Network


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

Hotspot


Dive into the research topics where Brian G. Till is active.

Publication


Featured researches published by Brian G. Till.


Blood | 2008

Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells

Brian G. Till; Michael C. Jensen; Jinjuan Wang; Eric Y. Chen; Brent L. Wood; Harvey A. Greisman; Xiaojun Qian; Scott E. James; Andrew Raubitschek; Stephen J. Forman; Ajay K. Gopal; John M. Pagel; Catherine G. Lindgren; Philip D. Greenberg; Stanley R. Riddell; Oliver W. Press

Adoptive immunotherapy with T cells expressing a tumor-specific chimeric T-cell receptor is a promising approach to cancer therapy that has not previously been explored for the treatment of lymphoma in human subjects. We report the results of a proof-of-concept clinical trial in which patients with relapsed or refractory indolent B-cell lymphoma or mantle cell lymphoma were treated with autologous T cells genetically modified by electroporation with a vector plasmid encoding a CD20-specific chimeric T-cell receptor and neomycin resistance gene. Transfected cells were immunophenotypically similar to CD8(+) effector cells and showed CD20-specific cytotoxicity in vitro. Seven patients received a total of 20 T-cell infusions, with minimal toxicities. Modified T cells persisted in vivo 1 to 3 weeks in the first 3 patients, who received T cells produced by limiting dilution methods, but persisted 5 to 9 weeks in the next 4 patients who received T cells produced in bulk cultures followed by 14 days of low-dose subcutaneous interleukin-2 (IL-2) injections. Of the 7 treated patients, 2 maintained a previous complete response, 1 achieved a partial response, and 4 had stable disease. These results show the safety, feasibility, and potential antitumor activity of adoptive T-cell therapy using this approach. This trial was registered at www.clinicaltrials.gov as #NCT00012207.


Journal of Immunology | 2008

Antigen Sensitivity of CD22-Specific Chimeric TCR Is Modulated by Target Epitope Distance from the Cell Membrane

Scott James; Philip D. Greenberg; Michael C. Jensen; Yukang Lin; Jinjuan Wang; Brian G. Till; Andrew Raubitschek; Stephen J. Forman; Oliver W. Press

We have targeted CD22 as a novel tumor-associated Ag for recognition by human CTL genetically modified to express chimeric TCR (cTCR) recognizing this surface molecule. CD22-specific cTCR targeting different epitopes of the CD22 molecule promoted efficient lysis of target cells expressing high levels of CD22 with a maximum lytic potential that appeared to decrease as the distance of the target epitope from the target cell membrane increased. Targeting membrane-distal CD22 epitopes with cTCR+ CTL revealed defects in both degranulation and lytic granule targeting. CD22-specific cTCR+ CTL exhibited lower levels of maximum lysis and lower Ag sensitivity than CTL targeting CD20, which has a shorter extracellular domain than CD22. This diminished sensitivity was not a result of reduced avidity of Ag engagement, but instead reflected weaker signaling per triggered cTCR molecule when targeting membrane-distal epitopes of CD22. Both of these parameters were restored by targeting a ligand expressing the same epitope, but constructed as a truncated CD22 molecule to approximate the length of a TCR:peptide-MHC complex. The reduced sensitivity of CD22-specific cTCR+ CTL for Ag-induced triggering of effector functions has potential therapeutic applications, because such cells selectively lysed B cell lymphoma lines expressing high levels of CD22, but demonstrated minimal activity against autologous normal B cells, which express lower levels of CD22. Thus, our results demonstrate that cTCR signal strength, and consequently Ag sensitivity, can be modulated by differential choice of target epitopes with respect to distance from the cell membrane, allowing discrimination between targets with disparate Ag density.


PLOS ONE | 2013

Combining a CD20 chimeric antigen receptor and an inducible caspase 9 suicide switch to improve the efficacy and safety of T cell adoptive immunotherapy for lymphoma.

Lihua E. Budde; Carolina Berger; Yukang Lin; Jinjuan Wang; Xubin Lin; Shani E. Frayo; Shaunda Brouns; David M. Spencer; Brian G. Till; Michael C. Jensen; Stanley R. Riddell; Oliver W. Press

Modification of T cells with chimeric antigen receptors (CAR) has emerged as a promising treatment modality for human malignancies. Integration of co-stimulatory domains into CARs can augment the activation and function of genetically targeted T cells against tumors. However, the potential for insertional mutagenesis and toxicities due to the infused cells have made development of safe methods for removing transferred cells an important consideration. We have genetically modified human T cells with a lentiviral vector to express a CD20-CAR containing both CD28 and CD137 co-stimulatory domains, a “suicide gene” relying on inducible activation of caspase 9 (iC9), and a truncated CD19 selectable marker. Rapid expansion (2000 fold) of the transduced T cells was achieved in 28 days after stimulation with artificial antigen presenting cells. Transduced T cells exhibited effective CD20-specific cytotoxic activity in vitro and in a mouse xenograft tumor model. Activation of the iC9 suicide switch resulted in efficient removal of transduced T cells both in vitro and in vivo. Our work demonstrates the feasibility and promise of this approach for treating CD20+ malignancies in a safe and more efficient manner. A phase I clinical trial using this approach in patients with relapsed indolent B-NHL is planned.


Journal of Clinical Oncology | 2011

Mantle Cell Lymphoma International Prognostic Index but Not Pretransplantation Induction Regimen Predicts Survival for Patients With Mantle-Cell Lymphoma Receiving High-Dose Therapy and Autologous Stem-Cell Transplantation

Lihua E. Budde; Katherine A. Guthrie; Brian G. Till; Oliver W. Press; Thomas R. Chauncey; John M. Pagel; Steven H. Petersdorf; William Bensinger; Leona Holmberg; Andrei R. Shustov; Damian J. Green; David G. Maloney; Ajay K. Gopal

PURPOSE High-dose therapy (HDT) and autologous stem-cell transplantation (ASCT) are frequently used in an attempt to improve outcome in patients with mantle-cell lymphoma (MCL); however, the importance of intensive induction regimens before transplantation is unknown. PATIENTS AND METHODS To address this question, we evaluated baseline characteristics, time to treatment, induction regimen, disease status at the time of transplantation, and MIPI score at diagnosis and their associations with survival in 118 consecutive patients with MCL who received HDT and ASCT at our centers. RESULTS The MIPI was independently associated with survival after transplantation in all 118 patients (hazard ratio [HR], 3.5; P < .001) and in the 85 patients who underwent ASCT as initial consolidation (HR, 7.2; P < .001). Overall survival rates were 93%, 60%, and 32% at 2.5 years from ASCT for all patients with low-, intermediate-, and high-risk MIPI, respectively. Low-risk MIPI scores were more common in the intensive induction group than the standard induction group in all patients (64% v 46%, respectively; P = .03) and in the initial consolidation group (66% v 45%, respectively; P = .03). After adjustment for the MIPI, an intensive induction regimen was not associated with improved survival after transplantation in all patients (HR, 0.5; P = .10), the initial consolidation group (HR, 1.1; P = .86), or patients ≤ 60 years old (HR, 0.6; P = .50). Observation of more than 3 months before initiating therapy did not yield inferior survival (HR, 2.1; P = .12) after adjustment for the MIPI in patients receiving ASCT. CONCLUSION An intensive induction regimen before HDT and ASCT was not associated with improved survival after adjusting for differences in MIPI scores at diagnosis.


Leukemia & Lymphoma | 2008

Effect of remission status and induction chemotherapy regimen on outcome of autologous stem cell transplantation for mantle cell lymphoma

Brian G. Till; Theodore A. Gooley; Nathan Crawford; Ajay K. Gopal; David G. Maloney; Stephen H. Petersdorf; John M. Pagel; Leona Holmberg; William Bensinger; Oliver W. Press

We analysed the outcomes of autologous stem cell transplantation (ASCT) following high-dose therapy with respect to remission status at the time of transplantation and induction regimen used in 56 consecutive patients with mantle cell lymphoma (MCL). Twenty-one patients received induction chemotherapy with HyperCVAD with or without rituximab (±R) followed by ASCT in first complete or partial remission (CR1/PR1), 15 received CHOP (±R) followed by ASCT in CR1/PR1 and 20 received ASCT following disease progression. Estimates of overall and progression-free survival (PFS) at 3 years among patients transplanted in CR1/PR1 were 93% and 63% compared with 46% and 36% for patients transplanted with relapsed/refractory disease, respectively. The hazard of mortality among patients transplanted with relapsed/refractory disease was 6.09 times that of patients transplanted in CR1/PR1 (P = 0.006). Patients in the CHOP (±R) group had a higher risk of failure for PFS compared with patients in the HyperCVAD (±R) group, though the difference did not reach statistical significance (hazard ratio 3.67, P = 0.11). These results suggest that ASCT in CR1/PR1 leads to improved survival outcomes for patients with MCL compared to ASCT with relapsed/refractory disease, and a HyperCVAD (±R) induction regimen may be associated with an improved PFS among patients transplanted in CR1/PR1.


Molecular Therapy | 2014

T-Cell Immunotherapy: Looking Forward

Jacqueline Corrigan-Curay; Hans Peter Kiem; David Baltimore; Marina O'Reilly; Renier J. Brentjens; Laurence J.N. Cooper; Stephen J. Forman; Stephen Gottschalk; Philip D. Greenberg; Richard P. Junghans; Helen E. Heslop; Michael C. Jensen; Crystal L. Mackall; Carl H. June; Oliver W. Press; Daniel J. Powell; Antoni Ribas; Steven A. Rosenberg; Michel Sadelain; Brian G. Till; Amy P. Patterson; Robert Jambou; Eugene Rosenthal; Linda Gargiulo; Maureen Montgomery; Donald B. Kohn

T rapidly expanding field of T-cell immunotherapy has experienced clinical successes along with some serious toxicities. “T Cell Immunotherapy: Optimizing Trial Design,” a workshop sponsored by the National Institutes of Health’s (NIH’s) Office of Biotechnology Activities (OBA), brought together researchers to discuss the scientific advances and share new data on key trial design issues, including the selection of new targets, optimizing the T-cell population, preconditioning regimens, strategies to promote persistence of cells, and analysis and management of acute reactions to T-cell infusions with the goal of identifying best practices and a research agenda that will facilitate further development and maximize the safety of this promising approach.The rapidly expanding field of T-cell immunotherapy has experienced clinical successes along with some serious toxicities. “T Cell Immunotherapy: Optimizing Trial Design,” a workshop sponsored by the National Institutes of Health’s (NIH’s) Office of Biotechnology Activities (OBA), brought together researchers to discuss the scientific advances and share new data on key trial design issues, including the selection of new targets, optimizing the T-cell population, preconditioning regimens, strategies to promote persistence of cells, and analysis and management of acute reactions to T-cell infusions with the goal of identifying best practices and a research agenda that will facilitate further development and maximize the safety of this promising approach.


Journal of Immunology | 2010

Mathematical Modeling of Chimeric TCR Triggering Predicts the Magnitude of Target Lysis and Its Impairment by TCR Downmodulation

Scott James; Philip D. Greenberg; Michael C. Jensen; Yukang Lin; Jinjuan Wang; Lihua E. Budde; Brian G. Till; Andrew Raubitschek; Stephen J. Forman; Oliver W. Press

We investigated relationships among chimeric TCR (cTCR) expression density, target Ag density, and cTCR triggering to predict lysis of target cells by cTCR+ CD8+ T human cells as a function of Ag density. Triggering of cTCR and canonical TCR by Ag could be quantified by the same mathematical equation, but cTCR represented a special case in which serial triggering was abrogated. The magnitude of target lysis could be predicted as a function of cTCR triggering, and the predicted minimum cTCR density required for maximal target lysis by CD20-specific cTCR was experimentally tested. cTCR density below ∼20,000 cTCR/cell impaired target lysis, but increasing cTCR expression above this density did not improve target lysis or Ag sensitivity. cTCR downmodulation to densities below this critical minimum by interaction with Ag-expressing targets limited the sequential lysis of targets in a manner that could be predicted based on the number of cTCRs remaining. In contrast, acute inhibition of lysis of primary, intended targets (e.g., leukemic B cells) due to the presence of an excess of secondary targets (e.g., normal B cells) was dependent on the Ag density of the secondary target but occurred at Ag densities insufficient to promote significant cTCR downmodulation, suggesting a role for functional exhaustion rather than insufficient cTCR density. This suggests increasing cTCR density above a critical threshold may enhance sequential lysis of intended targets in isolation, but will not overcome the functional exhaustion of cTCR+ T cells encountered in the presence of secondary targets with high Ag density.


Biology of Blood and Marrow Transplantation | 2013

Specific Features Identify Patients with Relapsed or Refractory Mantle Cell Lymphoma Benefitting from Autologous Hematopoietic Cell Transplantation

Ryan D. Cassaday; Katherine A. Guthrie; Elizabeth L. Budde; Leslie Thompson; Brian G. Till; Oliver W. Press; Thomas R. Chauncey; John M. Pagel; Stephen H. Petersdorf; Maria Corinna Palanca-Wessels; Mary Philip; William I. Bensinger; Leona Holmberg; Andrei R. Shustov; Damian J. Green; Edward N. Libby; David G. Maloney; Ajay K. Gopal

Outcomes with autologous hematopoietic cell transplantation (auto HCT) for relapsed and/or refractory mantle cell lymphoma (MCL) are typically poor. We hypothesized that certain factors could predict which patients experience a favorable outcome with this approach. We thus developed a predictive score from a cohort of 67 such patients using 3 factors independently associated with progression-free survival (PFS): (1) simplified Mantle Cell Lymphoma International Prognostic Index score before auto HCT (hazard ratio [HR], 2.9; P = .002); (2) B symptoms at diagnosis (HR, 2.7; P = .005); and (3) remission quotient, calculated by dividing the time, in months, from diagnosis to auto HCT by the number of prior treatments (HR, 1.4; P = .02). The estimated 5-year PFS for favorable-risk patients (n = 23) and unfavorable-risk patients (n = 44) were 58% (95% confidence interval [CI], 34% to 75%) and 15% (95% CI, 6% to 28%), respectively. These factors also independently predicted overall survival. In summary, we have defined 3 simple factors that can identify patients with relapsed/refractory MCL who derive a durable benefit from salvage auto HCT.


Blood | 2015

Astatine-211 conjugated to an anti-CD20 monoclonal antibody eradicates disseminated B-cell lymphoma in a mouse model

Damian J. Green; Mazyar Shadman; Jon C. Jones; Shani L. Frayo; Aimee L. Kenoyer; Mark D. Hylarides; Donald K. Hamlin; D. Scott Wilbur; Ethan R. Balkan; Yukang Lin; Brian W. Miller; Sofia H.L. Frost; Ajay K. Gopal; Johnnie J. Orozco; Theodore A. Gooley; Kelly L. Laird; Brian G. Till; Tom Bäck; John M. Pagel; Oliver W. Press

α-Emitting radionuclides deposit a large amount of energy within a few cell diameters and may be particularly effective for radioimmunotherapy targeting minimal residual disease (MRD). To evaluate this hypothesis, (211)At-labeled 1F5 monoclonal antibody (mAb) (anti-CD20) was studied in both bulky lymphoma tumor xenograft and MRD animal models. Superior treatment responses to (211)At-labeled 1F5 mAb were evident in the MRD setting. Lymphoma xenograft tumor-bearing animals treated with doses of up to 48 µCi of (211)At-labeled anti-CD20 mAb ([(211)At]1F5-B10) experienced modest responses (0% cures but two- to threefold prolongation of survival compared with negative controls). In contrast, 70% of animals in the MRD lymphoma model demonstrated complete eradication of disease when treated with (211)At-B10-1F5 at a radiation dose that was less than one-third (15 µCi) of the highest dose given to xenograft animals. Tumor progression among untreated control animals in both models was uniformly lethal. After 130 days, no significant renal or hepatic toxicity was observed in the cured animals receiving 15 µCi of [(211)At]1F5-B10. These findings suggest that α-emitters are highly efficacious in MRD settings, where isolated cells and small tumor clusters prevail.


Annals of Oncology | 2015

Maintenance rituximab after autologous stem cell transplantation in patients with mantle cell lymphoma

Solomon A. Graf; Philip A. Stevenson; Leona Holmberg; Brian G. Till; Oliver W. Press; Thomas R. Chauncey; Stephen D. Smith; Mary Philip; Johnnie J. Orozco; Andrei R. Shustov; Damian J. Green; Edward N. Libby; William I. Bensinger; John M. Pagel; David G. Maloney; Yi Zhou; Ryan D. Cassaday; Ajay K. Gopal

BACKGROUND High-dose therapy and autologous stem cell transplantation (ASCT) improves outcomes for patients with mantle cell lymphoma (MCL), but relapse ultimately occurs in most patients. Recently presented interim results from a phase III prospective trial suggest maintenance rituximab (MR) after ASCT for MCL improves progression-free survival (PFS). The maturation of these data and any benefit of MR on overall survival (OS) remain to be defined. PATIENTS AND METHODS In this retrospective study, we examined a cohort of consecutive patients with MCL that underwent ASCT for MCL at our center and evaluated their outcomes according to whether they received MR after ASCT (n = 50) or did not (n = 107). MR was treated as a time-dependent covariate to account for variation in timing of its initiation. RESULTS MR was associated with an improved PFS [hazard ratio (HR) 0.44; confidence interval (CI) (0.24-0.80), P = 0.007] and overall survival (OS; HR 0.46; CI 0.23-0.93, P = 0.03) following a multivariate adjustment for confounding factors with a median follow-up of ∼5 years. Grade 4 neutropenia was increased (34% versus 18%, P = 0.04) in the MR group, but no effect on the rate of mortality unrelated to relapse was observed. CONCLUSIONS These data support that MR after ASCT for MCL confers a benefit in PFS and additionally suggest it may improve OS. General application of this strategy will require confirmation of benefit in prospective randomized trials.

Collaboration


Dive into the Brian G. Till's collaboration.

Top Co-Authors

Avatar

Oliver W. Press

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Ajay K. Gopal

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Damian J. Green

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

David G. Maloney

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Leona Holmberg

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

John M. Pagel

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael C. Jensen

National Bureau of Economic Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lihua E. Budde

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge