Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Frederick L. Locke is active.

Publication


Featured researches published by Frederick L. Locke.


Nature Reviews Clinical Oncology | 2017

Chimeric antigen receptor T-cell therapy — assessment and management of toxicities

Sattva S. Neelapu; Sudhakar Tummala; Partow Kebriaei; William G. Wierda; Cristina Gutierrez; Frederick L. Locke; Krishna V. Komanduri; Yi Lin; Nitin Jain; Naval Daver; Jason R. Westin; Alison Gulbis; Monica Elena Loghin; John F. de Groot; Sherry Adkins; Suzanne E. Davis; Katayoun Rezvani; Patrick Hwu; Elizabeth J. Shpall

Immunotherapy using T cells genetically engineered to express a chimeric antigen receptor (CAR) is rapidly emerging as a promising new treatment for haematological and non-haematological malignancies. CAR-T-cell therapy can induce rapid and durable clinical responses, but is associated with unique acute toxicities, which can be severe or even fatal. Cytokine-release syndrome (CRS), the most commonly observed toxicity, can range in severity from low-grade constitutional symptoms to a high-grade syndrome associated with life-threatening multiorgan dysfunction; rarely, severe CRS can evolve into fulminant haemophagocytic lymphohistiocytosis (HLH). Neurotoxicity, termed CAR-T-cell-related encephalopathy syndrome (CRES), is the second most-common adverse event, and can occur concurrently with or after CRS. Intensive monitoring and prompt management of toxicities is essential to minimize the morbidity and mortality associated with this potentially curative therapeutic approach; however, algorithms for accurate and consistent grading and management of the toxicities are lacking. To address this unmet need, we formed a CAR-T-cell-therapy-associated TOXicity (CARTOX) Working Group, comprising investigators from multiple institutions and medical disciplines who have experience in treating patients with various CAR-T-cell therapy products. Herein, we describe the multidisciplinary approach adopted at our institutions, and provide recommendations for monitoring, grading, and managing the acute toxicities that can occur in patients treated with CAR-T-cell therapy.


Molecular Therapy | 2017

Phase 1 Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma

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.


Journal of Experimental Medicine | 2012

Transcriptional regulator early growth response gene 2 (Egr2) is required for T cell anergy in vitro and in vivo

Yan Zheng; Yuanyuan Zha; Gregory Driessens; Frederick L. Locke; Thomas F. Gajewski

Deletion of early growth response gene Egr2 prevents anergy induction through diacylglycerol kinase α and restores Ras/MAPK signaling in T cells.


Bone Marrow Transplantation | 2013

Race/ethnicity affects the probability of finding an HLA-A, -B, -C and -DRB1 allele-matched unrelated donor and likelihood of subsequent transplant utilization

Joseph Pidala; Jongphil Kim; Michael J. Schell; S.J. Lee; R Hillgruber; V Nye; Ernesto Ayala; Melissa Alsina; Brian C. Betts; Ryan Bookout; Hugo F. Fernandez; Teresa Field; Frederick L. Locke; Taiga Nishihori; Jose L Ochoa; Lia Perez; Janelle Perkins; J. Shapiro; C. Tate; Marcie Tomblyn; Claudio Anasetti

Factors relevant to finding a suitable unrelated donor and barriers to effective transplant utilization are incompletely understood. Among a consecutive series of unrelated searches (n=531), an 8/8 HLA-A, -B, -C and -DRB1-matched unrelated donor was available for 289 (54%) patients, 7/8 for 159 (30%) and no donor for 83 (16%). Patients of Caucasian race (P<0.0001) were more likely to find a donor. Younger age (P=0.01), Caucasian race (P=0.03), lower CIBMTR (Center for International Blood and Marrow Transplantation Research) risk (P=0.005), and 8/8 HLA matching (P=0.005) were associated with higher odds of reaching hematopoietic cell transplantation (HCT). In a univariate analysis of OS, finding a donor was associated with hazard ratio (HR) of 0.85 (95% CI 0.63–1.2), P=0.31. Karnofsky performance status (KPS) accounted for interaction between having a donor and survival. Patients with KPS 90–100 and a donor had significantly reduced hazard for death (HR 0.59, 95% CI 0.38–0.90, P=0.02). These data provide estimates of the probability to find an unrelated donor in the era of high-resolution HLA typing, and identify potentially modifiable barriers to reaching HCT. Further efforts are needed to enhance effective donor identification and transplant utilization, particularly in non-Caucasian ethnic groups.


Journal of Immunology | 2011

β-Catenin Inhibits T Cell Activation by Selective Interference with Linker for Activation of T Cells–Phospholipase C-γ1 Phosphorylation

Gregory Driessens; Yan Zheng; Frederick L. Locke; Fotini Gounari; Thomas F. Gajewski

Despite the defined function of the β-catenin pathway in thymocytes, its functional role in peripheral T cells is poorly understood. We report that in a mouse model, β-catenin protein is constitutively degraded in peripheral T cells. Introduction of stabilized β-catenin into primary T cells inhibited proliferation and cytokine secretion after TCR stimulation and blunted effector cell differentiation. Functional and biochemical studies revealed that β-catenin selectively inhibited linker for activation of T cells phosphorylation on tyrosine 136, which was associated with defective phospholipase C-γ1 phosphorylation and calcium signaling but normal ERK activation. Our findings indicate that β-catenin negatively regulates T cell activation by a previously undescribed mechanism and suggest that conditions under which β-catenin might be inducibly stabilized in vivo would be inhibitory for T cell-based immunity.


Biology of Blood and Marrow Transplantation | 2011

ATG Prevents Severe Acute Graft-versus-Host Disease in Mismatched Unrelated Donor Hematopoietic Cell Transplantation

Joseph Pidala; Marcie Tomblyn; Taiga Nishihori; Ernesto Ayala; Teresa Field; Hugo F. Fernandez; Lia Perez; Frederick L. Locke; Melissa Alsina; Jose L Ochoa; Janelle Perkins; C. Tate; J. Shapiro; Michelle Conwell; Ryan Bookout; Claudio Anasetti

Severe acute graft-versus-host disease (aGVHD) remains a major source of morbidity and mortality following mismatched unrelated donor hematopoietic cell transplantation (HCT). Through a retrospective analysis, we investigated the efficacy of GVHD prophylaxis with rabbit anti-thymocyte globulin (ATG) 7.5 mg/kg (1 mg/kg given on day -3, then 3.25 mg/kg/day on days -2 and -1 before stem cell infusion) followed by standard tacrolimus plus methotrexate in a consecutive series of 45 HLA partially matched unrelated donor HCT recipients. The cumulative incidence of grade III-IV aGVHD was 11% by 100 days (95% confidence interval [CI] 5%-25%). Moderate to severe chronic GVHD (per NIH consensus criteria) was 19% (95% CI 10%-36%) at 1 year, and 28% (95% CI 16%-48%) at 2 years. With a median follow-up time for surviving patients of 12 months (range: 5-39 months), overall survival was 55% (95% CI 39%-71%) at 1 year, and 45% (95% CI 27%-63%) at 2 years. Nonrelapse mortality was 11% (95% CI 5%-25%) by 100 days post-HCT, 26% (95% CI 16%-44%) by 1 year, and 30% (95% CI 18%-50%) by 2 years. The cumulative incidence of primary disease relapse was 23% (95% CI 13%-41%) at 1 year, and 33% (95% CI 20%-56%) by 2 years after HCT. Cytomegalovirus (CMV) infection or reactivation varied according to recipient and donor CMV serostatus. Epstein-Barr Virus (EBV) reactivation occurred in 54% (95% CI 40%-71%) of patients. Preemptive rituximab therapy was administered for EBV reactivation, however, posttransplant lymphoproliferative disorder was diagnosed in 5 (11%) cases, and was fatal in 1. A regimen of ATG 7.5 mg/kg total ending on day -1 effectively decreased the occurrence of grade III-IV aGVHD and severe chronic GVHD.


Bone Marrow Transplantation | 2010

Feasibility of clofarabine cytoreduction before allogeneic transplant conditioning for refractory AML

Frederick L. Locke; Andrew S. Artz; Elizabeth Rich; Yanming Zhang; K. van Besien; Wendy Stock

To control disease before allogeneic hematopoietic cell transplantation (HCT) for relapsed/refractory AML, we used clofarabine cytoreduction. Seventeen patients received clofarabine 30–40 mg/m2 i.v. daily for 5 days with plans to initiate conditioning during the nadir, 14 days later. Bone marrow biopsy 12 days after clofarabine showed effective cytoreduction (that is,<20% cellularity with <10% blasts) in 10 of 17 patients (59%). Ineffective cytoreduction correlated with lower PFS (3.8 vs 6.4 months; HR=2.7, 95% CI=1.10–14.29, P=0.035) and OS (5.1 vs 16.6 months; HR=2.5, 95% CI=0.98–12.17, P=0.053). Significant toxicities before HCT, attributable to clofarabine, were grade 1–2 hyperbilirubinemia (18%); grade 1–2 (59%) or grade 3–4 (18%) transaminitis; and grade 1–2 (18%) creatinine elevation. Sixteen patients proceeded to HCT infusion 22 days (median) after initiation of clofarabine. Day 100 and 2-year transplant-related mortality were 6 and 36%. Nine patients relapsed. One year PFS and OS were 25 and 38%, respectively. Two patients are alive in remission at 18 and 52 months. Clofarabine cytoreduction followed by immediate HCT is feasible with acceptable toxicity and TRM. Outcomes for this cohort of patients with refractory AML remain poor and we are studying this approach in a prospective manner.


Bone Marrow Transplantation | 2013

A novel clofarabine bridge strategy facilitates allogeneic transplantation in patients with relapsed/refractory leukemia and high-risk myelodysplastic syndromes

Frederick L. Locke; R Agarwal; Rangesh Kunnavakkam; K. Van Besien; Richard A. Larson; Olatoyosi Odenike; Lucy A. Godley; Hui Liu; M. Le Beau; Sandeep Gurbuxani; Michael J. Thirman; Dorothy A. Sipkins; C White; Andrew S. Artz; Wendy Stock

Patients with relapsed/refractory leukemias or advanced myelodysplastic syndrome (MDS) fare poorly following allogeneic hematopoietic cell transplant (HCT). We report prospective phase II study results of 29 patients given clofarabine 30 mg/m2/day i.v. × 5 days followed immediately by HCT conditioning while at the cytopenic nadir. A total of 15/29 patients (52%) were cytoreduced according to pre-defined criteria (cellularity <20% and blasts <10%). Marrow cellularity (P<0.0001) and blast% (P=0.03) were reduced. Toxicities were acceptable, with transient hyperbilirubinemia (48%) and gr3–4 infections (10%). In all, 28/29 proceeded to transplant; 27 received ATG or alemtuzumab. Post HCT, 180 day non-relapse mortality (NRM) was 7% (95% confidence interval (CI): 1–21), relapse was 29% (95% CI: 13–46) and OS was 71% (95% CI: 51–85), comparing favorably to published data for high-risk patients. Two-year graft vs host disease incidence was 40% (95% CI: 21–58) and 2 year OS was 31% (95% CI: 14–48). Disease at the nadir correlated with inferior OS after HCT (HR=1.22 for each 10% marrow blasts, 95% CI: 1.02–1.46). For AML/MDS patients, there was a suggestion that successful cytoreduction increased PFS (330 vs 171 days, P=0.3) and OS (375 vs 195 days, P=0.31). Clofarabine used as a bridge to HCT reduces disease burden, is well tolerated, and permits high-risk patients to undergo HCT with acceptable NRM. Late relapses are common; thus, additional strategies should be pursued. NCT-00724009.


Haematologica | 2011

Sirolimus demonstrates activity in the primary therapy of acute graft-versus-host disease without systemic glucocorticoids

Joseph Pidala; Marcie Tomblyn; Taiga Nishihori; Teresa Field; Ernesto Ayala; Janelle Perkins; Hugo F. Fernandez; Frederick L. Locke; Lia Perez; Jose L Ochoa; Melissa Alsina; Claudio Anasetti

Background Advances in acute graft-versus-host disease therapy are needed. Design and Methods We examined the efficacy of sirolimus as primary therapy for acute graft-versus-host disease in 32 patients. Results Acute graft-versus-host disease involved the skin in 53% of cases, gastrointestinal tract in 66%, liver in 16%. The syndrome was overall grade 1 in 12% cases, grade 2 in 75%, and grade 3 in 13%. Sirolimus was targeted to achieve serum trough levels of 5–14 ng/mL. Sixteen (50%) patients achieved sustained, complete resolution of acute graft-versus-host disease with sirolimus alone. In contrast, 19 of 32 (59%) matched historical controls treated with standard 1 mg/kg steroids achieved complete response (P=0.47). With median follow-up time for surviving patients of 16 (range 6–26) months, one year overall survival was 56% (95% CI 38–74%). The cumulative incidence of relapse at one year was 37% (95% CI 23–60%), and mortality in remission was 20% (95% CI 10–42%). The cumulative incidence of chronic graft-versus-host disease was 55% (95% CI 39–79%). Thrombotic microangiopathy occurred in 3 cases (grade 1 n=1; grade 2 n=2), and responded to dose reduction of calcineurin inhibitor. Conclusions In this retrospective series, sirolimus demonstrates activity comparable to that of high-dose glucocorticoids in the primary therapy of acute graft-versus-host disease. Confirmation of this activity requires prospective clinical trials.


Journal of Immunological Methods | 2008

Use of Cre-adenovirus and CAR transgenic mice for efficient deletion of genes in post-thymic T cells

Yuanyuan Zha; Ramila Shah; Frederick L. Locke; Austin Wong; Thomas F. Gajewski

Conditional gene deletion using lineage-specific transgenic expression of Cre has been useful for defining the role of specific gene products in mice in vivo. However, this technology has had limitations for studies of peripheral T cell biology, since the T-lineage promoters commonly used are active early in thymic development. As such, T cell development can be altered by the resulting genetic alterations, thus limiting the interpretation of the data in post-thymic T cell studies. Thus, new strategies are needed to delete targeted genes directly in peripheral T lymphocytes. The availability of transgenic mice expressing the CAR in the T cell compartment enabled testing of Cre-mediated recombination using an adenoviral vector in naïve peripheral T cells in vitro, even without cellular activation. Using Rosa26R reporterxCAR transgenic mice, we describe conditions by which Cre-mediated deletion of targeted genes can be achieved with primary T cells in vitro. These cells can also be adoptively transferred into defined recipient mice for study in vivo. We use conditional PTEN-deficient mice as proof of concept to confirm the value of this technique for deleting a negative regulator of T cell activation. This technology should be broadly applicable for studies of T cell-specific gene deletion to gain understanding of function in the post-thymic T cell compartment.

Collaboration


Dive into the Frederick L. Locke's collaboration.

Top Co-Authors

Avatar

Claudio Anasetti

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Taiga Nishihori

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ernesto Ayala

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Joseph Pidala

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Hugo F. Fernandez

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Teresa Field

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Lia Perez

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Sattva S. Neelapu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Brian C. Betts

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge