Armin Ghobadi
Washington University in St. Louis
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Armin Ghobadi.
The New England Journal of Medicine | 2016
John S. Welch; Allegra A. Petti; Christopher A. Miller; Catrina C. Fronick; Michelle O’Laughlin; Robert S. Fulton; Richard Wilson; Jack Baty; Eric J. Duncavage; Bevan Tandon; Yi-Shan Lee; Lukas D. Wartman; Geoffrey L. Uy; Armin Ghobadi; Michael H. Tomasson; Iskra Pusic; Rizwan Romee; Todd A. Fehniger; Keith Stockerl-Goldstein; Ravi Vij; Stephen T. Oh; Camille N. Abboud; Amanda F. Cashen; Mark A. Schroeder; Meagan A. Jacoby; Sharon Heath; Kierstin Luber; M R Janke; Andrew Hantel; Niloufer Khan
BACKGROUND The molecular determinants of clinical responses to decitabine therapy in patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) are unclear. METHODS We enrolled 84 adult patients with AML or MDS in a single-institution trial of decitabine to identify somatic mutations and their relationships to clinical responses. Decitabine was administered at a dose of 20 mg per square meter of body-surface area per day for 10 consecutive days in monthly cycles. We performed enhanced exome or gene-panel sequencing in 67 of these patients and serial sequencing at multiple time points to evaluate patterns of mutation clearance in 54 patients. An extension cohort included 32 additional patients who received decitabine in different protocols. RESULTS Of the 116 patients, 53 (46%) had bone marrow blast clearance (<5% blasts). Response rates were higher among patients with an unfavorable-risk cytogenetic profile than among patients with an intermediate-risk or favorable-risk cytogenetic profile (29 of 43 patients [67%] vs. 24 of 71 patients [34%], P<0.001) and among patients with TP53 mutations than among patients with wild-type TP53 (21 of 21 [100%] vs. 32 of 78 [41%], P<0.001). Previous studies have consistently shown that patients with an unfavorable-risk cytogenetic profile and TP53 mutations who receive conventional chemotherapy have poor outcomes. However, in this study of 10-day courses of decitabine, neither of these risk factors was associated with a lower rate of overall survival than the rate of survival among study patients with intermediate-risk cytogenetic profiles. CONCLUSIONS Patients with AML and MDS who had cytogenetic abnormalities associated with unfavorable risk, TP53 mutations, or both had favorable clinical responses and robust (but incomplete) mutation clearance after receiving serial 10-day courses of decitabine. Although these responses were not durable, they resulted in rates of overall survival that were similar to those among patients with AML who had an intermediate-risk cytogenetic profile and who also received serial 10-day courses of decitabine. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT01687400 .).
Molecular Therapy | 2017
Frederick L. Locke; Sattva S. Neelapu; Nancy L. Bartlett; Tanya Siddiqi; Julio C. Chavez; Chitra Hosing; Armin Ghobadi; Lihua E. Budde; Adrian Bot; John M. Rossi; Yizhou Jiang; Allen Xue; Meg Elias; Jeff Aycock; Jeff Wiezorek; William Y. Go
Outcomes for patients with refractory diffuse large B cell lymphoma (DLBCL) are poor. In the multicenter ZUMA-1 phase 1 study, we evaluated KTE-C19, an autologous CD3ζ/CD28-based chimeric antigen receptor (CAR) T cell therapy, in patients with refractory DLBCL. Patients received low-dose conditioning chemotherapy with concurrent cyclophosphamide (500 mg/m2) and fludarabine (30 mg/m2) for 3 days followed by KTE-C19 at a target dose of 2 × 106 CAR T cells/kg. The incidence of dose-limiting toxicity (DLT) was the primary endpoint. Seven patients were treated with KTE-C19 and one patient experienced a DLT of grade 4 cytokine release syndrome (CRS) and neurotoxicity. Grade ≥3 CRS and neurotoxicity were observed in 14% (n = 1/7) and 57% (n = 4/7) of patients, respectively. All other KTE-C19-related grade ≥3 events resolved within 1 month. The overall response rate was 71% (n = 5/7) and complete response (CR) rate was 57% (n = 4/7). Three patients have ongoing CR (all at 12+ months). CAR T cells demonstrated peak expansion within 2 weeks and continued to be detectable at 12+ months in patients with ongoing CR. This regimen of KTE-C19 was safe for further study in phase 2 and induced durable remissions in patients with refractory DLBCL.
Leukemia | 2018
Matthew L. Cooper; Jaebok Choi; Karl Staser; Julie Ritchey; Jessica M Devenport; Kayla Eckardt; Michael P. Rettig; Bing Wang; Linda Eissenberg; Armin Ghobadi; Leah N Gehrs; Julie L. Prior; Samuel Achilefu; Christopher A. Miller; Catrina C. Fronick; Julie O’Neal; Feng Gao; David M. Weinstock; Alejandro Gutierrez; Robert S. Fulton; John F. DiPersio
T cell malignancies represent a group of hematologic cancers with high rates of relapse and mortality in patients for whom no effective targeted therapies exist. The shared expression of target antigens between chimeric antigen receptor (CAR) T cells and malignant T cells has limited the development of CAR-T because of unintended CAR-T fratricide and an inability to harvest sufficient autologous T cells. Here, we describe a fratricide-resistant “off-the-shelf” CAR-T (or UCART7) that targets CD7+ T cell malignancies and, through CRISPR/Cas9 gene editing, lacks both CD7 and T cell receptor alpha chain (TRAC) expression. UCART7 demonstrates efficacy against human T cell acute lymphoblastic leukemia (T-ALL) cell lines and primary T-ALL in vitro and in vivo without the induction of xenogeneic GvHD. Fratricide-resistant, allo-tolerant “off-the-shelf” CAR-T represents a strategy for treatment of relapsed and refractory T-ALL and non-Hodgkin’s T cell lymphoma without a requirement for autologous T cells.
Transplant Infectious Disease | 2012
J.S. Chawla; Armin Ghobadi; James C. Mosley; Linda Verkruyse; K. Trinkaus; Camille N. Abboud; Amanda F. Cashen; Keith Stockerl-Goldstein; Geoffrey L. Uy; Peter Westervelt; John F. DiPersio; Ravi Vij
Cytomegalovirus (CMV) infection is an important cause of morbidity and mortality after allogeneic hematopoietic stem cell transplant (HSCT). This pilot prospective randomized clinical trial compares valganciclovir (VGV) to ganciclovir (GCV) as pre‐emptive therapy for CMV viremia in the post‐allogeneic HSCT population.
Molecular Therapy | 2015
Linda Eissenberg; Michael P. Rettig; Julie Ritchey; Julie L. Prior; Sally W. Schwarz; Jennifer Frye; Brian S. White; Robert S. Fulton; Armin Ghobadi; Matthew L. Cooper; Daniel R. Couriel; Muhammad Esa Seegulam; David Piwnica-Worms; Farrokh Dehdashti; Kenneth Cornetta; John F. DiPersio
Described herein is a first-in-man attempt to both genetically modify T cells with an imagable suicide gene and track these transduced donor T cells in allogeneic stem cell transplantation recipients using noninvasive positron emission tomography/computerized tomography (PET/CT) imaging. A suicide gene encoding a human CD34-Herpes Simplex Virus-1-thymidine kinase (CD34-TK75) fusion enabled enrichment of retrovirally transduced T cells (TdT), control of graft-versus-host disease and imaging of TdT migration and expansion in vivo in mice and man. Analysis confirmed that CD34-TK75-enriched TdT contained no replication competent γ-retrovirus, were sensitive to ganciclovir, and displayed characteristic retroviral insertion sites (by targeted sequencing). Affinity-purified CD34-TK75+-selected donor T cells (1.0-13 × 105)/kg were infused into eight patients who relapsed after allogeneic stem cell transplantation. Six patients also were administered 9-[4-(18F)fluoro-3-hydroxymethyl-butyl]guanine ([18F]FHBG) to specifically track the genetically modified donor T cells by PET/CT at several time points after infusion. All patients were assessed for graft-versus-host disease, response to ganciclovir, circulating TdT cells (using both quantitative polymerase chain reaction and [18F]FHBG PET/CT imaging), TdT cell clonal expansion, and immune response to the TdT. This phase 1 trial demonstrated that genetically modified T cells and [18F]FHBG can be safely infused in patients with relapsed hematologic malignancies after allogeneic stem cell transplantation.Described herein is a first-in-man attempt to both genetically modify T cells with an imagable suicide gene and track these transduced donor T cells in allogeneic stem cell transplantation recipients using noninvasive positron emission tomography/computerized tomography (PET/CT) imaging. A suicide gene encoding a human CD34-Herpes Simplex Virus-1-thymidine kinase (CD34-TK75) fusion enabled enrichment of retrovirally transduced T cells (TdT), control of graft-versus-host disease and imaging of TdT migration and expansion in vivo in mice and man. Analysis confirmed that CD34-TK75-enriched TdT contained no replication competent γ-retrovirus, were sensitive to ganciclovir, and displayed characteristic retroviral insertion sites (by targeted sequencing). Affinity-purified CD34-TK75(+)-selected donor T cells (1.0-13 × 10(5))/kg were infused into eight patients who relapsed after allogeneic stem cell transplantation. Six patients also were administered 9-[4-((18)F)fluoro-3-hydroxymethyl-butyl]guanine ([(18)F]FHBG) to specifically track the genetically modified donor T cells by PET/CT at several time points after infusion. All patients were assessed for graft-versus-host disease, response to ganciclovir, circulating TdT cells (using both quantitative polymerase chain reaction and [(18)F]FHBG PET/CT imaging), TdT cell clonal expansion, and immune response to the TdT. This phase 1 trial demonstrated that genetically modified T cells and [(18)F]FHBG can be safely infused in patients with relapsed hematologic malignancies after allogeneic stem cell transplantation.
Transplant Infectious Disease | 2016
Kristine M. Wylie; M. Blanco-Guzman; Todd Wylie; Steven J. Lawrence; Armin Ghobadi; John F. DiPersio; Gregory A. Storch
A 40‐year‐old man with chronic myelogenous leukemia presented multiple times over a period of 3 years with episodes of confusion, wide‐based gait and falls because of recurrent hydrocephalus despite repeated therapeutic lumbar punctures. These problems occurred in the context of persistent cerebrospinal fluid (CSF) pleocytosis and leptomeningeal enhancement. Extensive diagnostic workups and therapeutic trials had failed to identify a clinically plausible cause or produce any significant improvement in the CSF and neuroimaging abnormalities.
Leukemia Research | 2016
Armin Ghobadi; Jaebok Choi; Mark Fiala; Theresa Fletcher; Jingxia Liu; Linda Eissenberg; Camille N. Abboud; Amanda F. Cashen; Ravi Vij; Mark A. Schroeder; Iskra Pusic; Keith Stockerl-Goldstein; Meagan A. Jacoby; Geoffrey L. Uy; John F. DiPersio; Peter Westervelt
Donor lymphocyte infusion (DLI) without prophylactic immunosuppression has been used for relapsed AML after allogeneic stem cell transplant (allo-SCT). However DLI is associated with an increased incidence of acute Graft vs. Host Disease (aGVHD). In mice, administration of azacitidine (AzaC) on days 4, 6, 8, and 10 post DLI increases regulatory T cell (Treg) numbers and prevents GVHD without hindering Graft vs. Leukemia (GVL). Based on these findings, we conducted a phase 1 study of AzaC post DLI for AML relapse post allo-SCT. AzaC was administered on days 4, 6, 8 and 10 post-DLI. Dose escalation was done using a 3+3 design with three AzaC dose levels: 30mg/m(2) (level -1), 45mg/m(2) (level 1) and 75mg/m(2) (level 2). Three patients were treated in the 45mg/m(2) dose level and 5 patients were treated in the 75mg/m(2) dose level; no DLTs or grade 3-5 treatment related toxicities were observed. After a median follow-up of 5.2 months, no patients developed grade III-IV aGVHD and no patients died of aGVHD. Six out of 8 patients in the treatment group responded to treatment including two cytogenetic complete remissions, one hematologic complete remission, and three complete remissions with incomplete count recovery. In conclusion, administration of AzaC early post DLI is well tolerated and can potentially prevent GVHD after DLI. Further studies are required to evaluate the effect of azacitidine early post DLI on GVHD and GVL.
Blood | 2014
Armin Ghobadi; Michael P. Rettig; Matthew L. Cooper; Matthew Holt; Julie Ritchey; Linda Eissenberg; John F. DiPersio
To the editor: Mobilized hematopoietic stem and progenitor cells (HSPCs) collected from peripheral blood (PB) is the most common source of HSPCs for stem cell transplantation, and granulocyte colony-stimulating factor (G-CSF) is the most common agent used for stem cell mobilization. Unfortunately,
Bone Marrow Transplantation | 2015
Armin Ghobadi; Eric Nolley; Jingxia Liu; Ali McBride; Keith Stockerl-Goldstein; Amanda F. Cashen
Retrospective comparison of allogeneic vs autologous transplantation for diffuse large B-cell lymphoma with early relapse or primary induction failure
Current Research in Translational Medicine | 2018
Armin Ghobadi
Non-Hodgkin Lymphoma (NHL) is the most common hematologic malignancy. More than 20,000 people in United States, more than 37,000 people in Europe and more than 199,000 people worldwide die of NHL every year. Recent advances in immunotherapeutic approaches for cancer have resulted in development of new classes of very effective immunotherapeutic approaches including chimeric antigen receptor T (CAR-T) cell therapy that are designed to bypass cancer immune evasion. Here, we review recent advances in CAR-T cell therapy for NHL. US food and drug administration (FDA) recently approved axicabtagene ciloleucel (Yescarta) a CD19 CAR T cell therapy for treatment of relapsed refractory diffuse large B cell lymphoma (DLBCL), high grade lymphoma, and primary mediastinal B cell lymphoma (PMBCL). Approval of Yescarta and rapid development of other CAR T cell therapies at various stages of development are opening up the door for a new wave of CAR T cell therapies that will dramatically change the way we treat NHL and hopefully other malignancies in the near future.