanded in response to alloantigens in vivo, their ability to produce interleukin-2 and inflammatory cytokines was defective, and both CD4(+) and CD8(+) T cells failed to up-regulate selected effector molecules. Notch inhibition decreased the accumulation of alloreactive T cells in the intestine, a key GVHD target organ. However, Notch-deprived alloreactive CD4(+) T cells retained significant cytotoxic potential and antileukemic activity, leading to improved overall survival of the recipients. These results identify Notch as a novel essential regulator of pathogenic CD4(+) T-cell responses during acute GVHD and suggest that Notch signaling in T cells should be investigated as a therapeutic target after allogeneic HSCT.


Subject(s)
Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Receptors, Notch/metabolism , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cytokines/metabolism , Disease Models, Animal , Graft vs Host Disease/pathology , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transplantation, Homologous , Whole-Body Irradiation
14.
J Clin Oncol ; 41(10): 1864-1875, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36459673

ABSTRACT

PURPOSE: Chronic graft-versus-host disease (cGVHD) remains the major cause of late morbidity after allogeneic hematopoietic cell transplantation. Colony-stimulating factor 1 receptor (CSF-1R)-dependent macrophages promote cGVHD fibrosis, and their elimination in preclinical studies ameliorated cGVHD. Axatilimab is a humanized monoclonal antibody that inhibits CSF-1R signaling and restrains macrophage development. PATIENTS AND METHODS: This phase I (phI)/phase II (phII) open-label study (ClinicalTrials.gov identifier: NCT03604692) evaluated safety, tolerability, and efficacy of axatilimab in patients age ≥ 6 years with active cGVHD after ≥ 2 prior systemic therapy lines. Primary objectives in phI were to identify the optimal biologic and recommended phII dose and in phII to evaluate the overall (complete and partial) response rate (ORR) at the start of treatment cycle 7. RESULTS: Forty enrolled patients (17 phI; 23 phII) received at least one axatilimab dose. In phI, a dose of 3 mg/kg given once every 4 weeks met the optimal biologic dose definition. Two dose-limiting toxicities occurred at the 3 mg/kg dose given once every 2 weeks. At least one treatment-related adverse event (TRAE) was observed in 30 patients with grade ≥ 3 TRAEs in eight patients, the majority known on-target effects of CSF-1R inhibition. No cytomegalovirus reactivations occurred. With the 50% ORR at cycle 7 day 1, the phII cohort met the primary efficacy end point. Furthermore, the ORR in the first six cycles, an end point supporting regulatory approvals, was 82%. Responses were seen in all affected organs regardless of prior therapy. Fifty-eight percent of patients reported significant improvement in cGVHD-related symptoms using the Lee Symptom Scale. On-target activity of axatilimab was suggested by the decrease in skin CSF-1R-expressing macrophages. CONCLUSION: Targeting profibrotic macrophages with axatilimab is a therapeutically promising novel strategy with a favorable safety profile for refractory cGVHD.


Subject(s)
Biological Products , Bronchiolitis Obliterans Syndrome , Graft vs Host Disease , Humans , Child , Graft vs Host Disease/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Biological Products/therapeutic use , Chronic Disease
15.
J Immunol ; 184(2): 764-74, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19995899

ABSTRACT

Donor CD4+ T cells are thought to be essential for inducing delayed host tissue injury in chronic graft-versus-host disease (GVHD). However, the relative contributions of distinct effector CD4+ T cell subpopulations and the molecular pathways influencing their generation are not known. We investigated the role of the STAT3 pathway in a murine model of chronic sclerodermatous GVHD. This pathway integrates multiple signaling events during the differentiation of naive CD4+ T cells and impacts their homeostasis. We report that chimeras receiving an allograft containing STAT3-ablated donor CD4+ T cells do not develop classic clinical and pathological manifestations of alloimmune tissue injury. Analysis of chimeras showed that abrogation of STAT3 signaling reduced the in vivo expansion of donor-derived CD4+ T cells and their accumulation in GVHD target tissues without abolishing antihost alloreactivity. STAT3 ablation did not significantly affect Th1 differentiation while enhancing CD4+CD25+Foxp3+ T cell reconstitution through thymus-dependent and -independent pathways. Transient depletion of CD25+ T cells in chimeras receiving STAT3-deficient T cells resulted in delayed development of alloimmune gut and liver injury. This delayed de novo GVHD was associated with the emergence of donor hematopoietic stem cell-derived Th1 and Th17 cells. These results suggest that STAT3 signaling in graft CD4+ T cells links the alloimmune tissue injury of donor graft T cells and the emergence of donor hematopoietic stem cell-derived pathogenic effector cells and that both populations contribute, albeit in different ways, to the genesis of chronic GVHD after allogenic bone marrow transplantation in a murine model.


Subject(s)
Bone Marrow Transplantation/immunology , CD4-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/etiology , STAT3 Transcription Factor/metabolism , Scleroderma, Systemic/etiology , Animals , Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes/immunology , Chronic Disease , Disease Models, Animal , Mice , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Transplantation, Homologous
16.
J Clin Invest ; 132(21)2022 11 01.
Article in English | MEDLINE | ID: mdl-36099049

ABSTRACT

BackgroundImmune exhaustion and senescence are dominant dysfunctional states of effector T cells and major hurdles for the success of cancer immunotherapy. In the current study, we characterized how acute myeloid leukemia (AML) promotes the generation of senescent-like CD8+ T cells and whether they have prognostic relevance.METHODSWe analyzed NanoString, bulk RNA-Seq and single-cell RNA-Seq data from independent clinical cohorts comprising 1,896 patients treated with chemotherapy and/or immune checkpoint blockade (ICB).ResultsWe show that senescent-like bone marrow CD8+ T cells were impaired in killing autologous AML blasts and that their proportion negatively correlated with overall survival (OS). We defined what we believe to be new immune effector dysfunction (IED) signatures using 2 gene expression profiling platforms and reported that IED scores correlated with adverse-risk molecular lesions, stemness, and poor outcomes; these scores were a more powerful predictor of OS than 2017-ELN risk or leukemia stem cell (LSC17) scores. IED expression signatures also identified an ICB-unresponsive tumor microenvironment and predicted significantly shorter OS.ConclusionThe IED scores provided improved AML-risk stratification and could facilitate the delivery of personalized immunotherapies to patients who are most likely to benefit.TRIAL REGISTRATIONClinicalTrials.gov; NCT02845297.FUNDINGJohn and Lucille van Geest Foundation, Nottingham Trent University's Health & Wellbeing Strategic Research Theme, NIH/NCI P01CA225618, Genentech-imCORE ML40354, Qatar National Research Fund (NPRP8-2297-3-494).


Subject(s)
Immune System Diseases , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/drug therapy , Prognosis , Immunotherapy , Tumor Microenvironment , CD8-Positive T-Lymphocytes
17.
Transplant Cell Ther ; 27(10): 817-835, 2021 10.
Article in English | MEDLINE | ID: mdl-34217703

ABSTRACT

Chronic graft-versus-host disease (GVHD) can be associated with significant morbidity, in part because of nonreversible fibrosis, which impacts physical functioning (eye, skin, lung manifestations) and mortality (lung, gastrointestinal manifestations). Progress in preventing severe morbidity and mortality associated with chronic GVHD is limited by a complex and incompletely understood disease biology and a lack of prognostic biomarkers. Likewise, treatment advances for highly morbid manifestations remain hindered by the absence of effective organ-specific approaches targeting "irreversible" fibrotic sequelae and difficulties in conducting clinical trials in a heterogeneous disease with small patient numbers. The purpose of this document is to identify current gaps, to outline a roadmap of research goals for highly morbid forms of chronic GVHD including advanced skin sclerosis, fasciitis, lung, ocular and gastrointestinal involvement, and to propose strategies for effective trial design. The working group made the following recommendations: (1) Phenotype chronic GVHD clinically and biologically in future cohorts, to describe the incidence, prognostic factors, mechanisms of organ damage, and clinical evolution of highly morbid conditions including long-term effects in children; (2) Conduct longitudinal multicenter studies with common definitions and research sample collections; (3) Develop new approaches for early identification and treatment of highly morbid forms of chronic GVHD, especially biologically targeted treatments, with a special focus on fibrotic changes; and (4) Establish primary endpoints for clinical trials addressing each highly morbid manifestation in relationship to the time point of intervention (early versus late). Alternative endpoints, such as lack of progression and improvement in physical functioning or quality of life, may be suitable for clinical trials in patients with highly morbid manifestations. Finally, new approaches for objective response assessment and exploration of novel trial designs for small populations are required.


Subject(s)
Graft vs Host Disease , Chronic Disease , Consensus , Humans , Incidence , National Institutes of Health (U.S.) , Quality of Life , United States
18.
Cancer Immunol Immunother ; 59(1): 137-48, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19590872

ABSTRACT

Using a model of established malignancy, we found that cyclophosphamide (Cy), administered at a dose not requiring hematopoietic stem cell support, is superior to low-dose total body irradiation in augmenting antitumor immunity. We observed that Cy administration resulted in expansion of tumor antigen-specific T cells and transient depletion of CD4(+)Foxp3(+) regulatory T cells (Tregs). The antitumor efficacy of Cy was not improved by administration of anti-CD25 monoclonal antibody given to induce more profound Treg depletion. We found that Cy, through its myelosuppressive action, induced rebound myelopoiesis and perturbed dendritic cell (DC) homeostasis. The resulting DC turnover led to the emergence of tumor-infiltrating DCs that secreted more IL-12 and less IL-10 compared to those from untreated tumor-bearing animals. These newly recruited DCs, originating from proliferating early DC progenitors, were fully capable of priming T cell responses and ineffective in inducing expansion of Tregs. Together, our results show that Cy-mediated antitumor effects extend beyond the well-documented cytotoxicity and lymphodepletion and include resetting the DC homeostasis, thus providing an excellent platform for integration with other immunotherapeutic strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclophosphamide/pharmacology , Cytotoxicity, Immunologic/drug effects , Dendritic Cells/drug effects , Immunologic Factors/pharmacology , Neoplasms, Experimental/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclophosphamide/therapeutic use , Dendritic Cells/immunology , Homeostasis , Immunologic Factors/therapeutic use , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/radiotherapy , T-Lymphocytes, Regulatory/immunology , Whole-Body Irradiation
19.
J Clin Invest ; 129(6): 2189-2191, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063990

ABSTRACT

For high-risk and refractory hematological malignancies, allogeneic hematopoietic stem cell transplantation (alloHSCT) is the only available curative therapy, with benefits derived from the antigenic disparity between recipient cancer and the incoming immune system. This immunologic mismatch can also lead to lethal graft-versus-host disease (GVHD), and immunosuppression strategies, including high-dose posttransplantation cyclophosphamide (PTCy), have been developed to allow for safe alloHSCT delivery. In this issue of JCI, Wachsmuth et al. present the results of preclinical studies designed to evaluate the mechanisms that underlie efficacy of PTCy after alloHSCT. The results of this study challenge previous reports indicating that alloreactive T cell elimination and thymic clonal deletion are primary mediators of PTCy efficacy and provide strong evidence to support FoxP3+CD4+ Tregs as important effectors of PTCy benefits.


Subject(s)
Graft vs Host Disease , Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Cyclophosphamide , Humans
20.
Transplantation ; 83(5): 631-40, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17353785

ABSTRACT

BACKGROUND: There are limited data examining the effects of pharmacological immunosuppression on the in vivo fate of donor lymphocyte infusions (DLI)-derived T cells, their function, and their antitumor efficacy. METHODS: We addressed this question in a murine model in which DLI is given to stable mixed chimeras resulting in lymphohematopoietic graft-versus-host (LH-GVH) response. In this model, LH-GVH potency can be directly measured as the kinetics of conversion to full donor chimerism and can be correlated with associated graft-versus-leukemia (GVL) reactivity. RESULTS: We found discordance in DLI-mediated LH-GVH reactivity depending on the timing of rapamycin (RAPA) administration. Delayed administration of RAPA in contrast to its early administration at the time of adoptive transfer did not interfere with conversion to full donor chimerism. Moreover, delayed administration of RAPA preserved the GVL reactivity of DLI. Analysis of the long-term chimeras showed that regardless of RAPA administration, adoptively transferred T cells mediating the LH-GVH response contribute minimally to the reconstitution of the peripheral T-cell compartment and exhibit profound hyporesponsiveness and decreased production of interleukin (IL)-2 on restimulation in vitro. However, we observed only in the RAPA-treated chimeras that the remaining hyporesponsive DLI-derived CD4+ T cells secrete large amounts of IL-10, a known immunoregulatory cytokine. CONCLUSIONS: We conclude that delayed administration of RAPA after DLI does not interfere with their LH-GVH reactivity but promotes the emergence of IL-10-secreting DLI-derived CD4+ T cells that might contribute to the drug's known ability to promote bilateral donor host tolerance without interfering with GVL reactivity.


Subject(s)
Graft vs Leukemia Effect/physiology , Hematopoietic Stem Cells/immunology , Interleukin-10/metabolism , Sirolimus/pharmacology , Stem Cell Transplantation , T-Lymphocytes/immunology , Animals , Cell Differentiation , Hematopoietic Stem Cells/drug effects , Immunosuppressive Agents/pharmacology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transplantation Chimera , Transplantation Conditioning , Transplantation, Homologous/immunology
SELECTION OF CITATIONS
SEARCH DETAIL