224 results on '"Kean, Leslie S"'
Search Results
202. Enforced gut homing of murine regulatory T cells reduces early graft-versus-host disease severity.
- Author
-
Larson JH, Jin S, Loschi M, Bolivar Wagers S, Thangavelu G, Zaiken MC, McDonald-Hyman C, Saha A, Aguilar EG, Koehn B, Osborn MJ, Panoskaltsis-Mortari A, Macdonald KPA, Hill GR, Murphy WJ, Serody JS, Maillard I, Kean LS, Kim SV, Littman DR, and Blazar BR
- Subjects
- Animals, Mice, T-Lymphocytes, Regulatory, Intestine, Small, Inflammation, Graft vs Host Disease etiology, Graft vs Host Disease prevention & control, Graft vs Host Disease drug therapy, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Damage to the gastrointestinal tract following allogeneic hematopoietic stem cell transplantation is a significant contributor to the severity and perpetuation of graft-versus-host disease. In preclinical models and clinical trials, we showed that infusing high numbers of regulatory T cells reduces graft-versus-host disease incidence. Despite no change in in vitro suppressive function, transfer of ex vivo expanded regulatory T cells transduced to overexpress G protein-coupled receptor 15 or C-C motif chemokine receptor 9, specific homing receptors for colon or small intestine, respectively, lessened graft-versus-host disease severity in mice. Increased regulatory T cell frequency and retention within the gastrointestinal tissues of mice that received gut homing T cells correlated with lower inflammation and gut damage early post-transplant, decreased graft-versus-host disease severity, and prolonged survival compared with those receiving control transduced regulatory T cells. These data provide evidence that enforced targeting of ex vivo expanded regulatory T cells to the gastrointestinal tract diminishes gut injury and is associated with decreased graft-versus-host disease severity., (Copyright © 2023 American Society of Transplantation & American Society of Transplant Surgeons. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
203. Alemtuzumab and CXCL9 levels predict likelihood of sustained engraftment after reduced-intensity conditioning HCT.
- Author
-
Geerlinks AV, Scull B, Krupski C, Fleischmann R, Pulsipher MA, Eapen M, Connelly JA, Bollard CM, Pai SY, Duncan CN, Kean LS, Baker KS, Burroughs LM, Andolina JR, Shenoy S, Roehrs P, Hanna R, Talano JA, Schultz KR, Stenger EO, Lin H, Zoref-Lorenz A, McClain KL, Jordan MB, Man TK, Allen CE, and Marsh RA
- Subjects
- Humans, Alemtuzumab therapeutic use, Melphalan therapeutic use, Tissue Donors, Chemokine CXCL9, Antibodies, Monoclonal, Humanized therapeutic use, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Overall survival after reduced-intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (HCT) using alemtuzumab, fludarabine, and melphalan is associated with high rates of mixed chimerism (MC) and secondary graft failure (GF). We hypothesized that peritransplantation alemtuzumab levels or specific patterns of inflammation would predict these risks. We assessed samples from the Bone Marrow Transplant Clinical Trials Network 1204 (NCT01998633) to study the impact of alemtuzumab levels and cytokine patterns on MC and impending or established secondary GF (defined as donor chimerism <5% after initial engraftment and/or requirement of cellular intervention). Thirty-three patients with hemophagocytic lymphohistiocytosis (n = 25) and other IEIs (n = 8) who underwent HCTs with T-cell-replete grafts were included. Patients with day 0 alemtuzumab levels ≤0.32 μg/mL had a markedly lower incidence of MC, 14.3%, vs 90.9% in patients with levels >0.32 μg/mL (P = .008). Impending or established secondary GF was only observed in patients with day 0 alemtuzumab levels >0.32 μg/mL (P = .08). Unexpectedly, patients with impending or established secondary GF had lower CXCL9 levels. The cumulative incidence of impending or established secondary GF in patients with a day 14+ CXCL9 level ≤2394 pg/mL (day 14+ median) was 73.6% vs 0% in patients with a level >2394 pg/mL (P = .002). CXCL9 levels inversely correlated with alemtuzumab levels. These data suggest a model in which higher levels of alemtuzumab at day 0 deplete donor T cells, inhibit the graft-versus-marrow reaction (thereby suppressing CXCL9 levels), and adversely affect sustained engraftment in the nonmyeloablative HCT setting. This trial was registered at www.clinicaltrials.gov as #NCT01998633., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
204. Post-Transplantation Cyclophosphamide-Based Graft-versus-Host Disease Prophylaxis.
- Author
-
Bolaños-Meade J, Hamadani M, Wu J, Al Malki MM, Martens MJ, Runaas L, Elmariah H, Rezvani AR, Gooptu M, Larkin KT, Shaffer BC, El Jurdi N, Loren AW, Solh M, Hall AC, Alousi AM, Jamy OH, Perales MA, Yao JM, Applegate K, Bhatt AS, Kean LS, Efebera YA, Reshef R, Clark W, DiFronzo NL, Leifer E, Horowitz MM, Jones RJ, and Holtan SG
- Subjects
- Adult, Humans, Methotrexate administration & dosage, Mycophenolic Acid administration & dosage, Neoplasm Recurrence, Local drug therapy, Tacrolimus administration & dosage, Unrelated Donors, Bronchiolitis Obliterans Syndrome etiology, Bronchiolitis Obliterans Syndrome prevention & control, Cyclophosphamide administration & dosage, Graft vs Host Disease prevention & control, Graft vs Host Disease etiology, Hematopoietic Stem Cell Transplantation adverse effects, Hematopoietic Stem Cell Transplantation methods, Hematologic Neoplasms surgery, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
Background: In patients undergoing allogeneic hematopoietic stem-cell transplantation (HSCT), a calcineurin inhibitor plus methotrexate has been a standard prophylaxis against graft-versus-host disease (GVHD). A phase 2 study indicated the potential superiority of a post-transplantation regimen of cyclophosphamide, tacrolimus, and mycophenolate mofetil., Methods: In a phase 3 trial, we randomly assigned adults with hematologic cancers in a 1:1 ratio to receive cyclophosphamide-tacrolimus-mycophenolate mofetil (experimental prophylaxis) or tacrolimus-methotrexate (standard prophylaxis). The patients underwent HSCT from an HLA-matched related donor or a matched or 7/8 mismatched (i.e., mismatched at only one of the HLA-A , HLA-B , HLA-C , and HLA-DRB1 loci) unrelated donor, after reduced-intensity conditioning. The primary end point was GVHD-free, relapse-free survival at 1 year, assessed in a time-to-event analysis, with events defined as grade III or IV acute GVHD, chronic GVHD warranting systemic immunosuppression, disease relapse or progression, and death from any cause., Results: In a multivariate Cox regression analysis, GVHD-free, relapse-free survival was significantly more common among the 214 patients in the experimental-prophylaxis group than among the 217 patients in the standard-prophylaxis group (hazard ratio for grade III or IV acute GVHD, chronic GVHD, disease relapse or progression, or death, 0.64; 95% confidence interval [CI], 0.49 to 0.83; P = 0.001). At 1 year, the adjusted GVHD-free, relapse-free survival was 52.7% (95% CI, 45.8 to 59.2) with experimental prophylaxis and 34.9% (95% CI, 28.6 to 41.3) with standard prophylaxis. Patients in the experimental-prophylaxis group appeared to have less severe acute or chronic GVHD and a higher incidence of immunosuppression-free survival at 1 year. Overall and disease-free survival, relapse, transplantation-related death, and engraftment did not differ substantially between the groups., Conclusions: Among patients undergoing allogeneic HLA-matched HSCT with reduced-intensity conditioning, GVHD-free, relapse-free survival at 1 year was significantly more common among those who received cyclophosphamide-tacrolimus-mycophenolate mofetil than among those who received tacrolimus-methotrexate. (Funded by the National Heart, Lung, and Blood Institute and others; BMT CTN 1703 ClinicalTrials.gov number, NCT03959241.)., (Copyright © 2023 Massachusetts Medical Society.)
- Published
- 2023
- Full Text
- View/download PDF
205. Pre-existing immunity does not impair the engraftment of CRISPR-Cas9-edited cells in rhesus macaques conditioned with busulfan or radiation.
- Author
-
Essawi K, Hakami W, Naeem Khan MB, Martin R, Zeng J, Chu R, Uchida N, Bonifacino AC, Krouse AE, Linde NS, Donahue RE, Blobel GA, Gerdemann U, Kean LS, Maitland SA, Wolfe SA, Metais JY, Gottschalk S, Bauer DE, Tisdale JF, and Demirci S
- Abstract
CRISPR-Cas9-based therapeutic genome editing approaches hold promise to cure a variety of human diseases. Recent findings demonstrate pre-existing immunity for the commonly used Cas orthologs from Streptococcus pyogenes (SpCas9) and Staphylococcus aureus (SaCas9) in humans, which threatens the success of this powerful tool in clinical use. Thus, a comprehensive investigation and potential risk assessment are required to exploit the full potential of the system. Here, we investigated existence of immunity to SpCas9 and SaCas9 in control rhesus macaques ( Macaca mulatta) alongside monkeys transplanted with either lentiviral transduced or CRISPR-SpCas9 ribonucleoprotein (RNP)-edited cells. We observed significant levels of Cas9 antibodies in the peripheral blood of all transplanted and non-transplanted control animals. Transplantation of ex vivo transduced or SpCas9-mediated BCL11A enhancer-edited cells did not alter the levels of Cas9 antibodies in rhesus monkeys. Following stimulation of peripheral blood cells with SpCas9 or SaCas9, neither Cas9-specific T cells nor cytokine induction were detected. Robust and durable editing frequencies and expression of high levels of fetal hemoglobin in BCL11A enhancer-edited rhesus monkeys with no evidence of an immune response (>3 years) provide an optimistic outlook for the use of ex vivo CRISPR-SpCas9 (RNP)-edited cells., Competing Interests: S.G. has patent applications in the fields of cell and/or gene therapy for cancer. S.G. is a consultant of TESSA Therapeutics, a DSMB member of Immatics, and has received honoraria from Tidal, Catamaran Bio, Sanofi, and Novartis within the last 2 years.
- Published
- 2023
- Full Text
- View/download PDF
206. Engineering T cells to suppress acute GVHD and leukemia relapse after allogeneic hematopoietic stem cell transplantation.
- Author
-
Mo F, Watanabe N, Omdahl KI, Burkhardt PM, Ding X, Hayase E, Panoskaltsis-Mortari A, Jenq RR, Heslop HE, Kean LS, Brenner MK, Tkachev V, and Mamonkin M
- Subjects
- Humans, Animals, Mice, Leukocytes, Mononuclear pathology, Macaca mulatta, Herpesvirus 4, Human, Chronic Disease, Recurrence, Epstein-Barr Virus Infections complications, Graft vs Host Disease etiology, Leukemia complications, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Acute graft-versus-host disease (aGVHD) limits the therapeutic benefit of allogeneic hematopoietic stem cell transplantation (allo-HSCT) and requires immunosuppressive prophylaxis that compromises antitumor and antipathogen immunity. OX40 is a costimulatory receptor upregulated on circulating T cells in aGVHD and plays a central role in driving the expansion of alloreactive T cells. Here, we show that OX40 is also upregulated on T cells infiltrating GVHD target organs in a rhesus macaque model, supporting the hypothesis that targeted ablation of OX40+ T cells will mitigate GVHD pathogenesis. We thus created an OX40-specific cytotoxic receptor that, when expressed on human T cells, enables selective elimination of OX40+ T cells. Because OX40 is primarily upregulated on CD4+ T cells upon activation, engineered OX40-specific T cells mediated potent cytotoxicity against activated CD4+ T cells and suppressed alloreactive T-cell expansion in a mixed lymphocyte reaction model. OX40 targeting did not inhibit antiviral activity of memory T cells specific to Epstein-Barr virus, cytomegalovirus, and adenoviral antigens. Systemic administration of OX40-targeting T cells fully protected mice from fatal xenogeneic GVHD mediated by human peripheral blood mononuclear cells. Furthermore, combining OX40 targeting with a leukemia-specific chimeric antigen receptor in a single T cell product provides simultaneous protection against leukemia and aGVHD in a mouse xenograft model of residual disease posttransplant. These results underscore the central role of OX40+ T cells in mediating aGVHD pathogenesis and support the feasibility of a bifunctional engineered T-cell product derived from the stem cell donor to suppress both disease relapse and aGVHD following allo-HSCT., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
207. Retinoic acid signaling acts as a rheostat to balance Treg function.
- Author
-
Thangavelu G, Andrejeva G, Bolivar-Wagers S, Jin S, Zaiken MC, Loschi M, Aguilar EG, Furlan SN, Brown CC, Lee YC, Hyman CM, Feser CJ, Panoskaltsis-Mortari A, Hippen KL, MacDonald KP, Murphy WJ, Maillard I, Hill GR, Munn DH, Zeiser R, Kean LS, Rathmell JC, Chi H, Noelle RJ, and Blazar BR
- Subjects
- Animals, Autoimmunity, Immune Tolerance, Mice, Signal Transduction, T-Lymphocytes, Regulatory, Tretinoin pharmacology
- Abstract
Regulatory T cells (Tregs) promote immune homeostasis by maintaining self-tolerance and regulating inflammatory responses. Under certain inflammatory conditions, Tregs can lose their lineage stability and function. Previous studies have reported that ex vivo exposure to retinoic acid (RA) enhances Treg function and stability. However, it is unknown how RA receptor signaling in Tregs influences these processes in vivo. Herein, we employed mouse models in which RA signaling is silenced by the expression of the dominant negative receptor (DN) RARα in all T cells. Despite the fact that DNRARα conventional T cells are hypofunctional, Tregs had increased CD25 expression, STAT5 pathway activation, mTORC1 signaling and supersuppressor function. Furthermore, DNRARα Tregs had increased inhibitory molecule expression, amino acid transporter expression, and metabolic fitness and decreased antiapoptotic proteins. Supersuppressor function was observed when wild-type mice were treated with a pharmacologic pan-RAR antagonist. Unexpectedly, Treg-specific expression of DNRARα resulted in distinct phenotypes, such that a single allele of DNRARα in Tregs heightened their suppressive function, and biallelic expression led to loss of suppression and autoimmunity. The loss of Treg function was not cell intrinsic, as Tregs that developed in a noninflammatory milieu in chimeric mice reconstituted with DNRARα and wild-type bone marrow maintained the enhanced suppressive capacity. Fate mapping suggested that maintaining Treg stability in an inflammatory milieu requires RA signaling. Our findings indicate that RA signaling acts as a rheostat to balance Treg function in inflammatory and noninflammatory conditions in a dose-dependent manner., (© 2022. The Author(s), under exclusive licence to CSI and USTC.)
- Published
- 2022
- Full Text
- View/download PDF
208. Case Report: The First Case Report of Visceral Leishmaniasis in Cambodia.
- Author
-
Lyvannak S, Sreynich K, Heng S, Thyl M, Chandna A, Chanpheaktra N, Pises N, Farrilend P, Jarzembowski J, Leventaki V, Davick J, Neunert C, Keller F, Kean LS, Camitta B, Tarlock K, and Watkins B
- Subjects
- Child, Humans, Cambodia, Spleen, Fever complications, Leishmaniasis, Visceral diagnosis, Leishmaniasis, Visceral drug therapy, Leishmaniasis, Visceral etiology, Leishmania, Leishmaniasis complications
- Abstract
Leishmaniasis is considered a neglected tropical disease that is commonly found in Asia, Africa, South America, and Mediterranean countries. Visceral leishmaniasis (VL) is the most severe form of the disease and is almost universally fatal if left untreated. The symptoms of VL overlap with many infectious diseases, malignancies, and other blood disorders. The most common findings include fever, cytopenias, and splenomegaly. Given the nonspecific symptoms, the diagnosis requires detailed laboratory investigations, including bone marrow examination, that can be challenging in low- and middle-income countries. Diagnostic limitations likely lead to the underdiagnosis or delay in diagnosis of VL. We describe, to our knowledge, the first case report of VL in Cambodia in a child presenting with fever, anemia, and thrombocytopenia. The diagnosis required a liver biopsy and multiple bone marrow biopsies to visualize intracellular Leishmania spp. Our case illustrates the diagnostic challenges and the importance of timely diagnosis. This case also highlights the need for heightened awareness of the diagnostic findings of VL and improved reporting of tropical diseases.
- Published
- 2022
- Full Text
- View/download PDF
209. BET-bromodomain and EZH2 inhibitor-treated chronic GVHD mice have blunted germinal centers with distinct transcriptomes.
- Author
-
Zaiken MC, Flynn R, Paz KG, Rhee SY, Jin S, Mohamed FA, Saha A, Thangavelu G, Park PMC, Hemming ML, Sage PT, Sharpe AH, DuPage M, Bluestone JA, Panoskaltsis-Mortari A, Cutler CS, Koreth J, Antin JH, Soiffer RJ, Ritz J, Luznik L, Maillard I, Hill GR, MacDonald KPA, Munn DH, Serody JS, Murphy WJ, Kean LS, Zhang Y, Bradner JE, Qi J, and Blazar BR
- Subjects
- Animals, B-Lymphocytes drug effects, B-Lymphocytes metabolism, B-Lymphocytes pathology, Chronic Disease, Enzyme Inhibitors pharmacology, Humans, Mice, Transcriptome, Bronchiolitis Obliterans genetics, Bronchiolitis Obliterans metabolism, Bronchiolitis Obliterans pathology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Germinal Center drug effects, Germinal Center pathology, Graft vs Host Disease drug therapy, Graft vs Host Disease genetics, Graft vs Host Disease pathology, Proteins metabolism
- Abstract
Despite advances in the field, chronic graft-versus-host-disease (cGVHD) remains a leading cause of morbidity and mortality following allogenic hematopoietic stem cell transplant. Because treatment options remain limited, we tested efficacy of anticancer, chromatin-modifying enzyme inhibitors in a clinically relevant murine model of cGVHD with bronchiolitis obliterans (BO). We observed that the novel enhancer of zeste homolog 2 (EZH2) inhibitor JQ5 and the BET-bromodomain inhibitor JQ1 each improved pulmonary function; impaired the germinal center (GC) reaction, a prerequisite in cGVHD/BO pathogenesis; and JQ5 reduced EZH2-mediated H3K27me3 in donor T cells. Using conditional EZH2 knockout donor cells, we demonstrated that EZH2 is obligatory for the initiation of cGVHD/BO. In a sclerodermatous cGVHD model, JQ5 reduced the severity of cutaneous lesions. To determine how the 2 drugs could lead to the same physiological improvements while targeting unique epigenetic processes, we analyzed the transcriptomes of splenic GCB cells (GCBs) from transplanted mice treated with either drug. Multiple inflammatory and signaling pathways enriched in cGVHD/BO GCBs were reduced by each drug. GCBs from JQ5- but not JQ1-treated mice were enriched for proproliferative pathways also seen in GCBs from bone marrow-only transplanted mice, likely reflecting their underlying biology in the unperturbed state. In conjunction with in vivo data, these insights led us to conclude that epigenetic targeting of the GC is a viable clinical approach for the treatment of cGVHD, and that the EZH2 inhibitor JQ5 and the BET-bromodomain inhibitor JQ1 demonstrated clinical potential for EZH2i and BETi in patients with cGVHD/BO., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
210. Standardizing Definitions of Hematopoietic Recovery, Graft Rejection, Graft Failure, Poor Graft Function, and Donor Chimerism in Allogeneic Hematopoietic Cell Transplantation: A Report on Behalf of the American Society for Transplantation and Cellular Therapy.
- Author
-
Kharfan-Dabaja MA, Kumar A, Ayala E, Aljurf M, Nishihori T, Marsh R, Burroughs LM, Majhail N, Al-Homsi AS, Al-Kadhimi ZS, Bar M, Bertaina A, Boelens JJ, Champlin R, Chaudhury S, DeFilipp Z, Dholaria B, El-Jawahri A, Fanning S, Fraint E, Gergis U, Giralt S, Hamilton BK, Hashmi SK, Horn B, Inamoto Y, Jacobsohn DA, Jain T, Johnston L, Kanate AS, Kansagra A, Kassim A, Kean LS, Kitko CL, Knight-Perry J, Kurtzberg J, Liu H, MacMillan ML, Mahmoudjafari Z, Mielcarek M, Mohty M, Nagler A, Nemecek E, Olson TS, Oran B, Perales MA, Prockop SE, Pulsipher MA, Pusic I, Riches ML, Rodriguez C, Romee R, Rondon G, Saad A, Shah N, Shaw PJ, Shenoy S, Sierra J, Talano J, Verneris MR, Veys P, Wagner JE, Savani BN, Hamadani M, and Carpenter PA
- Subjects
- Adult, Child, Graft Rejection prevention & control, Humans, Transplantation Conditioning, Transplantation, Homologous, United States, Chimerism, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is potentially curative for certain hematologic malignancies and nonmalignant diseases. The field of allo-HCT has witnessed significant advances, including broadening indications for transplantation, availability of alternative donor sources, less toxic preparative regimens, new cell manipulation techniques, and novel GVHD prevention methods, all of which have expanded the applicability of the procedure. These advances have led to clinical practice conundrums when applying traditional definitions of hematopoietic recovery, graft rejection, graft failure, poor graft function, and donor chimerism, because these may vary based on donor type, cell source, cell dose, primary disease, graft-versus-host disease (GVHD) prophylaxis, and conditioning intensity, among other variables. To address these contemporary challenges, we surveyed a panel of allo-HCT experts in an attempt to standardize these definitions. We analyzed survey responses from adult and pediatric transplantation physicians separately. Consensus was achieved for definitions of neutrophil and platelet recovery, graft rejection, graft failure, poor graft function, and donor chimerism, but not for delayed engraftment. Here we highlight the complexities associated with the management of mixed donor chimerism in malignant and nonmalignant hematologic diseases, which remains an area for future research. We recognize that there are multiple other specific, and at times complex, clinical scenarios for which clinical management must be individualized., (Copyright © 2021 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
211. Insights from integrating clinical and preclinical studies advance understanding of graft-versus-host disease.
- Author
-
Socié G, Kean LS, Zeiser R, and Blazar BR
- Subjects
- Acute Disease, Animals, Chronic Disease, Humans, Transplantation, Homologous, Graft vs Host Disease immunology, Graft vs Host Disease pathology, Graft vs Host Disease therapy, Hematopoietic Stem Cell Transplantation, Translational Research, Biomedical
- Abstract
As a result of impressive increases in our knowledge of rodent and human immunology, the understanding of the pathophysiologic mechanisms underlying graft-versus-host disease (GVHD) has dramatically improved in the past 15 years. Despite improved knowledge, translation to clinical care has not proceeded rapidly, and results from experimental models have been inconsistent in their ability to predict the clinical utility of new therapeutic agents. In parallel, new tools in immunology have allowed in-depth analyses of the human system and have recently been applied in the field of clinical GVHD. Notwithstanding these advances, there is a relative paucity of mechanistic insights into human translational research, and this remains an area of high unmet need. Here we review selected recent advances in both preclinical experimental transplantation and translational human studies, including new insights into human immunology, the microbiome, and regenerative medicine. We focus on the fact that both approaches can interactively improve our understanding of both acute and chronic GVHD biology and open the door to improved therapeutics and successes.
- Published
- 2021
- Full Text
- View/download PDF
212. Repurposing a novel anti-cancer RXR agonist to attenuate murine acute GVHD and maintain graft-versus-leukemia responses.
- Author
-
Thangavelu G, Wang C, Loschi M, Saha A, Osborn MJ, Furlan SN, Aoyama K, McDonald-Hyman C, Aguilar EG, Janesick AS, Chandraratna RA, Refaeli Y, Panoskaltsis-Mortari A, MacDonald KP, Hill GR, Zeiser R, Maillard I, Serody JS, Murphy WJ, Munn DH, Blumberg B, Brown C, Kuchroo V, Kean LS, Hippen KL, Noelle RJ, and Blazar BR
- Subjects
- Animals, Drug Repositioning, Female, Graft vs Host Disease pathology, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory pathology, Bone Marrow Transplantation adverse effects, Cyclopropanes therapeutic use, Graft vs Host Disease drug therapy, Graft vs Leukemia Effect drug effects, Retinoid X Receptors agonists
- Abstract
The nuclear receptor (NR) subclass, retinoid X receptors (RXRs), exert immunomodulatory functions that control inflammation and metabolism via homodimers and heterodimers, with several other NRs, including retinoic acid receptors. IRX4204 is a novel, highly specific RXR agonist in clinical trials that potently and selectively activates RXR homodimers, but not heterodimers. In this study, in vivo IRX4204 compared favorably with FK506 in abrogating acute graft-versus-host disease (GVHD), which was associated with inhibiting allogeneic donor T-cell proliferation, reducing T-helper 1 differentiation, and promoting regulatory T-cell (Treg) generation. Recipient IRX4204 treatment reduced intestinal injury and decreased IFN-γ and TNF-α serum levels. Transcriptional analysis of donor T cells isolated from intestines of GVHD mice treated with IRX4204 revealed significant decreases in transcripts regulating proinflammatory pathways. In vitro, inducible Treg differentiation from naive CD4+ T cells was enhanced by IRX4204. In vivo, IRX4204 increased the conversion of donor Foxp3- T cells into peripheral Foxp3+ Tregs in GVHD mice. Using Foxp3 lineage-tracer mice in which both the origin and current FoxP3 expression of Tregs can be tracked, we demonstrated that IRX4204 supports Treg stability. Despite favoring Tregs and reducing Th1 differentiation, IRX4204-treated recipients maintained graft-versus-leukemia responses against both leukemia and lymphoma cells. Notably, IRX4204 reduced in vitro human T-cell proliferation and enhanced Treg generation in mixed lymphocyte reaction cultures. Collectively, these beneficial effects indicate that targeting RXRs with IRX4204 could be a novel approach to preventing acute GVHD in the clinic., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
213. Robust expansion of HIV CAR T cells following antigen boosting in ART-suppressed nonhuman primates.
- Author
-
Rust BJ, Kean LS, Colonna L, Brandenstein KE, Poole NH, Obenza W, Enstrom MR, Maldini CR, Ellis GI, Fennessey CM, Huang ML, Keele BF, Jerome KR, Riley JL, Kiem HP, and Peterson CW
- Subjects
- Animals, Antiretroviral Therapy, Highly Active adverse effects, Antiretroviral Therapy, Highly Active methods, Disease Models, Animal, HIV Infections drug therapy, HIV Infections virology, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Proteins genetics, Immune Checkpoint Proteins metabolism, Macaca mulatta, Simian Immunodeficiency Virus immunology, T-Lymphocytes drug effects, Antigens, Viral immunology, HIV Infections immunology, HIV-1 immunology, Immunocompromised Host, Receptors, Antigen, T-Cell immunology, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19+ hematologic malignancies have rapidly emerged as a promising, novel therapy. In contrast, results from the few CAR T-cell studies for infectious diseases such as HIV-1 have been less convincing. These challenges are likely due to the low level of antigen present in antiretroviral therapy (ART)-suppressed patients in contrast to those with hematologic malignancies. Using our well-established nonhuman primate model of ART-suppressed HIV-1 infection, we tested strategies to overcome these limitations and challenges. We first optimized CAR T-cell production to maintain central memory subsets, consistent with current clinical paradigms. We hypothesized that additional exogenous antigen might be required in an ART-suppressed setting to aid expansion and persistence of CAR T cells. Thus, we studied 4 simian/HIV-infected, ART-suppressed rhesus macaques infused with virus-specific CD4CAR T cells, followed by supplemental infusion of cell-associated HIV-1 envelope (Env). Env boosting led to significant and unprecedented expansion of virus-specific CAR+ T cells in vivo; after ART treatment interruption, viral rebound was significantly delayed compared with controls (P = .014). In 2 animals with declining CAR T cells, rhesusized anti-programmed cell death protein 1 (PD-1) antibody was administered to reverse PD-1-dependent immune exhaustion. Immune checkpoint blockade triggered expansion of exhausted CAR T cells and concordantly lowered viral loads to undetectable levels. These results show that supplemental cell-associated antigen enables robust expansion of CAR T cells in an antigen-sparse environment. To our knowledge, this is the first study to show expansion of virus-specific CAR T cells in infected, suppressed hosts, and delay/control of viral recrudescence., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
214. Isolation of a Highly Purified HSC-enriched CD34 + CD90 + CD45RA - Cell Subset for Allogeneic Transplantation in the Nonhuman Primate Large-animal Model.
- Author
-
Radtke S, Colonna L, Perez AM, Hoffman M, Kean LS, and Kiem HP
- Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HCT) is a common treatment for patients suffering from different hematological disorders. Allo-HCT in combination with hematopoietic stem cell (HSC) gene therapy is considered a promising treatment option for millions of patients with HIV+ and acute myeloid leukemia. Most currently available HSC gene therapy approaches target CD34-enriched cell fractions, a heterogeneous mix of mostly progenitor cells and only very few HSCs with long-term multilineage engraftment potential. As a consequence, gene therapy approaches are currently limited in their HSC targeting efficiency, very expensive consuming huge quantities of modifying reagents, and can lead to unwanted side effects in nontarget cells. We have previously shown that purified CD34
+ CD90+ CD45RA- cells are enriched for multipotent HSCs with long-term multilineage engraftment potential, which can reconstitute the entire hematopoietic system in an autologous nonhuman primate transplant model. Here, we tested the feasibility of transplantation with purified CD34+ CD90+ CD45RA- cells in the allogeneic setting in a nonhuman primate model., Methods: To evaluate the feasibility of this approach, CD34+ CD90+ CD45RA- cells from 2 fully major histocompatibility complex-matched, full sibling rhesus macaques were sort-purified, quality controlled, and transplanted. Engraftment and donor chimerism were evaluated in the peripheral blood and bone marrow of both animals., Results: Despite limited survival due to infectious complications, we show that the large-scale sort-purification and transplantation of CD34+ CD90+ CD45RA- cells is technically feasible and leads to rapid engraftment of cells in bone marrow in the allogeneic setting and absence of cotransferred T cells., Conclusions: We show that purification of an HSC-enriched CD34+ subset can serve as a potential stem cell source for allo-HCTs. Most importantly, the combination of allo-HCT and HSC gene therapy has the potential to treat a wide array of hematologic and nonhematologic disorders., Competing Interests: S.R. is a consultant to Forty Seven Inc (Gilead Sciences). H.-P.K. is a consultant to and has ownership interests with Rocket Pharma and Homology Medicines. H.-P.K. is a consultant to CSL Behring and Magenta Therapeutics. S.R. and H.-P.K. are inventors on patent applications (nos. 62/351 761, 62/428 994, and PCT/US2017/037967) submitted by the Fred Hutchinson Cancer Research Center that covers the selection and use of cell populations for research and therapeutic purposes as well as strategies to assess and/or produce cell populations with predictive engraftment potential. The other authors declare no conflicts of interest., (Copyright © 2020 The Author(s). Transplantation Direct. Published by Wolters Kluwer Health, Inc.)- Published
- 2020
- Full Text
- View/download PDF
215. Risk factors for transplant-associated thrombotic microangiopathy and mortality in a pediatric cohort.
- Author
-
Schoettler M, Lehmann LE, Margossian S, Lee M, Kean LS, Kao PC, Ma C, and Duncan CN
- Subjects
- Child, Female, Humans, Male, Retrospective Studies, Risk Factors, Graft vs Host Disease etiology, Hematopoietic Stem Cell Transplantation adverse effects, Thrombotic Microangiopathies diagnosis, Thrombotic Microangiopathies epidemiology, Thrombotic Microangiopathies etiology
- Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA) is a well-recognized complication of hematopoietic cell transplantation (HCT). Diagnosis is challenging and in the absence of a tissue biopsy, TA-TMA is provisionally diagnosed by meeting clinical criteria. In this study, we describe the prevalence, outcomes, and risk factors for meeting 2 different diagnostic criteria for TA-TMA and for increased transplant-related mortality (TRM). In this retrospective study of 307 pediatric HCT patients, records were reviewed for the first 100 days after HCT. Patients who were diagnosed with TA-TMA by a provider during this time were included. In addition, the Cho et al criteria (2010) and Jodele et al (2014) TA-TMA criteria were applied retrospectively. Eight patients (2.6%) were diagnosed with TA-TMA by their provider. However, on retrospective review, 20% and 36% met the Cho and Jodele criteria for TA-TMA, respectively. Overall survival was significantly worse (P < .0001) and TRM was significantly higher in patients who met criteria for TA-TMA (MC-TA-TMA) (P < .0001). After controlling for comorbid conditions, MC-TA-TMA (hazard ratio [HR], 10.9; P = .0001) and grade 3/4 acute graft-versus-host-disease (aGVHD) (HR 3.5; P = .01) remained independently associated with increased TRM. Among allogeneic HCT recipients, features associated with an increased risk for MC-TA-TMA included ≥2 HCT, concurrent grade 3/4 aGVHD and concurrent infections. Among patients who MC-TA-TMA, LDH ≥2 times the upper limit of normal (P = .001), the need for ≥2 antihypertensive medications (P < .0001), and acute kidney injury (P = .003) were associated with significantly increased TRM., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
216. IL-2 enhances ex vivo-expanded regulatory T-cell persistence after adoptive transfer.
- Author
-
Furlan SN, Singh K, Lopez C, Tkachev V, Hunt DJ, Hibbard J, Betz KM, Blazar BR, Trapnell C, and Kean LS
- Subjects
- Adoptive Transfer, Animals, Interleukin-2 Receptor alpha Subunit, Sirolimus pharmacology, Interleukin-2, T-Lymphocytes, Regulatory
- Abstract
As regulatory T cell (Treg) adoptive therapy continues to develop clinically, there is a need to determine which immunomodulatory agents pair most compatibly with Tregs to enable persistence and stabilize suppressor function. Prior work has shown that mechanistic target of rapamycin inhibition can increase the stability of thymic Tregs. In this study, we investigated the transcriptomic signatures of ex vivo-expanded Tregs after adoptive transfer in the setting of clinically relevant immunosuppression using a nonhuman primate (NHP) model as a prelude to future transplant studies. Here, we found that adding interleukin-2 (IL-2) to rapamycin in vivo supported a logarithmic increase in the half-life of adoptively transferred carboxyfluorescein diacetate succinimidyl ester-labeled, autologous NHP Tregs, effectively doubling the number of cells in the peripheral blood Treg compartment compared with Treg infusion when rapamycin was given alone. Using single-cell transcriptomics, we found that transferred ex vivo-expanded Tregs initially exhibit a gene expression signature consistent with an activated state. Moreover, those cells with the highest levels of activation also expressed genes associated with p53-mediated apoptosis. In contrast, transferred Tregs interrogated at day +20 posttransfer demonstrated a gene signature more similar to published profiles of resting Tregs. Together, these preclinical data further support combining IL-2 and rapamycin in vivo as adjunctive therapy for ex vivo-expanded adoptively transferred Tregs and suggest that the activation status of ex vivo-expanded Tregs is critical to their persistence., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
217. Busulfan Combined with Immunosuppression Allows Efficient Engraftment of Gene-Modified Cells in a Rhesus Macaque Model.
- Author
-
Uchida N, Nassehi T, Drysdale CM, Gamer J, Yapundich M, Bonifacino AC, Krouse AE, Linde N, Hsieh MM, Donahue RE, Dunbar CE, Kean LS, and Tisdale JF
- Subjects
- Animals, Gene Expression, Genes, Reporter, Genetic Therapy methods, Genetic Vectors genetics, Macaca mulatta, Models, Animal, Transduction, Genetic, gamma-Globins genetics, Busulfan pharmacology, Hematopoietic Stem Cell Transplantation methods, Hematopoietic Stem Cells metabolism, Immunosuppressive Agents pharmacology, Transgenes, Transplantation Conditioning
- Abstract
Busulfan conditioning is utilized for hematopoietic stem cell (HSC) depletion in the context of HSC gene-therapy conditioning but may result in insufficient immunosuppression. In this study, we evaluated whether additional immunosuppression is required for efficient engraftment of gene-modified cells using a rhesus HSC lentiviral gene-therapy model. We transduced half of rhesus CD34
+ cells with an enhanced green fluorescent protein (GFP)-encoding vector (immunogenic) and the other half with a γ-globin-encoding vector (no predicted immunogenicity). After autologous transplantation of both transduced cell populations following myeloablative busulfan conditioning (5.5 mg/kg/day for 4 days), we observed immunological rejection of GFP-transduced cells up to 3 months post-transplant and stable engraftment of γ-globin-transduced cells in two animals, demonstrating that ablative busulfan conditioning is sufficient for engraftment of gene-modified cells producing non-immunogenic proteins but insufficient to permit engraftment of immunogenic proteins. We then added immunosuppression with abatacept and sirolimus to busulfan conditioning and observed engraftment of both GFP- and γ-globin-transduced cells in two animals, demonstrating that additional immunosuppression allows for engraftment of gene-modified cells expressing immunogenic proteins. In conclusion, myeloablative busulfan conditioning should permit engraftment of gene-modified cells producing non-immunogenic proteins, while additional immunosuppression is required to prevent immunological rejection of a neoantigen., (Published by Elsevier Inc.)- Published
- 2019
- Full Text
- View/download PDF
218. Evidence for persistence of the SHIV reservoir early after MHC haploidentical hematopoietic stem cell transplantation.
- Author
-
Colonna L, Peterson CW, Schell JB, Carlson JM, Tkachev V, Brown M, Yu A, Reddy S, Obenza WM, Nelson V, Polacino PS, Mack H, Hu SL, Zeleski K, Hoffman M, Olvera J, Furlan SN, Zheng H, Taraseviciute A, Hunt DJ, Betz K, Lane JF, Vogel K, Hotchkiss CE, Moats C, Baldessari A, Murnane RD, English C, Astley CA, Wangari S, Agricola B, Ahrens J, Iwayama N, May A, Stensland L, Huang MW, Jerome KR, Kiem HP, and Kean LS
- Subjects
- Animals, Antiretroviral Therapy, Highly Active, CD8-Positive T-Lymphocytes immunology, DNA, Viral metabolism, Macaca mulatta, RNA, Viral metabolism, Simian Acquired Immunodeficiency Syndrome drug therapy, Simian Acquired Immunodeficiency Syndrome immunology, Simian Acquired Immunodeficiency Syndrome virology, Transplantation, Homologous, Disease Reservoirs virology, Hematopoietic Stem Cell Transplantation, Major Histocompatibility Complex, Simian Immunodeficiency Virus physiology, Transplantation, Haploidentical
- Abstract
Allogeneic transplantation (allo-HCT) has led to the cure of HIV in one individual, raising the question of whether transplantation can eradicate the HIV reservoir. To test this, we here present a model of allo-HCT in SHIV-infected, cART-suppressed nonhuman primates. We infect rhesus macaques with SHIV-1157ipd3N4, suppress them with cART, then transplant them using MHC-haploidentical allogeneic donors during continuous cART. Transplant results in ~100% myeloid donor chimerism, and up to 100% T-cell chimerism. Between 9 and 47 days post-transplant, terminal analysis shows that while cell-associated SHIV DNA levels are reduced in the blood and in lymphoid organs post-transplant, the SHIV reservoir persists in multiple organs, including the brain. Sorting of donor-vs.-recipient cells reveals that this reservoir resides in recipient cells. Moreover, tetramer analysis indicates a lack of virus-specific donor immunity post-transplant during continuous cART. These results suggest that early post-transplant, allo-HCT is insufficient for recipient reservoir eradication despite high-level donor chimerism and GVHD.
- Published
- 2018
- Full Text
- View/download PDF
219. CD28 blockade controls T cell activation to prevent graft-versus-host disease in primates.
- Author
-
Watkins BK, Tkachev V, Furlan SN, Hunt DJ, Betz K, Yu A, Brown M, Poirier N, Zheng HB, Taraseviciute A, Colonna L, Mary C, Blancho G, Soulillou JP, Panoskaltsis-Mortari A, Sharma P, Garcia A, Strobert E, Hamby K, Garrett A, Deane T, Blazar BR, Vanhove B, and Kean LS
- Subjects
- Abatacept administration & dosage, Animals, Antibodies, Monoclonal administration & dosage, Disease Models, Animal, Graft vs Host Disease immunology, Hematopoietic Stem Cell Transplantation adverse effects, Lymphocyte Activation, Macaca mulatta, Sirolimus administration & dosage, Systems Biology, CD28 Antigens antagonists & inhibitors, Graft vs Host Disease prevention & control, T-Lymphocytes immunology
- Abstract
Controlling graft-versus-host disease (GVHD) remains a major unmet need in stem cell transplantation, and new, targeted therapies are being actively developed. CD28-CD80/86 costimulation blockade represents a promising strategy, but targeting CD80/CD86 with CTLA4-Ig may be associated with undesired blockade of coinhibitory pathways. In contrast, targeted blockade of CD28 exclusively inhibits T cell costimulation and may more potently prevent GVHD. Here, we investigated FR104, an antagonistic CD28-specific pegylated-Fab', in the nonhuman primate (NHP) GVHD model and completed a multiparameter interrogation comparing it with CTLA4-Ig, with and without sirolimus, including clinical, histopathologic, flow cytometric, and transcriptomic analyses. We document that FR104 monoprophylaxis and combined prophylaxis with FR104/sirolimus led to enhanced control of effector T cell proliferation and activation compared with the use of CTLA4-Ig or CTLA4-Ig/sirolimus. Importantly, FR104/sirolimus did not lead to a beneficial impact on Treg reconstitution or homeostasis, consistent with control of conventional T cell activation and IL-2 production needed to support Tregs. While FR104/sirolimus had a salutary effect on GVHD-free survival, overall survival was not improved, due to death in the absence of GVHD in several FR104/sirolimus recipients in the setting of sepsis and a paralyzed INF-γ response. These results therefore suggest that effectively deploying CD28 in the clinic will require close scrutiny of both the benefits and risks of extensively abrogating conventional T cell activation after transplant.
- Published
- 2018
- Full Text
- View/download PDF
220. Defining success with cellular therapeutics: the current landscape for clinical end point and toxicity analysis.
- Author
-
Kean LS
- Subjects
- Animals, Hematologic Diseases immunology, Hematologic Diseases pathology, Hematologic Neoplasms immunology, Hematologic Neoplasms pathology, Hematopoietic Stem Cell Transplantation adverse effects, Humans, Immunotherapy, Adoptive adverse effects, T-Lymphocytes immunology, T-Lymphocytes transplantation, Hematologic Diseases therapy, Hematologic Neoplasms therapy, Hematopoietic Stem Cell Transplantation methods, Immunotherapy, Adoptive methods
- Abstract
Cellular therapies play a major and expanding role in the treatment of hematologic diseases. For each of these therapies, a narrow therapeutic window exists, where efficacy is maximized and toxicities minimized. This review focuses on one of the most established cellular therapies, hematopoietic stem cell transplant, and one of the newest cellular therapies, chimeric antigen receptor-T cells. In this review, I will discuss the current state of the field for clinical end point analysis with each of these therapeutics, including their critical toxicities, and focus on the major elements of success for each of these complex treatments for hematologic disease., (© 2018 by The American Society of Hematology.)
- Published
- 2018
- Full Text
- View/download PDF
221. Human CD19-Targeted Mouse T Cells Induce B Cell Aplasia and Toxicity in Human CD19 Transgenic Mice.
- Author
-
Pennell CA, Barnum JL, McDonald-Hyman CS, Panoskaltsis-Mortari A, Riddle MJ, Xiong Z, Loschi M, Thangavelu G, Campbell HM, Storlie MD, Refaeli Y, Furlan SN, Jensen MC, Kean LS, Miller JS, Tolar J, Osborn MJ, and Blazar BR
- Subjects
- Animals, Female, Humans, Immunotherapy, Adoptive, Interferon-gamma metabolism, Interleukin-6 metabolism, Male, Mice, Mice, Transgenic, Antigens, CD19 metabolism, B-Lymphocytes metabolism
- Abstract
The clinical success of chimeric antigen receptor (CAR) T cell therapy for CD19
+ B cell malignancies can be limited by acute toxicities and immunoglobulin replacement needs due to B cell aplasia from persistent CAR T cells. Life-threatening complications include cytokine release syndrome and neurologic adverse events, the exact etiologies of which are unclear. To elucidate the underlying toxicity mechanisms and test potentially safer CAR T cells, we developed a mouse model in which human CD19 (hCD19)-specific mouse CAR T cells were adoptively transferred into mice whose normal B cells express a hCD19 transgene at hemizygous levels. Compared to homozygous hCD19 transgenic mice that have ∼75% fewer circulating B cells, hemizygous mice had hCD19 frequencies and antigen density more closely simulating human B cells. Hemizygous mice given a lethal dose of hCD19 transgene-expressing lymphoma cells and treated with CAR T cells had undetectable tumor levels. Recipients experienced B cell aplasia and antigen- and dose-dependent acute toxicities mirroring patient complications. Interleukin-6 (IL-6), interferon γ (IFN-γ), and inflammatory pathway transcripts were enriched in affected tissues. As in patients, antibody-mediated neutralization of IL-6 (and IFN-γ) blunted toxicity. Apparent behavioral abnormalities associated with decreased microglial cells point to CAR-T-cell-induced neurotoxicity. This model will prove useful in testing strategies designed to improve hCD19-specific CAR T cell safety., (Copyright © 2018 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
222. Advances in targeting co-inhibitory and co-stimulatory pathways in transplantation settings: the Yin to the Yang of cancer immunotherapy.
- Author
-
Kean LS, Turka LA, and Blazar BR
- Subjects
- Animals, Costimulatory and Inhibitory T-Cell Receptors immunology, Humans, Immune Tolerance, Immunomodulation, Lymphocyte Activation, Neoplasms immunology, Receptors, Antigen, T-Cell genetics, Recombinant Fusion Proteins genetics, Signal Transduction, Antibodies, Monoclonal therapeutic use, Costimulatory and Inhibitory T-Cell Receptors metabolism, Graft Rejection prevention & control, Immunotherapy methods, Neoplasms therapy, Organ Transplantation, T-Lymphocytes physiology
- Abstract
In the past decade, the power of harnessing T-cell co-signaling pathways has become increasingly understood to have significant clinical importance. In cancer immunotherapy, the field has concentrated on two related modalities: First, targeting cancer antigens through highly activated chimeric antigen T cells (CAR-Ts) and second, re-animating endogenous quiescent T cells through checkpoint blockade. In each of these strategies, the therapeutic goal is to re-ignite T-cell immunity, in order to eradicate tumors. In transplantation, there is also great interest in targeting T-cell co-signaling, but with the opposite goal: in this field, we seek the Yin to cancer immunotherapy's Yang, and focus on manipulating T-cell co-signaling to induce tolerance rather than activation. In this review, we discuss the major T-cell signaling pathways that are being investigated for tolerance induction, detailing preclinical studies and the path to the clinic for many of these molecules. These include blockade of co-stimulation pathways and agonism of coinhibitory pathways, in order to achieve the delicate state of balance that is transplant tolerance: a state which guarantees lifelong transplant acceptance without ongoing immunosuppression, and with preservation of protective immune responses. In the context of the clinical translation of immune tolerance strategies, we discuss the significant challenge that is embodied by the fact that targeted pathway modulators may have opposing effects on tolerance based on their impact on effector vs regulatory T-cell biology. Achieving this delicate balance holds the key to the major challenge of transplantation: lifelong control of alloreactivity while maintaining an otherwise intact immune system., (© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
223. In vivo T cell costimulation blockade with abatacept for acute graft-versus-host disease prevention: a first-in-disease trial.
- Author
-
Koura DT, Horan JT, Langston AA, Qayed M, Mehta A, Khoury HJ, Harvey RD, Suessmuth Y, Couture C, Carr J, Grizzle A, Johnson HR, Cheeseman JA, Conger JA, Robertson J, Stempora L, Johnson BE, Garrett A, Kirk AD, Larsen CP, Waller EK, and Kean LS
- Subjects
- Abatacept, Acute Disease, Adolescent, Adult, Aged, Feasibility Studies, Female, Humans, Male, Middle Aged, Transplantation, Homologous, Young Adult, Graft vs Host Disease prevention & control, Hematopoietic Stem Cell Transplantation methods, Immunoconjugates therapeutic use, Immunosuppressive Agents therapeutic use, T-Lymphocytes immunology, Transplantation Conditioning methods
- Abstract
We performed a first-in-disease trial of in vivo CD28:CD80/86 costimulation blockade with abatacept for acute graft-versus-host disease (aGVHD) prevention during unrelated-donor hematopoietic cell transplantation (HCT). All patients received cyclosporine/methotrexate plus 4 doses of abatacept (10 mg/kg/dose) on days -1, +5, +14, +28 post-HCT. The feasibility of adding abatacept, its pharmacokinetics, pharmacodynamics, and its impact on aGVHD, infection, relapse, and transplantation-related mortality (TRM) were assessed. All patients received the planned abatacept doses, and no infusion reactions were noted. Compared with a cohort of patients not receiving abatacept (the StdRx cohort), patients enrolled in the study (the ABA cohort) demonstrated significant inhibition of early CD4(+) T cell proliferation and activation, affecting predominantly the effector memory (Tem) subpopulation, with 7- and 10-fold fewer proliferating and activated CD4(+) Tem cells, respectively, at day+28 in the ABA cohort compared with the StdRx cohort (P < .01). The ABA patients demonstrated a low rate of aGVHD, despite robust immune reconstitution, with 2 of 10 patients diagnosed with grade II-IV aGVHD before day +100, no deaths from infection, no day +100 TRM, and with 7 of 10 evaluable patients surviving (median follow-up, 16 months). These results suggest that costimulation blockade with abatacept can significantly affect CD4(+) T cell proliferation and activation post-transplantation, and may be an important adjunct to standard immunoprophylaxis for aGVHD in patients undergoing unrelated-donor HCT., (Copyright © 2013 American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
224. Expansion of effector memory TCR Vbeta4+ CD8+ T cells is associated with latent infection-mediated resistance to transplantation tolerance.
- Author
-
Stapler D, Lee ED, Selvaraj SA, Evans AG, Kean LS, Speck SH, Larsen CP, and Gangappa S
- Subjects
- Animals, Bone Marrow Transplantation immunology, Bone Marrow Transplantation pathology, CD8-Positive T-Lymphocytes pathology, Cell Division genetics, Cell Division immunology, Graft Survival genetics, Graft Survival immunology, Herpesviridae Infections pathology, Herpesviridae Infections virology, Immunity, Innate genetics, Mice, Mice, Inbred BALB C, Mice, Inbred C3H, Mice, Inbred C57BL, Radiation Chimera immunology, Skin Transplantation immunology, Skin Transplantation pathology, Transplantation Tolerance genetics, Virus Latency genetics, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes virology, Genes, T-Cell Receptor beta, Herpesviridae Infections immunology, Immunologic Memory genetics, Rhadinovirus immunology, Transplantation Tolerance immunology, Virus Latency immunology
- Abstract
Therapies that control largely T cell-dependent allograft rejection in humans also possess the undesirable effect of impairing T cell function, leaving transplant recipients susceptible to opportunistic viruses. Prime among these opportunists are the ubiquitous herpesviruses. To date, studies are lacking that address the effect of viruses that establish a true latent state on allograft tolerance or the effect of tolerance protocols on the immune control of latent viruses. By using a mixed chimerism-based tolerance-induction protocol, we found that mice undergoing latent infection with gammaHV68, a murine gamma-herpesvirus closely related to human gamma-herpesviruses such as EBV and Kaposi's sarcoma-associated herpesvirus, significantly resist tolerance to allografts. Limiting the degree of virus reactivation or innate immune response did not reconstitute chimerism in latently infected mice. However, gammaHV68-infected mice showed increased frequency of CD8+ T cell alloreactivity and, interestingly, expansion of virus-induced, alloreactive, "effector/effector memory" TCR Vbeta4+CD8+ T cells driven by the gammaHV68-M1 gene was associated with resistance to tolerance induction in studies using gammaHV68-M1 mutant virus. These results define the viral gene and immune cell types involved in latent infection-mediated resistance to allograft tolerance and underscore the influence of latent herpesviruses on allograft survival.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.