161 results on '"Melenhorst JJ"'
Search Results
2. Flow cytometric quantitation and characterization of the T-lymphocyte memory response to CMV in healthy donors.
- Author
-
Hensel, N, Melenhorst, JJ, Bradstock, K, Schwarer, AP, Eniafe, R, Nakamura, R, and Barrett, AJ
- Subjects
- *
FLOW cytometry , *CYTOLOGICAL techniques , *T cells , *CYTOMEGALOVIRUSES , *INTERFERONS - Abstract
Background Levels of circulating CMV Ag-specific lymphocytes determine CMV reactivation risk in immunocompromised individuals. Methods Frequencies of T cells producing cytokines after stimulation by CMV Ag were measured in hematopoietic stem-cell donors using flow cytometry. Results In seropositive individuals (n = 75) the mean number of CD8[sup +] (CD8[sup bright], CD8[sup dim]) and CD4[sup +] cells producing IFN-γ was respectively 3.1% (12.6/μL) and 0.38% (3.2/μL), over 10-fold higher than in seronegative subjects (n = 22). CMV stimulation induced tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) in both CD4[sup +] and CD8[sup +] cells (usually together), with a shift from memory- to effector-cell phenotype, while only a small proportion of CD4[sup +] cells produced IL-4. Although the normal range was wide, neither age, sex nor HLA type affected the frequency. Discussion These quantitative studies and the recognition of CD4[sup +] cells as potential effectors of CMV immunity are of relevance for immunotherapeutic approaches to prevent CMV disease after stem-cell transplantation. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
3. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells
- Author
-
Fraietta, JA, primary, Nobels, CL, additional, Sammons, MA, additional, Lundh, S, additional, Carty, SA, additional, Reich, TJ, additional, Cogdill, AP, additional, Morrissette, JJD, additional, DeNizio, JE, additional, Reddy, S, additional, Hwang, Y, additional, Gohil, M, additional, Kulikovskaya, I, additional, Nazimuddin, F, additional, Gupta, M, additional, Chen, F, additional, Everett, JK, additional, Alexander, KA, additional, Lin-Shiao, E, additional, Gee, MH, additional, Liu, X, additional, Young, RM, additional, Ambrose, D, additional, Wang, Y, additional, Xu, J, additional, Jordan, MS, additional, Marcucci, KT, additional, Levine, BL, additional, Garcia, KC, additional, Zhao, Y, additional, Kalos, M, additional, Porter, DL, additional, Kohli, RM, additional, Lacey, SF, additional, Berger, SL, additional, Bushman, FD, additional, June, CH, additional, and Melenhorst, JJ, additional
- Full Text
- View/download PDF
4. Induction of Resistance to Chimeric Antigen Receptor (CAR) T cell Therapy by Transduction of a Single Leukemic B Cell
- Author
-
Ruella, M, primary, Xu, J, additional, Barrett, DM, additional, Kulikovskaya, I, additional, Nazimuddin, F, additional, Bhoj, VG, additional, Orlando, E, additional, Fry, TJ, additional, Bitter, H, additional, Maude, SL, additional, Levine, BL, additional, Nobles, CL, additional, Bushman, FD, additional, Young, RM, additional, Scholler, J, additional, Gill, SI, additional, June, CH, additional, Grupp, SA, additional, Lacey, SF, additional, and Melenhorst, JJ, additional
- Full Text
- View/download PDF
5. Single-cell CAR T atlas reveals type 2 function in 8-year leukaemia remission.
- Author
-
Bai Z, Feng B, McClory SE, de Oliveira BC, Diorio C, Gregoire C, Tao B, Yang L, Zhao Z, Peng L, Sferruzza G, Zhou L, Zhou X, Kerr J, Baysoy A, Su G, Yang M, Camara PG, Chen S, Tang L, June CH, Melenhorst JJ, Grupp SA, and Fan R
- Subjects
- Animals, Child, Humans, Mice, Cytokines blood, Cytokines metabolism, Disease Models, Animal, Epigenomics, Gene Expression Profiling, Interleukin-4 metabolism, Proteomics, Recurrence, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes transplantation, Time Factors, Mice, Inbred NOD, Mice, SCID, Immunotherapy, Adoptive, Multiomics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Receptors, Chimeric Antigen therapeutic use, Remission Induction, Single-Cell Analysis
- Abstract
Despite a high response rate in chimeric antigen receptor (CAR) T cell therapy for acute lymphocytic leukaemia (ALL)
1-3 , approximately 50% of patients relapse within the first year4-6 , representing an urgent question to address in the next stage of cellular immunotherapy. Here, to investigate the molecular determinants of ultralong CAR T cell persistence, we obtained a single-cell multi-omics atlas from 695,819 pre-infusion CAR T cells at the basal level or after CAR-specific stimulation from 82 paediatric patients with ALL enrolled in the first two CAR T ALL clinical trials and 6 healthy donors. We identified that elevated type 2 functionality in CAR T infusion products is significantly associated with patients maintaining a median B cell aplasia duration of 8.4 years. Analysis of ligand-receptor interactions revealed that type 2 cells regulate a dysfunctional subset to maintain whole-population homeostasis, and the addition of IL-4 during antigen-specific activation alleviates CAR T cell dysfunction while enhancing fitness at both transcriptomic and epigenomic levels. Serial proteomic profiling of sera after treatment revealed a higher level of circulating type 2 cytokines in 5-year or 8-year relapse-free responders. In a leukaemic mouse model, type 2high CAR T cell products demonstrated superior expansion and antitumour activity, particularly after leukaemia rechallenge. Restoring antitumour efficacy in type 2low CAR T cells was attainable by enhancing their type 2 functionality, either through incorporating IL-4 into the manufacturing process or by priming manufactured CAR T products with IL-4 before infusion. Our findings provide insights into the mediators of durable CAR T therapy response and suggest potential therapeutic strategies to sustain long-term remission by boosting type 2 functionality in CAR T cells., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
6. The type 2 cytokine Fc-IL-4 revitalizes exhausted CD8 + T cells against cancer.
- Author
-
Feng B, Bai Z, Zhou X, Zhao Y, Xie YQ, Huang X, Liu Y, Enbar T, Li R, Wang Y, Gao M, Bonati L, Peng MW, Li W, Tao B, Charmoy M, Held W, Melenhorst JJ, Fan R, Guo Y, and Tang L
- Subjects
- Animals, Female, Humans, Mice, Cell Line, Tumor, Glycolysis drug effects, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Immunotherapy, Adoptive, Lactate Dehydrogenase 5 metabolism, Mice, Inbred C57BL, NAD metabolism, Signal Transduction drug effects, STAT6 Transcription Factor metabolism, TOR Serine-Threonine Kinases metabolism, Xenograft Model Antitumor Assays, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Immunoglobulin Fc Fragments genetics, Immunoglobulin Fc Fragments immunology, Immunoglobulin Fc Fragments metabolism, Immunotherapy methods, Interleukin-4 genetics, Interleukin-4 immunology, Interleukin-4 pharmacology, Interleukin-4 therapeutic use, Neoplasms drug therapy, Neoplasms immunology, Neoplasms therapy, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Recombinant Fusion Proteins pharmacology, Recombinant Fusion Proteins therapeutic use
- Abstract
Current cancer immunotherapy predominately focuses on eliciting type 1 immune responses fighting cancer; however, long-term complete remission remains uncommon
1,2 . A pivotal question arises as to whether type 2 immunity can be orchestrated alongside type 1-centric immunotherapy to achieve enduring response against cancer3,4 . Here we show that an interleukin-4 fusion protein (Fc-IL-4), a typical type 2 cytokine, directly acts on CD8+ T cells and enriches functional terminally exhausted CD8+ T (CD8+ TTE ) cells in the tumour. Consequently, Fc-IL-4 enhances antitumour efficacy of type 1 immunity-centric adoptive T cell transfer or immune checkpoint blockade therapies and induces durable remission across several syngeneic and xenograft tumour models. Mechanistically, we discovered that Fc-IL-4 signals through both signal transducer and activator of transcription 6 (STAT6) and mammalian target of rapamycin (mTOR) pathways, augmenting the glycolytic metabolism and the nicotinamide adenine dinucleotide (NAD) concentration of CD8+ TTE cells in a lactate dehydrogenase A-dependent manner. The metabolic modulation mediated by Fc-IL-4 is indispensable for reinvigorating intratumoural CD8+ TTE cells. These findings underscore Fc-IL-4 as a potent type 2 cytokine-based immunotherapy that synergizes effectively with type 1 immunity to elicit long-lasting responses against cancer. Our study not only sheds light on the synergy between these two types of immune responses, but also unveils an innovative strategy for advancing next-generation cancer immunotherapy by integrating type 2 immune factors., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
7. Advancing Cholangiocarcinoma Care: Insights and Innovations in T Cell Therapy.
- Author
-
Dadgar N, Arunachalam AK, Hong H, Phoon YP, Arpi-Palacios JE, Uysal M, Wehrle CJ, Aucejo F, Ma WW, and Melenhorst JJ
- Abstract
Cholangiocarcinoma (CCA) is a rare and aggressive malignancy originating from the bile ducts, with poor prognosis and limited treatment options. Traditional therapies, such as surgery, chemotherapy, and radiation, have shown limited efficacy, especially in advanced cases. Recent advancements in immunotherapy, particularly T cell-based therapies like chimeric antigen receptor T (CAR T) cells, tumor-infiltrating lymphocytes (TILs), and T cell receptor (TCR)-based therapies, have opened new avenues for improving outcomes in CCA. This review provides a comprehensive overview of the current state of T cell therapies for CCA, focusing on CAR T cell therapy. It highlights key challenges, including the complex tumor microenvironment and immune evasion mechanisms, and the progress made in preclinical and clinical trials. The review also discusses ongoing clinical trials targeting specific CCA antigens, such as MUC1, EGFR, and CD133, and the evolving role of precision immunotherapy in enhancing treatment outcomes. Despite significant progress, further research is needed to optimize these therapies for solid tumors like CCA. By summarizing the most recent clinical results and future directions, this review underscores the promising potential of T cell therapies in revolutionizing CCA treatment.
- Published
- 2024
- Full Text
- View/download PDF
8. Advancing CAR T-cell therapies: Preclinical insights and clinical translation for hematological malignancies.
- Author
-
Arunachalam AK, Grégoire C, Coutinho de Oliveira B, and Melenhorst JJ
- Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved significant success in achieving durable and potentially curative responses in patients with hematological malignancies. CARs are tailored fusion proteins that direct T cells to a specific antigen on tumor cells thereby eliciting a targeted immune response. The approval of several CD19-targeted CAR T-cell therapies has resulted in a notable surge in clinical trials involving CAR T cell therapies for hematological malignancies. Despite advancements in understanding response mechanisms, resistance patterns, and adverse events associated with CAR T-cell therapy, the translation of these insights into robust clinical efficacy has shown modest outcomes in both clinical trials and real-world scenarios. Therefore, the assessment of CAR T-cell functionality through rigorous preclinical studies plays a pivotal role in refining therapeutic strategies for clinical applications. This review provides an overview of the various in vitro and animal models used to assess the functionality of CAR T-cells. We discuss the findings from preclinical research involving approved CAR T-cell products, along with the implications derived from recent preclinical studies aiming to optimize the functionality of CAR T-cells. The review underscores the importance of robust preclinical evaluations and the need for models that accurately replicate human disease to bridge the gap between preclinical success and clinical efficacy., Competing Interests: Declaration of competing interest Dr. Melenhorst has patents related to Biomarkers and manufacturing of chimeric antigen receptor-engineered T cells. The remaining authors declare no conflicts of interest., (Copyright © 2024. Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
9. Chimeric antigen receptor T-cell therapy for haematological malignancies: Insights from fundamental and translational research to bedside practice.
- Author
-
Grégoire C, Coutinho de Oliveira B, Caimi PF, Caers J, and Melenhorst JJ
- Abstract
Autologous chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of lymphoid malignancies, leading to the approval of CD19-CAR T cells for B-cell lymphomas and acute leukaemia, and more recently, B-cell maturation antigen-CAR T cells for multiple myeloma. The long-term follow-up of patients treated in the early clinical trials demonstrates the possibility for long-term remission, suggesting a cure. This is associated with a low incidence of significant long-term side effects and a rapid improvement in the quality of life for responders. In contrast, other types of immunotherapies require prolonged treatments or carry the risk of long-term side effects impairing the quality of life. Despite impressive results, some patients still experience treatment failure or ultimately relapse, underscoring the imperative to improve CAR T-cell therapies and gain a better understanding of their determinants of efficacy to maximize positive outcomes. While the next-generation of CAR T cells will undoubtingly be more potent, there are already opportunities for optimization when utilizing the currently available CAR T cells. This review article aims to summarize the current evidence from clinical, translational and fundamental research, providing clinicians with insights to enhance their understanding and use of CAR T cells., (© 2024 The Author(s). British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
10. T-cell dysfunction in CLL is mediated through expression of Siglec-10 ligands CD24 and CD52 on CLL cells.
- Author
-
van Bruggen JAC, Peters FS, Mes M, Rietveld JM, Cerretani E, Cretenet G, van Kampen R, Jongejan A, Moerland PD, Melenhorst JJ, van der Windt GJW, Eldering E, and Kater AP
- Subjects
- Humans, Lymphocyte Activation immunology, Ligands, Receptors, Chimeric Antigen metabolism, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell therapy, CD52 Antigen metabolism, T-Lymphocytes metabolism, T-Lymphocytes immunology, CD24 Antigen metabolism
- Abstract
Abstract: Autologous T-cell-based therapies, such as chimeric antigen receptor (CAR) T-cell therapy, exhibit low success rates in chronic lymphocytic leukemia (CLL) and correlate with a dysfunctional T-cell phenotype observed in patients. Despite various proposed mechanisms of T-cell dysfunction in CLL, the specific CLL-derived factors responsible remain unidentified. This study aimed to investigate the mechanisms through which CLL cells suppress CAR T-cell activation and function. We found that CLL-derived T cells get activated, albeit in a delayed fashion, and specifically that restimulation of CAR T cells in the presence of CLL cells causes impaired cytokine production and reduced proliferation. Notably, coculture of T cells with CD40-activated CLL cells did not lead to T-cell dysfunction, and this required direct cell contact between the CD40-stimulated CLL cells and T cells. Inhibition of kinases involved in the CD40 signaling cascade revealed that the Spare Respiratory Capacity (SRC) kinase inhibitor dasatinib prevented rescue of T-cell function independent of CD40-mediated increased levels of costimulatory and adhesion ligands on CLL cells. Transcriptome profiling of CD40-stimulated CLL cells with or without dasatinib identified widespread differential gene expression. Selecting for surface receptor genes revealed CD40-mediated downregulation of the Sialic acid-binding Ig-like lectin 10 (Siglec-10) ligands CD24 and CD52, which was prevented by dasatinib, suggesting a role for these ligands in functional T-cell suppression in CLL. Indeed, blocking CD24 and/or CD52 markedly reduced CAR T-cell dysfunction upon coculture with resting CLL cells. These results demonstrated that T cells derived from CLL patients can be reinvigorated by manipulating CLL-T-cell interactions. Targeting CD24- and CD52-mediated CLL-T-cell interaction could be a promising therapeutic strategy to enhance T-cell function in CLL., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Autologous human preclinical modeling of melanoma interpatient clinical responses to immunotherapeutics.
- Author
-
Phoon YP, Lopes JE, Pfannenstiel LW, Marcela Diaz-Montero C, Tian YF, Ernstoff MS, Funchain P, Ko JS, Winquist R, Losey HC, Melenhorst JJ, and Gastman BR
- Subjects
- Humans, Animals, Mice, Autografts, Mice, Inbred NOD, Melanoma therapy, Xenograft Model Antitumor Assays, Immune Checkpoint Inhibitors administration & dosage, Immune Checkpoint Inhibitors therapeutic use, Interleukin-2 administration & dosage, Interleukin-2 therapeutic use, Skin Neoplasms therapy, Immunotherapy methods
- Abstract
Background: Despite recent advances in immunotherapy, a substantial population of late-stage melanoma patients still fail to achieve sustained clinical benefit. Lack of translational preclinical models continues to be a major challenge in the field of immunotherapy; thus, more optimized translational models could strongly influence clinical trial development. To address this unmet need, we designed a preclinical model reflecting the heterogeneity in melanoma patients' clinical responses that can be used to evaluate novel immunotherapies and synergistic combinatorial treatment strategies. Using our all-autologous humanized melanoma mouse model, we examined the efficacy of a novel engineered interleukin 2 (IL-2)-based cytokine variant immunotherapy., Methods: To study immune responses and antitumor efficacy for human melanoma tumors, we developed an all-autologous humanized melanoma mouse model using clinically annotated, matched patient tumor cells and peripheral blood mononuclear cells (PBMCs). After inoculating immunodeficient NSG mice with patient tumors and an adoptive cell transfer of autologous PBMCs, mice were treated with anti-PD-1, a novel investigational engineered IL-2-based cytokine (nemvaleukin), or recombinant human IL-2 (rhIL-2). The pharmacodynamic effects and antitumor efficacy of these treatments were then evaluated. We used tumor cells and autologous PBMCs from patients with varying immunotherapy responses to both model the diversity of immunotherapy efficacy observed in the clinical setting and to recapitulate the heterogeneous nature of melanoma., Results: Our model exhibited long-term survival of engrafted human PBMCs without developing graft-versus-host disease. Administration of an anti-PD-1 or nemvaleukin elicited antitumor responses in our model that were patient-specific and were found to parallel clinical responsiveness to checkpoint inhibitors. An evaluation of nemvaleukin-treated mice demonstrated increased tumor-infiltrating CD4
+ and CD8+ T cells, preferential expansion of non-regulatory T cell subsets in the spleen, and significant delays in tumor growth compared with vehicle-treated controls or mice treated with rhIL-2., Conclusions: Our model reproduces differential effects of immunotherapy in melanoma patients, capturing the inherent heterogeneity in clinical responses. Taken together, these data demonstrate our model's translatability for novel immunotherapies in melanoma patients. The data are also supportive for the continued clinical investigation of nemvaleukin as a novel immunotherapeutic for the treatment of melanoma., Competing Interests: Competing interests: JL, RW and HL hold stock in Alkermes. Research performed at the Cleveland Clinic in the laboratory of BRG was funded by Alkermes., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
12. Allogeneic CAR T Cells: Complex Cellular Therapy Designs Test the Limits of Our Preclinical Models.
- Author
-
Caimi PF and Melenhorst JJ
- Subjects
- Humans, Receptors, Antigen, T-Cell genetics, T-Lymphocytes, Immunotherapy, Adoptive methods, Multiple Myeloma genetics, Multiple Myeloma therapy, Hematopoietic Stem Cell Transplantation
- Abstract
All chimeric antigen receptor (CAR) T-cell products currently approved by the FDA are autologous, which poses several challenges for widespread use. In this issue, Degagné and colleagues present their preclinical research on creating off-the-shelf CAR T cells for multiple myeloma. They utilized the CRISPR/Cas12a genome editing platform and gene knock-in techniques to eliminate alloreactivity and decrease susceptibility to natural killer (NK)-cell elimination. This work has led to an ongoing phase I trial of off-the-shelf CAR T cells for multiple myeloma treatment. See related article by Degagné et al., p. 462 (2)., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
13. Key role of CD4+ T cells in determining CD8 function during CAR-T cell manufacture.
- Author
-
Melenhorst JJ and Oliveira BC
- Subjects
- Humans, Receptors, Antigen, T-Cell, CD8-Positive T-Lymphocytes, CD4-Positive T-Lymphocytes, Receptors, Chimeric Antigen genetics
- Abstract
Competing Interests: Competing interests: No, there are no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
14. Two cases of severe pulmonary toxicity from highly active mesothelin-directed CAR T cells.
- Author
-
Haas AR, Golden RJ, Litzky LA, Engels B, Zhao L, Xu F, Taraszka JA, Ramones M, Granda B, Chang WJ, Jadlowsky J, Shea KM, Runkle A, Chew A, Dowd E, Gonzalez V, Chen F, Liu X, Fang C, Jiang S, Davis MM, Sheppard NC, Zhao Y, Fraietta JA, Lacey SF, Plesa G, Melenhorst JJ, Mansfield K, Brogdon JL, Young RM, Albelda SM, June CH, and Tanyi JL
- Subjects
- Humans, GPI-Linked Proteins genetics, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, T-Lymphocytes, Mesothelin, Neoplasms therapy
- Abstract
Multiple clinical studies have treated mesothelin (MSLN)-positive solid tumors by administering MSLN-directed chimeric antigen receptor (CAR) T cells. Although these products are generally safe, efficacy is limited. Therefore, we generated and characterized a potent, fully human anti-MSLN CAR. In a phase 1 dose-escalation study of patients with solid tumors, we observed two cases of severe pulmonary toxicity following intravenous infusion of this product in the high-dose cohort (1-3 × 10
8 T cells per m2 ). Both patients demonstrated progressive hypoxemia within 48 h of infusion with clinical and laboratory findings consistent with cytokine release syndrome. One patient ultimately progressed to grade 5 respiratory failure. An autopsy revealed acute lung injury, extensive T cell infiltration, and accumulation of CAR T cells in the lungs. RNA and protein detection techniques confirmed low levels of MSLN expression by benign pulmonary epithelial cells in affected lung and lung samples obtained from other inflammatory or fibrotic conditions, indicating that pulmonary pneumocyte and not pleural expression of mesothelin may lead to dose-limiting toxicity. We suggest patient enrollment criteria and dosing regimens of MSLN-directed therapies consider the possibility of dynamic expression of mesothelin in benign lung with a special concern for patients with underlying inflammatory or fibrotic conditions., Competing Interests: Declaration of interests C.H.J., R.M.Y., and M.M.D. are inventors on patents and/or patent applications licensed to Novartis Institutes of Biomedical Research and receive license revenue from such licenses. J.L.B., B.E., K.M., and L.Z. are holders of stock options and patents with Novartis Institutes for Biomedical Research. R.M.Y. and M.M.D. are inventors on patents and/or patent applications licensed to Tmunity Therapeutics and receive license revenue from such licenses. C.H.J. and A.C. are scientific cofounders of Tmunity Therapeutics. C.H.J. is a scientific cofounder of Capstan Therapeutics and is a member of the scientific advisory boards of AC Immune, Alaunos, BluesphereBio, Cabaletta, Carisma, Cartography, Cellares, Celldex, Decheng, Poseida, Verismo, and WIRB-Copernicus. B.E. and R.J.G. completed work on this study before becoming employees at Miltenyi Biotec and AstraZeneca, respectively. R.J.G. holds or may hold AstraZeneca stock. N.C.S. holds equity in Fate Therapeutics and Pfizer. J.A.F. has received grants and personal fees from Cartography Bio., grants from Tmunity Therapeutics, and personal fees from Retro Bio and Shennon Bio outside the submitted work. Additionally, J.A.F. holds patents related to CAR T cells for cancer that are licensed and associated with royalties. S.F.L. is an inventor on patents in the areas of CAR T and biomarkers at Penn that were assigned to Novartis; received research funding from Novartis, Tmunity, and Cabaletta; and consults for Kite/Gilead. M.M.D. is a consultant for Tmunity Therapeutics and is a Consultant and Member on the Scientific Advisory Board of Cellares Corporation., (Copyright © 2023 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. Positively charged patches: tonic for CAR fitness.
- Author
-
Khan MH and Melenhorst JJ
- Subjects
- Signal Transduction
- Published
- 2023
- Full Text
- View/download PDF
16. Spatial immunosampling of MRI-defined glioblastoma regions reveals immunologic fingerprint of non-contrast enhancing, infiltrative tumor margins.
- Author
-
Grabowski MM, Watson DC, Chung K, Lee J, Bayik D, Lauko A, Alban T, Melenhorst JJ, Chan T, Lathia JD, Ahluwalia MS, and Mohammadi AM
- Abstract
Glioblastoma (GBM) treatment includes maximal safe resection of the core and MRI contrast-enhancing (CE) tumor. Complete resection of the infiltrative non-contrast-enhancing (NCE) tumor rim is rarely achieved. We established a safe, semi-automated workflow for spatially-registered sampling of MRI-defined GBM regions in 19 patients with downstream analysis and biobanking, enabling studies of NCE, wherefrom recurrence/progression typically occurs. Immunophenotyping revealed underrepresentation of myeloid cell subsets and CD8+ T cells in the NCE. While NCE T cells phenotypically and functionally resembled those in matching CE tumor, subsets of activated (CD69
hi ) effector memory CD8+ T cells were overrepresented. Contrarily, CD25hi Tregs and other subsets were underrepresented. Overall, our study demonstrated that MRI-guided, spatially-registered, intraoperative immunosampling is feasible as part of routine GBM surgery. Further elucidation of the shared and spatially distinct microenvironmental biology of GBM will enable development of therapeutic approaches targeting the NCE infiltrative tumor to decrease GBM recurrence., Competing Interests: NICO Corporation provided the NICO Myriad and Tissue Preservation System devices at no cost. MSA declares the following: Research funding from Seagen; Consultation for Bayer, Novocure, Kiyatec, Insightec, GSK, Xoft, Nuvation, Cellularity, SDP Oncology, Apollomics, Prelude, Janssen, Tocagen, Voyager Therapeutics, Viewray, Caris Lifesciences, Pyramid Biosciences, Anheart Therapeutics, Varian Medical Systems, Theraguix; Scientific Advisory Board for Cairn Therapeutics, Pyramid Biosciences, Modifi Biosciences; Stock shareholder of Mimivax, Cytodyn, MedInnovate Advisors LLC. The authors have declared that no other potential conflict of interest exists.- Published
- 2023
- Full Text
- View/download PDF
17. Challenges and opportunities of CAR T-cell therapies for CLL.
- Author
-
Zhao Z, Grégoire C, Oliveira B, Chung K, and Melenhorst JJ
- Subjects
- Humans, Immunotherapy, Adoptive, T-Lymphocytes, Treatment Outcome, Leukemia, Lymphocytic, Chronic, B-Cell therapy
- Abstract
Chimeric antigen receptor (CAR) T-cell therapies have transformed the treatment landscape of blood cancers. These engineered receptors which endow T cells with antibody-like target cell recognition combined with the typical T cell target cell lysis abilities. Introduced into the clinic in the 2010s, CAR T-cells have shown efficacy in chronic B lymphocytic leukemia (CLL), but a majority of patients do not achieve sustained remission. Here we discuss the current treatment landscape in CLL using small molecules and allogeneic stem cell transplantation, the niche CAR T-cells filled in this context, and what we have learned from biomarker and mechanistic studies. Several product parameters and improvements are introduced as examples of how the bedside-to-bench is translated into improved CAR T-cells for CLL. We hope to convey to our readers the crucial role translational medicine plays in transforming the treatment outcomes for patients with CLL and how this line of research is an essential component of modern medicine., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Author Correction: Decade-long leukaemia remissions with persistence of CD4 + CAR T cells.
- Author
-
Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, Gao P, Bandyopadhyay S, Sun H, Zhao Z, Lundh S, Pruteanu-Malinici I, Nobles CL, Maji S, Frey NV, Gill SI, Loren AW, Tian L, Kulikovskaya I, Gupta M, Ambrose DE, Davis MM, Fraietta JA, Brogdon JL, Young RM, Chew A, Levine BL, Siegel DL, Alanio C, Wherry EJ, Bushman FD, Lacey SF, Tan K, and June CH
- Published
- 2022
- Full Text
- View/download PDF
19. Anti-CD19 CAR T cells in combination with ibrutinib for the treatment of chronic lymphocytic leukemia.
- Author
-
Gill S, Vides V, Frey NV, Hexner EO, Metzger S, O'Brien M, Hwang WT, Brogdon JL, Davis MM, Fraietta JA, Gaymon AL, Gladney WL, Lacey SF, Lamontagne A, Mato AR, Maus MV, Melenhorst JJ, Pequignot E, Ruella M, Shestov M, Byrd JC, Schuster SJ, Siegel DL, Levine BL, June CH, and Porter DL
- Subjects
- Humans, Antigens, CD19, Disease-Free Survival, Neoplasm, Residual drug therapy, Prospective Studies, Pyrazoles therapeutic use, Pyrimidines therapeutic use, T-Lymphocytes, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy
- Abstract
In chronic lymphocytic leukemia (CLL) patients who achieve a complete remission (CR) to anti-CD19 chimeric antigen receptor T cells (CART-19), remissions are remarkably durable. Preclinical data suggesting synergy between CART-19 and the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib prompted us to conduct a prospective single-center phase 2 trial in which we added autologous anti-CD19 humanized binding domain T cells (huCART-19) to ibrutinib in patients with CLL not in CR despite ≥6 months of ibrutinib. The primary endpoints were safety, feasibility, and achievement of a CR within 3 months. Of 20 enrolled patients, 19 received huCART-19. The median follow-up for all infused patients was 41 months (range, 0.25-58 months). Eighteen patients developed cytokine release syndrome (CRS; grade 1-2 in 15 of 18 subjects), and 5 developed neurotoxicity (grade 1-2 in 4 patients, grade 4 in 1 patient). While the 3-month CR rate among International Working Group on CLL (iwCLL)-evaluable patients was 44% (90% confidence interval [CI], 23-67%), at 12 months, 72% of patients tested had no measurable residual disease (MRD). The estimated overall and progression-free survival at 48 months were 84% and 70%, respectively. Of 15 patients with undetectable MRD at 3 or 6 months, 13 remain in ongoing CR at the last follow-up. In patients with CLL not achieving a CR despite ≥6 months of ibrutinib, adding huCART-19 mediated a high rate of deep and durable remissions. ClinicalTrials.gov number, NCT02640209., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Changes in Bone Marrow Tumor and Immune Cells Correlate with Durability of Remissions Following BCMA CAR T Therapy in Myeloma.
- Author
-
Dhodapkar KM, Cohen AD, Kaushal A, Garfall AL, Manalo RJ, Carr AR, McCachren SS, Stadtmauer EA, Lacey SF, Melenhorst JJ, June CH, Milone MC, and Dhodapkar MV
- Subjects
- Humans, B-Cell Maturation Antigen genetics, Bone Marrow pathology, Neoplasm Recurrence, Local, T-Lymphocytes immunology, Tumor Microenvironment, Receptors, Chimeric Antigen genetics, Multiple Myeloma immunology, Bone Marrow Neoplasms
- Abstract
Chimeric antigen-receptor (CAR) T cells lead to high response rates in myeloma, but most patients experience recurrent disease. We combined several high-dimensional approaches to study tumor/immune cells in the tumor microenvironment (TME) of myeloma patients pre- and post-B-cell maturation antigen (BCMA)-specific CAR T therapy. Lower diversity of pretherapy T-cell receptor (TCR) repertoire, presence of hyperexpanded clones with exhaustion phenotype, and BAFF+PD-L1+ myeloid cells in the marrow correlated with shorter progression-free survival (PFS) following CAR T therapy. In contrast, longer PFS was associated with an increased proportion of CLEC9A+ dendritic cells (DC), CD27+TCF1+ T cells with diverse T-cell receptors, and emergence of T cells expressing marrow-residence genes. Residual tumor cells at initial response express stemlike genes, and tumor recurrence was associated with the emergence of new dominant clones. These data illustrate a dynamic interplay between endogenous T, CAR T, myeloid/DC, and tumor compartments that affects the durability of response following CAR T therapy in myeloma., Significance: There is an unmet need to identify determinants of durable responses following BCMA CAR T therapy of myeloma. High-dimensional analysis of the TME was performed to identify features of immune and tumor cells that correlate with survival and suggest several strategies to improve outcomes following CAR T therapy. See related commentary by Graham and Maus, p. 478. This article is highlighted in the In This Issue feature, p. 476., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
21. Enhanced Costimulatory Signaling Improves CAR T-cell Effector Responses in CLL.
- Author
-
Collins MA, Jung IY, Zhao Z, Apodaca K, Kong W, Lundh S, Fraietta JA, Kater AP, Sun C, Wiestner A, and Melenhorst JJ
- Subjects
- Humans, T-Lymphocytes, B-Lymphocytes, CD40 Ligand genetics, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Receptors, Chimeric Antigen genetics
- Abstract
CD19-redirected chimeric antigen receptor (CAR) T cells have shown remarkable activity against B-cell cancers. While second-generation CARs induce complete remission in >80% of patients with acute lymphoblastic leukemia, similar monotherapy induces long-term remissions in only 26% of patients with chronic lymphocytic leukemia (CLL). This disparity is attributed to cell-intrinsic effector defects in autologous CLL-derived T cells. However, the mechanisms by which leukemic cells impact CAR T-cell potency are poorly understood. Herein we describe an in vitro assay that recapitulates endogenous CLL-mediated T-cell defects in healthy donor CAR T cells. Contact with CLL cells insufficiently activates, but does not irreversibly impair, CAR T-cell function. This state is rescuable by strong antigenic stimulation or IL2, and is not driven by immune suppression. Rather, this activation defect is attributable to low levels of costimulatory molecules on CLL cells, and exogenous costimulation enhanced CAR T-cell activation. We next assessed the stimulatory phenotype of CLL cells derived from different niches within the same patient. Lymph node (LN)-derived CLL cells had a strong costimulatory phenotype and promoted better CAR T-cell degranulation and cytokine production than matched peripheral blood CLL cells. Finally, in vitro CD40L-activated CLL cells acquired a costimulatory phenotype similar to the LN-derived tumor and stimulated improved CAR T-cell proliferation, cytokine production, and cytotoxicity. Together, these data identify insufficient activation as a driver of poor CAR T-cell responses in CLL. The costimulatory phenotype of CLL cells drives differential CAR T-cell responses, and can be augmented by improving costimulatory signaling., Significance: CLL cells insufficiently activate CAR T cells, driven by low levels of costimulatory molecules on the tumor. LN-derived CLL cells are more costimulatory and mediate enhanced CAR T-cell killing. This costimulatory phenotype can be modeled via CD40 L activation, and the activated tumor promotes stronger CAR T-cell responses., Competing Interests: M.A. Collins reports grants from Parker Institute for Cancer Immunotherapy during the conduct of the study. A.P. Kater reports grants and other from Abbvie, AstraZeneca, BMS; and other from Janssen and LAVA outside the submitted work; in addition, A.P. Kater has a patent to Janssen pending and a patent to LAVA pending. C. Sun reports grants from Genmab outside the submitted work. A. Wiestner reports grants from Pharmacyclics, Acerta, Merck, Nurix, Verastem, and Genmab outside the submitted work. J.J. Melenhorst reports grants from Parker Institute for Cancer Immunotherapy during the conduct of the study; personal fees from IASO Biotherapeutics, Poseida Therapeutics, and Kite Pharma outside the submitted work; in addition, J.J. Melenhorst has a patent to Methods for improving the efficacy and expansion of immune cells issued, a patent to Biomarkers predictive of therapeutic responsiveness to chimeric antigen: issued, a patent to Methods of making chimeric antigen receptor - expressing cells issued, a patent to Methods for improving the efficacy and expansion of chimeric antigen receptor: issued, a patent to CAR T-cell therapies with enhanced efficacy issued, and a patent to Biomarkers predictive of cytokine release syndrome issued. No disclosures were reported by the other authors., (© 2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
22. Comprehensive Serum Proteome Profiling of Cytokine Release Syndrome and Immune Effector Cell-Associated Neurotoxicity Syndrome Patients with B-Cell ALL Receiving CAR T19.
- Author
-
Diorio C, Shraim R, Myers R, Behrens EM, Canna S, Bassiri H, Aplenc R, Burudpakdee C, Chen F, DiNofia AM, Gill S, Gonzalez V, Lambert MP, Leahy AB, Levine BL, Lindell RB, Maude SL, Melenhorst JJ, Newman H, Perazzelli J, Seif AE, Lacey SF, June CH, Barrett DM, Grupp SA, and Teachey DT
- Subjects
- Biomarkers, Child, Cytokine Release Syndrome etiology, Cytokines metabolism, Humans, Immunotherapy, Adoptive, Interleukin-18, Proteome, Proteomics, Neurotoxicity Syndromes drug therapy, Neurotoxicity Syndromes etiology, Receptors, Chimeric Antigen
- Abstract
Purpose: To study the biology and identify markers of severe cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) in children after chimeric antigen receptor T-cell (CAR T) treatment., Experimental Design: We used comprehensive proteomic profiling to measure over 1,400 serum proteins at multiple serial timepoints in a cohort of patients with B-cell acute lymphoblastic leukemia treated with the CD19-targeted CAR T CTL019 on two clinical trials., Results: We identified fms-like tyrosine kinase 3 (FLT3) and mast cell immunoglobulin-like receptor 1 (MILR1) as preinfusion predictive biomarkers of severe CRS. We demonstrated that CRS is an IFNγ-driven process with a protein signature overlapping with hemophagocytic lymphohistiocytosis (HLH). We identified IL18 as a potentially targetable cytokine associated with the development of ICANS., Conclusions: We identified preinfusion biomarkers that can be used to predict severe CRS with a sensitivity, specificity, and accuracy superior to the current gold standard of disease burden. We demonstrated the fundamental role of the IFNγ pathway in driving CRS, suggesting CRS and carHLH are overlapping rather than distinct phenomena, an observation with important treatment implications. We identified IL18 as a possible targetable cytokine in ICANS, providing rationale for IL18 blocking therapies to be translated into clinical trials in ICANS., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
23. Adaptable Leukemia Cells Resisting CAR T-cell Attack via B-cell Activation.
- Author
-
Zhao Z and Melenhorst JJ
- Subjects
- Antigens, CD19 immunology, Germinal Center immunology, Humans, Immunotherapy, Adoptive, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology, Lymphoma, B-Cell immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved remarkable milestones in the treatment of B-cell malignancies. However, cancer cells frequently survive CAR T-cell killing in a large cohort of patients. Relapse oftentimes is associated with antigen loss. In this issue, Im and colleagues report a new mechanism of leukemic-cell resistance to anti-CD19 CAR T cells: Leukemic cells can enable a B-cell activation and germinal center reaction signature, which causes CD19 transcriptional downregulation and survival from CAR exposure. See related article by Im et al., p. 1055 (5)., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
24. B cell targeting in CAR T cell therapy: Side effect or driver of CAR T cell function?
- Author
-
Chen GM, Melenhorst JJ, and Tan K
- Subjects
- Antigens, CD19, B-Lymphocytes, Humans, Receptors, Antigen, T-Cell, T-Lymphocytes, Drug-Related Side Effects and Adverse Reactions, Immunotherapy, Adoptive adverse effects
- Abstract
Chimeric antigen receptor (CAR) T cell therapies targeting CD19 and CD22 have been successful for treating B cell cancers, but CAR T cells targeting non-B cell cancers remain unsuccessful. We propose that rather than being strictly a side effect of therapy, the large number of CAR interactions with normal B cells may be a key contributor to clinical CAR T cell responses.
- Published
- 2022
- Full Text
- View/download PDF
25. Single-cell antigen-specific landscape of CAR T infusion product identifies determinants of CD19-positive relapse in patients with ALL.
- Author
-
Bai Z, Woodhouse S, Zhao Z, Arya R, Govek K, Kim D, Lundh S, Baysoy A, Sun H, Deng Y, Xiao Y, Barrett DM, Myers RM, Grupp SA, June CH, Fan R, Camara PG, and Melenhorst JJ
- Subjects
- Antigens, CD19, Humans, Proteomics, Receptors, Chimeric Antigen metabolism, Recurrence, Immunotherapy, Adoptive, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
A notable number of acute lymphoblastic leukemia (ALL) patients develop CD19-positive relapse within 1 year after receiving chimeric antigen receptor (CAR) T cell therapy. It remains unclear if the long-term response is associated with the characteristics of CAR T cells in infusion products, hindering the identification of biomarkers to predict therapeutic outcomes. Here, we present 101,326 single-cell transcriptomes and surface protein landscape from the infusion products of 12 ALL patients. We observed substantial heterogeneity in the antigen-specific activation states, among which a deficiency of T helper 2 function was associated with CD19-positive relapse compared with durable responders (remission, >54 months). Proteomic data revealed that the frequency of early memory T cells, rather than activation or coinhibitory signatures, could distinguish the relapse. These findings were corroborated by independent functional profiling of 49 patients, and an integrative model was developed to predict the response. Our data unveil the molecular mechanisms that may inform strategies to boost specific T cell function to maintain long-term remission.
- Published
- 2022
- Full Text
- View/download PDF
26. PD1 Expression in EGFRvIII-Directed CAR T Cell Infusion Product for Glioblastoma Is Associated with Clinical Response.
- Author
-
Tang OY, Tian L, Yoder T, Xu R, Kulikovskaya I, Gupta M, Melenhorst JJ, Lacey SF, O'Rourke DM, and Binder ZA
- Subjects
- ErbB Receptors, Humans, Neoplasm Recurrence, Local metabolism, T-Lymphocytes, Glioblastoma pathology, Receptors, Chimeric Antigen
- Abstract
The epidermal growth factor receptor variant III (EGFRvIII) has been investigated as a therapeutic target for chimeric antigen receptor (CAR) T cell therapy in glioblastoma. Earlier research demonstrated that phenotypic and genotypic characteristics in T cells and CAR T product predicted therapeutic success in hematologic malignancies, to date no determinants for clinical response in solid tumors have been identified. We analyzed apheresis and infusion products from the first-in-human trial of EGFRvIII-directed CAR T for recurrent glioblastoma (NCT02209376) by flow cytometry. Clinical response was quantified via engraftment in peripheral circulation and progression-free survival (PFS), as determined by the time from CAR T infusion to first radiographic evidence of progression. The CD4
+ CAR T cell population in patient infusion products demonstrated PD1 expression which positively correlated with AUC engraftment and PFS. On immune checkpoint inhibitor analysis, CTLA-4, TIM3, and LAG3 did not exhibit significant associations with engraftment or PFS. The frequencies of PD1+ GZMB+ and PD1+ HLA-DR+ CAR T cells in the CD4+ infusion products were directly proportional to AUC and PFS. No significant associations were observed within the apheresis products. In summary, PD1 in CAR T infusion products predicted peripheral engraftment and PFS in recurrent glioblastoma., Competing Interests: DO’R receives laboratory support from Tmunity Therapeutics. DO’R and ZB are on patents relating to CAR T cell therapy for GBM. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Tang, Tian, Yoder, Xu, Kulikovskaya, Gupta, Melenhorst, Lacey, O’Rourke and Binder.)- Published
- 2022
- Full Text
- View/download PDF
27. Temperature Trajectory Sub-phenotypes and the Immuno-Inflammatory Response in Pediatric Sepsis.
- Author
-
Yehya N, Fitzgerald JC, Hayes K, Zhang D, Bush J, Koterba N, Chen F, Tuluc F, Teachey DT, Balamuth F, Lacey SF, Melenhorst JJ, and Weiss SL
- Subjects
- Biomarkers, Child, Critical Illness, Cytokines, Humans, Phenotype, Prospective Studies, Temperature, Hypothermia, Sepsis
- Abstract
Objective: Heterogeneity has hampered sepsis trials, and sub-phenotyping may assist with enrichment strategies. However, biomarker-based strategies are difficult to operationalize. Four sub-phenotypes defined by distinct temperature trajectories in the first 72 h have been reported in adult sepsis. Given the distinct epidemiology of pediatric sepsis, the existence and relevance of temperature trajectory-defined sub-phenotypes in children is unknown. We aimed to classify septic children into de novo sub-phenotypes derived from temperature trajectories in the first 72 h, and compare cytokine, immune function, and immunometabolic markers across subgroups., Methods: This was a secondary analysis of a prospective cohort of 191 critically ill septic children recruited from a single academic pediatric intensive care unit. We performed group-based trajectory modeling using temperatures over the first 72 h of sepsis to identify latent profiles. We then used mixed effects regression to determine if temperature trajectory-defined sub-phenotypes were associated with cytokine levels, immune function, and mitochondrial respiration., Results: We identified four temperature trajectory-defined sub-phenotypes: hypothermic, normothermic, hyperthermic fast-resolvers, and hyperthermic slow-resolvers. Hypothermic patients were less often previously healthy and exhibited lower levels of pro- and anti-inflammatory cytokines and chemokines. Hospital mortality did not differ between hypothermic children (17%) and other sub-phenotypes (3-11%; P = 0.26)., Conclusions: Critically ill septic children can be categorized into temperature trajectory-defined sub-phenotypes that parallel adult sepsis. Hypothermic children exhibit a blunted cytokine and chemokine profile. Group-based trajectory modeling has utility for identifying subtypes of clinical syndromes by incorporating readily available longitudinal data, rather than relying on inputs from a single timepoint., Competing Interests: The authors report no conflicts of interest., (Copyright © 2022 by the Shock Society.)
- Published
- 2022
- Full Text
- View/download PDF
28. Do CARs finally hit the CLL road?
- Author
-
Kater AP and Melenhorst JJ
- Subjects
- Humans, Leukemia, Lymphocytic, Chronic, B-Cell therapy
- Published
- 2022
- Full Text
- View/download PDF
29. STAT3 Role in T-Cell Memory Formation.
- Author
-
Kaminskiy Y and Melenhorst JJ
- Subjects
- Humans, STAT3 Transcription Factor metabolism, Signal Transduction, Memory T Cells, Neoplasms metabolism
- Abstract
Along with the clinical success of immuno-oncology drugs and cellular therapies, T-cell biology has attracted considerable attention in the immunology community. Long-term immunity, traditionally analyzed in the context of infection, is increasingly studied in cancer. Many signaling pathways, transcription factors, and metabolic regulators have been shown to participate in the formation of memory T cells. There is increasing evidence that the signal transducer and activator of transcription-3 (STAT3) signaling pathway is crucial for the formation of long-term T-cell immunity capable of efficient recall responses. In this review, we summarize what is currently known about STAT3 role in the context of memory T-cell formation and antitumor immunity.
- Published
- 2022
- Full Text
- View/download PDF
30. Decade-long leukaemia remissions with persistence of CD4 + CAR T cells.
- Author
-
Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, Gao P, Bandyopadhyay S, Sun H, Zhao Z, Lundh S, Pruteanu-Malinici I, Nobles CL, Maji S, Frey NV, Gill SI, Loren AW, Tian L, Kulikovskaya I, Gupta M, Ambrose DE, Davis MM, Fraietta JA, Brogdon JL, Young RM, Chew A, Levine BL, Siegel DL, Alanio C, Wherry EJ, Bushman FD, Lacey SF, Tan K, and June CH
- Subjects
- Antigens, CD19 immunology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Cell Separation, Humans, Time Factors, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes immunology, Immunotherapy, Adoptive, Leukemia immunology, Leukemia therapy, Receptors, Chimeric Antigen immunology
- Abstract
The adoptive transfer of T lymphocytes reprogrammed to target tumour cells has demonstrated potential for treatment of various cancers
1-7 . However, little is known about the long-term potential and clonal stability of the infused cells. Here we studied long-lasting CD19-redirected chimeric antigen receptor (CAR) T cells in two patients with chronic lymphocytic leukaemia1-4 who achieved a complete remission in 2010. CAR T cells remained detectable more than ten years after infusion, with sustained remission in both patients. Notably, a highly activated CD4+ population emerged in both patients, dominating the CAR T cell population at the later time points. This transition was reflected in the stabilization of the clonal make-up of CAR T cells with a repertoire dominated by a small number of clones. Single-cell profiling demonstrated that these long-persisting CD4+ CAR T cells exhibited cytotoxic characteristics along with ongoing functional activation and proliferation. In addition, longitudinal profiling revealed a population of gamma delta CAR T cells that prominently expanded in one patient concomitant with CD8+ CAR T cells during the initial response phase. Our identification and characterization of these unexpected CAR T cell populations provide novel insight into the CAR T cell characteristics associated with anti-cancer response and long-term remission in leukaemia., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
31. The Safety of Bridging Radiation with Anti-BCMA CAR T-Cell Therapy for Multiple Myeloma.
- Author
-
Manjunath SH, Cohen AD, Lacey SF, Davis MM, Garfall AL, Melenhorst JJ, Maxwell R, Arscott WT, Maity A, Jones JA, Plastaras JP, Stadtmauer EA, Levine BL, June CH, Milone MC, and Paydar I
- Subjects
- B-Cell Maturation Antigen, Humans, Receptors, Chimeric Antigen, Retrospective Studies, Immunotherapy, Adoptive adverse effects, Multiple Myeloma drug therapy
- Abstract
Purpose: B-cell maturation antigen (BCMA)-targeted chimeric antigen receptor (CAR) T cells (CART-BCMA) are a promising treatment for relapsed/refractory multiple myeloma (r/rMM). We evaluated the safety and feasibility of bridging radiation (RT) in subjects treated on a phase I trial of CART-BCMA., Experimental Design: Twenty-five r/rMM subjects were treated in three cohorts with two doses of CART-BCMA cells ± cyclophosphamide. We retrospectively analyzed toxicity, response, and CART manufacturing data based on RT receipt., Results: Thirteen subjects received no RT <1 year before CART infusion (Group A). Eight subjects received RT <1 year before CART infusion (Group B) with median time from RT to apheresis of 114 days (range 40-301). Four subjects received bridging-RT (Group C) with a median dose of 22 Gy and time from RT to infusion of 25 days (range 18-35). Group C had qualitatively lower rates of grade 4 (G4) hematologic toxicities (25%) versus A (61.5%) and B (62.5%). G3-4 neurotoxicity occurred in 7.7%, 25%, and 25% in Group A, B, and C, respectively. G3-4 cytokine release syndrome was observed in 38.5%, 25%, and 25% in Group A, B, and C, respectively. Partial response or better was observed in 54%, 38%, and 50% of Group A, B, and C, respectively. RT administered <1 year ( P = 0.002) and <100 days ( P = 0.069) before apheresis was associated with lower in vitro proliferation during manufacturing; however, in vivo CART-BCMA expansion appeared similar across groups., Conclusions: Bridging-RT appeared safe and feasible with CART-BCMA therapy in our r/rMM patients, though larger future studies are needed to draw definitive conclusions., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
32. Cancer stem cells: advances in biology and clinical translation-a Keystone Symposia report.
- Author
-
Cable J, Pei D, Reid LM, Wang XW, Bhatia S, Karras P, Melenhorst JJ, Grompe M, Lathia JD, Song E, Kuo CJ, Zhang N, White RM, Ma SK, Ma L, Chin YR, Shen MM, Ng IOL, Kaestner KH, Zhou L, Sikandar S, Schmitt CA, Guo W, Wong CC, Ji J, Tang DG, Dubrovska A, Yang C, Wiedemeyer WR, and Weissman IL
- Subjects
- Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Humans, Neoplasms metabolism, Translational Research, Biomedical methods, Congresses as Topic trends, Neoplasms genetics, Neoplastic Stem Cells physiology, Research Report, Translational Research, Biomedical trends, Tumor Microenvironment physiology
- Abstract
The test for the cancer stem cell (CSC) hypothesis is to find a target expressed on all, and only CSCs in a patient tumor, then eliminate all cells with that target that eliminates the cancer. That test has not yet been achieved, but CSC diagnostics and targets found on CSCs and some other cells have resulted in a few clinically relevant therapies. However, it has become apparent that eliminating the subset of tumor cells characterized by self-renewal properties is essential for long-term tumor control. CSCs are able to regenerate and initiate tumor growth, recapitulating the heterogeneity present in the tumor before treatment. As great progress has been made in identifying and elucidating the biology of CSCs as well as their interactions with the tumor microenvironment, the time seems ripe for novel therapeutic strategies that target CSCs to find clinical applicability. On May 19-21, 2021, researchers in cancer stem cells met virtually for the Keystone eSymposium "Cancer Stem Cells: Advances in Biology and Clinical Translation" to discuss recent advances in the understanding of CSCs as well as clinical efforts to target these populations., (© 2021 New York Academy of Sciences.)
- Published
- 2021
- Full Text
- View/download PDF
33. CAR T-cell immunotherapy: a powerful weapon for fighting hematological B-cell malignancies.
- Author
-
Mi JQ, Xu J, Zhou J, Zhao W, Chen Z, Melenhorst JJ, and Chen S
- Subjects
- Humans, Immunotherapy adverse effects, T-Lymphocytes, Hematologic Neoplasms therapy, Neoplasms, Receptors, Chimeric Antigen
- Abstract
The current standard of care in hematological malignancies has brought considerable clinical benefits to patients. However, important bottlenecks still limit optimal achievements following a current medical practice. The genetic complexity of the diseases and the heterogeneity of tumor clones cause difficulty in ensuring long-term efficacy of conventional treatments for most hematological disorders. Consequently, new treatment strategies are necessary to improve clinical outcomes. Chimeric antigen receptor T-cell (CAR T) immunotherapy opens a new path for targeted therapy of hematological malignancies. In this review, through a representative case study, we summarize the current experience of CAR T-cell therapy, the management of common side effects, the causative mechanisms of therapy resistance, and new strategies to improve the efficacy of CAR T-cell therapy., (© 2021. Higher Education Press.)
- Published
- 2021
- Full Text
- View/download PDF
34. Next-generation CAR T cells to overcome current drawbacks.
- Author
-
Lundh S, Maji S, and Melenhorst JJ
- Subjects
- Animals, Combined Modality Therapy, Disease Management, Disease Susceptibility, Humans, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive economics, Immunotherapy, Adoptive trends, Neoplasms etiology, Neoplasms metabolism, Prognosis, Receptors, Antigen, T-Cell immunology, Receptors, Chimeric Antigen immunology, Treatment Outcome, Immunotherapy, Adoptive methods, Neoplasms therapy, Receptors, Antigen, T-Cell metabolism, Receptors, Chimeric Antigen metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
As a rapidly emerging treatment in the oncology field, adoptive transfer of autologous, genetically modified chimeric antigen receptor (CAR) T cells has shown striking efficacy and is curative in certain relapsed/refractory patients with hematologic malignancy. This treatment modality of using a "living drug" offers many tantalizing and novel therapeutic strategies for cancer patients whose remaining treatment options may have otherwise been limited. Despite the early success of CAR T cells in hematologic malignancies, many barriers remain for widespread adoption. General barriers include cellular manufacturing limitations, baseline quality of the T cells, adverse events post-infusion such as cytokine release syndrome (CRS) and neurotoxicity, and host rejection of non-human CARs. Additionally, each hematologic disease presents unique mechanisms of relapse which have to be addressed in future clinical trials if we are to augment the efficacy of CAR T treatment. In this review, we will describe current barriers to hindering efficacy of CAR T-cell treatment for hematologic malignancies in a disease-specific manner and review recent innovations aimed at enhancing the potency and applicability of CAR T cells, with the overall goal of building a framework to begin incorporating this form of therapy into the standard medical management of blood cancers., (© 2020. Japanese Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
35. The 2020 BMT CTN Myeloma Intergroup Workshop on Immune Profiling and Minimal Residual Disease Testing in Multiple Myeloma.
- Author
-
Holstein SA, Bahlis N, Bergsagel PL, Bhutani M, Bolli N, Brownstein C, Demolis P, Foureau D, Gay F, Ghobrial IM, Gormley N, Hillengass J, Kaiser M, Maus MV, Melenhorst JJ, Merz M, Dwyer MO, Paiva B, Pasquini MC, Shah N, Wong SW, Usmani SZ, and McCarthy PL
- Subjects
- Humans, Bone Marrow, Diterpenes, High-Throughput Nucleotide Sequencing, Neoplasm, Residual, Multiple Myeloma diagnosis
- Abstract
The fifth annual Blood and Marrow Transplant Clinical Trials Network Myeloma Intergroup Workshop on Immune Profiling and Minimal Residual Disease Testing in Multiple Myeloma was conducted as one of the American Society of Hematology Annual Meeting Scientific Workshops on Thursday December 3, 2020. This workshop focused on four main topics: (1) integrating minimal residual disease into clinical trial design and practice; (2) the molecular and immunobiology of disease evolution and progression in myeloma; (3) adaptation of next-generation sequencing, next-generation flow cytometry, and cytometry by time of flight techniques; and (4) chimeric antigen receptor T-cell and other cellular therapies for myeloma. In this report, we provide a summary of the workshop presentations and discuss future directions in the field., (Copyright © 2021 The American Society for Transplantation and Cellular Therapy. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
36. Author Correction: Engineering-enhanced CAR T cells for improved cancer therapy.
- Author
-
Milone MC, Xu J, Chen SJ, Collins MA, Zhou J, Powell DJ Jr, and Melenhorst JJ
- Published
- 2021
- Full Text
- View/download PDF
37. BET bromodomain protein inhibition reverses chimeric antigen receptor extinction and reinvigorates exhausted T cells in chronic lymphocytic leukemia.
- Author
-
Kong W, Dimitri A, Wang W, Jung IY, Ott CJ, Fasolino M, Wang Y, Kulikovskaya I, Gupta M, Yoder T, DeNizio JE, Everett JK, Williams EF, Xu J, Scholler J, Reich TJ, Bhoj VG, Haines KM, Maus MV, Melenhorst JJ, Young RM, Jadlowsky JK, Marcucci KT, Bradner JE, Levine BL, Porter DL, Bushman FD, Kohli RM, June CH, Davis MM, Lacey SF, Vahedi G, and Fraietta JA
- Subjects
- Antigens, CD19 immunology, Azepines pharmacology, Epigenesis, Genetic, Glycolysis drug effects, Humans, Immune Tolerance, Immunologic Memory, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Oxidative Phosphorylation drug effects, Receptors, Chimeric Antigen genetics, T-Lymphocytes drug effects, T-Lymphocytes metabolism, Triazoles pharmacology, Immunotherapy, Adoptive, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Proteins antagonists & inhibitors, Proteins immunology, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology
- Abstract
Chimeric antigen receptor (CAR) T cells have induced remarkable antitumor responses in B cell malignancies. Some patients do not respond because of T cell deficiencies that hamper the expansion, persistence, and effector function of these cells. We used longitudinal immune profiling to identify phenotypic and pharmacodynamic changes in CD19-directed CAR T cells in patients with chronic lymphocytic leukemia (CLL). CAR expression maintenance was also investigated because this can affect response durability. CAR T cell failure was accompanied by preexisting T cell-intrinsic defects or dysfunction acquired after infusion. In a small subset of patients, CAR silencing was observed coincident with leukemia relapse. Using a small molecule inhibitor, we demonstrated that the bromodomain and extra-terminal (BET) family of chromatin adapters plays a role in downregulating CAR expression. BET protein blockade also ameliorated CAR T cell exhaustion as manifested by inhibitory receptor reduction, enhanced metabolic fitness, increased proliferative capacity, and enriched transcriptomic signatures of T cell reinvigoration. BET inhibition decreased levels of the TET2 methylcytosine dioxygenase, and forced expression of the TET2 catalytic domain eliminated the potency-enhancing effects of BET protein targeting in CAR T cells, providing a mechanism linking BET proteins and T cell dysfunction. Thus, modulating BET epigenetic readers may improve the efficacy of cell-based immunotherapies.
- Published
- 2021
- Full Text
- View/download PDF
38. Engineering enhanced CAR T-cells for improved cancer therapy.
- Author
-
Milone MC, Xu J, Chen SJ, Collins MA, Zhou J, Powell DJ Jr, and Melenhorst JJ
- Subjects
- Antigens, CD19, Humans, Immunotherapy, Adoptive, T-Lymphocytes, Neoplasms therapy, Receptors, Chimeric Antigen
- Abstract
Chimeric antigen receptor (CAR) T-cell therapies have evolved from a research tool to a paradigm-shifting therapy with impressive responses in B cell malignancies. This review summarizes the current state of the CAR T-cell field, focusing on CD19- and B cell maturation antigen-directed CAR T-cells, the most developed of the CAR T-cell therapies. We discuss the many challenges to CAR-T therapeutic success and innovations in CAR design and T-cell engineering aimed at extending this therapeutic platform beyond hematologic malignancies.
- Published
- 2021
- Full Text
- View/download PDF
39. Case Report: Prolonged Survival Following EGFRvIII CAR T Cell Treatment for Recurrent Glioblastoma.
- Author
-
Durgin JS, Henderson F Jr, Nasrallah MP, Mohan S, Wang S, Lacey SF, Melenhorst JJ, Desai AS, Lee JYK, Maus MV, June CH, Brem S, O'Connor RS, Binder Z, and O'Rourke DM
- Abstract
Autologous chimeric antigen receptor (CAR) T cells targeted to epidermal growth factor receptor variant III (CAR T-EGFRvIII) have been developed and administered experimentally to treat patients with IDH1 wildtype recurrent glioblastoma (rGBM) (NCT02209376). We report the case of a 59-year-old patient who received a single peripheral infusion of CAR T-EGFRvIII cells and survived 36 months after disease recurrence, exceeding expected survival for recurrent glioblastoma. Post-infusion histopathologic analysis of tissue obtained during a second stage surgical resection revealed immunosuppressive adaptive changes in the tumor tissue as well as reduced EGFRvIII expression. Serial brain imaging demonstrated a significant reduction in relative cerebral blood volume (rCBV), a measure strongly associated with tumor proliferative activity, at early time points following CAR T treatment. Notably, CAR T-EGFRvIII cells persisted in her peripheral circulation during 29 months of follow-up, the longest period of CAR T persistence reported in GBM trials to date. These findings in a long-term survivor show that peripherally administered CAR T-EGFRvIII cells can persist for years in the circulation and suggest that this cell therapy approach could be optimized to achieve broader efficacy in recurrent GBM patients., Competing Interests: DO’R and ZB are inventors on patents related to CAR T cells that have been filed by the University of Pennsylvania. JJM consults with or serves on the board of directors of several companies developing CAR T technology. JM and SL are inventors of intellectual property related to CAR T cells that is licensed by the University of Pennsylvania to Novartis. CHJ reports receiving grants from Tmunity Therapeutics and holds founders stock in Tmunity Therapeutics and DeCART Therapeutics. CHJ also receives personal income from BluesphereBio, Cabaletta, Carisma, Cellares, Celldex Therapeutics, Viracta Therapeutics, Ziopharm and WIRB-Copernicus Group as well as royalties from Novartis. MCM is an inventor on patent applications related to CAR technology and has received licensing royalties from Novartis corporation. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Durgin, Henderson, Nasrallah, Mohan, Wang, Lacey, Melenhorst, Desai, Lee, Maus, June, Brem, O’Connor, Binder and O’Rourke.)
- Published
- 2021
- Full Text
- View/download PDF
40. B-cell maturation antigen chimeric antigen receptor T-cell re-expansion in a patient with myeloma following salvage programmed cell death protein 1 inhibitor-based combination therapy.
- Author
-
Bernabei L, Tian L, Garfall AL, Melenhorst JJ, Lacey SF, Stadtmauer EA, Vogl DT, Gonzalez VE, Plesa G, Young RM, Waxman A, Levine BL, June CH, Milone MC, and Cohen AD
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized administration & dosage, Combined Modality Therapy, Dexamethasone administration & dosage, Disease-Free Survival, Female, Humans, Immune Checkpoint Inhibitors administration & dosage, Lenalidomide administration & dosage, Male, Middle Aged, Survival Rate, Thalidomide administration & dosage, Thalidomide analogs & derivatives, Antineoplastic Combined Chemotherapy Protocols administration & dosage, B-Cell Maturation Antigen blood, Immunotherapy, Adoptive, Multiple Myeloma blood, Multiple Myeloma mortality, Multiple Myeloma therapy, Neoplasm Proteins blood, Programmed Cell Death 1 Receptor blood, Receptors, Chimeric Antigen
- Published
- 2021
- Full Text
- View/download PDF
41. Single-cell multiomics dissection of basal and antigen-specific activation states of CD19-targeted CAR T cells.
- Author
-
Bai Z, Lundh S, Kim D, Woodhouse S, Barrett DM, Myers RM, Grupp SA, Maus MV, June CH, Camara PG, Melenhorst JJ, and Fan R
- Subjects
- Animals, Antigens, CD19 immunology, Antigens, CD19 metabolism, Case-Control Studies, Cell Line, Tumor, Coculture Techniques, Cytotoxicity, Immunologic genetics, Humans, Mice, NIH 3T3 Cells, Phenotype, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, RNA-Seq, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism, Antigens, CD19 genetics, Gene Expression Profiling, Immunotherapy, Adoptive, Lymphocyte Activation genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Chimeric Antigen genetics, Single-Cell Analysis, T-Lymphocytes transplantation, Transcriptome
- Abstract
Background: Autologous T cells engineered to express a chimeric antigen receptor (CAR) specific for CD19 molecule have transformed the therapeutic landscape in patients with highly refractory leukemia and lymphoma, and the use of donor-generated allogeneic CAR T is paving the way for further breakthroughs in the treatment of cancer. However, it remains unknown how the intrinsic heterogeneities of these engineered cells mediate therapeutic efficacy and whether allogeneic products match the effectiveness of autologous therapies., Methods: Using single-cell mRNA sequencing in conjunction with CITE-seq, we performed multiomics characterization of CAR T cells generated from healthy donor and patients with acute lymphoblastic leukemia. CAR T cells used in this study were manufactured at the University of Pennsylvania through lentiviral transduction with a CD19-4-1BB-CD3ζ construct. Besides the baseline condition, we engineered NIH-3T3 cells with human CD19 or mesothelin expression to conduct ex vivo antigen-specific or non-antigen stimulation of CAR T cells through 6-hour coculture at a 1:1 ratio., Results: We delineated the global cellular and molecular CAR T landscape and identified that transcriptional CAR tonic signaling was regulated by a mixture of early activation, exhaustion signatures, and cytotoxic activities. On CD19 stimulation, we illuminated the disparities of CAR T cells derived from different origins and found that donor CAR T had more pronounced activation level in correlation with the upregulation of major histocompatibility complex class II genes compared with patient CAR T cells. This finding was independently validated in additional datasets from literature. Furthermore, GM-CSF( CSF2 ) expression was found to be associated with functional gene productions, but it induced little impact on the CAR T activation., Conclusions: Through integrated multiomics profiling and unbiased canonical pathway analyses, our results unveil heterogeneities in the transcriptional, phenotypic, functional, and metabolic profiles of donor and patient CAR T cells, providing mechanistic basis for ameliorating clinical outcomes and developing next-generation 'off- the-shelf' allogeneic products., Competing Interests: Competing interests: RF is a cofounder of IsoPlexis, Singleron Biotechnologies and AtlasXomics and a member of their scientific advisory boards with financial interests, which could affect or have the perception of affecting the author’s objectivity. The interests of RF were reviewed and managed by Yale University Provost’s Office in accordance with the University’s conflict of interest policies., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
42. CT103A, a forward step in multiple myeloma immunotherapies.
- Author
-
Xu J and Melenhorst JJ
- Abstract
Competing Interests: Dr. Melenhorst conflicts of interest disclosure: JJM receives research funding from IASO Biotherapeutics and Kite Pharma, a Gilead company. He furthermore served as speaker for Novartis and Johnson & Johnson and consults for Simcere of America, Shanghai Unicar Therapy, Janssen Research & Development, LLC, Poseida, Allogene, and IASO Biotherapeutics, and is on the Medical and Scientific Advisory Board of IASO Biotherapeutics. Lastly, JJM holds patents related to CAR T cell manufacturing and biomarkers.
- Published
- 2021
- Full Text
- View/download PDF
43. Author Correction: Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia.
- Author
-
Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, Boesteanu AC, Wang Y, O'Connor RS, Hwang WT, Pequignot E, Ambrose DE, Zhang C, Wilcox N, Bedoya F, Dorfmeier C, Chen F, Tian L, Parakandi H, Gupta M, Young RM, Johnson FB, Kulikovskaya I, Liu L, Xu J, Kassim SH, Davis MM, Levine BL, Frey NV, Siegel DL, Huang AC, Wherry EJ, Bitter H, Brogdon JL, Porter DL, June CH, and Melenhorst JJ
- Published
- 2021
- Full Text
- View/download PDF
44. Improving and Maintaining Responses in Pediatric B-Cell Acute Lymphoblastic Leukemia Chimeric Antigen Receptor-T Cell Therapy.
- Author
-
Arya R, Barrett DM, Grupp SA, and Melenhorst JJ
- Subjects
- Cell- and Tissue-Based Therapy, Child, Humans, Receptors, Antigen, T-Cell genetics, Immunotherapy, Adoptive, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Chimeric Antigen genetics
- Abstract
Abstract: Chimeric antigen receptor T therapy has heralded a new era in the treatment of acute lymphoblastic leukemia (ALL) and other hematologic malignancies. In this autologous immunotherapy, patient-derived T cells are genetically engineered and then infused back to kill the leukemia cells. The observed response rates in ALL are a testament to the success of this therapy. However, there have been instances where the patients either did not respond or relapsed after initial response. Emergence of resistance due to antigen loss and T-cell exhaustion has been observed. This poses a challenge in making this therapy successful for every ALL patient and warrants deeper understanding of emergence of resistance and potential approaches to overcome them. Here we discuss current perspectives and advances in this area., Competing Interests: Conflicts of Interest and Source of Funding: The authors have disclosed that they have no significant relationships with, or financial interest in, any commercial companies pertaining to this article. S.A.G. reports research support from Novartis, Servier, CRISPR/Vertex, and Kite/Gillead, and SAB/SSC membership and/or consulting from Novartis, CRISPR/Vertex, Allogene, CBMG, Adaptimmune, TCR2, Juno, Jazz, Cellectis, Roche, GSK, Cabaletta, and Janssen/J&J. J.J.M. is supported by the NIH grant R01 CA241762., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
45. High-Dimensional Immune Monitoring for Chimeric Antigen Receptor T Cell Therapies.
- Author
-
Sharma S, Quinn D, Melenhorst JJ, and Pruteanu-Malinici I
- Subjects
- Animals, Humans, Receptors, Chimeric Antigen analysis, Software, T-Lymphocytes cytology, Flow Cytometry methods, Immunotherapy, Adoptive methods, Monitoring, Immunologic methods
- Abstract
Purpose of Review: High-dimensional flow cytometry experiments have become a method of choice for high-throughput integration and characterization of cell populations. Here, we present a summary of state-of-the-art R-based pipelines used for differential analyses of cytometry data, largely based on chimeric antigen receptor (CAR) T cell therapies. These pipelines are based on publicly available R libraries, put together in a systematic and functional fashion, therefore free of cost., Recent Findings: In recent years, existing tools tailored to analyze complex high-dimensional data such as single-cell RNA sequencing (scRNAseq) have been successfully ported to cytometry studies due to the similar nature of flow cytometry and scRNAseq platforms. Existing environments like Cytobank (Kotecha et al., 2010), FlowJo (FlowJo™ Software) and FCS Express (https://denovosoftware.com) already offer a variety of these ported tools, but they either come at a premium or are fairly complicated to manage by an inexperienced user. To mitigate these limitations, experienced cytometrists and bioinformaticians usually incorporate these functions into an RShiny (https://shiny.rstudio.com) application that ultimately offers a user-friendly, intuitive environment that can be used to analyze flow cytometry data. Computational tools and Shiny-based tools are the perfect answer to the ever-growing dimensionality and complexity of flow cytometry data, by offering a dynamic, yet user-friendly exploratory space, tailored to bridge the space between the lab experimental world and the computational, machine learning space.
- Published
- 2021
- Full Text
- View/download PDF
46. Endothelial Biomarkers Are Associated With Indirect Lung Injury in Sepsis-Associated Pediatric Acute Respiratory Distress Syndrome.
- Author
-
Whitney JE, Feng R, Koterba N, Chen F, Bush J, Graham K, Lacey SF, Melenhorst JJ, Parikh SM, Weiss SL, and Yehya N
- Abstract
Objectives: Acute respiratory distress syndrome occurring in the setting of direct versus indirect lung injury may reflect different pathobiologies amenable to different treatment strategies. We sought to test whether a panel of plasma biomarkers differed between children with sepsis-associated direct versus indirect acute respiratory distress syndrome. We hypothesized that a biomarker profile indicative of endothelial activation would be associated with indirect acute respiratory distress syndrome., Design: Observational cohort., Setting: Academic PICU., Subjects: Patients less than 18 years old with sepsis-associated direct (pneumonia, n = 52) or indirect (extrapulmonary sepsis, n = 46) acute respiratory distress syndrome., Interventions: None., Measurements and Main Results: Of 58 biomarkers examined, 33 differed by acute respiratory distress syndrome subtype. We used classification and regression tree methodology to examine associations between clinical and biochemical markers and acute respiratory distress syndrome subtype. The classification and regression tree model using only clinical variables (age, sex, race, oncologic comorbidity, and Pediatric Risk of Mortality-III score) performed worse than the classification and regression tree model using five clinical variables and 58 biomarkers. The best classification and regression tree model used only four endothelial biomarkers, including elevated angiopoietin-2/angiopoietin-1 ratio, vascular cell-adhesion molecule, and von Willebrand factor, to identify indirect acute respiratory distress syndrome. Test characteristics were 89% (80-97%) sensitivity, 80% (69-92%) specificity, positive predictive value 84% (74-93%), and negative predictive value 86% (76-96%)., Conclusions: Indirect lung injury in children with acute respiratory distress syndrome is characterized by a biomarker profile indicative of endothelial activation, excess inflammation, and worse outcomes. A model using four biomarkers has the potential to be useful for more precisely identifying patients with acute respiratory distress syndrome whose pathobiology may respond to endothelial-targeted therapies in future trials., Competing Interests: The authors have disclosed that they do not have any potential conflicts of interest., (Copyright © 2020 The Authors. Published by Wolters Kluwer Health, Inc. on behalf of the Society of Critical Care Medicine.)
- Published
- 2020
- Full Text
- View/download PDF
47. Dual Targeting of Mesothelin and CD19 with Chimeric Antigen Receptor-Modified T Cells in Patients with Metastatic Pancreatic Cancer.
- Author
-
Ko AH, Jordan AC, Tooker E, Lacey SF, Chang RB, Li Y, Venook AP, Tempero M, Damon L, Fong L, O'Hara MH, Levine BL, Melenhorst JJ, Plesa G, June CH, and Beatty GL
- Subjects
- Humans, Lymphocyte Depletion methods, Mesothelin, Neoplasm Metastasis, Neoplasm Staging, Pancreatic Neoplasms immunology, Pancreatic Neoplasms pathology, Pilot Projects, T-Lymphocytes metabolism, Treatment Outcome, Antigens, CD19 immunology, GPI-Linked Proteins antagonists & inhibitors, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Pancreatic Neoplasms therapy, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology
- Abstract
B cells infiltrate pancreatic ductal adenocarcinoma (PDAC) and in preclinical cancer models, can suppress T cell immunosurveillance in cancer. Here, we conducted a pilot study to assess the safety and feasibility of administering lentiviral-transduced chimeric antigen receptor (CAR)-modified autologous T cells redirected against mesothelin to target tumor cells along with CART cells redirected against CD19 to deplete B cells. Both CARs contained 4-1BB and CD3ζ signaling domains. Three patients with chemotherapy-refractory PDAC received 1.5 g/m
2 cyclophosphamide prior to separate infusions of lentiviral-transduced T cells engineered to express chimeric anti-mesothelin immunoreceptor SS1 (CART-Meso, 3 × 107 /m2 ) and chimeric anti-CD19 immunoreceptor (CART-19, 3 × 107 /m2 ). Treatment was well tolerated without dose-limiting toxicities. Best response was stable disease (1 of 3 patients). CART-19 (compared to CART-Meso) cells showed the greatest expansion in the blood, although persistence was transient. B cells were successfully depleted in all subjects, became undetectable by 7-10 days post-infusion, and remained undetectable for at least 28 days. Together, concomitant delivery of CART-Meso and CART-19 cells in patients with PDAC is safe. CART-19 cells deplete normal B cells but at the dose tested in these 3 subjects did not improve CART-Meso cell persistence., (Copyright © 2020 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
48. Clinical practice: chimeric antigen receptor (CAR) T cells: a major breakthrough in the battle against cancer.
- Author
-
Lundh S, Jung IY, Dimitri A, Vora A, Melenhorst JJ, Jadlowsky JK, and Fraietta JA
- Subjects
- Genetic Engineering methods, Humans, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive economics, Neoplasms therapy, Hematologic Neoplasms therapy, Immunotherapy, Adoptive methods, Receptors, Chimeric Antigen genetics
- Abstract
Chimeric antigen receptor (CAR) T cell therapy has come of age, offering a potentially curative option for patients who are refractory to standard anti-cancer treatments. The success of CAR T cell therapy in the setting of acute lymphoblastic leukemia and specific types of B cell lymphoma led to rapid regulatory approvals of CD19-directed CAR T cells, first in the United States and subsequently across the globe. Despite these major milestones in the field of immuno-oncology, growing experience with CAR T cells has also highlighted the major limitations of this strategy, namely challenges associated with manufacturing a bespoke patient-specific product, intrinsic immune cell defects leading to poor CAR T cell function as well as persistence, and/or tumor cell resistance resulting from loss or modulation of the targeted antigen. In addition, both on- and off-tumor immunotoxicities and the financial burden inherent in conventional cellular biomanufacturing often hamper the success of CAR T cell-based treatment approaches. Herein, we provide an overview of the opportunities and challenges related to the first form of gene transfer therapy to gain commercial approval in the United States. Ongoing advances in the areas of genetic engineering, precision genome editing, toxicity mitigation methods and cell manufacturing will improve the efficacy and safety of CAR T cells for hematologic malignancies and expand the use of this novel class of therapeutics to reach solid tumors.
- Published
- 2020
- Full Text
- View/download PDF
49. Diagnostic biomarkers to differentiate sepsis from cytokine release syndrome in critically ill children.
- Author
-
Diorio C, Shaw PA, Pequignot E, Orlenko A, Chen F, Aplenc R, Barrett DM, Bassiri H, Behrens E, DiNofia AM, Gonzalez V, Koterba N, Levine BL, Maude SL, Meyer NJ, Moore JH, Paessler M, Porter DL, Bush JL, Siegel DL, Davis MM, Zhang D, June CH, Grupp SA, Melenhorst JJ, Lacey SF, Weiss SL, and Teachey DT
- Subjects
- Child, Critical Illness, Cytokine Release Syndrome, Humans, Receptors, Antigen, T-Cell, Precursor Cell Lymphoblastic Leukemia-Lymphoma, Sepsis diagnosis
- Abstract
Chimeric antigen receptor (CAR) T-cells directed against CD19 have drastically altered outcomes for children with relapsed and refractory acute lymphoblastic leukemia (r/r ALL). Pediatric patients with r/r ALL treated with CAR-T are at increased risk of both cytokine release syndrome (CRS) and sepsis. We sought to investigate the biologic differences between CRS and sepsis and to develop predictive models which could accurately differentiate CRS from sepsis at the time of critical illness. We identified 23 different cytokines that were significantly different between patients with sepsis and CRS. Using elastic net prediction modeling and tree classification, we identified cytokines that were able to classify subjects as having CRS or sepsis accurately. A markedly elevated interferon γ (IFNγ) or a mildly elevated IFNγ in combination with a low IL1β were associated with CRS. A normal to mildly elevated IFNγ in combination with an elevated IL1β was associated with sepsis. This combination of IFNγ and IL1β was able to categorize subjects as having CRS or sepsis with 97% accuracy. As CAR-T therapies become more common, these data provide important novel information to better manage potential associated toxicities., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
50. Transdifferentiation of lymphoma into sarcoma associated with profound reprogramming of the epigenome.
- Author
-
Zhang Q, Orlando EJ, Wang HY, Bogusz AM, Liu X, Lacey SF, Strauser HT, Nunez-Cruz S, Nejati R, Zhang P, Brooks S, Watt C, Melenhorst JJ, June CH, Schuster SJ, and Wasik MA
- Subjects
- Adenine administration & dosage, Adenine adverse effects, Adenine analogs & derivatives, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cellular Reprogramming genetics, Cellular Reprogramming immunology, Cellular Reprogramming physiology, Combined Modality Therapy, Epigenesis, Genetic physiology, Epigenome immunology, Gene Rearrangement, Genes, Immunoglobulin Heavy Chain genetics, Humans, Immunotherapy methods, Lymph Nodes pathology, Lymphoma, Mantle-Cell genetics, Lymphoma, Mantle-Cell immunology, Neoplasms, Second Primary diagnosis, Piperidines administration & dosage, Piperidines adverse effects, Receptors, Antigen, T-Cell therapeutic use, Rituximab administration & dosage, Rituximab adverse effects, Sarcoma genetics, Sarcoma immunology, Sarcoma pathology, Transplantation, Autologous adverse effects, Tumor Cells, Cultured, Cell Transdifferentiation genetics, Cell Transdifferentiation immunology, Immunotherapy adverse effects, Lymphoma, Mantle-Cell therapy, Neoplasms, Second Primary etiology, Sarcoma etiology
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.