Barbara Pender
Fred Hutchinson Cancer Research Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Barbara Pender.
Journal of Clinical Investigation | 2016
Cameron J. Turtle; Laïla Aïcha Hanafi; Carolina Berger; Theodore A. Gooley; Sindhu Cherian; Michael Hudecek; Daniel Sommermeyer; Katherine Melville; Barbara Pender; Tanya M Budiarto; Emily Robinson; Natalia N Steevens; Colette Chaney; Lorinda Soma; Xueyan Chen; Cecilia Yeung; Brent L. Wood; Daniel Li; Jianhong Cao; Shelly Heimfeld; Michael C. Jensen; Stanley R. Riddell; David G. Maloney
BACKGROUND T cells that have been modified to express a CD19-specific chimeric antigen receptor (CAR) have antitumor activity in B cell malignancies; however, identification of the factors that determine toxicity and efficacy of these T cells has been challenging in prior studies in which phenotypically heterogeneous CAR-T cell products were prepared from unselected T cells. METHODS We conducted a clinical trial to evaluate CD19 CAR-T cells that were manufactured from defined CD4+ and CD8+ T cell subsets and administered in a defined CD4+:CD8+ composition to adults with B cell acute lymphoblastic leukemia after lymphodepletion chemotherapy. RESULTS The defined composition product was remarkably potent, as 27 of 29 patients (93%) achieved BM remission, as determined by flow cytometry. We established that high CAR-T cell doses and tumor burden increase the risks of severe cytokine release syndrome and neurotoxicity. Moreover, we identified serum biomarkers that allow testing of early intervention strategies in patients at the highest risk of toxicity. Risk-stratified CAR-T cell dosing based on BM disease burden decreased toxicity. CD8+ T cell-mediated anti-CAR transgene product immune responses developed after CAR-T cell infusion in some patients, limited CAR-T cell persistence, and increased relapse risk. Addition of fludarabine to the lymphodepletion regimen improved CAR-T cell persistence and disease-free survival. CONCLUSION Immunotherapy with a CAR-T cell product of defined composition enabled identification of factors that correlated with CAR-T cell expansion, persistence, and toxicity and facilitated design of lymphodepletion and CAR-T cell dosing strategies that mitigated toxicity and improved disease-free survival. TRIAL REGISTRATION ClinicalTrials.gov NCT01865617. FUNDING R01-CA136551; Life Science Development Fund; Juno Therapeutics; Bezos Family Foundation.
Science Translational Medicine | 2016
Cameron J. Turtle; Laïla Aïcha Hanafi; Carolina Berger; Michael Hudecek; Barbara Pender; Emily Robinson; Reed M. Hawkins; Colette Chaney; Sindhu Cherian; Xueyan Chen; Lorinda Soma; Brent L. Wood; Daniel Li; Shelly Heimfeld; Stanley R. Riddell; David G. Maloney
A CD19 chimeric antigen receptor–modified T cell product with defined composition has potent antitumor activity. Standardizing the CAR assembly line Chimeric antigen receptor (CAR)–modified T cells are engineered to recognize specific tumor antigens. They have shown promising results in clinical trials, primarily in leukemia so far, but it has been difficult to predict therapeutic efficacy and toxicity for individual patients. To address this issue, Turtle et al. treated non-Hodgkin’s lymphoma patients with CAR-T cells prepared from strictly defined subsets. By carefully controlling the ratio of CD4 to CD8 T cells, the authors were able to identify some of the treatment characteristics that correlate with therapeutic response and toxicity, including the role of the drug regimen used for lymphodepletion before CAR-T cell treatment. CD19-specific chimeric antigen receptor (CAR)–modified T cells have antitumor activity in B cell malignancies, but factors that affect toxicity and efficacy have been difficult to define because of differences in lymphodepletion and heterogeneity of CAR-T cells administered to individual patients. We conducted a clinical trial in which CD19 CAR-T cells were manufactured from defined T cell subsets and administered in a 1:1 CD4+/CD8+ ratio of CAR-T cells to 32 adults with relapsed and/or refractory B cell non-Hodgkin’s lymphoma after cyclophosphamide (Cy)–based lymphodepletion chemotherapy with or without fludarabine (Flu). Patients who received Cy/Flu lymphodepletion had increased CAR-T cell expansion and persistence, and higher response rates [50% complete remission (CR), 72% overall response rate (ORR)] than patients who received Cy-based lymphodepletion without Flu (8% CR, 50% ORR). The CR rate in patients treated with Cy/Flu at the maximally tolerated dose was 64% (82% ORR; n = 11). Cy/Flu minimized the effects of an immune response to the murine single-chain variable fragment component of the CAR, which limited CAR-T cell expansion and clinical efficacy in patients who received Cy-based lymphodepletion without Flu. Severe cytokine release syndrome (sCRS) and grade ≥3 neurotoxicity were observed in 13 and 28% of all patients, respectively. Serum biomarkers, one day after CAR-T cell infusion, correlated with subsequent sCRS and neurotoxicity. Immunotherapy with CD19 CAR-T cells in a defined CD4+/CD8+ ratio allowed identification of correlative factors for CAR-T cell expansion, persistence, and toxicity, and facilitated optimization of lymphodepletion that improved disease response and overall and progression-free survival.
Cancer immunology research | 2016
Gregory A. Rufener; Oliver W. Press; Philip Olsen; Sang Yun Lee; Michael C. Jensen; Ajay K. Gopal; Barbara Pender; Lihua E. Budde; Jeffrey K. Rossow; Damian J. Green; David G. Maloney; Stanley R. Riddell; Brian G. Till
Most patients with B-cell lymphoma are treated with mAbs to CD20, which might interfere with subsequent chimeric antigen receptor (CAR) T cells against CD20. However, function was preserved in vivo in the presence of clinically relevant rituximab concentrations and only modestly impaired in vitro. CD20 is an attractive immunotherapy target for B-cell non-Hodgkin lymphomas, and adoptive transfer of T cells genetically modified to express a chimeric antigen receptor (CAR) targeting CD20 is a promising strategy. A theoretical limitation is that residual serum rituximab might block CAR binding to CD20 and thereby impede T cell–mediated anti-lymphoma responses. The activity of CD20 CAR-modified T cells in the presence of various concentrations of rituximab was tested in vitro and in vivo. CAR-binding sites on CD20+ tumor cells were blocked by rituximab in a dose-dependent fashion, although at 37°C blockade was incomplete at concentrations up to 200 μg/mL. T cells with CD20 CARs also exhibited modest dose-dependent reductions in cytokine secretion and cytotoxicity, but not proliferation, against lymphoma cell lines. At rituximab concentrations of 100 μg/mL, CAR T cells retained ≥50% of baseline activity against targets with high CD20 expression, but were more strongly inhibited when target cells expressed low CD20. In a murine xenograft model using a rituximab-refractory lymphoma cell line, rituximab did not impair CAR T-cell activity, and tumors were eradicated in >85% of mice. Clinical residual rituximab serum concentrations were measured in 103 lymphoma patients after rituximab therapy, with the median level found to be only 38 μg/mL (interquartile range, 19–72 μg/mL). Thus, despite modest functional impairment in vitro, the in vivo activity of CD20-targeted CAR T cells remains intact at clinically relevant levels of rituximab, making use of these T cells clinically feasible. Cancer Immunol Res; 4(6); 509–19. ©2016 AACR. See related Spotlight by Sadelain, p. 473.
Leukemia & Lymphoma | 2016
Mazyar Shadman; Ajay K. Gopal; Britt Kammerer; Pamela S. Becker; David G. Maloney; Barbara Pender; Andrei R. Shustov; Oliver W. Press; John M. Pagel
Despite initial responses to chemoimmunotherapy, relapse and minimal residual disease (MRD) remain major issues in treatment of chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL) patients. We administered 131I-tositumomab to patients in complete response (CR) or partial response (PR) after induction chemotherapy. Toxicities and rate of PR to CR conversion and MRD elimination were assessed three months later. The study stopped prematurely after enrolling 16 patients. Four (25%) were in CR, 12 (75%) in PR, and 12 (75%) had MRD. Three months after treatment with 131I-tositumomab, CR was achieved (n = 8; 50%) or sustained (n = 4; 25%) in 12 patients and MRD was eliminated in four of 12 patients (33%). Hematologic toxicities were anemia in one patient (6%), neutropenia in 13 (81%), and thrombocytopenia in eight (50%). Two patients (12%) developed MDS 17 and 20 months after consolidation. Consolidation with 131I-tositumomab for CLL/SLL patients in first remission is feasible and may provide the benefit of converting PR to CR and/or eliminating MRD.
Blood | 2015
Cameron J. Turtle; Carolina Berger; Daniel Sommermeyer; Laila-Aicha Hanafi; Barbara Pender; Emily Robinson; Katherine Melville; Tanya M Budiarto; Natalia N Steevens; Colette Chaney; Sindhu Cherian; Brent L. Wood; Lorinda Soma; Xueyan Chen; Shelly Heimfeld; Michael C. Jensen; Stanley R. Riddell; David G. Maloney
Blood | 2015
Cameron J. Turtle; Laila-Aicha Hanafi; Carolina Berger; Daniel Sommermeyer; Barbara Pender; Emily Robinson; Katherine Melville; Tanya M Budiarto; Natalia N Steevens; Colette Chaney; Sindhu Cherian; Brent L. Wood; Lorinda Soma; Xueyan Chen; Shelly Heimfeld; Michael C. Jensen; Stanley R. Riddell; David G. Maloney
Journal of Clinical Oncology | 2015
Cameron J. Turtle; Carolina Berger; Daniel Sommermeyer; Tanya M Budiarto; Laila-Aicha Hanafi; Katherine Melville; Barbara Pender; Natalia N Steevens; Colette Chaney; Shelly Heimfeld; Sindhu Cherian; Brent L. Wood; Lori Soma; Xueyan Chen; Michael C. Jensen; Stan R. Riddell; David G. Maloney
Journal of Clinical Oncology | 2018
Jordan Gauthier; Alexandre V. Hirayama; Kevin A. Hay; Daniel Li; Alyssa Sheih; Vicky Wu; Jenna M. Voutsinas; Sindhu Cherian; Xueyan Chen; Barbara Pender; Reed M. Hawkins; Aesha Vakil; Tinh-Doan Phi; Rachel N. Steinmetz; Stanley R. Riddell; David G. Maloney; Cameron J. Turtle
Blood | 2007
David G. Maloney; Barbara Pender; Erin McCarthy; Daniel P. Gold
Journal of Clinical Oncology | 2018
Kevin Anthony Hay; Jordan Gauthier; Alexandre V. Hirayama; Daniel Li; Alyssa Sheih; Vicky Wu; Jenna M. Voutsinas; Sindhu Cherian; Xueyan Chen; Barbara Pender; Reed M. Hawkins; Aesha Vakil; Rachel N. Steinmetz; Tinh-Doan Phi; Stanley R. Riddell; David G. Maloney; Cameron J. Turtle