Current Role at Stanford


Director, CCT Regulatory Operations and Procedures (CROP)

Education & Certifications


  • B.A., Northwestern University, Chemistry; Biological Sciences (2004)
  • PhD, Stanford University, Chemical & Systems Biology (2012)

All Publications


  • CD22 CAR T cells demonstrate high response rates and safety in pediatric and adult B-ALL: Phase 1b results. Leukemia Schultz, L. M., Jeyakumar, N., Kramer, A. M., Sahaf, B., Srinagesh, H., Shiraz, P., Agarwal, N., Hamilton, M., Erickson, C., Jacobs, A., Moon, J., Baggott, C., Arai, S., Bharadwaj, S., Johnston, L. J., Liedtke, M., Lowsky, R., Meyer, E., Negrin, R., Rezvani, A., Shizuru, J., Sidana, S., Egeler, E., Mavroukakis, S., Tunuguntla, R., Gkitsas-Long, N., Retherford, A., Brown, A. K., Gramstrap-Petersen, A. L., Ibañez, R. M., Feldman, S. A., Miklos, D. B., Mackall, C. L., Davis, K. L., Frank, M., Ramakrishna, S., Muffly, L. 2024

    Abstract

    Chimeric antigen receptor (CAR) T cells targeting CD22 (CD22-CAR) provide a therapeutic option for patients with CD22+ malignancies with progression after CD19-directed therapies. Using on-site, automated, closed-loop manufacturing, we conducted parallel Phase 1b clinical trials investigating a humanized CD22-CAR with 41BB costimulatory domain in children and adults with heavily treated, relapsed/refractory (r/r) B-ALL. Of 19 patients enrolled, 18 had successful CD22-CAR manufacturing, and 16 patients were infused. High grade (3-4) cytokine release syndrome (CRS) and immune effector-cell-associated neurotoxicity syndrome (ICANS) each occurred in only one patient; however, three patients experienced immune-effector-cell-associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS). Twelve of 16 patients (75%) achieved CR with an overall 56% MRD-negative CR rate. Duration of response was overall limited (median 77 days), and CD22 expression was downregulated in 4/12 (33%) available samples at relapse. In summary, we demonstrate that closed-loop manufacturing of CD22-CAR T cells is feasible and is associated with a favorable safety profile and high CR rates in pediatric and adult r/r B-ALL, a cohort with limited CD22-CAR reporting.

    View details for DOI 10.1038/s41375-024-02220-y

    View details for PubMedID 38491306

    View details for PubMedCentralID 4993814

  • GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature Majzner, R. G., Ramakrishna, S., Yeom, K. W., Patel, S., Chinnasamy, H., Schultz, L. M., Richards, R. M., Jiang, L., Barsan, V., Mancusi, R., Geraghty, A. C., Good, Z., Mochizuki, A. Y., Gillespie, S. M., Toland, A. M., Mahdi, J., Reschke, A., Nie, E., Chau, I. J., Rotiroti, M. C., Mount, C. W., Baggott, C., Mavroukakis, S., Egeler, E., Moon, J., Erickson, C., Green, S., Kunicki, M., Fujimoto, M., Ehlinger, Z., Reynolds, W., Kurra, S., Warren, K. E., Prabhu, S., Vogel, H., Rasmussen, L., Cornell, T. T., Partap, S., Fisher, P. G., Campen, C. J., Filbin, M. G., Grant, G., Sahaf, B., Davis, K. L., Feldman, S. A., Mackall, C. L., Monje, M. 2022

    Abstract

    Diffuse intrinsic pontine glioma (DIPG) and other H3K27M-mutated diffuse midline gliomas (DMG) are universally lethal paediatric central nervous system tumours1. We previously discovered that the disialoganglioside GD2 is highly expressed on H3K27M-mutant glioma cells and demonstrated promising preclinical efficacy of GD2-directed chimeric antigen receptor (CAR) T cells2, providing the rationale for a first-in-human Phase 1 clinical trial (NCT04196413). Because CAR T-cell-induced brainstem inflammation can result in obstructive hydrocephalus, increased intracranial pressure, and dangerous tissue shifts, neurocritical care precautions were incorporated. Here we present the clinical experience from the first four patients with H3K27M-mutant DIPG/DMG treated with GD2-CAR T cells (GD2-CART) at dose level 1 (1e6 GD2-CAR T cells/kg administered intravenously). Patients who exhibited clinical benefit were eligible for subsequent GD2-CAR T infusions administered intracerebroventricularly3. Toxicity was largely related to tumor location and reversible with intensive supportive care. On-target, off-tumor toxicity was not observed. Three of four patients exhibited clinical and radiographic improvement. Proinflammatory cytokines were increased in plasma and cerebrospinal fluid (CSF). Transcriptomic analyses of 65,598 single cells from CAR T cell products and CSF elucidate heterogeneity in response between subjects and administration routes. These early results underscore the promise of this approach for H3K27M+ DIPG/DMG therapy.

    View details for DOI 10.1038/s41586-022-04489-4

    View details for PubMedID 35130560

  • Risk Factors and Outcomes for Hematotoxicity Following Tisagenlecleucel for Pediatric BAcute Lymphoblastic Leukemia McNerney, K. O., Fabrizio, V. A., Talleur, A. C., Lim, S., Dreyzin, A., Baggott, C., Vatsayan, A., Rossoff, J., Prabhu, S., Pacenta, H. L., Phillips, C. L., Talano, J., Moskop, A., Verneris, M. R., Myers, D., Karras, N., Bonifant, C. L., Qayed, M., Hermiston, M. L., Satwani, P., Krupski, C., Keating, A., Baumeister, S. C., Chinnabhandar, V., Egeler, E., Curran, K. J., Mackall, C. L., Laetsch, T. W., Schultz, L., Naik, S. AMER SOC HEMATOLOGY. 2023
  • HLH-like toxicities predict poor survival following use of tisagenlecleucel in children and young adults with B-ALL. Blood advances McNerney, K. O., Si Lim, S., Ishikawa, K., Dreyzin, A., Vatsayan, A., Chen, J. J., Baggott, C., Prabhu, S., Pacenta, H. L., Phillips, C. L., Rossoff, J., Stefanski, H. E., Talano, J. A., Moskop, A., Verneris, M. R., Myers, D., Karras, N. A., Brown, P. A., Bonifant, C. L., Qayed, M., Hermiston, M. L., Satwani, P., Krupski, C., Keating, A. K., Baumeister, S. H., Fabrizio, V. A., Chinnabhandar, V., Egeler, E., Mavroukakis, S., Curran, K. J., Mackall, C. L., Laetsch, T. W., Schultz, L. M. 2023

    Abstract

    Chimeric antigen-receptor (CAR)-associated hemophagocytic lymphohistiocytosis (HLH)-like toxicities involving hyperferritinemia, multi-organ dysfunction, coagulopathy, and/or hemophagocytosis are described as occurring in a subset of patients with cytokine release syndrome (CRS). Case series report poor outcomes for those with B-acute lymphoblastic leukemia (B-ALL) who develop HLH-like toxicities, although larger outcomes analyses of children and young adults (CAYA) with B-ALL who develop these toxicities following commercial tisagenlecleucel are not described. Using a multi-institutional database of 185 CAYA with B-ALL, we conducted a retrospective cohort study including groups that developed HLH-like toxicities, high grade CRS without HLH-like toxicities, or no to low grade CRS without HLH-like toxicities. Primary objectives included characterizing the incidence, outcomes, and pre-infusion factors associated with HLH-like toxicities. Among 185 CAYA infused with tisagenlecleucel, 26 (14.1%) met criteria for HLH-like toxicities. One-year overall survival and relapse-free survival were 25.7% and 4.7% in those with HLH-like toxicities, compared with 80.1% and 57.6% in those without. In multivariable analysis for death, meeting criteria for HLH-like toxicities carried a hazard ratio of 4.61 (95% confidence interval: 2.41-8.83), controlling for disease burden, age, and sex. Patients who developed HLH-like toxicities had higher pre-tisagenlecleucel disease burden, ferritin, C-reactive protein levels, and lower platelet and absolute neutrophil counts than patients with high grade or no/low grade CRS without HLH-like toxicities. Overall, CAYA with B-ALL who developed HLH-like toxicities following tisagenlecleucel experienced high rates of relapse and non-relapse mortality, indicating the urgent need for further investigations into prevention and optimal management of patients who develop HLH-like toxicities following tisagenlecleucel.

    View details for DOI 10.1182/bloodadvances.2022008893

    View details for PubMedID 36857419

  • CAR-ASSOCIATED HEMOPHAGOCYTIC LYMPHOHISTIOCYTOSIS (carHLH) IS ASSOCIATED WITH PRE-INFUSION INFLAMMATION AND HIGH DISEASE BURDEN AND PREDICTS POOR OUTCOMES FOLLOWING TISAGENLECLEUCEL IN PEDIATRIC B-ALL McNerney, K. O., Lim, S., Ishikawa, K., Dreyzin, A., Vatsayan, A., Chen, J. J., Baggott, C., Prabhu, S., Pacenta, H., Philips, C., Rossoff, J., Stefanski, H., Talano, J., Moskop, A., Verneris, M., Myers, D., Karras, N. A., Brown, P., Qayed, M., Hermiston, M., Satwani, P., Krupski, C., Keating, A. K., Wilcox, R., Baumeister, S., Fabrizio, V. A., Chinnabhandar, V., Egeler, E., Mavroukakis, S., Curran, K. J., Mackall, C. L., Laetsch, T. W., Schultz, L. M. WILEY. 2023: S32-S33
  • Outcomes After Nonresponse and Relapse Post-Tisagenlecleucel in Children, Adolescents, and Young Adults With B-Cell Acute Lymphoblastic Leukemia. Journal of clinical oncology : official journal of the American Society of Clinical Oncology Schultz, L. M., Eaton, A., Baggott, C., Rossoff, J., Prabhu, S., Keating, A. K., Krupski, C., Pacenta, H., Philips, C. L., Talano, J., Moskop, A., Baumeister, S. H., Myers, G. D., Karras, N. A., Brown, P. A., Qayed, M., Hermiston, M., Satwani, P., Wilcox, R., Rabik, C. A., Fabrizio, V. A., Chinnabhandar, V., Kunicki, M., Mavroukakis, S., Egeler, E., Li, Y., Mackall, C. L., Curran, K. J., Verneris, M. R., Laetsch, T. W., Stefanski, H. 2022: JCO2201076

    Abstract

    PURPOSE: Nonresponse and relapse after CD19-chimeric antigen receptor (CAR) T-cell therapy continue to challenge survival outcomes. Phase II landmark data from the ELIANA trial demonstrated nonresponse and relapse rates of 14.5% and 28%, respectively, whereas use in the real-world setting showed nonresponse and relapse rates of 15% and 37%. Outcome analyses describing fate after post-CAR nonresponse and relapse remain limited. Here, we aim to establish survival outcomes after nonresponse and both CD19+ and CD19- relapses and explore treatment variables associated with inferior survival.METHODS: We conducted a retrospective multi-institutional study of 80 children and young adults with B-cell acute lymphoblastic leukemia experiencing nonresponse (n = 23) or relapse (n = 57) after tisagenlecleucel. We analyze associations between baseline characteristics and these outcomes and establish survival rates and salvage approaches.RESULTS: The overall survival (OS) at 12 months was 19% across nonresponders (n = 23; 95% CI, 7 to 50). Ninety-five percent of patients with nonresponse had high preinfusion disease burden. Among 156 morphologic responders, the cumulative incidence of relapse was 37% (95% CI, 30 to 47) at 12 months (CD19+; 21% [15 to 29], CD19-; 16% [11 to 24], median follow-up; 380 days). Across 57 patients experiencing relapse, the OS was 52% (95% CI, 38 to 71) at 12 months after time of relapse. Notably, CD19- relapse was associated with significantly decreased OS as compared with patients who relapsed with conserved CD19 expression (CD19- 12-month OS; 30% [14 to 66], CD19+ 12-month OS; 68% [49 to 92], P = .0068). Inotuzumab, CAR reinfusion, and chemotherapy were used as postrelapse salvage therapy with greatest frequency, yet high variability in treatment sequencing and responses limits efficacy analysis across salvage approaches.CONCLUSION: We describe poor survival across patients experiencing nonresponse to tisagenlecleucel. In the post-tisagenlecleucel relapse setting, patients can be salvaged; however, CD19- relapse is distinctly associated with decreased survival outcomes.

    View details for DOI 10.1200/JCO.22.01076

    View details for PubMedID 36108252

  • Higher doses of tisagenlecleucel associate with improved outcomes: a report from the pediatric real-world CAR consortium. Blood advances Stefanski, H., Eaton, A., Baggott, C., Rossoff, J., Verneris, M. R., Keating, A. K., Prabhu, S., Pacenta, H. L., Phillips, C. L., Talano, J., Moskop, A., Margossian, S. P., Myers, G. D., Karras, N. A., Brown, P. A., Qayed, M., Hermiston, M. L., Satwani, P., Krupski, C., Wilcox, R., Rabik, C. A., Fabrizio, V. A., Chinnabhandar, V., Goksenin, A. Y., Egeler, E., Mavroukakis, S., Curran, K. J., Mackall, C. L., Laetsch, T. W., Schultz, L. M. 2022

    Abstract

    Tisagenlecleucel, chimeric antigen receptor T cell (CART) therapy targeting CD19, has shown remarkable complete response (CR) rates for patients up to the age of 26 with refractory/relapsed B-cell acute lymphoblastic leukemia (B-ALL), and is FDA-approved for this indication. Currently, patients receive a single dose of tisagenlecleucel across a wide dose range of 0.2- 5.0 x 106 CAR T cells/kg for patients ≤ 50 kg, or 0.1- 2.5 x 108 CAR T cells for patients > 50 kg. The effect of cell dose on survival and remission has yet to be well-established. Our primary goal was to determine if CART cell dose impacts overall survival (OS), event-free survival (EFS) and relapse-free-survival (RFS) in tisagenlecleucel-recipients. Retrospective data were collected from the Pediatric Real World CAR Consortium (PRWCC) member institutions and included 185 patients infused with commercial tisagenlecleucel. The median dose of viable transduced CAR T cells was 1.7 x106 CAR T cells/kg. To assess the impact of cell dose, we divided the responders into dose quartiles: 0.134-1.300 x 106 (D1; n=48 (27%), 1.301-1.700 x 106 (D2; n=46 (26%), 1.701-2.400 x 106(D3; n=43 (24%) and 2.401-5.100 x 106 (D4; n=43 (24%). OS, EFS, and RFS were improved in patients that received higher doses of Tisagenlecleucel (p=0.031, 0.0079 and 0.0045 respectively). Moreover, higher doses of tisagenlecleucel were not associated with increased toxicity. As the current tisagenlecleucel package insert dose-range remains broad, this work has implications in regard to targeting higher cell doses to optimize patients for long-standing remission.

    View details for DOI 10.1182/bloodadvances.2022007246

    View details for PubMedID 35938863

  • MAJOR TUMOR REGRESSIONS IN H3K27M-MUTATED DIFFUSE MIDLINE GLIOMA (DMG) FOLLOWING SEQUENTIAL INTRAVENOUS (IV) AND INTRACEREBROVENTRICULAR (ICV) DELIVERY OF GD2-CAR T-CELLS Monje, M., Majzner, R., Mahdi, J., Ramakrishna, S., Patel, S., Chinnasamy, H., Yeom, K., Schultz, L., Barsan, V., Richards, R., Campen, C., Reschke, A., Toland, A., Baggott, C., Mavroukakis, S., Egeler, E., Moon, J., Jacobs, A., Yamabe-Kwong, K., Rasmussen, L., Nie, E., Green, S., Kunicki, M., Fujimoto, M., Ehlinger, Z., Reynolds, W., Prabhu, S., Warren, K. E., Cornell, T., Partap, S., Fisher, P., Grant, G., Vogel, H., Sahaf, B., Davis, K., Feldman, S., Mackall, C. OXFORD UNIV PRESS INC. 2022: 20-21
  • Disease Burden Affects Outcomes in Pediatric and Young Adult B-Cell Lymphoblastic Leukemia After Commercial Tisagenlecleucel: A Pediatric Real-World Chimeric Antigen Receptor Consortium Report. Journal of clinical oncology : official journal of the American Society of Clinical Oncology Schultz, L. M., Baggott, C., Prabhu, S., Pacenta, H. L., Phillips, C. L., Rossoff, J., Stefanski, H. E., Talano, J., Moskop, A., Margossian, S. P., Verneris, M. R., Myers, G. D., Karras, N. A., Brown, P. A., Qayed, M., Hermiston, M., Satwani, P., Krupski, C., Keating, A. K., Wilcox, R., Rabik, C. A., Fabrizio, V. A., Rouce, R. H., Chinnabhandar, V., Kunicki, M., Barsan, V. V., Goksenin, A. Y., Li, Y., Mavroukakis, S., Egeler, E., Curran, K. J., Mackall, C. L., Laetsch, T. W. 2021: JCO2003585

    Abstract

    PURPOSE: Tisagenlecleucel is a CD19-specific chimeric antigen receptor T-cell therapy, US Food and Drug Administration-approved for children, adolescents, and young adults (CAYA) with relapsed and/or refractory (RR) B-cell acute lymphoblastic leukemia (B-ALL). The US Food and Drug Administration registration for tisagenlecleucel was based on a complete response (CR) rate of 81%, 12-month overall survival (OS) of 76%, and event-free survival (EFS) of 50%. We report clinical outcomes and analyze covariates of outcomes after commercial tisagenlecleucel.METHODS: We conducted a retrospective, multi-institutional study of CAYA with RR B-ALL across 15 US institutions, who underwent leukapheresis shipment to Novartis for commercial tisagenlecleucel. A total of 200 patients were included in an intent-to-treat response analysis, and 185 infused patients were analyzed for survival and toxicity.RESULTS: Intent-to-treat analysis demonstrates a 79% morphologic CR rate (95% CI, 72 to 84). The infused cohort had an 85% CR (95% CI, 79 to 89) and 12-month OS of 72% and EFS of 50%, with 335 days of median follow-up. Notably, 48% of patients had low-disease burden (< 5% bone marrow lymphoblasts, no CNS3, or other extramedullary disease), or undetectable disease, pretisagenlecleucel. Univariate and multivariate analyses associate high-disease burden (HB, ≥ 5% bone marrow lymphoblasts, CNS3, or non-CNS extramedullary) with inferior outcomes, with a 12-month OS of 58% and EFS of 31% compared with low-disease burden (OS; 85%, EFS; 70%) and undetectable disease (OS; 95%, EFS; 72%; P < .0001 for OS and EFS). Grade ≥ 3 cytokine release syndrome and neurotoxicity rates were 21% and 7% overall and 35% and 9% in patients with HB, respectively.CONCLUSION: Commercial tisagenlecleucel in CAYA RR B-ALL demonstrates efficacy and tolerability. This first analysis of commercial tisagenlecleucel stratified by disease burden identifies HB preinfusion to associate with inferior OS and EFS and increased toxicity.

    View details for DOI 10.1200/JCO.20.03585

    View details for PubMedID 34882493

  • CD22-CAR T-Cell Therapy Mediates High Durable Remission Rates in Adults with Large B-Cell Lymphoma Who Have Relapsed after CD19-CAR T-Cell Therapy Frank, M. J., Baird, J. H., Patel, S., Craig, J., Spiegel, J. Y., Ehlinger, Z., Chinnasamy, H., Younes, S. F., Oak, J. S., Natkunam, Y., Reynolds, W. D., Iglesias, M., Crawford, E., Srinagesh, H. K., Egeler, E. L., Arai, S., Johnston, L. J., Lowsky, R., Negrin, R. S., Rezvani, A. R., Shiraz, P., Sidana, S., Weng, W., Schultz, L. M., Ramakrishna, S., Davis, K. L., Sahaf, B., Feldman, S. A., Mackall, C. L., Miklos, D. B., Muffl, L. AMER SOC HEMATOLOGY. 2021
  • Tisagenlecleucel Outcomes in Relapsed/Refractory Extramedullary ALL: A Pediatric Real World CAR Consortium Report. Blood advances Fabrizio, V. A., Phillips, C. L., Lane, A., Baggott, C., Prabhu, S., Egeler, E., Mavroukakis, S., Pacenta, H. L., Rossoff, J., Stefanski, H., Talano, J. A., Moskop, A., Margossian, S. P., Verneris, M. R., Myers, G. D., Karras, N. A., Brown, P. A., Qayed, M., Hermiston, M. L., Satwani, P., Krupski, C., Keating, A. K., Wilcox, R., Rabik, C. A., Chinnabhandar, V., Kunicki, M., Goksenin, A. Y., Curran, K. J., Mackall, C. L., Laetsch, T. W., Schultz, L. M. 2021

    Abstract

    Chimeric antigen receptor (CAR) T-cells have transformed the therapeutic options for relapsed/refractory (R/R) B-cell ALL. Data for CAR therapy in extramedullary (EM) involvement is limited. Retrospective data was abstracted from the Pediatric Real World CAR Consortium (PRWCC) of 184 infused patients from 15 US institutions. Response (CR) rate, overall survival (OS), relapse-free survival (RFS), and duration of B-cell aplasia (BCA) in patients referred for tisagenlecleucel with EM disease (both CNS3 and non-CNS EM) were compared to bone marrow (BM) only. Patients with CNS disease were further stratified for comparison. Outcomes are reported on 55 patients with EM disease prior to CAR (n=40 CNS3; n=15 non-CNS EM). The median age at infusion in CNS cohort was 10 years (range <1-25) and the non-CNS EM cohort was 13 years (2-26). In patients with CNS disease, 88% (35/40) achieved a CR, versus only 66% (10/15) with non-CNS EM disease. Patients with CNS disease (both with and without marrow involvement) had comparable 24-month OS outcomes to non-CNS EM or BM only (p=0.41). There was no difference in 12-month RFS between CNS, non-CNS EM, or BM only patients (p=0.92). No increased toxicity was seen with CNS or non-CNS EM disease (p=0.3). Active CNS disease at time of infusion did not impact outcomes. Isolated (iCNS) disease trended towards improved OS when compared to combined CNS and BM (p=0.12). R/R EM disease can be effectively treated with tisagenlecleucel with toxicity, relapse rates and survival rates comparable to patients with BM only. Outcomes for iCNS relapse are encouraging.

    View details for DOI 10.1182/bloodadvances.2021005564

    View details for PubMedID 34794180

  • Optimal fludarabine lymphodepletion is associated with improved outcomes following CAR T-cell Therapy. Blood advances Fabrizio, V. A., Boelens, J. J., Mauguen, A., Baggott, C., Prabhu, S., Egeler, E., Mavroukakis, S., Pacenta, H. L., Phillips, C. L., Rossoff, J., Stefanski, H., Talano, J. A., Moskop, A., Margossian, S. P., Verneris, M. R., Myers, G. D., Karras, N. A., Brown, P. A., Qayed, M., Hermiston, M. L., Satwani, P., Krupski, C., Keating, A. K., Wilcox, R., Rabik, C. A., Chinnabhandar, V., Kunicki, M., Goksenin, A. Y., Mackall, C. L., Laetsch, T. W., Schultz, L. M., Curran, K. J. 2021

    Abstract

    Chimeric antigen receptor (CAR) T-cells provide a therapeutic option in hematologic malignancies. However, treatment failure after initial response approaches 50%. In allogeneic hematopoietic cell transplantation, optimal fludarabine exposure improves immune reconstitution, resulting in lower nonrelapse mortality and increased survival. We hypothesized that optimal fludarabine exposure in lymphodepleting chemotherapy prior to CAR T-cell therapy would improve outcomes. In a retrospective analysis of relapsed/refractory B-cell acute lymphoblastic leukemia patients undergoing CAR T-cell (tisagenlecleucel) infusion after cyclophosphamide/fludarabine lymphodepleting chemotherapy, we estimated the fludarabine exposure as area-under-the-curve (AUC;mg*hr/L) using a validated population-pharmacokinetic model. Fludarabine exposure was related to overall survival (OS), cumulative incidence of relapse (CIR), and a composite endpoint (loss of B-cell aplasia (BCA) or relapse). Eligible patients (n=152) had a median age of 12.5 years (range <1-26), response rate of 86% (131/152), 12-month OS of 75.1% (95%-CI: 67.6-82.6%), and 12-month CIR of 36.4% (95%-CI: 27.5-45.2%). Optimal fludarabine-exposure was determined as an AUC≥13.8mg*hr/L. In multivariable analyses patients with an AUC<13.8mg*hr/L had a 2.5-fold higher CIR (HR=2.45 [1.34-4.48]; P=0.005) and a twofold higher risk of relapse or loss of BCA (HR=1.96 [1.19-3.23]; P=0.01) compared to those with optimal fludarabine exposure. High preinfusion disease burden was also associated with an increased risk of relapse (HR=2.66 [1.45-4.87]; P=0.001) and death (HR=4.77 [2.10-10.9]; p<0.001). Personalized PK-directed dosing to achieve optimal fludarabine exposure should be tested in prospective trials and based on this analysis may reduce disease relapse after CAR T-cell therapy.

    View details for DOI 10.1182/bloodadvances.2021006418

    View details for PubMedID 34788386

  • GD2 CAR T cells mediate clinical activity and manageable toxicity in children and young adults with DIPG and H3K27M-mutated diffuse midline gliomas. Majzner, R. G., Ramakrishna, S., Mochizuki, A., Patel, S., Chinnasamy, H., Yeom, K., Schultz, L., Richards, R., Campen, C., Reschke, A., Mahdi, J., Toland, A., Baggott, C., Mavroukakis, S., Egeler, E., Moon, J., Landrum, K., Erickson, C., Rasmussen, L., Barsan, V., Tamaresis, J. S., Marcy, A., Kunicki, M., Fujimoto, M., Ehlinger, Z., Kurra, S., Cornell, T., Partap, S., Fisher, P., Grant, G., Vogel, H., Sahaf, B., Davis, K., Feldman, S., Mackall, C. L., Monje, M. AMER ASSOC CANCER RESEARCH. 2021
  • SINGLE CELL RNA SEQUENCING FROM THE CSF OF SUBJECTS WITH H3K27M+DIPG/DMG TREATED WITH GD2 CAR T-CELLULAR THERAPY Mochizuki, A., Ramakrishna, S., Good, Z., Patel, S., Chinnasamy, H., Yeom, K., Schultz, L., Richards, R., Campen, C., Reschke, A., Mahdi, J., Toland, A., Baggot, C., Mavroukakis, S., Egeler, E., Moon, J., Landrum, K., Erickson, C., Rasmussen, L., Barsan, V., Tamaresis, J., Marcy, A., Kunicki, M., Celones, M., Ehlinger, Z., Kurra, S., Cornell, T., Partap, S., Fisher, P., Grant, G., Vogel, H., Davis, K., Feldman, S., Sahaf, B., Majzner, R., Mackall, C., Monje, M. OXFORD UNIV PRESS INC. 2021: 39
  • GD2 CAR T-CELLS MEDIATE CLINICAL ACTIVITY AND MANAGEABLE TOXICITY IN CHILDREN AND YOUNG ADULTS WITH H3K27M-MUTATED DIPG AND SPINAL CORD DMG Majzner, R., Ramakrishna, S., Mochizuki, A., Patel, S., Chinnasamy, H., Yeom, K., Schultz, L., Richards, R., Campen, C., Reschke, A., Mahdi, J., Martin, A., Toland, S., Baggott, C., Mavroukakis, S., Egeler, E., Moon, J., Landrum, K., Erickson, C., Rasmussen, L., Barsan, V., Tamaresis, J., Marcy, A., Kunicki, M., Fujimoto, M., Ehlinger, Z., Kurra, S., Cornell, T., Partap, S., Fisher, P., Grant, G., Vogel, H., Sahaf, B., Davis, K., Feldman, S., Mackall, C., Monje, M. OXFORD UNIV PRESS INC. 2021: 49-50
  • Intracellular Context Affects Levels of a Chemically Dependent Destabilizing Domain PLOS ONE Sellmyer, M. A., Chen, L., Egeler, E. L., Rakhit, R., Wandless, T. J. 2012; 7 (9)

    Abstract

    The ability to regulate protein levels in live cells is crucial to understanding protein function. In the interest of advancing the tool set for protein perturbation, we developed a protein destabilizing domain (DD) that can confer its instability to a fused protein of interest. This destabilization and consequent degradation can be rescued in a reversible and dose-dependent manner with the addition of a small molecule that is specific for the DD, Shield-1. Proteins encounter different local protein quality control (QC) machinery when targeted to cellular compartments such as the mitochondrial matrix or endoplasmic reticulum (ER). These varied environments could have profound effects on the levels and regulation of the cytoplasmically derived DD. Here we show that DD fusions in the cytoplasm or nucleus can be efficiently degraded in mammalian cells; however, targeting fusions to the mitochondrial matrix or ER lumen leads to accumulation even in the absence of Shield-1. Additionally, we characterize the behavior of the DD with perturbants that modulate protein production, degradation, and local protein QC machinery. Chemical induction of the unfolded protein response in the ER results in decreased levels of an ER-targeted DD indicating the sensitivity of the DD to the degradation environment. These data reinforce that DD is an effective tool for protein perturbation, show that the local QC machinery affects levels of the DD, and suggest that the DD may be a useful probe for monitoring protein quality control machinery.

    View details for DOI 10.1371/journal.pone.0043297

    View details for Web of Science ID 000308738500009

    View details for PubMedID 22984418

    View details for PubMedCentralID PMC3440426

  • Ligand-switchable Substrates for a Ubiquitin-Proteasome System JOURNAL OF BIOLOGICAL CHEMISTRY Egeler, E. L., Urner, L. M., Rakhit, R., Liu, C. W., Wandless, T. J. 2011; 286 (36): 31328-31336

    Abstract

    Cellular maintenance of protein homeostasis is essential for normal cellular function. The ubiquitin-proteasome system (UPS) plays a central role in processing cellular proteins destined for degradation, but little is currently known about how misfolded cytosolic proteins are recognized by protein quality control machinery and targeted to the UPS for degradation in mammalian cells. Destabilizing domains (DDs) are small protein domains that are unstable and degraded in the absence of ligand, but whose stability is rescued by binding to a high affinity cell-permeable ligand. In the work presented here, we investigate the biophysical properties and cellular fates of a panel of FKBP12 mutants displaying a range of stabilities when expressed in mammalian cells. Our findings correlate observed cellular instability to both the propensity of the protein domain to unfold in vitro and the extent of ubiquitination of the protein in the non-permissive (ligand-free) state. We propose a model in which removal of stabilizing ligand causes the DD to unfold and be rapidly ubiquitinated by the UPS for degradation at the proteasome. The conditional nature of DD stability allows a rapid and non-perturbing switch from stable protein to unstable UPS substrate unlike other methods currently used to interrogate protein quality control, providing tunable control of degradation rates.

    View details for DOI 10.1074/jbc.M111.264101

    View details for Web of Science ID 000294487500028

    View details for PubMedID 21768107

    View details for PubMedCentralID PMC3173072

  • Regulating protein stability in mammalian cells using small molecules. Cold Spring Harbor protocols Hagan, E. L., Banaszynski, L. A., Chen, L., Maynard-Smith, L. A., Wandless, T. J. 2009; 2009 (3): pdb prot5172-?

    View details for DOI 10.1101/pdb.prot5172

    View details for PubMedID 20147107

    View details for PubMedCentralID PMC3213858