Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Lancet Haematol ; 11(5): e358-e367, 2024 May.
Article En | MEDLINE | ID: mdl-38555923

BACKGROUND: Chimeric antigen receptor (CAR) T cells targeting CD30 are safe and have promising activity when preceded by lymphodepleting chemotherapy. We aimed to determine the safety of anti-CD30 CAR T cells as consolidation after autologous haematopoietic stem-cell transplantation (HSCT) in patients with CD30+ lymphoma at high risk of relapse. METHODS: This phase 1 dose-escalation study was performed at two sites in the USA. Patients aged 3 years and older, with classical Hodgkin lymphoma or non-Hodgkin lymphoma with CD30+ disease documented by immunohistochemistry, and a Karnofsky performance score of more than 60% planned for autologous HSCT were eligible if they were considered high risk for relapse as defined by primary refractory disease or relapse within 12 months of initial therapy or extranodal involvement at the start of pre-transplantation salvage therapy. Patients received a single infusion of CAR T cells (2 × 107 CAR T cells per m2, 1 × 108 CAR T cells per m2, or 2 × 108 CAR T cells per m2) as consolidation after trilineage haematopoietic engraftment (defined as absolute neutrophil count ≥500 cells per µL for 3 days, platelet count ≥25 × 109 platelets per L without transfusion for 5 days, and haemoglobin ≥8 g/dL without transfusion for 5 days) following carmustine, etoposide, cytarabine, and melphalan (BEAM) and HSCT. The primary endpoint was the determination of the maximum tolerated dose, which was based on the rate of dose-limiting toxicity in patients who received CAR T-cell infusion. This study is registered with ClinicalTrials.gov (NCT02663297) and enrolment is complete. FINDINGS: Between June 7, 2016, and Nov 30, 2020, 21 patients were enrolled and 18 patients (11 with Hodgkin lymphoma, six with T-cell lymphoma, one with grey zone lymphoma) were infused with anti-CD30 CAR T cells at a median of 22 days (range 16-44) after autologous HSCT. There were no dose-limiting toxicities observed, so the highest dose tested, 2 × 108 CAR T cells per m2, was determined to be the maximum tolerated dose. One patient had grade 1 cytokine release syndrome. The most common grade 3-4 adverse events were lymphopenia (two [11%] of 18) and leukopenia (two [11%] of 18). There were no treatment-related deaths. Two patients developed secondary malignancies approximately 2 years and 2·5 years following treatment (one stage 4 non-small cell lung cancer and one testicular cancer), but these were judged unrelated to treatment. At a median follow-up of 48·2 months (IQR 27·5-60·7) post-infusion, the median progression-free survival for all treated patients (n=18) was 32·3 months (95% CI 4·6 months to not estimable) and the median progression-free survival for treated patients with Hodgkin lymphoma (n=11) has not been reached. The median overall survival for all treated patients has not been reached. INTERPRETATION: Anti-CD30 CAR T-cell infusion as consolidation after BEAM and autologous HSCT is safe, with low rates of toxicity and encouraging preliminary activity in patients with Hodgkin lymphoma at high risk of relapse, highlighting the need for larger studies to confirm these findings. FUNDING: National Heart Lung and Blood Institute, University Cancer Research Fund at the Lineberger Comprehensive Cancer Center.


Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Ki-1 Antigen , Transplantation, Autologous , Humans , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Female , Middle Aged , Adult , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Aged , Adolescent , Hodgkin Disease/therapy , Hodgkin Disease/immunology , Young Adult , Child , Receptors, Chimeric Antigen/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melphalan/therapeutic use , Melphalan/administration & dosage , Lymphoma, Non-Hodgkin/therapy , Lymphoma, Non-Hodgkin/immunology , Carmustine/therapeutic use , Carmustine/administration & dosage , Etoposide/therapeutic use , Etoposide/administration & dosage , Child, Preschool , Cytarabine/therapeutic use , Cytarabine/administration & dosage
2.
Blood Adv ; 5(22): 4578-4589, 2021 11 23.
Article En | MEDLINE | ID: mdl-34619767

Acute graft-versus-host disease (aGVHD), mediated by the recognition of host major histocompatibility complex/peptide polymorphisms by donor T cells, remains a significant complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). aGVHD most commonly involves the gastrointestinal tract, liver, and skin; symptomatic aGVHD is treated with corticosteroids. Steroid-nonresponsive aGVHD is a significant problem for patients undergoing allo-HSCT, with <15% of these patients alive 1 year after diagnosis. Previously, we found that the infusion of donor innate lymphoid type 2 (ILC2) cells could prevent and treat aGVHD of the lower gastrointestinal tract with no effect on the graft-versus-leukemia response. This approach for clinical translation is cumbersome, as it would require the generation of donor-derived ILC2 cells for each recipient. Thus, the ability to use third-party ILC2 cells would provide an "off-the-shelf" reagent that could be used to treat and/or prevent aGVHD. Here, we show that third-party ILC2 cells enhance the survival of allo-HSCT recipients. Treatment required at least 4 weekly infusions of ILC2 cells. Mechanistically, we show that ILC2 cell function was completely lost if the cells could not express both interleukin-13 (IL-13) and amphiregulin. Finally, we show that the activity of IL-13 has a greater dependence on the expression of the IL-13R on host rather than donor bone marrow cells. The ability to generate third-party ILC2 cells offers a new avenue for the prevention of aGVHD.


Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Gastrointestinal Tract , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunity, Innate , Lymphocytes
3.
Int J Mol Sci ; 21(3)2020 Jan 29.
Article En | MEDLINE | ID: mdl-32013193

Tacrolimus exhibits high inter-patient pharmacokinetics (PK) variability, as well as a narrow therapeutic index, and therefore requires therapeutic drug monitoring. Germline mutations in cytochrome P450 isoforms 4 and 5 genes (CYP3A4/5) and the ATP-binding cassette B1 gene (ABCB1) may contribute to interindividual tacrolimus PK variability, which may impact clinical outcomes among allogeneic hematopoietic stem cell transplantation (HSCT) patients. In this study, 252 adult patients who received tacrolimus for acute graft versus host disease (aGVHD) prophylaxis after allogeneic HSCT were genotyped to evaluate if germline genetic variants associated with tacrolimus PK and pharmacodynamic (PD) variability. Significant associations were detected between germline variants in CYP3A4/5 and ABCB1 and PK endpoints (e.g., median steady-state tacrolimus concentrations and time to goal tacrolimus concentration). However, significant associations were not observed between CYP3A4/5 or ABCB1 germline variants and PD endpoints (e.g., aGVHD and treatment-emergent nephrotoxicity). Decreased age and CYP3A5*1/*1 genotype were independently associated with subtherapeutic tacrolimus trough concentrations while CYP3A5*1*3 or CYP3A5*3/*3 genotypes, myeloablative allogeneic HSCT conditioning regimen (MAC) and increased weight were independently associated with supratherapeutic tacrolimus trough concentrations. Future lines of prospective research inquiry are warranted to use both germline genetic and clinical data to develop precision dosing tools that will optimize both tacrolimus dosing and clinical outcomes among adult HSCT patients.


ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Cytochrome P-450 CYP3A/genetics , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/pharmacokinetics , Tacrolimus/pharmacokinetics , Adult , Aged , Databases, Genetic , Female , Genotype , Germ-Line Mutation , Graft vs Host Disease/genetics , Graft vs Host Disease/prevention & control , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Logistic Models , Male , Middle Aged , Odds Ratio , Polymorphism, Single Nucleotide , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Transplantation, Homologous , Young Adult
4.
Am J Transplant ; 19(7): 1941-1954, 2019 07.
Article En | MEDLINE | ID: mdl-30748092

Acute graft-versus-host disease (aGVHD) remains a barrier to the success of allogeneic hematopoietic stem cell transplantation (HSCT). Previously, we demonstrated that CC-chemokine receptor 7 (CCR7) is critical for aGVHD pathogenesis but dispensable for beneficial graft-versus-leukemia responses. As a result, we evaluated a fully human anti-CCR7-blocking antibody as a new approach to prevent aGVHD in preclinical models. Here we report that antibody R707 is able to block human CCR7 signaling and function in vitro in response to its 2 natural ligands. The antibody was less active against the murine orthologue, however, and failed to substantially limit aGVHD in a standard murine allogeneic HSCT model. Nevertheless, R707 significantly reduced xenogeneic aGVHD induced by human peripheral blood mononuclear cells (PBMCs). R707 limited CD4+ and in particular CD8+ T cell expansion during the period of antibody administration. These effects were transient, however, and T cell numbers recovered after antibody cessation. R707 did not substantially impair the antitumor potential of the PBMC inoculum as antibody-treated mice retained their capacity to reject a human acute myeloid leukemia cell line. Collectively, these data indicate for the first time that an antibody directed against CCR7 might represent a viable new approach for aGVHD prevention.


Antibodies, Monoclonal/pharmacology , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Leukocytes, Mononuclear/immunology , Receptors, CCR7/immunology , T-Lymphocytes/immunology , Acute Disease , Animals , Antibodies, Monoclonal/immunology , Graft vs Host Disease/genetics , Hematopoietic Stem Cell Transplantation , Heterografts , Humans , Mice , Receptors, CCR7/genetics , T-Lymphocytes/transplantation , Tissue Donors , Transplantation, Homologous
5.
Biol Blood Marrow Transplant ; 25(6): 1062-1074, 2019 06.
Article En | MEDLINE | ID: mdl-30668984

Despite recent advances in therapy, allogeneic hematopoietic stem cell transplantation (HSCT) remains the only curative option for a range of high-risk hematologic malignancies. However, acute graft-versus-host disease (aGVHD) continues to limit the long-term success of HSCT, and new therapies are still needed. We previously demonstrated that aGVHD depends on the ability of donor conventional T cells (Tcons) to express the lymph node trafficking receptor, CC-Chemokine Receptor 7 (CCR7). Consequently, we examined the ability of cosalane, a recently identified CCR7 small-molecule antagonist, to attenuate aGVHD in mouse HSCT model systems. Here we show that the systemic administration of cosalane to transplant recipients after allogeneic HSCT did not prevent aGVHD. However, we were able to significantly reduce aGVHD by briefly incubating donor Tcons with cosalane ex vivo before transplantation. Cosalane did not result in Tcon toxicity and did not affect their activation or expansion. Instead, cosalane prevented donor Tcon trafficking into host secondary lymphoid tissues very early after transplantation and limited their subsequent accumulation within the liver and colon. Cosalane did not appear to impair the intrinsic ability of donor Tcons to produce inflammatory cytokines. Furthermore, cosalane-treated Tcons retained their graft-versus-leukemia (GVL) potential and rejected a murine P815 inoculum after transplantation. Collectively, our data indicate that a brief application of cosalane to donor Tcons before HSCT significantly reduces aGVHD in relevant preclinical models while generally sparing beneficial GVL effects, and that cosalane might represent a viable new approach for aGVHD prophylaxis.


Aurintricarboxylic Acid/analogs & derivatives , Graft vs Host Disease/genetics , Graft vs Leukemia Effect/genetics , Hematopoietic Stem Cell Transplantation/methods , Receptors, CCR7/metabolism , T-Lymphocytes/metabolism , Transplantation Conditioning/methods , Acute Disease , Animals , Aurintricarboxylic Acid/pharmacology , Aurintricarboxylic Acid/therapeutic use , Humans , Mice , Tissue Donors
6.
Biol Blood Marrow Transplant ; 25(2): 391-397, 2019 02.
Article En | MEDLINE | ID: mdl-30244102

Targeted busulfan dosing helps limit chemotherapy-related toxicity and optimize disease outcomes in hematopoietic stem cell transplantation (HCT). The objective of this study was to evaluate busulfan exposure from a pharmacokinetic (PK)-guided dosing strategy using a test dose. This retrospective evaluation included adult patients who underwent HCT at our institution with busulfan-based myeloablative (>9 mg/kg) conditioning between January 2014 and October 2015. A weight-based test dose of 0.8 mg/kg was used with PK assessments to predict area under the curve (AUCpred) achieved with weight-based dosing, with a target AUC of 4800 µM*minute (AUCtarget). PK from the test dose was then used to calculate a PK-guided first myeloablative busulfan dose. PK assessments were also done after the first dose to assess if the goal area under the curve (AUC) had been achieved (AUCfirst). A PK-guided first dose resulted in achievement of target AUC with target ranges of ±10% in 50% of patients, ±15% in 75%, and ±20% in 94%. This was an improved rate of target achievement compared with the 33%, 44%, and 63% of patients who achieved the desired AUC for these respective target ranges when using weight-based dosing (P = .12, .004, and <.001, respectively). The PK-guided strategy also decreased the variability of AUC from 3.6-fold in AUCpred from the weight-based test doses (2700.8 to 9631 µM*minute; SD, 1211.6 µM*minute) to 1.8-fold in AUCfirst from the PK-guided first doses (3672.1 to 6609.8 µM*minute; SD, 574.7 µM*minute). This reflects a 2-fold improvement in AUC variability with a PK-guided dosing strategy. This is also improved from the 3-fold variability in AUC reported in other studies. Weight and body surface area were significantly associated with the likelihood of AUCfirst being within the ±10% target range (P = .04 for both associations). There was no significant association between AUCfirst and death, relapse, or a composite of the two. These results demonstrate a significant improvement in target AUC attainment and less interpatient variability with PK-guided dosing using a test dose strategy compared with weight-based dosing.


Busulfan , Hematopoietic Stem Cell Transplantation , Myeloablative Agonists , Transplantation Conditioning , Adult , Busulfan/administration & dosage , Busulfan/pharmacokinetics , Disease-Free Survival , Female , Humans , Male , Middle Aged , Myeloablative Agonists/administration & dosage , Myeloablative Agonists/pharmacokinetics
7.
Blood Adv ; 2(18): 2307-2319, 2018 09 25.
Article En | MEDLINE | ID: mdl-30228128

Chronic graft-versus-host disease (cGVHD) causes significant morbidity and mortality in patients after allogeneic bone marrow (BM) or stem cell transplantation (allo-SCT). Recent work has indicated that both T and B lymphocytes play an important role in the pathophysiology of cGVHD. Previously, our group showed a critical role for the germinal center response in the function of B cells using a bronchiolitis obliterans (BO) model of cGVHD. Here, we demonstrated for the first time that cGVHD is associated with severe defects in the generation of BM B lymphoid and uncommitted common lymphoid progenitor cells. We found an increase in the number of donor CD4+ T cells in the BM of mice with cGVHD that was negatively correlated with B-cell development and the frequency of osteoblasts and Prrx-1-expressing perivascular stromal cells, which are present in the B-cell niche. Use of anti-DR3 monoclonal antibodies to enhance the number of donor regulatory T cells (Tregs) in the donor T-cell inoculum ameliorated the pathology associated with BO in this model. This correlated with an increased number of endosteal osteoblastic cells and significantly improved the generation of B-cell precursors in the BM after allo-SCT. Our work indicates that donor Tregs play a critical role in preserving the generation of B-cell precursors in the BM after allo-SCT. Approaches to enhance the number and/or function of donor Tregs that do not enhance conventional T-cell activity may be important to decrease the incidence and severity of cGVHD in part through normal B-cell lymphopoiesis.


B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Bronchiolitis Obliterans/etiology , Cell Differentiation , Graft vs Host Disease/etiology , Animals , B-Lymphocytes/metabolism , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bronchiolitis Obliterans/pathology , Bronchiolitis Obliterans/physiopathology , Cell Differentiation/immunology , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Graft vs Host Disease/pathology , Immunophenotyping , Lymphocyte Depletion , Mice , Mice, Transgenic , Osteoblasts/immunology , Osteoblasts/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
8.
Biol Blood Marrow Transplant ; 24(11): 2184-2189, 2018 11.
Article En | MEDLINE | ID: mdl-29981461

Acute graft-versus-host disease (aGVHD) remains a barrier to the success of allogeneic hematopoietic stem cell transplantation. In mice, studies have demonstrated that donor conventional T cells traffic into host secondary lymphoid tissues early after transplant, and that this process is critical for the development of disease. As a result, the measurement of cellular proliferation within lymphoid sites early after transplant might be a useful approach for predicting aGVHD in humans. 18F-3'-deoxy-3'-fluorothymidine (FLT) positron emission tomography (PET) imaging has recently emerged as a functional imaging modality in oncology patients. FLT, a thymidine analog, is incorporated into replicating DNA and is thus an indirect marker of cellular proliferation. Here we report that FLT PET imaging can differentiate mice receiving alloreactive T cells and destined to develop lethal aGVHD from control mice. Mice receiving allogeneic T cells demonstrated a stronger FLT signal within the peripheral lymph nodes compared with control mice at all time points after transplant. In addition, allogeneic T cell recipients transiently demonstrated stronger FLT uptake within the spleen. Importantly, these differences were apparent before the development of clinical disease. In contrast, the FLT signal within the host bowel, an important aGVHD target organ, was more variable after transplant and was not consistently different between aGVHD mice and control mice. Collectively, these findings suggest that the imaging of patient lymphoid sites using existing FLT PET technology might be useful for predicting aGVHD in the clinical setting.


Fluorodeoxyglucose F18/therapeutic use , Graft vs Host Disease/diagnostic imaging , Hematopoietic Stem Cell Transplantation/methods , Positron-Emission Tomography/methods , Transplantation Conditioning/methods , Acute Disease , Animals , Disease Models, Animal , Graft vs Host Disease/pathology , Mice
9.
SLAS Discov ; 23(10): 1083-1091, 2018 12.
Article En | MEDLINE | ID: mdl-29958052

CC-chemokine receptor 7 (CCR7) is a G protein-coupled receptor expressed on a variety of immune cells. CCR7 plays a critical role in the migration of lymphocytes into secondary lymphoid tissues. CCR7 expression, however, has been linked to numerous disease states. Due to its therapeutic relevance and absence of available CCR7 inhibitors, we undertook a high-throughput screen (HTS) to identify small-molecule antagonists of the receptor. Here, we describe a robust HTS approach using a commercially available ß-galactosidase enzyme fragment complementation system and confirmatory transwell chemotaxis assays. This work resulted in the identification of several compounds with activity against CCR7. The most potent of these was subsequently determined to be cosalane, a cholesterol derivative previously designed as a therapeutic for human immunodeficiency virus. Cosalane inhibited both human and murine CCR7 in response to both CCL19 and CCL21 agonists at physiologic concentrations. Furthermore, cosalane produced durable inhibition of the receptor following a cellular incubation period with subsequent washout. Overall, our work describes the development of an HTS-compatible assay, completion of a large HTS campaign, and demonstration for the first time that cosalane is a validated CCR7 antagonist. These efforts could pave the way for new approaches to address CCR7-associated disease processes.


Aurintricarboxylic Acid/analogs & derivatives , High-Throughput Screening Assays , Receptors, CCR7/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , Cell Line , Chemotaxis/drug effects , Drug Design , Humans , Ligands , Mice , Molecular Structure , Receptors, CCR7/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
Blood ; 130(3): 348-359, 2017 07 20.
Article En | MEDLINE | ID: mdl-28550042

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that affects the function and development of immune cells. Here, we show that recipient mice receiving AhR-/- T cells have improved survival and decreased acute graft-versus-host disease (aGVHD) in 2 different murine allogeneic bone marrow transplant (BMT) models. We also show that CD4+ T cells lacking AhR demonstrate reduced accumulation in secondary lymphoid tissue because of low levels of proliferation 4 days after BMT. Additionally, we found a significant increase in the quantity of peripherally induced regulatory donor T (pTreg) cells in the colon of recipients transplanted with AhR-/- T cells 14 days after transplant. Blockade of AhR using a clinically available AhR antagonist greatly enhanced the in vitro generation of inducible Treg (iTreg) cells from naïve CD4+ human T cells. We have identified AhR as a novel target on donor T cells that is critical to the pathogenesis of aGVHD.


Basic Helix-Loop-Helix Transcription Factors/immunology , Bone Marrow Transplantation , Colon/immunology , Graft vs Host Disease/prevention & control , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Colon/pathology , Gene Expression Regulation , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Purines/pharmacology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Sirolimus/pharmacology , Survival Analysis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/transplantation , Transplantation, Heterologous
11.
J Clin Invest ; 127(5): 1813-1825, 2017 May 01.
Article En | MEDLINE | ID: mdl-28375154

Acute graft-versus-host disease (aGVHD) is the most common complication for patients undergoing allogeneic stem cell transplantation. Despite extremely aggressive therapy targeting donor T cells, patients with grade III or greater aGVHD of the lower GI tract, who do not respond to therapy with corticosteroids, have a dismal prognosis. Thus, efforts to improve understanding of the function of local immune and non-immune cells in regulating the inflammatory process in the GI tract during aGVHD are needed. Here, we demonstrate, using murine models of allogeneic BMT, that type 2 innate lymphoid cells (ILC2s) in the lower GI tract are sensitive to conditioning therapy and show very limited ability to repopulate from donor bone marrow. Infusion of donor ILC2s was effective in reducing the lethality of aGVHD and in treating lower GI tract disease. ILC2 infusion was associated with reduced donor proinflammatory Th1 and Th17 cells, accumulation of donor myeloid-derived suppressor cells (MDSCs) mediated by ILC2 production of IL-13, improved GI tract barrier function, and a preserved graft-versus-leukemia (GVL) response. Collectively, these findings suggest that infusion of donor ILC2s to restore gastrointestinal tract homeostasis may improve treatment of severe lower GI tract aGVHD.


Bone Marrow Transplantation , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/therapy , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Lymphocytes/immunology , Acute Disease , Allografts , Animals , Gastrointestinal Diseases/pathology , Graft vs Host Disease/pathology , Lymphocytes/pathology , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/pathology
12.
Biol Blood Marrow Transplant ; 23(4): 569-580, 2017 Apr.
Article En | MEDLINE | ID: mdl-28161607

Idiopathic pneumonia syndrome (IPS) is a noninfectious inflammatory disorder of the lungs that occurs most often after fully myeloablative allogeneic hematopoietic stem cell transplantation (HSCT). IPS can be severe and is associated with high 1-year mortality rates despite existing therapies. The canonical nuclear factor-(NF) κB signaling pathway has previously been linked to several inflammatory disorders of the lung, including asthma and lung allograft rejection. It has never been specifically targeted as a novel IPS treatment approach, however. Here, we report that the IκB kinase 2 (IKK2) antagonist BAY 65-5811 or "compound A," a highly potent and specific inhibitor of the NF-κB pathway, was able to improve median survival times and recipient oxygenation in a well-described mouse model of IPS. Compound A impaired the production of the proinflammatory chemokines CCL2 and CCL5 within the host lung after transplantation. This resulted in significantly lower numbers of donor lung infiltrating CD4+ and CD8+ T cells and reduced pulmonary inflammatory cytokine production after allograft. Compound A's beneficial effects appeared to be specific for limiting pulmonary injury, as the drug was unable to improve outcomes in a B6 into B6D2 haplotype-matched murine HSCT model in which recipient mice succumb to lethal acute graft-versus-host disease of the gastrointestinal tract. Collectively, our data suggest that the targeting of the canonical NF-κB pathway with a small molecule IKK2 antagonist may represent an effective and novel therapy for the specific management of acute lung injury that can occur after allogeneic HSCT.


Hematopoietic Stem Cell Transplantation/adverse effects , I-kappa B Kinase/antagonists & inhibitors , Lung Injury/drug therapy , Molecular Targeted Therapy/methods , NF-kappa B/metabolism , Pneumonia/drug therapy , Animals , Lung Injury/etiology , Mice , Treatment Outcome
13.
Cancer Immunol Res ; 3(3): 228-35, 2015 Mar.
Article En | MEDLINE | ID: mdl-25576336

Testing of T cell-based cancer therapeutics often involves measuring cancer antigen-specific T-cell populations with the assumption that they arise from in vivo clonal expansion. This analysis, using peptide/MHC tetramers, is often ambiguous. From a leukemia cell line, we identified a CDK4-derived peptide epitope, UNC-CDK4-1 (ALTPVVVTL), that bound HLA-A*02:01 with high affinity and could induce CD8⁺ T-cell responses in vitro. We identified UNC-CDK4-1/HLA-A*02:01 tetramer⁺ populations in 3 of 6 patients with acute myeloid leukemia who had undergone allogeneic stem cell transplantation. Using tetramer-based, single-cell sorting and T-cell receptor ß (TCRß) sequencing, we identified recurrent UNC-CDK4-1 tetramer-associated TCRß clonotypes in a patient with a UNC-CDK4-1 tetramer⁺ population, suggesting in vivo T-cell expansion to UNC-CDK4-1. In parallel, we measured the patient's TCRß repertoire and found it to be highly restricted/oligoclonal. The UNC-CDK4-1 tetramer-associated TCRß clonotypes represented >17% of the entire TCRß repertoire-far in excess of the UNC-CDK4-1 tetramer⁺ frequency-indicating that the recurrent TCRß clonotypes identified from UNC-CDK-4-1 tetramer⁺ cells were likely a consequence of the extremely constrained T-cell repertoire in the patient and not in vivo UNC-CDK4-1-driven clonal T-cell expansion. Mapping recurrent TCRß clonotype sequences onto TCRß repertoires can help confirm or refute antigen-specific T-cell expansion in vivo.


Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , Female , HLA-A2 Antigen/immunology , Humans , Leukemia/immunology , Male , Middle Aged , Peptides/immunology , U937 Cells
14.
Eur J Immunol ; 44(6): 1662-71, 2014 Jun.
Article En | MEDLINE | ID: mdl-24752751

Acute graft-versus-host disease (aGvHD) is a major limitation to the use of allogeneic stem cell transplantation for the treatment of patients with relapsed malignant disease. Previous work using animals lacking secondary lymphoid tissue (SLT) suggested that activation of donor T cells in SLT is critically important for the pathogenesis of aGvHD. However, these studies did not determine if impaired migration into, and more importantly, out of SLT, would ameliorate aGvHD. Here, we show that T cells from mice lacking Coronin 1A (Coro 1A(-/-)), an actin-associated protein shown to be important for thymocyte egress, do not mediate acute GvHD. The attenuation of aGvHD was associated with decreased expression of the critical trafficking proteins C-C chemokines receptor type 7 (CCR7) and sphingosine 1 phosphate receptor on donor T cells. This was mediated in part by impaired activation of the canonical NF-κB pathway in the absence of Coro 1A. As a result of these alterations, donor T cells from Coro 1A(-/-) mice were not able to initially traffic to SLT or exit SLT after BM transplantation. However, this alteration did not abrogate the graft-versus-leukemia response. Our data suggest that blocking T-cell migration into and out of SLT is a valid approach to prevent aGvHD.


Bone Marrow Transplantation , Cell Movement/immunology , Graft vs Host Disease/immunology , Microfilament Proteins/immunology , T-Lymphocytes/immunology , Acute Disease , Allografts , Animals , Cell Movement/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Mice , Mice, Inbred BALB C , Mice, Knockout , Microfilament Proteins/isolation & purification , NF-kappa B/genetics , NF-kappa B/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/pathology
15.
Blood ; 123(10): 1604-14, 2014 Mar 06.
Article En | MEDLINE | ID: mdl-24415540

Graft-versus-host disease (GVHD) is a systemic inflammatory response due to the recognition of major histocompatibility complex disparity between donor and recipient after hematopoietic stem cell transplantation (HSCT). T-cell activation is critical to the induction of GVHD, and data from our group and others have shown that regulatory T cells (Tregs) prevent GVHD when given at the time of HSCT. Using multiphoton laser scanning microscopy, we examined the single cell dynamics of donor T cells and dendritic cells (DCs) with or without Tregs postallogeneic transplantation. We found that donor conventional T cells (Tcons) spent very little time screening host DCs. Tcons formed stable contacts with DCs very early after transplantation and only increased velocity in the lymph node at 20 hours after transplant. We also observed that Tregs reduced the interaction time between Tcons and DCs, which was dependent on the generation of interleukin 10 by Tregs. Imaging using inducible Tregs showed similar disruption of Tcon-DC contact. Additionally, we found that donor Tregs induce host DC death and down-regulate surface proteins required for donor T-cell activation. These data indicate that Tregs use multiple mechanisms that affect host DC numbers and function to mitigate acute GVHD.


Dendritic Cells/immunology , Graft vs Host Disease/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-2 Antigen/metabolism , Cell Communication/immunology , Cell Death/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
16.
Blood ; 122(5): 825-36, 2013 Aug 01.
Article En | MEDLINE | ID: mdl-23798714

The infusion of donor regulatory T cells (Tregs) has been used to prevent acute graft-versus-host disease (GVHD) in mice and has shown promise in phase 1 clinical trials. Previous work suggested that early Treg migration into lymphoid tissue was important for GVHD prevention. However, it is unclear how and where Tregs function longitudinally to affect GVHD. To better understand their mechanism of action, we studied 2 Treg-associated chemokine receptors in murine stem cell transplant models. CC chemokine receptor (CCR) 4 was dispensable for donor Treg function in the transplant setting. Donor Tregs lacking CCR8 (CCR8(-/-)), however, were severely impaired in their ability to prevent lethal GVHD because of increased cell death. By itself, CCR8 stimulation was unable to rescue Tregs from apoptosis. Instead, CCR8 potentiated Treg survival by promoting critical interactions with dendritic cells. In vivo, donor bone marrow-derived CD11c(+) antigen-presenting cells (APCs) were important for promoting donor Treg maintenance after transplant. In contrast, host CD11c(+) APCs appeared to be dispensable for early activation and expansion of donor Tregs. Collectively, our data indicate that a sustained donor Treg presence is critical for their beneficial properties, and that their survival depends on CCR8 and donor but not host CD11c(+) APCs.


Graft vs Host Disease/genetics , Graft vs Host Disease/prevention & control , Receptors, CCR8/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/physiology , CD11c Antigen/metabolism , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Graft Survival/genetics , Graft Survival/immunology , Graft vs Host Disease/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Receptors, CCR8/genetics , Receptors, CCR8/metabolism , T-Lymphocytes, Regulatory/metabolism , Tissue Donors
17.
J Immunol ; 189(4): 1765-72, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22778391

Graft-versus-host disease (GVHD) remains the most significant complication after allogeneic stem cell transplantation. Previously, acute GVHD had been considered to be mediated predominantly by Th1-polarized T cells. Recently, investigators have identified a second proinflammatory lineage of T cells termed Th17 that is critically dependent on the transcription factor retinoic acid-related orphan receptor (ROR)γt. In this study, we have evaluated the role of Th17 cells in murine acute GVHD by infusing donor T cells lacking RORC and as a consequence the isoform RORγt. Recipients given donor CD4(+) and CD8(+) T cells lacking RORC had significantly attenuated acute GVHD and markedly decreased tissue pathology in the colon, liver, and lung. Using a clinically relevant haploidentical murine transplantation model, we showed that RORC(-/-) CD4(+) T cells alone diminished the severity and lethality of acute GVHD. This was not found when CD4(+) T cells from RORC(-/-) mice were given to completely mismatched BALB/c mice, and it was correlated with absolute differences in the generation of TNF in the colon after transplant. Thus, CD4(+) T cell expression of RORC is important in the pathogenesis of acute GVHD.


Graft vs Host Disease/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Real-Time Polymerase Chain Reaction
18.
Blood ; 117(12): 3268-76, 2011 Mar 24.
Article En | MEDLINE | ID: mdl-21245483

GVHD is a syndrome that results from minor and major histocompatibility complex incompatibilities between the donor and recipient. More than 50 years after its initial description, the pathophysiology of GVHD remains poorly understood. Nonetheless, donor T cells have been shown to be critical to the pathophysiology of acute and chronic GVHD, yet precisely how they function remains unclear. The effector mechanisms by which donor T cells mediate tissue inflammation is even less well understood. Identification of several new lineages of CD4(+) T cells made in the past decade and their roles in the pathophysiology of T cell-mediated diseases has shed new light on these effector mechanisms. In this review, we summarize the recent descriptions of these T-cell lineages and the current data supporting their role in acute and to a lesser extent chronic GVHD. Investigations into the activity of these new T-cell lineages may provide more rationale approaches to the treatment or prevention of GVHD.


CD4-Positive T-Lymphocytes/physiology , Cytokines/metabolism , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Lineage/immunology , Humans , Models, Biological , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/physiology , Transplantation Immunology/physiology
19.
Blood ; 115(23): 4914-22, 2010 Jun 10.
Article En | MEDLINE | ID: mdl-20185583

CC-chemokine receptor 7 (CCR7) is expressed on the surface of naive T cells, and plays a critical role in their movement into secondary lymphoid tissue. Here, we show that murine T cells lacking CCR7 (CCR7(-/-)) generate attenuated graft-versus-host disease (GVHD) responses compared with wild-type (WT) cells, with the difference varying inversely with the degree of major histocompatibility complex (MHC) disparity between the donor and recipient. CCR7(-/-) T cells exhibited an impaired ability to traffic to recipient lymph nodes, with an increased capacity to home to the spleen. CCR7(-/-) T cells, however, demonstrated a reduced ability to undergo in vivo expansion in the spleen due to impaired interactions with splenic antigen-presenting cells. On a cellular level, CCR7(-/-) T cells were functionally competent, demonstrating a normal in vitro proliferative capacity and a preserved ability to produce inflammatory cytokines. Importantly, CCR7(-/-) T cells were capable of generating robust graft-versus-leukemia (GVL) responses in vivo, as well as complete donor T-cell reconstitution. CCR7(-/-) regulatory T cells were able to protect against lethal GVHD when administered before WT conventional T cells. Our data suggest that CCR7 inhibition in the early posttransplantation period may represent a feasible new therapeutic approach for acute GVHD attenuation without compromising GVL responses.


Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/immunology , Receptors, CCR7/immunology , T-Lymphocytes/immunology , Animals , Cell Proliferation , Cytokines/immunology , Graft vs Host Disease/genetics , Graft vs Leukemia Effect/genetics , Humans , Inflammation Mediators/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, CCR7/genetics , Spleen/immunology , T-Lymphocytes/transplantation
20.
Blood ; 113(6): 1365-74, 2009 Feb 05.
Article En | MEDLINE | ID: mdl-18957685

The morbidity and mortality associated with graft-host-disease (GVHD) is a significant obstacle to the greater use of allogeneic stem cell transplantation. Donor T cells that predominantly differentiate into TH1/Tc1 T cells and generate pro-inflammatory cytokines such as interferon-gamma (IFN-gamma) mediate GVHD. Although numerous studies have described a pathogenic role for IFN-gamma, multiple reports have demonstrated that the lack of IFN-gamma paradoxically exacerbated GVHD lethality. This has led to speculation that another subset of T cells may significantly contribute to GVHD mortality. Several groups have demonstrated a new lineage of CD4+ T helper cell development distinct from TH1 or TH2 differentiation. This lineage is characterized by production of interleukin (IL)-17A, IL-17F, IL-22, and IL-21 and has been termed TH17 cells. Here, we demonstrate that a highly purified population of TH17 cells is capable of inducing lethal GVHD, hallmarked by extensive pathologic cutaneous and pulmonary lesions. Upon transfer, these cells migrate to and expand in GVHD target organs and secondary lymphoid tissues. Finally, we demonstrate differential roles for tumor necrosis factor-alpha (TNF-alpha) and IL-17A in the clinical manifestations of GVHD induced by TH17 cells. Our studies demonstrate that cells other than TH1/Tc1 can mediate acute GVHD.


Cell Differentiation , Graft vs Host Disease/etiology , Lung Diseases/etiology , Skin Diseases/etiology , T-Lymphocytes, Helper-Inducer/immunology , Acute Disease , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Flow Cytometry , Interferon-gamma/physiology , Interleukin-17/immunology , Leukocytes/cytology , Leukocytes/metabolism , Lung Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Diseases/pathology , Survival Rate , T-Lymphocytes, Helper-Inducer/transplantation , Tumor Necrosis Factor-alpha/blood
...