5 results on '"Mueller KP"'
Search Results
2. FOXO1 is a master regulator of memory programming in CAR T cells.
- Author
-
Doan AE, Mueller KP, Chen AY, Rouin GT, Chen Y, Daniel B, Lattin J, Markovska M, Mozarsky B, Arias-Umana J, Hapke R, Jung IY, Wang A, Xu P, Klysz D, Zuern G, Bashti M, Quinn PJ, Miao Z, Sandor K, Zhang W, Chen GM, Ryu F, Logun M, Hall J, Tan K, Grupp SA, McClory SE, Lareau CA, Fraietta JA, Sotillo E, Satpathy AT, Mackall CL, and Weber EW
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Chromatin metabolism, Chromatin genetics, Gene Editing, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Forkhead Box Protein O1 metabolism, Immunologic Memory, Immunotherapy, Adoptive, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Receptors, Chimeric Antigen genetics, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes cytology
- Abstract
A major limitation of chimeric antigen receptor (CAR) T cell therapies is the poor persistence of these cells in vivo
1 . The expression of memory-associated genes in CAR T cells is linked to their long-term persistence in patients and clinical efficacy2-6 , suggesting that memory programs may underpin durable CAR T cell function. Here we show that the transcription factor FOXO1 is responsible for promoting memory and restraining exhaustion in human CAR T cells. Pharmacological inhibition or gene editing of endogenous FOXO1 diminished the expression of memory-associated genes, promoted an exhaustion-like phenotype and impaired the antitumour activity of CAR T cells. Overexpression of FOXO1 induced a gene-expression program consistent with T cell memory and increased chromatin accessibility at FOXO1-binding motifs. CAR T cells that overexpressed FOXO1 retained their function, memory potential and metabolic fitness in settings of chronic stimulation, and exhibited enhanced persistence and tumour control in vivo. By contrast, overexpression of TCF1 (encoded by TCF7) did not enforce canonical memory programs or enhance the potency of CAR T cells. Notably, FOXO1 activity correlated with positive clinical outcomes of patients treated with CAR T cells or tumour-infiltrating lymphocytes, underscoring the clinical relevance of FOXO1 in cancer immunotherapy. Our results show that overexpressing FOXO1 can increase the antitumour activity of human CAR T cells, and highlight memory reprogramming as a broadly applicable approach for optimizing therapeutic T cell states., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
3. FOXO1 is a master regulator of CAR T memory programming.
- Author
-
Doan A, Mueller KP, Chen A, Rouin GT, Daniel B, Lattin J, Chen Y, Mozarsky B, Markovska M, Arias-Umana J, Hapke R, Jung I, Xu P, Klysz D, Bashti M, Quinn PJ, Sandor K, Zhang W, Hall J, Lareau C, Grupp SA, Fraietta JA, Sotillo E, Satpathy AT, Mackall CL, and Weber EW
- Abstract
Poor CAR T persistence limits CAR T cell therapies for B cell malignancies and solid tumors
1,2 . The expression of memory-associated genes such as TCF7 (protein name TCF1) is linked to response and long-term persistence in patients3-7 , thereby implicating memory programs in therapeutic efficacy. Here, we demonstrate that the pioneer transcription factor, FOXO1, is responsible for promoting memory programs and restraining exhaustion in human CAR T cells. Pharmacologic inhibition or gene editing of endogenous FOXO1 in human CAR T cells diminished the expression of memory-associated genes, promoted an exhaustion-like phenotype, and impaired antitumor activity in vitro and in vivo . FOXO1 overexpression induced a gene expression program consistent with T cell memory and increased chromatin accessibility at FOXO1 binding motifs. FOXO1-overexpressing cells retained function, memory potential, and metabolic fitness during settings of chronic stimulation and exhibited enhanced persistence and antitumor activity in vivo . In contrast, TCF1 overexpression failed to enforce canonical memory programs or enhance CAR T cell potency. Importantly, endogenous FOXO1 activity correlated with CAR T and TIL responses in patients, underscoring its clinical relevance in cancer immunotherapy. Our results demonstrate that memory reprogramming through FOXO1 can enhance the persistence and potency of human CAR T cells and highlights the utility of pioneer factors, which bind condensed chromatin and induce local epigenetic remodeling, for optimizing therapeutic T cell states., Competing Interests: Competing interests: C.A.L. is a consultant to Cartography Biosciences. S.A.G. receives research funding from Novartis, Kite, Vertex, and Servier and consults for Novartis, Roche, GSK, Humanigen, CBMG, Eureka, Janssen/JNJ, and Jazz Pharmaceuticals and has advised for Novartis, Adaptimmune, TCR2, Cellctis, Juno, Vertex, Allogene, Jazz Pharmaceuticals, and Cabaletta. J.A.F. receives research funding from Tceleron (formerly Tmunity Therapeutics) and Danaher Corporation and consults for Retro Biosciences, and is a member of the Scientific Advisory Boards of Cartography Bio and Shennon Biotechnologies Inc. A.T.S. is a founder of Immunai and Cartography Biosciences and receives research funding from Allogene Therapeutics and Merck Research Laboratories. C.L.M. is a co-founder of and holds equity in Link Cell Therapies, is a co-founder of and holds equity in Cargo Therapeutics (formerly Syncopation Life Sciences), is a co-founder of and holds equity in Lyell Immunopharma, holds equity and consults for Mammoth and Ensoma, consults for Immatics, Nektar, and receives research funding from Tune Therapeutics. E.W.W. is a consultant for and holds equity in Lyell Immunopharma and consults for Umoja Immunopharma.- Published
- 2023
- Full Text
- View/download PDF
4. Label-free in vitro assays predict the potency of anti-disialoganglioside chimeric antigen receptor T-cell products.
- Author
-
Logun M, Colonna MB, Mueller KP, Ventarapragada D, Rodier R, Tondepu C, Piscopo NJ, Das A, Chvatal S, Hayes HB, Capitini CM, Brat DJ, Kotanchek T, Edison AS, Saha K, and Karumbaiah L
- Subjects
- Humans, CD8-Positive T-Lymphocytes, Antibodies, Cytokines, Immunotherapy, Adoptive methods, Receptors, Antigen, T-Cell, Receptors, Chimeric Antigen genetics, Glioblastoma
- Abstract
Background Aims: Chimeric antigen receptor (CAR) T cells have demonstrated remarkable efficacy against hematological malignancies; however, they have not experienced the same success against solid tumors such as glioblastoma (GBM). There is a growing need for high-throughput functional screening platforms to measure CAR T-cell potency against solid tumor cells., Methods: We used real-time, label-free cellular impedance sensing to evaluate the potency of anti-disialoganglioside (GD2) targeting CAR T-cell products against GD2+ patient-derived GBM stem cells over a period of 2 days and 7 days in vitro. We compared CAR T products using two different modes of gene transfer: retroviral transduction and virus-free CRISPR-editing. Endpoint flow cytometry, cytokine analysis and metabolomics data were acquired and integrated to create a predictive model of CAR T-cell potency., Results: Results indicated faster cytolysis by virus-free CRISPR-edited CAR T cells compared with retrovirally transduced CAR T cells, accompanied by increased inflammatory cytokine release, CD8+ CAR T-cell presence in co-culture conditions and CAR T-cell infiltration into three-dimensional GBM spheroids. Computational modeling identified increased tumor necrosis factor α concentrations with decreased glutamine, lactate and formate as being most predictive of short-term (2 days) and long-term (7 days) CAR T cell potency against GBM stem cells., Conclusions: These studies establish impedance sensing as a high-throughput, label-free assay for preclinical potency testing of CAR T cells against solid tumors., Competing Interests: Declaration of Competing Interest KS is a member of the scientific advisory boards of Andson Biotech and Notch Therapeutics and receives research support from Synthego Corporation, Spotlight Therapeutics, and the Center for Cell Manufacturing Technologies. LK received in-kind contributions of assay consumables from Axion Biosystems Inc., and research support from the Center for Cell Manufacturing Technologies. SC is employed by Axion Biosystems Inc. TK is owner and employee of Evolved Analytics LLC., licensor of DataModelor., (Copyright © 2023 International Society for Cell & Gene Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. Production and characterization of virus-free, CRISPR-CAR T cells capable of inducing solid tumor regression.
- Author
-
Mueller KP, Piscopo NJ, Forsberg MH, Saraspe LA, Das A, Russell B, Smerchansky M, Cappabianca D, Shi L, Shankar K, Sarko L, Khajanchi N, La Vonne Denne N, Ramamurthy A, Ali A, Lazzarotto CR, Tsai SQ, Capitini CM, and Saha K
- Subjects
- Humans, Gangliosides metabolism, Xenograft Model Antitumor Assays, Receptors, Antigen, T-Cell, Antigens, CD19, T-Lymphocytes, Immunotherapy, Adoptive, Neuroblastoma pathology
- Abstract
Background: Chimeric antigen receptor (CAR) T cells have demonstrated high clinical response rates against hematological malignancies (e.g., CD19+ cancers) but have shown limited activity in patients with solid tumors. Recent work showed that precise insertion of a CAR at a defined locus improves treatment outcomes in the context of a CD19 CAR; however, it is unclear if such a strategy could also affect outcomes in solid tumors. Furthermore, CAR manufacturing generally relies on viral vectors for gene delivery, which comprise a complex and resource-intensive part of the manufacturing supply chain., Methods: Anti-GD2 CAR T cells were generated using CRISPR/Cas9 within 9 days using recombinant Cas9 protein and nucleic acids, without any viral vectors. The CAR was specifically targeted to the T cell receptor alpha constant gene ( TRAC ). T cell products were characterized at the level of the genome, transcriptome, proteome, and secretome using CHANGE-seq, targeted next-generation sequencing, scRNA-seq, spectral cytometry, and ELISA assays, respectively. Functionality was evaluated in vivo in an NSG™ xenograft neuroblastoma model., Results: In comparison to retroviral CAR T cells, virus-free CRISPR CAR (VFC-CAR) T cells exhibit TRAC -targeted genomic integration of the CAR transgene, elevation of transcriptional and protein characteristics associated with a memory-like phenotype, and low tonic signaling prior to infusion arising in part from the knockout of the T cell receptor. On exposure to the GD2 target antigen, anti-GD2 VFC-CAR T cells exhibit specific cytotoxicity against GD2+ cells in vitro and induce solid tumor regression in vivo . VFC-CAR T cells demonstrate robust homing and persistence and decreased exhaustion relative to retroviral CAR T cells against a human neuroblastoma xenograft model., Conclusions: This study leverages virus-free genome editing technology to generate CAR T cells featuring a TRAC -targeted CAR, which could inform manufacturing of CAR T cells to treat cancers, including solid tumors., Competing Interests: Competing interests: KPM, NJP, MHF, LAS, AD, CMC and KSa are inventors on a patent application related to this manuscript. CMC receives honoraria for advisory board membership for Bayer, Elephas Bioscience, Nektar Therapeutics and Novartis. No other conflicts of interest are reported., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.