Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Blood ; 143(16): 1656-1669, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38295333

ABSTRACT

ABSTRACT: Autologous stem cell transplantation (ASCT) is the standard of care consolidation therapy for eligible patients with myeloma but most patients eventually progress, an event associated with features of immune escape. Novel approaches to enhance antimyeloma immunity after ASCT represent a major unmet need. Here, we demonstrate that patient-mobilized stem cell grafts contain high numbers of effector CD8 T cells and immunosuppressive regulatory T cells (Tregs). We showed that bone marrow (BM)-residing T cells are efficiently mobilized during stem cell mobilization (SCM) and hypothesized that mobilized and highly suppressive BM-derived Tregs might limit antimyeloma immunity during SCM. Thus, we performed ASCT in a preclinical myeloma model with or without stringent Treg depletion during SCM. Treg depletion generated SCM grafts containing polyfunctional CD8 T effector memory cells, which dramatically enhanced myeloma control after ASCT. Thus, we explored clinically tractable translational approaches to mimic this scenario. Antibody-based approaches resulted in only partial Treg depletion and were inadequate to recapitulate this effect. In contrast, a synthetic interleukin-2 (IL-2)/IL-15 mimetic that stimulates the IL-2 receptor on CD8 T cells without binding to the high-affinity IL-2Ra used by Tregs efficiently expanded polyfunctional CD8 T cells in mobilized grafts and protected recipients from myeloma progression after ASCT. We confirmed that Treg depletion during stem cell mobilization can mitigate constraints on tumor immunity and result in profound myeloma control after ASCT. Direct and selective cytokine signaling of CD8 T cells can recapitulate this effect and represent a clinically testable strategy to improve responses after ASCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Humans , Multiple Myeloma/pathology , T-Lymphocytes, Regulatory , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Mobilization/methods , Transplantation, Autologous , Stem Cell Transplantation
2.
Blood ; 137(14): 1970-1979, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33512442

ABSTRACT

We determined the efficacy of tocilizumab (TCZ) in preventing grade 2-4 acute graft-versus-host disease (aGVHD) in patients with acute leukemia or myelodysplasia undergoing matched sibling donor (MSD) or volunteer unrelated donor (VUD) allogeneic stem cell transplantation after myeloablative or reduced-intensity conditioning across 5 Australian centers. A total of 145 patients (50 MSD, 95 VUD) were randomly assigned to placebo or TCZ on day -1. All patients received T-cell-replete peripheral blood stem cell grafts and graft-versus-host disease (GVHD) prophylaxis with cyclosporin/methotrexate. A planned substudy analyzed the VUD cohort. With a median follow-up of 746 days, the incidence of grade 2-4 aGVHD at day 100 for the entire cohort was 36% for placebo vs 27% for TCZ (hazard ratio [HR], 0.69; 95% confidence interval [CI], 0.38-1.26; P = .23) and 45% vs 32% (HR, 0.61; 95% CI, 0.31-1.22; P = .16) for the VUD subgroup. The incidence of grade 2-4 aGVHD at day 180 for the entire cohort was 40% for placebo vs 29% for TCZ (HR, 0.68; 95% CI, 0.38-1.22; P = .19) and 48% vs 32% (HR, 0.59; 95% CI, 0.30-1.16; P = .13) for the VUD subgroup. Reductions in aGVHD were predominantly in grade 2 disease. For the entire cohort, transplant-related mortality occurred in 8% vs 11% of placebo-treated vs TCZ-treated patients, respectively (P = .56), and overall survival was 79% vs 71% (P = .27). Median day to neutrophil and platelet engraftment was delayed by 2 to 3 days in TCZ-treated patients, whereas liver toxicity and infectious complications were similar between groups. In this phase 3 randomized double-blind trial, TCZ showed nonsignificant trends toward reduced incidence of grade 2-4 aGVHD in recipients from HLA-matched VUDs but no improvements in long term-survival.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Cyclosporine/therapeutic use , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/therapeutic use , Methotrexate/therapeutic use , Adult , Double-Blind Method , Female , Humans , Leukemia/therapy , Male , Middle Aged , Myelodysplastic Syndromes/therapy , Placebo Effect , Transplantation, Homologous , Treatment Outcome
3.
Blood ; 138(26): 2874-2885, 2021 12 30.
Article in English | MEDLINE | ID: mdl-34115118

ABSTRACT

Donor and recipient cytomegalovirus (CMV) serostatus correlate with transplant-related mortality that is associated with reduced survival following allogeneic stem cell transplant (SCT). Prior epidemiologic studies have suggested that CMV seronegative recipients (R-) receiving a CMV-seropositive graft (D+) experience inferior outcomes compared with other serostatus combinations, an observation that appears independent of viral reactivation. We therefore investigated the hypothesis that prior donor CMV exposure irreversibly modifies immunologic function after SCT. We identified a CD4+/CD57+/CD27- T-cell subset that was differentially expressed between D+ and D- transplants and validated results with 120 patient samples. This T-cell subset represents an average of 2.9% (D-/R-), 18% (D-/R+), 12% (D+/R-), and 19.6% (D+/R+) (P < .0001) of the total CD4+ T-cell compartment and stably persists for at least several years post-SCT. Even in the absence of CMV reactivation post-SCT, D+/R- transplants displayed a significant enrichment of these cells compared with D-/R- transplants (P = .0078). These are effector memory cells (CCR7-/CD45RA+/-) that express T-bet, Eomesodermin, granzyme B, secrete Th1 cytokines, and are enriched in CMV-specific T cells. These cells are associated with decreased T-cell receptor diversity (P < .0001) and reduced proportions of major histocompatibility class (MHC) II expressing classical monocytes (P < .0001), myeloid (P = .024), and plasmacytoid dendritic cells (P = .0014). These data describe a highly expanded CD4+ T-cell population and putative mechanisms by which prior donor or recipient CMV exposure may create a lasting immunologic imprint following SCT, providing a rationale for using D- grafts for R- transplant recipients.


Subject(s)
CD4 Antigens/immunology , CD57 Antigens/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Memory T Cells/immunology , CD4 Antigens/analysis , CD4-Positive T-Lymphocytes/immunology , CD57 Antigens/analysis , Cells, Cultured , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/diagnosis , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Humans , Tissue Donors , Transplantation, Homologous/adverse effects
4.
J Immunol ; 200(10): 3547-3555, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29626088

ABSTRACT

IL-6 mediates broad physiological and pathological effects through its receptor signal transducing unit gp130. Due to the reportedly wide cellular expression of gp130, IL-6 is thought to signal ubiquitously via gp130 complex formation with membrane-bound IL-6Rα or soluble IL-6Rα. gp130 signaling primarily induces p-STAT3 and p-STAT1. In contrast to the previous dogma, we show in this article that circulating mouse and human granulocytes are unable to induce p-STAT3 or p-STAT1 after stimulation with IL-6 or an IL-6/soluble IL-6R complex. Furthermore, we demonstrate that this is due to a lack of gp130 expression on mouse and human granulocytes, despite their expression of membrane-bound IL-6R. Importantly, the absence of gp130 is not only a feature of mature granulocytes in healthy individuals, it is also observed after allogeneic stem cell transplantation. Moreover, granulocyte gp130 expression is lost during maturation, because granulocyte-monocyte progenitor cells express gp130 and respond to IL-6. Given that granulocytes constitute 50-70% of circulating leukocytes, this indicates a significantly smaller scope of IL-6 signaling than previously anticipated and has important implications for therapeutic IL-6 inhibition and the mechanisms of action thereof.


Subject(s)
Cytokine Receptor gp130/metabolism , Granulocytes/metabolism , Interleukin-6/metabolism , Animals , Female , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Neutrophils/metabolism , Receptors, Interleukin-6/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology
5.
Blood ; 126(13): 1609-20, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26206951

ABSTRACT

IL-17-producing cells are important mediators of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Here we demonstrate that a distinct CD8(+) Tc17 population develops rapidly after SCT but fails to maintain lineage fidelity such that they are unrecognizable in the absence of a fate reporter. Tc17 differentiation is dependent on alloantigen presentation by host dendritic cells (DCs) together with IL-6. Tc17 cells express high levels of multiple prototypic lineage-defining transcription factors (eg, RORγt, T-bet) and cytokines (eg, IL-17A, IL-22, interferon-γ, granulocyte macrophage colony-stimulating factor, IL-13). Targeted depletion of Tc17 early after transplant protects from lethal acute GVHD; however, Tc17 cells are noncytolytic and fail to mediate graft-versus-leukemia (GVL) effects. Thus, the Tc17 differentiation program during GVHD culminates in a highly plastic, hyperinflammatory, poorly cytolytic effector population, which we term "inflammatory iTc17" (iTc17). Because iTc17 cells mediate GVHD without contributing to GVL, therapeutic inhibition of iTc17 development in a clinical setting represents an attractive approach for separating GVHD and GVL.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Graft vs Host Disease/pathology , Graft vs Leukemia Effect , Interleukin-17/immunology , Stem Cell Transplantation/adverse effects , Th17 Cells/pathology , Animals , Bone Marrow Transplantation/adverse effects , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Graft vs Host Disease/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Th17 Cells/immunology
6.
Blood ; 125(15): 2435-44, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25673640

ABSTRACT

Idiopathic pneumonia syndrome (IPS) is a relatively common, frequently fatal clinical entity, characterized by noninfectious acute lung inflammation following allogeneic stem cell transplantation (SCT), the mechanisms of which are unclear. In this study, we demonstrate that immune suppression with cyclosporin after SCT limits T-helper cell (Th) 1 differentiation and interferon-γ secretion by donor T cells, which is critical for inhibiting interleukin (IL)-6 generation from lung parenchyma during an alloimmune response. Thereafter, local IL-6 secretion induces donor alloantigen-specific Th17 cells to preferentially expand within the lung, and blockade of IL-17A or transplantation of grafts lacking the IL-17 receptor prevents disease. Studies using IL-6(-/-) recipients or IL-6 blockade demonstrate that IL-6 is the critical driver of donor Th17 differentiation within the lung. Importantly, IL-6 is also dysregulated in patients undergoing clinical SCT and is present at very high levels in the plasma of patients with IPS compared with SCT recipients without complications. Furthermore, at the time of diagnosis, plasma IL-6 levels were higher in a subset of IPS patients who were nonresponsive to steroids and anti-tumor necrosis factor therapy. In sum, pulmonary-derived IL-6 promotes IPS via the induction of Th17 differentiation, and strategies that target these cytokines represent logical therapeutic approaches for IPS.


Subject(s)
Acute Lung Injury/etiology , Cyclosporine/adverse effects , Immunosuppressive Agents/adverse effects , Interleukin-17/immunology , Interleukin-6/immunology , Lung/pathology , Stem Cell Transplantation/adverse effects , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Cyclosporine/therapeutic use , Female , Immunosuppressive Agents/therapeutic use , Interferon-gamma/immunology , Lung/drug effects , Lung/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Th17 Cells/drug effects , Th17 Cells/immunology , Transplantation, Homologous
7.
Blood ; 121(17): 3511-20, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23430112

ABSTRACT

Donor T cells play pivotal roles in graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) effects following bone marrow transplantation (BMT). DNAX accessory molecule 1 (DNAM-1) is a costimulatory and adhesion molecule, expressed mainly by natural killer cells and CD8(+) T cells at steady state to promote adhesion to ligand-expressing targets and enhance cytolysis. We have analyzed the role of this pathway in GVHD and GVL. The absence of DNAM-1 on the donor graft attenuated GVHD in major histocompatibility complex (MHC)-mismatched and MHC-matched BMT following conditioning with lethal and sublethal irradiation. In contrast, DNAM-1 was not critical for GVL effects against ligand (CD155) expressing and nonexpressing leukemia. The effects on GVHD following myeloablative conditioning were independent of CD8(+) T cells and dependent on CD4(+) T cells, and specifically donor FoxP3(+) regulatory T cells (Treg). The absence of DNAM-1 promoted the expansion and suppressive function of Treg after BMT. These findings provide support for therapeutic DNAM-1 inhibition to promote tolerance in relevant inflammatory-based diseases characterized by T-cell activation.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Bone Marrow Transplantation , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/immunology , Leukemia, Experimental/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Leukemia, Experimental/etiology , Leukemia, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Transplantation Conditioning , Tumor Cells, Cultured , Whole-Body Irradiation
8.
J Immunol ; 191(10): 5291-303, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24123683

ABSTRACT

Natural regulatory T cells (nTregs) play an important role in tolerance; however, the small numbers of cells obtainable potentially limit the feasibility of clinical adoptive transfer. Therefore, we studied the feasibility and efficacy of using murine-induced regulatory T cells (iTregs) for the induction of tolerance after bone marrow transplantation. iTregs could be induced in large numbers from conventional donor CD4 and CD8 T cells within 1 wk and were highly suppressive. During graft-versus-host disease (GVHD), CD4 and CD8 iTregs suppressed the proliferation of effector T cells and the production of proinflammatory cytokines. However, unlike nTregs, both iTreg populations lost Foxp3 expression within 3 wk in vivo, reverted to effector T cells, and exacerbated GVHD. The loss of Foxp3 in iTregs followed homeostatic and/or alloantigen-driven proliferation and was unrelated to GVHD. However, the concurrent administration of rapamycin, with or without IL-2/anti-IL-2 Ab complexes, to the transplant recipients significantly improved Foxp3 stability in CD4 iTregs (and, to a lesser extent, CD8 iTregs), such that they remained detectable 12 wk after transfer. Strikingly, CD4, but not CD8, iTregs could then suppress Teff proliferation and proinflammatory cytokine production and prevent GVHD in an equivalent fashion to nTregs. However, at high numbers and when used as GVHD prophylaxis, Tregs potently suppress graft-versus-leukemia effects and so may be most appropriate as a therapeutic modality to treat GVHD. These data demonstrate that CD4 iTregs can be produced rapidly in large, clinically relevant numbers and, when transferred in the presence of systemic rapamycin and IL-2, induce tolerance in transplant recipients.


Subject(s)
Graft vs Host Disease/immunology , Immune Tolerance/immunology , Interleukin-2/metabolism , Sirolimus/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation , Cytokines/biosynthesis , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/metabolism , Graft vs Host Disease/prevention & control , Immune Tolerance/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects
9.
Lancet Oncol ; 15(13): 1451-1459, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25456364

ABSTRACT

BACKGROUND: Interleukin 6 mediates graft-versus-host disease (GVHD) in experimental allogeneic stem-cell transplantation (allogeneic SCT) and represents an attractive therapeutic target. We aimed to assess whether the humanised anti-interleukin-6 receptor monoclonal antibody, tocilizumab, could attenuate the incidence of acute GVHD. METHODS: We undertook a single-group, single-institution phase 1/2 study at the Royal Brisbane and Women's Hospital Bone Marrow Transplantation unit, QLD, Australia. Eligible patients were 18-65 years old and underwent T-replete HLA-matched allogeneic SCT with either total body irradiation-based myeloablative or reduced-intensity conditioning from unrelated or sibling donors. One intravenous dose of tocilizumab (8 mg/kg, capped at 800 mg, over 60 mins' infusion) was given the day before allogeneic SCT along with standard GVHD prophylaxis (cyclosporin [5 mg/kg per day on days -1 to +1, then 3 mg/kg per day to maintain therapeutic levels (trough levels of 140-300 ng/mL) for 100 days plus methotrexate [15 mg/m(2) on day 1, then 10 mg/m(2) on days 3, 6, and 11]). The primary endpoint was incidence of grade 2-4 acute GVHD at day 100, assessed and graded as per the Seattle criteria. Immunological profiles were compared with a non-randomised group of patients receiving allogeneic SCT, but not treated with tocilizumab. This trial is registered with the Australian and New Zealand Clinical Trials Registry, number ACTRN12612000726853. FINDINGS: Between Jan 19, 2012, and Aug 27, 2013, 48 eligible patients receiving cyclosporin and methotrexate as GVHD prophylaxis were enrolled into the study. The incidence of grade 2-4 acute GVHD in patients treated with tocilizumab at day 100 was 12% (95% CI 5-24), and the incidence of grade 3-4 acute GVHD was 4% (1-13). Grade 2-4 acute GVHD involving the skin developed in five (10%) patients of 48 treated with tocilizumab, involving the gastrointestinal tract in four (8%) patients; there were no reported cases involving the liver. Low incidences of grade 2-4 acute GVHD were noted in patients receiving both myeloablative total body irradiation-based conditioning (12% [95% CI 2-34) and fludarabine and melphalan reduced-intensity conditioning (12% [4-27]). Immune reconstitution was preserved in recipients of interleukin-6 receptor inhibition, but qualitatively modified with suppression of known pathogenic STAT3-dependent pathways. INTERPRETATION: Interleukin 6 is the main detectable and dysregulated cytokine secreted after allogeneic SCT and its inhibition is a potential new and simple strategy to protect from acute GVHD despite robust immune reconstitution; a randomised, controlled trial assessing tocilizumab in addition to standard GVHD prophylaxis in these patients is warranted. FUNDING: National Health and Medical Research Council and Queensland Health.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Graft vs Host Disease/drug therapy , Hematologic Neoplasms/complications , Interleukin-6/antagonists & inhibitors , Stem Cell Transplantation/adverse effects , Adolescent , Adult , Aged , Female , Follow-Up Studies , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Humans , Interleukin-6/immunology , Male , Middle Aged , Neoplasm Staging , Prognosis , Prospective Studies , Survival Rate , Transplantation, Homologous , Young Adult
10.
Blood ; 119(24): 5898-908, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22538855

ABSTRACT

FoxP3(+) confers suppressive properties and is confined to regulatory T cells (T(reg)) that potently inhibit autoreactive immune responses. In the transplant setting, natural CD4(+) T(reg) are critical in controlling alloreactivity and the establishment of tolerance. We now identify an important CD8(+) population of FoxP3(+) T(reg) that convert from CD8(+) conventional donor T cells after allogeneic but not syngeneic bone marrow transplantation. These CD8(+) T(reg) undergo conversion in the mesenteric lymph nodes under the influence of recipient dendritic cells and TGF-ß. Importantly, this population is as important for protection from GVHD as the well-studied natural CD4(+)FoxP3(+) population and is more potent in exerting class I-restricted and antigen-specific suppression in vitro and in vivo. Critically, CD8(+)FoxP3(+) T(reg) are exquisitely sensitive to inhibition by cyclosporine but can be massively and specifically expanded in vivo to prevent GVHD by coadministering rapamycin and IL-2 antibody complexes. CD8(+)FoxP3(+) T(reg) thus represent a new regulatory population with considerable potential to preferentially subvert MHC class I-restricted T-cell responses after bone marrow transplantation.


Subject(s)
Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/cytology , Forkhead Transcription Factors/metabolism , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/cytology , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes/immunology , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Immune Tolerance/drug effects , Interleukin-2/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Phenotype , Sirolimus/administration & dosage , Sirolimus/pharmacology , Survival Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/pharmacology , Transplantation, Homologous
11.
Blood ; 119(24): 5918-30, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22415754

ABSTRACT

Alloreactivity after transplantation is associated with profound immune suppression, and consequent opportunistic infection results in high morbidity and mortality. This immune suppression is most profound during GVHD after bone marrow transplantation where an inflammatory cytokine storm dominates. Contrary to current dogma, which avers that this is a T-cell defect, we demonstrate that the impairment lies within conventional dendritic cells (cDCs). Significantly, exogenous antigens can only be presented by the CD8(-) cDC subset after bone marrow transplantation, and inflammation during GVHD specifically renders the MHC class II presentation pathway in this population incompetent. In contrast, both classic and cross-presentation within MHC class I remain largely intact. Importantly, this defect in antigen processing can be partially reversed by TNF inhibition or the adoptive transfer of donor cDCs generated in the absence of inflammation.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Graft vs Host Disease/immunology , Immunosuppression Therapy , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cross-Priming/immunology , Graft vs Host Disease/pathology , Histocompatibility Antigens Class II/immunology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/metabolism , Isoantigens/immunology , Mice , Mice, Transgenic , Peptides/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/metabolism
12.
Blood ; 118(12): 3399-409, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21719602

ABSTRACT

Although the effects of type II-IFN (IFN-γ) on GVHD and leukemia relapse are well studied, the effects of type I-interferon (type I-IFN, IFN-α/ß) remain unclear. We investigated this using type I-IFN receptor-deficient mice and exogenous IFN-α administration in established models of GVHD and GVL. Type I-IFN signaling in host tissue prevented severe colon-targeted GVHD in CD4-dependent models of GVHD directed toward either major histocompatibility antigens or multiple minor histocompatibility antigens. This protection was the result of suppression of donor CD4(+) T-cell proliferation and differentiation. Studies in chimeric recipients demonstrated this was due to type I-IFN signaling in hematopoietic tissue. Consistent with this finding, administration of IFN-α during conditioning inhibited donor CD4(+) proliferation and differentiation. In contrast, CD8-dependent GVHD and GVL effects were enhanced when type I-IFN signaling was intact in the host or donor, respectively. This finding reflected the ability of type I-IFN to both sensitize host target tissue/leukemia to cell-mediated cytotoxicity and augment donor CTL function. These data confirm that type I-IFN plays an important role in defining the balance of GVHD and GVL responses and suggests that administration of the cytokine after BM transplantation could be studied prospectively in patients at high risk of relapse.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Interferon-alpha , Leukemia/immunology , Receptor, Interferon alpha-beta/deficiency , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Female , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/drug effects , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Interferon-beta/immunology , Leukemia/mortality , Leukemia/pathology , Leukemia/therapy , Lymphocyte Activation/immunology , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Receptor, Interferon alpha-beta/immunology , Signal Transduction , Survival Rate , Transplantation, Homologous , Whole-Body Irradiation
13.
J Clin Invest ; 133(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36512425

ABSTRACT

Autologous stem cell transplantation (ASCT) with subsequent lenalidomide maintenance is standard consolidation therapy for multiple myeloma, and a subset of patients achieve durable progression-free survival that is suggestive of long-term immune control. Nonetheless, most patients ultimately relapse, suggesting immune escape. TIGIT appears to be a potent inhibitor of myeloma-specific immunity and represents a promising new checkpoint target. Here we demonstrate high expression of TIGIT on activated CD8+ T cells in mobilized peripheral blood stem cell grafts from patients with myeloma. To guide clinical application of TIGIT inhibition, we evaluated identical anti-TIGIT antibodies that do or do not engage FcγR and demonstrated that anti-TIGIT activity is dependent on FcγR binding. We subsequently used CRBN mice to investigate the efficacy of anti-TIGIT in combination with lenalidomide maintenance after transplantation. Notably, the combination of anti-TIGIT with lenalidomide provided synergistic, CD8+ T cell-dependent, antimyeloma efficacy. Analysis of bone marrow (BM) CD8+ T cells demonstrated that combination therapy suppressed T cell exhaustion, enhanced effector function, and expanded central memory subsets. Importantly, these immune phenotypes were specific to the BM tumor microenvironment. Collectively, these data provide a logical rationale for combining TIGIT inhibition with immunomodulatory drugs to prevent myeloma progression after ASCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Lenalidomide , Multiple Myeloma , Receptors, Immunologic , Animals , Mice , Immunity/drug effects , Immunity/genetics , Lenalidomide/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Neoplasm Recurrence, Local , Receptors, IgG , Stem Cell Transplantation/adverse effects , Transplantation, Autologous , Tumor Microenvironment , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism
14.
Blood ; 116(5): 819-28, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20435882

ABSTRACT

The recent shift to the use of stem cells mobilized by granulocyte colony-stimulating factor (G-CSF) for hematopoietic transplantation has increased chronic graftversus-host disease (GVHD), although the mechanisms of this are unclear. We have found that G-CSF invokes potent type 17 rather than type 1 or type 2 differentiation. The amplification of interleukin-17 (IL-17) production by G-CSF occurs in both CD4 and CD8 conventional T cells and is dependent on, and downstream of, G-CSF-induced IL-21 signaling. Importantly, donor IL-17A controls the infiltration of macrophages into skin and cutaneous fibrosis, manifesting late after transplantation as scleroderma. Interestingly, donor CD8 T cells were the predominant source of IL-17A after transplantation and could mediate scleroderma independently of CD4 T cells. This study provides a logical explanation for the propensity of allogeneic stem cell transplantation to invoke sclerodermatous GVHD and suggests a therapeutic strategy for intervention.


Subject(s)
Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Mobilization/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Interleukin-17/physiology , Interleukins/physiology , Scleroderma, Localized/etiology , T-Lymphocyte Subsets/transplantation , Animals , Cell Differentiation/drug effects , Crosses, Genetic , Cytokines/biosynthesis , Female , Fibrosis , Humans , Interleukin-17/metabolism , Macrophages/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Radiation Chimera , Signal Transduction , Skin/immunology , Skin/pathology , Transplantation, Homologous/adverse effects
15.
Blood ; 116(2): 287-96, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20435883

ABSTRACT

Suppressor of cytokine signaling-3 (SOCS3) is the main intracellular regulator of signaling by granulocyte colony-stimulating factor, an immune-modulatory cytokine used to mobilize stem cells for transplantation. We have therefore studied the contribution of SOCS3 to the spectrum of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Grafts from SOCS3(-/Deltavav) donor mice in which SOCS3 deficiency is restricted to the hematopoietic compartment had an augmented capacity to induce acute GVHD. With the use of SOCS3(-/DeltaLysM) and SOCS3(-/Deltalck) donors in which SOCS3 deficiency was restricted to the myeloid or T-cell lineage, respectively, we confirmed SOCS3 deficiency promoted acute GVHD mortality and histopathology within the gastrointestinal tract by effects solely within the donor T cell. SOCS3(-/Deltalck) donor T cells underwent enhanced alloantigen-dependent proliferation and generation of interleukin-10 (IL-10), IL-17, and interferon-gamma (IFNgamma) after SCT. The enhanced capacity of the SOCS3(-/Deltalck) donor T cell to induce acute GVHD was dependent on IFNgamma but independent of IL-10 or IL-17. Surprisingly, SOCS3(-/Deltalck) donor T cells also induced severe, transforming growth factor beta- and IFNgamma-dependent, sclerodermatous GVHD. Thus, the delivery of small molecule SOCS3 mimetics may prove to be useful for the inhibition of both acute and chronic GVHD.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes/immunology , Animals , Cell Separation , Cytokines/biosynthesis , Female , Flow Cytometry , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Mice, Inbred C57BL , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes/metabolism , Transplantation, Homologous
16.
Blood ; 115(1): 122-32, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19789388

ABSTRACT

Tumor necrosis factor (TNF) is a key cytokine in the effector phase of graft-versus-host disease (GVHD) after bone marrow transplantation, and TNF inhibitors have shown efficacy in clinical and experimental GVHD. TNF signals through the TNF receptors (TNFR), which also bind soluble lymphotoxin (LTalpha3), a TNF family member with a previously unexamined role in GVHD pathogenesis. We have used preclinical models to investigate the role of LT in GVHD. We confirm that grafts deficient in LTalpha have an attenuated capacity to induce GVHD equal to that seen when grafts lack TNF. This is not associated with other defects in cytokine production or T-cell function, suggesting that LTalpha3 exerts its pathogenic activity directly via TNFR signaling. We confirm that donor-derived LTalpha is required for graft-versus-leukemia (GVL) effects, with equal impairment in leukemic clearance seen in recipients of LTalpha- and TNF-deficient grafts. Further impairment in tumor clearance was seen using Tnf/Lta(-/-) donors, suggesting that these molecules play nonredundant roles in GVL. Importantly, donor TNF/LTalpha were only required for GVL where the recipient leukemia was susceptible to apoptosis via p55 TNFR signaling. These data suggest that antagonists neutralizing both TNF and LTalpha3 may be effective for treatment of GVHD, particularly if residual leukemia lacks the p55 TNFR.


Subject(s)
Graft vs Host Disease/immunology , Lymphotoxin-alpha/immunology , Animals , Apoptosis , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Graft vs Host Disease/pathology , Inflammation Mediators/metabolism , Mice , Protein Multimerization , Receptors, Antigen, T-Cell/immunology , Receptors, Immunologic/administration & dosage , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/immunology , Solubility , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/immunology
17.
EBioMedicine ; 75: 103784, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34968760

ABSTRACT

BACKGROUND: Protective malarial antibodies are acquired more rapidly in adults than children, independently of cumulative exposure, however the cellular responses mediating these differences are unknown. CD4 T-follicular helper (Tfh) cells have key roles in inducing antibodies, with Th2-Tfh cell activation associated with antibody development in malaria. Whether Tfh cell activation in malaria is age dependent is unknown and no studies have compared Tfh cell activation in children and adults with malaria. METHODS: We undertook a comprehensive study of Tfh cells, along with B cells and antibody induction in children and adults with malaria. Activation and proliferation of circulating Tfh (cTfh) cell subsets was measured ex vivo and parasite-specific Tfh cell frequencies and functions studied with Activation Induced Marker (AIM) assays and intracellular cytokine staining. FINDINGS: During acute malaria, the magnitude of cTfh cell activation was higher in adults than in children and occurred across all cTfh cell subsets in adults but was restricted only to the Th1-cTfh subset in children. Further, adults had higher levels of parasite-specific cTfh cells, and cTfh cells which produced more Th2-Tfh associated cytokine IL-4. Consistent with a role of higher Tfh cell activation in rapid immune development in adults, adults had higher activation of B cells during infection and higher induction of antibodies 7 and 28 days after malaria compared to children. INTERPRETATION: Our data provide evidence that age impacts Tfh cell activation during malaria, and that these differences may influence antibody induction after treatment. Findings have important implications for vaccine development in children. FUNDING: This word was supported by the National Health and Medical Research Council of Australia, Wellcome Trust, Charles Darwin University Menzies School of Health Research, Channel 7 Children's Research Foundation, and National Health Institute.


Subject(s)
Malaria, Falciparum , T Follicular Helper Cells , Adult , Australia , B-Lymphocytes , Child , Humans
18.
Blood ; 113(22): 5644-9, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19336758

ABSTRACT

We have quantified the relative contribution of donor antigen-presenting cell populations to alloantigen presentation after bone marrow transplantation (BMT) by using transgenic T cells that can respond to host-derived alloantigen presented within the donor major histocompatibility complex. We also used additional transgenic/knockout donor mice and/or monoclonal antibodies that allowed conditional depletion of conventional dendritic cells (cDCs), plasmacytoid DC (pDCs), macrophages, or B cells. Using these systems, we demonstrate that donor cDCs are the critical population presenting alloantigen after BMT, whereas pDCs and macrophages do not make a significant contribution in isolation. In addition, alloantigen presentation was significantly enhanced in the absence of donor B cells, confirming a regulatory role for these cells early after transplantation. These data have major implications for the design of therapeutic strategies post-BMT, and suggest that cDC depletion and the promotion of B-cell reconstitution may be beneficial tools for the control of alloreactivity.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Transplantation/immunology , Dendritic Cells/immunology , Isoantigens/immunology , Animal Experimentation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD11 Antigens/genetics , Dendritic Cells/physiology , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
19.
J Immunol ; 182(2): 912-20, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124734

ABSTRACT

The role of Ag presenting cell subsets in graft-versus-host disease (GVHD) remains unclear. We have thus examined the ability of plasmacytoid dendritic cells (pDC) to modulate transplant outcome. Surprisingly, host pDC were exquisitely sensitive to total body irradiation and were depleted before transplantation, thus allowing us to focus on donor pDC [corrected]. The depletion of all pDC from bone marrow grafts resulted in an acceleration of GVHD mortality while the depletion of mature pDC from G-CSF mobilized splenic grafts had no effect. Thus, donor bone marrow pDC, but not mature pDC contained within stem cell grafts attenuate acute GVHD. In the presence of GVHD, donor pDC completely failed to reconstitute although a CD11clow120G8+ precursor DC reconstituted in an exaggerated and transient manner. These cells expressed Flt-3, the macrophage colony stimulating factor receptor and, consistent with a common dendritic cell (DC) precursor, were capable of differentiation into pDC and conventional DC in vivo in the absence of GVHD. These precursors were MHC class II+ and CD80/86+ but lacked CD40, were actively presenting host Ag and inhibited GVHD and T cell proliferation in a contact-dependent fashion. These data demonstrate that GVHD prevents the maturation of pDC and instead promotes the generation of a suppressive precursor DC, further contributing to the state of immune paralysis after transplantation.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Cells, Cultured , Dendritic Cells/transplantation , Female , Graft vs Host Disease/diagnosis , Immunophenotyping , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Spleen/immunology , Spleen/pathology , Stem Cell Transplantation , Stem Cells/immunology , Stem Cells/pathology
20.
Clin Cancer Res ; 25(6): 1749-1755, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30765390

ABSTRACT

PURPOSE: Inducible caspase 9 (iCasp9) is a cellular safety switch that can make T-cell therapy safer. The purpose of this phase I trial was to investigate the use of iCasp9-transduced T-cell addback in adult patients undergoing haploidentical stem cell transplantation for high-risk hematologic malignancies. PATIENTS AND METHODS: Patients undergoing myeloablative, CD34-selected haploidentical stem cell transplantation were treated with 0.5-1.0 × 106/kg donor-derived iCasp9-transduced T cells on day +25 or 26 post-transplant, with additional doses allowed for disease relapse, infection, or mixed chimerism. RESULTS: Three patients were enrolled. iCasp9-transduced T cells were readily detectable by 4 weeks post-infusion in all patients and remained at high level (114 cells/µL, 11% of T cells) in 1 patient alive at 3.6 years. One patient developed donor-derived Epstein-Barr virus-associated post-transplant lymphoproliferative disease (EBV-PTLD), which was followed by a marked expansion of iCasp9 T cells and cytokine release syndrome (CRS). These iCasp9-transduced T cells infiltrated the affected lymph nodes and secreted IFNγ and IL-10. They peaked at 1,848 cells/µL and were found to be monoclonal by T-cell receptor (TCR) clonotype and oligoclonal by viral integrant analysis, representing a 6-log in vivo expansion of the dominant T-cell clone. These T cells were not autonomous and contracted with the resolution of EBV-PTLD, which did not recur. CONCLUSIONS: iCasp9-transduced T cells could persist long-term. They retained very high in vivo clonotypic proliferative capacity and function, and could cause CRS in response to de novo lymphoma development.


Subject(s)
Caspase 9/metabolism , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Myeloablative Agonists/administration & dosage , T-Lymphocytes/transplantation , Adolescent , Adult , Caspase 9/genetics , Caspase 9/immunology , Female , Hematologic Neoplasms/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Lymphocyte Depletion/adverse effects , Lymphocyte Depletion/methods , Male , Middle Aged , Myeloablative Agonists/adverse effects , Neoplasm Recurrence, Local , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transplantation Conditioning/methods , Transplantation, Haploidentical/adverse effects , Transplantation, Haploidentical/methods , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL