110 results on '"Milone MC"'
Search Results
2. A pilot study of reduced toxicity conditioning with BU, fludarabine and alemtuzumab before the allogeneic hematopoietic SCT in children and adolescents
- Author
-
Styczynski, J, Tallamy, B, Waxman, I, van de Ven, C, Milone, MC, Shaw, LM, Harrison, L, Morris, E, Satwani, P, Bhatia, M, George, D, Bradley, MB, Garvin, JH, Schwartz, J, Baxter-Lowe, LA, and Cairo, MS
- Published
- 2011
- Full Text
- View/download PDF
3. Methotrexate clearance by high-flux hemodialysis and peritoneal dialysis: a case report.
- Author
-
Murashima M, Adamski J, Milone MC, Shaw L, Tsai DE, and Bloom RD
- Abstract
We report a case of a patient on maintenance peritoneal dialysis therapy treated with a high-dose methotrexate regimen for central nervous system lymphoma. For the initial methotrexate cycles, he had received temporary daily high-flux hemodialysis starting 24 hours after the infusion of methotrexate to avoid toxicity. However, on account of issues with vascular access, he was treated with continuous multiple-exchange peritoneal dialysis for the last 2 cycles of chemotherapy. Time-averaged clearances (dose divided by area under the curve, combination of endogenous and dialysis clearance) during treatment with high-flux hemodialysis and continuous multiple-exchange peritoneal dialysis were 0.77 mL/min/kg (0.013 mL/s/kg) and 0.65 mL/min/kg (0.011 mL/s/kg), respectively. Peritoneal clearance of methotrexate was estimated to be 0.124 +/- 0.037 mL/min/kg (0.00207 +/- 0.00062 mL/s/kg). Despite lower clearance by means of peritoneal dialysis compared with hemodialysis, the patient did not develop clinical evidence of methotrexate toxicity. Copyright © 2009 National Kidney Foundation, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
4. Hyponatremia associated with levamisole-adulterated cocaine use in emergency department patients.
- Author
-
Friend K, Milone MC, and Perrone J
- Abstract
An increasing percentage of US cocaine has been adulterated with levamisole, an immunomodulator associated with agranulocytosis. We describe 3 emergency department patients with hyponatremia and cocaine use. Despite extensive evaluation, the cause of the hyponatremia was not elucidated but resolved during hospitalization. Because hyponatremia has not previously been associated with cocaine, we sought to uncover a plausible explanation that might be contributing to this new finding. Levamisole was detected in all 3 patients. Although we are unable to confirm causality, we propose that levamisole-adulterated cocaine may have contributed to the hyponatremia described in these patients. [ABSTRACT FROM AUTHOR]
- Published
- 2012
5. The next GME accreditation system.
- Author
-
Milone MC and Milone, Michael C
- Published
- 2012
- Full Text
- View/download PDF
6. Author Correction: Precision targeting of autoantigen-specific B cells in muscle-specific tyrosine kinase myasthenia gravis with chimeric autoantibody receptor T cells.
- Author
-
Oh S, Mao X, Manfredo-Vieira S, Lee J, Patel D, Choi EJ, Alvarado A, Cottman-Thomas E, Maseda D, Tsao PY, Ellebrecht CT, Khella SL, Richman DP, O'Connor KC, Herzberg U, Binder GK, Milone MC, Basu S, and Payne AS
- Published
- 2024
- Full Text
- View/download PDF
7. Bendamustine lymphodepletion before axicabtagene ciloleucel is safe and associates with reduced inflammatory cytokines.
- Author
-
Ghilardi G, Paruzzo L, Svoboda J, Chong EA, Shestov AA, Chen L, Cohen IJ, Gabrielli G, Nasta SD, Porazzi P, Landsburg DJ, Gerson JN, Carter J, Barta SK, Yelton R, Pajarillo R, Patel V, White G, Ballard HJ, Weber E, Napier E, Chong ER, Fraietta JA, Garfall AL, Porter DL, Milone MC, O'Connor R, Schuster SJ, and Ruella M
- Subjects
- Humans, Bendamustine Hydrochloride adverse effects, CD28 Antigens, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Cyclophosphamide, Cytokines, Lymphoma, Follicular, Biological Products
- Abstract
Abstract: Lymphodepletion (LD) is an integral component of chimeric antigen receptor T-cell (CART) immunotherapies. In this study, we compared the safety and efficacy of bendamustine (Benda) to standard fludarabine/cyclophosphamide (Flu/Cy) LD before CD19-directed, CD28-costimulated CART axicabtagene ciloleucel (axi-cel) for patients with large B-cell lymphoma (LBCL) and follicular lymphoma (FL). We analyzed 59 patients diagnosed with LBCL (n = 48) and FL (n = 11) consecutively treated with axi-cel at the University of Pennsylvania. We also analyzed serum samples for cytokine levels and metabolomic changes before and after LD. Flu/Cy and Benda demonstrated similar efficacy, with complete remission rates of 51.4% and 50.0% (P = .981), respectively, and similar progression-free and overall survivals. Any-grade cytokine-release syndrome occurred in 91.9% of patients receiving Flu/Cy vs 72.7% of patients receiving Benda (P = .048); any-grade neurotoxicity after Flu/Cy occurred in 45.9% of patients and after Benda in 18.2% of patients (P = .031). In addition, Flu/Cy was associated with a higher incidence of grade ≥3 neutropenia (100% vs 54.5%; P < .001), infections (78.4% vs 27.3%; P < .001), and neutropenic fever (78.4% vs 13.6%; P < .001). These results were confirmed both in patients with LBCL and those with FL. Mechanistically, patients with Flu/Cy had a greater increase in inflammatory cytokines associated with neurotoxicity and reduced levels of metabolites critical for redox balance and biosynthesis. This study suggests that Benda LD may be a safe alternative to Flu/Cy for CD28-based CART CD19-directed immunotherapy with similar efficacy and reduced toxicities. Benda is associated with reduced levels of inflammatory cytokines and increased anabolic metabolites., (© 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
8. Immunotherapy targeting B cells and long-lived plasma cells effectively eliminates pre-existing donor-specific allo-antibodies.
- Author
-
Zhang Z, Markmann C, Yu M, Agarwal D, Rostami S, Wang W, Liu C, Zhao H, Ochoa T, Parvathaneni K, Xu X, Li E, Gonzalez V, Khadka R, Hoffmann J, Knox JJ, Scholler J, Marcellus B, Allman D, Fraietta JA, Samelson-Jones B, Milone MC, Monos D, Garfall AL, Naji A, and Bhoj VG
- Subjects
- Humans, Animals, Mice, Plasma Cells, B-Cell Maturation Antigen, T-Lymphocytes, Immunotherapy, Antibodies, Receptors, Chimeric Antigen
- Abstract
Pre-existing anti-human leukocyte antigen (HLA) allo-antibodies constitute a major barrier to transplantation. Current desensitization approaches fail due to ineffective depletion of allo-specific memory B cells (Bmems) and long-lived plasma cells (LLPCs). We evaluate the efficacy of chimeric antigen receptor (CAR) T cells targeting CD19 and B cell maturation antigen (BCMA) to eliminate allo-antibodies in a skin pre-sensitized murine model of islet allo-transplantation. We find that treatment of allo-sensitized hosts with CAR T cells targeting Bmems and LLPCs eliminates donor-specific allo-antibodies (DSAs) and mitigates hyperacute rejection of subsequent islet allografts. We then assess the clinical efficacy of the CAR T therapy for desensitization in patients with multiple myeloma (MM) with pre-existing HLA allo-antibodies who were treated with the combination of CART-BCMA and CART-19 (ClinicalTrials.gov: NCT03549442) and observe clinically meaningful allo-antibody reduction. These findings provide logical rationale for clinical evaluation of CAR T-based immunotherapy in highly sensitized candidates to promote successful transplantation., Competing Interests: Declaration of interests J.A.F. is an inventor on patents in the field of T cell therapy for cancer, from which he has received royalties. J.A.F. is also a member of the scientific advisory boards of Cartography Bio. and Shennon Biotechnologies, Inc. M.C.M. is an inventor on patents pending or granted related to CAR T cell technology that are assigned to the University of Pennsylvania and licensed to Novartis Ag, from which he receives royalties; these include patents on the CAR T cell therapies described in this study (PCT/US2011/064191, PCT/US2018/061239, PCT/US2018/063255). M.C.M. is also a founder, stockholder, and co-chair of the scientific advisory boards for Cabaletta Bio and Verismo Therapeutics. A.L.G. has received grants from Novartis, grants from National Institutes of Health, and grants from the Leukemia & Lymphoma Society while this study was conducted, grants and personal fees from Janssen, personal fees from GlaxoSmithKline, personal fees from Legend Biotech, grants from CRISPR Therapeutics, grants from Tmunity Therapeutics, personal fees from Amgen, grants from the Leukemia & Lymphoma Society, and grants from National Institutes of Health outside the submitted work; in addition, A.L.G. has a patent for US15/757,123 pending and licensed to Novartis, a patent for US16/764,459 pending, and a patent for US16/768,260 pending and stock ownership in Cabaletta Bio. V.G.B. is an inventor on patents pending or granted related to CAR T cell technology that are assigned to the University of Pennsylvania and licensed to Cabaletta Bio and Tmunity, from which he receives royalties., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. F77 antigen is a promising target for adoptive T cell therapy of prostate cancer.
- Author
-
Grover P, Nunez-Cruz S, Leferovich J, Wentz T, Bagchi A, Milone MC, and Greene MI
- Subjects
- Male, Animals, Mice, Humans, Cell Line, Tumor, Immunotherapy, Adoptive, Carbohydrates, Cell- and Tissue-Based Therapy, Receptors, Antigen, T-Cell genetics, Xenograft Model Antitumor Assays, Androgens, Prostatic Neoplasms pathology
- Abstract
Adoptive immunotherapy using chimeric antigen receptor (CAR) T cells has made significant success in treating hematological malignancies, paving the way for solid tumors like prostate cancer. However, progress is impeded by a paucity of suitable target antigens. A novel carbohydrate antigen, F77, is expressed on both androgen-dependent and androgen-independent prostate cancer cells, making it a potential immunotherapy target. This study entails the generation and evaluation of a second-generation CAR against a carbohydrate antigen on malignant prostate cancer cells. Using a single chain fragment variable (scFv) from an F77-specific mouse monoclonal antibody, we created second-generation CARs with CD28 and CD137 (4-1BB) costimulatory signals. F77 expressing lentiviral CAR T cells produce cytokines and kill tumor cells in a F77 expression-dependent manner. These F77-specific CAR T cells eradicate prostate tumors in a human xenograft model employing PC3 cells. These findings validate F77 as a promising immunotherapeutic target for prostate cancer and other malignancies with this aberrant carbohydrate structure., Competing Interests: Declaration of competing interest None declared., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Precision targeting of autoantigen-specific B cells in muscle-specific tyrosine kinase myasthenia gravis with chimeric autoantibody receptor T cells.
- Author
-
Oh S, Mao X, Manfredo-Vieira S, Lee J, Patel D, Choi EJ, Alvarado A, Cottman-Thomas E, Maseda D, Tsao PY, Ellebrecht CT, Khella SL, Richman DP, O'Connor KC, Herzberg U, Binder GK, Milone MC, Basu S, and Payne AS
- Subjects
- Humans, Mice, Animals, Autoantigens therapeutic use, T-Lymphocytes, Autoantibodies therapeutic use, Immunoglobulin G, Protein-Tyrosine Kinases therapeutic use, Muscles, Receptors, Cholinergic therapeutic use, Myasthenia Gravis, Autoimmune, Experimental drug therapy
- Abstract
Muscle-specific tyrosine kinase myasthenia gravis (MuSK MG) is an autoimmune disease that causes life-threatening muscle weakness due to anti-MuSK autoantibodies that disrupt neuromuscular junction signaling. To avoid chronic immunosuppression from current therapies, we engineered T cells to express a MuSK chimeric autoantibody receptor with CD137-CD3ζ signaling domains (MuSK-CAART) for precision targeting of B cells expressing anti-MuSK autoantibodies. MuSK-CAART demonstrated similar efficacy as anti-CD19 chimeric antigen receptor T cells for depletion of anti-MuSK B cells and retained cytolytic activity in the presence of soluble anti-MuSK antibodies. In an experimental autoimmune MG mouse model, MuSK-CAART reduced anti-MuSK IgG without decreasing B cells or total IgG levels, reflecting MuSK-specific B cell depletion. Specific off-target interactions of MuSK-CAART were not identified in vivo, in primary human cell screens or by high-throughput human membrane proteome array. These data contributed to an investigational new drug application and phase 1 clinical study design for MuSK-CAART for the treatment of MuSK autoantibody-positive MG., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
11. CD38 as a pan-hematologic target for chimeric antigen receptor T cells.
- Author
-
Glisovic-Aplenc T, Diorio C, Chukinas JA, Veliz K, Shestova O, Shen F, Nunez-Cruz S, Vincent TL, Miao F, Milone MC, June CH, Teachey DT, Tasian SK, Aplenc R, and Gill S
- Subjects
- Adult, Animals, Child, Humans, Mice, T-Lymphocytes, Hematologic Neoplasms therapy, Hematologic Neoplasms metabolism, Leukemia, Myeloid, Acute pathology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism
- Abstract
Many hematologic malignancies are not curable with chemotherapy and require novel therapeutic approaches. Chimeric antigen receptor (CAR) T-cell therapy is 1 such approach that involves the transfer of T cells engineered to express CARs for a specific cell-surface antigen. CD38 is a validated tumor antigen in multiple myeloma (MM) and T-cell acute lymphoblastic leukemia (T-ALL) and is also overexpressed in acute myeloid leukemia (AML). Here, we developed human CD38-redirected T cells (CART-38) as a unified approach to treat 3 different hematologic malignancies that occur across the pediatric-to-adult age spectrum. Importantly, CD38 expression on activated T cells did not impair CART-38 cells expansion or in vitro function. In xenografted mice, CART-38 mediated the rejection of AML, T-ALL, and MM cell lines and primary samples and prolonged survival. In a xenograft model of normal human hematopoiesis, CART-38 resulted in the expected reduction of hematopoietic progenitors, which warrants caution and careful monitoring of this potential toxicity when translating this new immunotherapy into the clinic. Deploying CART-38 against multiple CD38-expressing malignancies is significant because it expands the potential for this novel therapy to affect diverse patient populations., (© 2023 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
- 2023
- Full Text
- View/download PDF
12. Association of Urine Fentanyl Concentration With Severity of Opioid Withdrawal Among Patients Presenting to the Emergency Department.
- Author
-
Thakrar AP, Faude S, Perrone J, Milone MC, Lowenstein M, Snider CK, Spadaro A, Delgado MK, Nelson LS, and Kilaru AS
- Subjects
- Humans, Female, Retrospective Studies, Cross-Sectional Studies, Fentanyl adverse effects, Narcotics, Emergency Service, Hospital, Analgesics, Opioid adverse effects, Analgesics, Opioid urine, Drug Overdose
- Abstract
Background and Aims: Fentanyl is involved in most US drug overdose deaths and its use can complicate opioid withdrawal management. Clinical applications of quantitative urine fentanyl testing have not been demonstrated previously. The aim of this study was to determine whether urine fentanyl concentration is associated with severity of opioid withdrawal., Design: This is a retrospective cross-sectional study., Setting: This study was conducted in 3 emergency departments in an urban, academic health system from January 1, 2020, to December 31, 2021., Participants: This study included patients with opioid use disorder, detectable urine fentanyl or norfentanyl, and Clinical Opiate Withdrawal Scale (COWS) recorded within 6 hours of urine drug testing., Measurements: The primary exposure was urine fentanyl concentration stratified as high (>400 ng/mL), medium (40-399 ng/mL), or low (<40 ng/mL). The primary outcome was opioid withdrawal severity measured with COWS within 6 hours before or after urine specimen collection. We used a generalized linear model with γ distribution and log-link function to estimate the adjusted association between COWS and the exposures., Findings: For the 1127 patients in our sample, the mean age (SD) was 40.0 (10.7), 384 (34.1%) identified as female, 332 (29.5%) reported their race/ethnicity as non-Hispanic Black, and 658 (58.4%) reported their race/ethnicity as non-Hispanic White. For patients with high urine fentanyl concentrations, the adjusted mean COWS (95% confidence interval) was 4.4 (3.9-4.8) compared with 5.5 (5.1-6.0) among those with medium and 7.7 (6.8-8.7) among those with low fentanyl concentrations., Conclusions: Lower urine fentanyl concentration was associated with more severe opioid withdrawal, suggesting potential clinical applications for quantitative urine measurements in evolving approaches to fentanyl withdrawal management., Competing Interests: The authors report no conflicts of interest., (Copyright © 2023 American Society of Addiction Medicine.)
- Published
- 2023
- Full Text
- View/download PDF
13. Anti-BCMA/CD19 CAR T Cells with Early Immunomodulatory Maintenance for Multiple Myeloma Responding to Initial or Later-Line Therapy.
- Author
-
Garfall AL, Cohen AD, Susanibar-Adaniya SP, Hwang WT, Vogl DT, Waxman AJ, Lacey SF, Gonzalez VE, Fraietta JA, Gupta M, Kulikovskaya I, Tian L, Chen F, Koterba N, Bartoszek RL, Patchin M, Xu R, Plesa G, Siegel DL, Brennan A, Nelson AM, Ferthio R, Cosey A, Shea KM, Leskowitz R, Four M, Wilson WV, Miao F, Lancaster E, Carreno BM, Linette GP, Hexner EO, Young RM, Bu D, Mansfield KG, Brogdon JL, June CH, Milone MC, and Stadtmauer EA
- Subjects
- Humans, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Lenalidomide therapeutic use, Antigens, CD19 therapeutic use, T-Lymphocytes, Multiple Myeloma therapy, Receptors, Chimeric Antigen therapeutic use
- Abstract
We conducted a phase I clinical trial of anti-BCMA chimeric antigen receptor T cells (CART-BCMA) with or without anti-CD19 CAR T cells (huCART19) in multiple myeloma (MM) patients responding to third- or later-line therapy (phase A, N = 10) or high-risk patients responding to first-line therapy (phase B, N = 20), followed by early lenalidomide or pomalidomide maintenance. We observed no high-grade cytokine release syndrome (CRS) and only one instance of low-grade neurologic toxicity. Among 15 subjects with measurable disease, 10 exhibited partial response (PR) or better; among 26 subjects responding to prior therapy, 9 improved their response category and 4 converted to minimal residual disease (MRD)-negative complete response/stringent complete response. Early maintenance therapy was safe, feasible, and coincided in some patients with CAR T-cell reexpansion and late-onset, durable clinical response. Outcomes with CART-BCMA + huCART19 were similar to CART-BCMA alone. Collectively, our results demonstrate favorable safety, pharmacokinetics, and antimyeloma activity of dual-target CAR T-cell therapy in early lines of MM treatment., Significance: CAR T cells in early lines of MM therapy could be safer and more effective than in the advanced setting, where prior studies have focused. We evaluated the safety, pharmacokinetics, and efficacy of CAR T cells in patients with low disease burden, responding to current therapy, combined with standard maintenance therapy. This article is highlighted in the In This Issue feature, p. 101., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
14. The IAP antagonist birinapant enhances chimeric antigen receptor T cell therapy for glioblastoma by overcoming antigen heterogeneity.
- Author
-
Song EZ, Wang X, Philipson BI, Zhang Q, Thokala R, Zhang L, Assenmacher CA, Binder ZA, Ming GL, O'Rourke DM, Song H, and Milone MC
- Abstract
Antigen heterogeneity that results in tumor antigenic escape is one of the major obstacles to successful chimeric antigen receptor (CAR) T cell therapies in solid tumors including glioblastoma multiforme (GBM). To address this issue and improve the efficacy of CAR T cell therapy for GBM, we developed an approach that combines CAR T cells with inhibitor of apoptosis protein (IAP) antagonists, a new class of small molecules that mediate the degradation of IAPs, to treat GBM. Here, we demonstrated that the IAP antagonist birinapant could sensitize GBM cell lines and patient-derived primary GBM organoids to apoptosis induced by CAR T cell-derived cytokines, such as tumor necrosis factor. Therefore, birinapant could enhance CAR T cell-mediated bystander death of antigen-negative GBM cells, thus preventing tumor antigenic escape in antigen-heterogeneous tumor models in vitro and in vivo . In addition, birinapant could promote the activation of NF-κB signaling pathways in antigen-stimulated CAR T cells, and with a birinapant-resistant tumor model we showed that birinapant had no deleterious effect on CAR T cell functions in vitro and in vivo . Overall, we demonstrated the potential of combining the IAP antagonist birinapant with CAR T cells as a novel and feasible approach to overcoming tumor antigen heterogeneity and enhancing CAR T cell therapy for GBM., Competing Interests: R.T., Z.A.B., D.M.O., and M.C.M. are inventors on multiple patents relating to CAR T cell therapy for GBM. M.C.M. is also a founder and scientific advisor for Verismo Therapeutics., (© 2022.)
- Published
- 2022
- Full Text
- View/download PDF
15. Generation of non-human primate CAR Tregs using artificial antigen-presenting cells, simian tropic lentiviral vectors, and antigen-specific restimulation.
- Author
-
Ellis GI, Deng MZ, Winn DW, Coker KE, Shukla D, Bhoj V, Milone MC, Duran-Struuck R, and Riley JL
- Subjects
- Animals, Antigen-Presenting Cells, T-Lymphocytes, Regulatory, Primates, Transplantation Tolerance, Receptors, Chimeric Antigen genetics
- Abstract
It is technically challenging to generate large doses of regulatory T cells (Tregs) engineered to express a chimeric antigen receptor (CAR) in non-human primates (NHP). Here, we have optimized the manufacturing of CAR Tregs by stringent sorting of Tregs, stimulation by artificial antigen-presenting cells, transduction by simian tropic lentiviral vectors, and antigen-specific expansion. The result of this method is highly suppressive CAR Tregs for use in a pre-clinical, large animal model of transplant tolerance. For complete details on the use and execution of this protocol, please refer to Ellis et al. (2022)., Competing Interests: J.L.R. and G.I.E. have submitted patents related to use of the Bw6 CAR and the aAPCs described in this manuscript. J.L.R. is a founder and equity holder of Tmunity Therapeutics., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
16. 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
17. An Epidemic within a Pandemic: An 8-Year-Old Boy with Xylazine-Complicating Fentanyl Poisoning, and Drug Detection Challenges.
- Author
-
Gill EL, Mack EA, Osterhoudt KC, Shaw LM, and Milone MC
- Subjects
- Male, Humans, Child, Fentanyl, Xylazine, Pandemics
- Abstract
Competing Interests: Authors’ Disclosures or Potential Conflicts of Interest: No authors declared any potential conflicts of interest.
- Published
- 2022
- Full Text
- View/download PDF
18. CRISPR/Cas9-mediated Gene Knockout Followed by Negative Selection Leads to a Complete TCR Depletion in ortho CAR19 T Cells.
- Author
-
Zhang Q, Yang J, Manoharan ENEA, Yu AB, and Milone MC
- Abstract
Genome-editing technologies, especially CRISPR (clustered regularly interspaced short palindrome repeats)/Cas9 (CRISPR-associated protein 9), endows researchers the ability to make efficient, simple , and precise genomic DNA changes in many eukaryotic cell types. CRISPR/Cas9-mediated efficient gene knockout holds huge potential to improve the efficacy and safety of chimeric antigen receptor (CAR) T cell-based immunotherapies. Here, we describe an optimized approach for a complete loss of endogenous T cell receptor (TCR) protein expression, by CRISPR/Cas9-mediated TCR α constant (TRAC) and TCR β constant (TRBC) gene knockout, followed by subsequent CD3 negative selection in engineered human ortho CAR19 T cells. We believe this method can be expanded beyond CAR T cell application, and target other cell surface receptors. Graphical abstract: Schematic overview of the two-step process of endogenous TCR depletion in engineered human ortho CAR19 T cells using (1) CRISPR/Cas9-mediated gene knockout followed by (2) CD3 negative selection., Competing Interests: Competing interests The authors have no financial and/or non-financial competing interests to disclose., (Copyright © 2022 The Authors; exclusive licensee Bio-protocol LLC.)
- Published
- 2022
- Full Text
- View/download PDF
19. Targeting PARP11 to avert immunosuppression and improve CAR T therapy in solid tumors.
- Author
-
Zhang H, Yu P, Tomar VS, Chen X, Atherton MJ, Lu Z, Zhang HG, Li S, Ortiz A, Gui J, Leu NA, Yan F, Blanco A, Meyer-Ficca ML, Meyer RG, Beiting DP, Li J, Nunez-Cruz S, O'Connor RS, Johnson LR, Minn AJ, George SS, Koumenis C, Diehl JA, Milone MC, Zheng H, and Fuchs SY
- Subjects
- Humans, Immunosuppression Therapy, Immunotherapy, Adoptive, Tumor Microenvironment, Neoplasms drug therapy, Poly(ADP-ribose) Polymerases metabolism, Receptors, Chimeric Antigen genetics
- Abstract
Evasion of antitumor immunity and resistance to therapies in solid tumors are aided by an immunosuppressive tumor microenvironment (TME). We found that TME factors, such as regulatory T cells and adenosine, downregulated type I interferon receptor IFNAR1 on CD8
+ cytotoxic T lymphocytes (CTLs). These events relied upon poly-ADP ribose polymerase-11 (PARP11), which was induced in intratumoral CTLs and acted as a key regulator of the immunosuppressive TME. Ablation of PARP11 prevented loss of IFNAR1, increased CTL tumoricidal activity and inhibited tumor growth in an IFNAR1-dependent manner. Accordingly, genetic or pharmacologic inactivation of PARP11 augmented the therapeutic benefits of chimeric antigen receptor T cells. Chimeric antigen receptor CTLs engineered to inactivate PARP11 demonstrated a superior efficacy against solid tumors. These findings highlight the role of PARP11 in the immunosuppressive TME and provide a proof of principle for targeting this pathway to optimize immune therapies., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
20. Trafficking and persistence of alloantigen-specific chimeric antigen receptor regulatory T cells in Cynomolgus macaque.
- Author
-
Ellis GI, Coker KE, Winn DW, Deng MZ, Shukla D, Bhoj V, Milone MC, Wang W, Liu C, Naji A, Duran-Struuck R, and Riley JL
- Subjects
- Adoptive Transfer, Animals, Macaca, T-Lymphocytes, Regulatory, Isoantigens, Receptors, Chimeric Antigen genetics
- Abstract
Adoptive transfer of chimeric antigen receptor regulatory T cells (CAR Tregs) is a promising way to prevent allograft loss without the morbidity associated with current therapies. Non-human primates (NHPs) are a clinically relevant model to develop transplant regimens, but manufacturing and engraftment of NHP CAR Tregs have not been demonstrated yet. Here, we describe a culture system that massively expands CAR Tregs specific for the Bw6 alloantigen. In vitro, these Tregs suppress in an antigen-specific manner without pro-inflammatory cytokine secretion or cytotoxicity. In vivo, Bw6-specific CAR Tregs preferentially traffic to and persist in bone marrow for at least 1 month. Following transplant of allogeneic Bw6
+ islets and autologous CAR Tregs into the bone marrow of diabetic recipients, CAR Tregs traffic to the site of islet transplantation and maintain a phenotype of suppressive Tregs. Our results establish a framework for the optimization of CAR Treg therapy in NHP disease models., Competing Interests: Declaration of interests J.L.R. and G.I.E. have submitted patents related to use of the HLA-A2 and Bw6 CARs described in this manuscript. J.L.R. is an equity holder of and receives sponsored research funding from Tmunity Therapeutics. The remaining authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
21. Rapid manufacturing of non-activated potent CAR T cells.
- Author
-
Ghassemi S, Durgin JS, Nunez-Cruz S, Patel J, Leferovich J, Pinzone M, Shen F, Cummins KD, Plesa G, Cantu VA, Reddy S, Bushman FD, Gill SI, O'Doherty U, O'Connor RS, and Milone MC
- Subjects
- Animals, Humans, Immunotherapy, Adoptive methods, Mice, T-Lymphocytes, Leukemia, Receptors, Chimeric Antigen
- Abstract
Chimaeric antigen receptor (CAR) T cells can generate durable clinical responses in B-cell haematologic malignancies. The manufacturing of these T cells typically involves their activation, followed by viral transduction and expansion ex vivo for at least 6 days. However, the activation and expansion of CAR T cells leads to their progressive differentiation and the associated loss of anti-leukaemic activity. Here we show that functional CAR T cells can be generated within 24 hours from T cells derived from peripheral blood without the need for T-cell activation or ex vivo expansion, and that the efficiency of viral transduction in this process is substantially influenced by the formulation of the medium and the surface area-to-volume ratio of the culture vessel. In mouse xenograft models of human leukaemias, the rapidly generated non-activated CAR T cells exhibited higher anti-leukaemic in vivo activity per cell than the corresponding activated CAR T cells produced using the standard protocol. The rapid manufacturing of CAR T cells may reduce production costs and broaden their applicability., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
22. A human orthogonal IL-2 and IL-2Rβ system enhances CAR T cell expansion and antitumor activity in a murine model of leukemia.
- Author
-
Zhang Q, Hresko ME, Picton LK, Su L, Hollander MJ, Nunez-Cruz S, Zhang Z, Assenmacher CA, Sockolosky JT, Garcia KC, and Milone MC
- Subjects
- Animals, Antigens, CD19 metabolism, Cell Proliferation, Disease Models, Animal, Humans, Immunotherapy, Adoptive, Mice, T-Lymphocytes, Xenograft Model Antitumor Assays, Interleukin-2 metabolism, Leukemia metabolism
- Abstract
Interleukin-2 (IL-2) is a central T cell cytokine that promotes T cell proliferation and effector function; however, toxicity due to its pluripotency limits its application to enhance CAR T cell immunotherapy. Previously, mouse IL-2 and its cognate receptor were engineered to create an orthogonal ( ortho ) cytokine-cytokine receptor pair capable of delivering an IL-2 signal without toxicity. Here, we engineered a human orthogonal IL-2 ( ortho- hIL-2) and human orthogonal IL-2Rβ ( ortho- hIL-2Rβ) pair, containing human-specific mutations. Ortho- hIL-2 is selective toward ortho- hIL-2Rβ–expressing cells with no appreciable signaling on wild-type T cells. Ortho- hIL-2 induces IL-2 receptor signaling and supports proliferation of both an IL-2–dependent cell line and primary T cells transduced to express the ortho- hIL-2Rβ. Using CD19-specific chimeric antigen receptor (CAR) T cells, we show that ortho -hIL-2 induces a dose-dependent increase in ortho -hIL-2Rβ
+ CAR T cell expansion in vivo by as much as 1000-fold at 2 weeks after adoptive transfer into immunodeficient mice bearing CD19+ Nalm6 leukemia xenografts. Ortho -hIL-2 can rescue the antileukemic effect of an otherwise suboptimal CAR T cell dose. In addition, ortho -hIL-2 administration initiated at the time of leukemic relapse after CAR T cell therapy can rescue an otherwise failed antileukemic response. These data highlight the potential of combining an orthogonal cytokine approach with T cell–based immunotherapies to augment the antitumor efficacy of engineered T cells.- Published
- 2021
- Full Text
- View/download PDF
23. 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
24. Evaluation of a Nanoparticle-Based Busulfan Immunoassay for Rapid Analysis on Routine Clinical Analyzers.
- Author
-
Hilaire MR, Gill RV, Courtney JB, Baburina I, Gardiner J, Milone MC, Shaw LM, Meng QH, and Salamone SJ
- Subjects
- Chromatography, High Pressure Liquid methods, Drug Monitoring methods, Humans, Immunoassay methods, Reproducibility of Results, Tandem Mass Spectrometry methods, Busulfan, Nanoparticles
- Abstract
Background: Busulfan is an alkylating agent used in allogeneic hematopoietic stem cell transplantation for various malignant and nonmalignant disorders. Therapeutic drug monitoring of busulfan is common because busulfan exposure has been linked to veno-occlusive disease, disease relapse, and failed engraftment. The authors developed an automated immunoassay, along with stable calibrators and controls, and quantified busulfan in sodium heparin plasma., Methods: The authors evaluated a homogenous nanoparticle immunoassay, the MyCare Oncology Busulfan Assay Kit (Saladax Biomedical, Inc), for precision, sensitivity, accuracy, and linearity on an open channel clinical chemistry analyzer; they compared the method with 2 mass spectrometry methods (liquid chromatography-tandem mass spectrometry and gas chromatography/mass spectrometry), using anonymized, remnant patient samples., Results: The coefficients of variation for repeatability and within-laboratory precision were ≤9.0%. The linear range was 150-2000 ng/mL; samples up to 6000 ng/mL can be measured with sample dilution. Measured values deviated by ≤14% from assigned values. Comparison between validated mass spectrometry methods resulted in a correlation coefficient R ≥ 0.995., Conclusions: The MyCare Busulfan Assay Kit shows the precision, accuracy, linearity, and test range for performing busulfan concentration measurements in sodium heparin plasma on routine clinical chemistry analyzers., Competing Interests: The authors declare no conflict of interest., (Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the International Association of Therapeutic Drug Monitoring and Clinical Toxicology.)
- Published
- 2021
- Full Text
- View/download PDF
25. Multi-Site Evaluation of Immunoassays for Antipsychotic Drug Measurement in Clinical Samples.
- Author
-
Clarke WA, Salyer B, Hussey C, Gardiner J, Johnson-Davis K, and Milone MC
- Subjects
- Chromatography, Liquid, Humans, Immunoassay, Tandem Mass Spectrometry, Antipsychotic Agents therapeutic use, Clozapine
- Abstract
Background: Atypical antipsychotic drugs are frequently used in the treatment of serious mental illness (SMI), specifically schizophrenia and bipolar disorder. Adherence to these prescribed drug regimens is a challenge to successful treatment with these drugs. For some of the more common drugs in this class, novel turbidimetric immunoassays have been developed for therapeutic drug monitoring (TDM) to aid in the management of patients prescribed these drugs., Methods: Immunoassays for aripiprazole, clozapine, olanzapine, paliperidone, quetiapine, and risperidone were set up at 2 centers: Johns Hopkins Hospital (JHH) on the Roche Cobas® c501, and the Hospital of the University of Pennsylvania (HUP) on the Beckman AU480. Assay imprecision, limit of quantification (LOQ), functional sensitivity, linearity, and recovery were assessed. Remnant clinical samples were obtained from a reference laboratory (ARUP), and immunoassay results were compared with those obtained by LC-MS/MS., Results: Imprecision at both sites for all analytes and concentrations tested was <10%. The manufacturer's LOQ was confirmed for each assay, and the functional sensitivity for each assay was found to be lower than the LOQ. All assays were found to be linear over the measuring range, with recoveries ranging from 91% to 123%. For method comparison, Deming regression slopes were found to be between 0.84 to 1.28., Conclusion: The immunoassays evaluated here are suitable for quantifying drug concentrations to be used in TDM for all 6 drugs. Commercialization of these assays will enable increased access for TDM in psychiatric patient management., (© American Association for Clinical Chemistry 2021. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
26. 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
27. High-Affinity Chimeric Antigen Receptor With Cross-Reactive scFv to Clinically Relevant EGFR Oncogenic Isoforms.
- Author
-
Thokala R, Binder ZA, Yin Y, Zhang L, Zhang JV, Zhang DY, Milone MC, Ming GL, Song H, and O'Rourke DM
- Abstract
Tumor heterogeneity is a key reason for therapeutic failure and tumor recurrence in glioblastoma (GBM). Our chimeric antigen receptor (CAR) T cell (2173 CAR T cells) clinical trial (NCT02209376) against epidermal growth factor receptor (EGFR) variant III (EGFRvIII) demonstrated successful trafficking of T cells across the blood-brain barrier into GBM active tumor sites. However, CAR T cell infiltration was associated only with a selective loss of EGFRvIII+ tumor, demonstrating little to no effect on EGFRvIII
- tumor cells. Post-CAR T-treated tumor specimens showed continued presence of EGFR amplification and oncogenic EGFR extracellular domain (ECD) missense mutations, despite loss of EGFRvIII. To address tumor escape, we generated an EGFR-specific CAR by fusing monoclonal antibody (mAb) 806 to a 4-1BB co-stimulatory domain. The resulting construct was compared to 2173 CAR T cells in GBM, using in vitro and in vivo models. 806 CAR T cells specifically lysed tumor cells and secreted cytokines in response to amplified EGFR, EGFRvIII, and EGFR-ECD mutations in U87MG cells, GBM neurosphere-derived cell lines, and patient-derived GBM organoids. 806 CAR T cells did not lyse fetal brain astrocytes or primary keratinocytes to a significant degree. They also exhibited superior antitumor activity in vivo when compared to 2173 CAR T cells. The broad specificity of 806 CAR T cells to EGFR alterations gives us the potential to target multiple clones within a tumor and reduce opportunities for tumor escape via antigen loss., Competing Interests: The described work involves patent applications owned by the University of Pennsylvania. MM is an inventor on multiple issued and pending patents related to CAR T cell technology used in this study. These patents are assigned to the University of Pennsylvania and have been licensed to third parties for which royalties have or may be received. 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 Thokala, Binder, Yin, Zhang, Zhang, Zhang, Milone, Ming, Song and O’Rourke.)- Published
- 2021
- Full Text
- View/download PDF
28. CAR T-Cells Depend on the Coupling of NADH Oxidation with ATP Production.
- Author
-
Garcia-Canaveras JC, Heo D, Trefely S, Leferovich J, Xu C, Philipson BI, Ghassemi S, Milone MC, Moon EK, Snyder NW, June CH, Rabinowitz JD, and O'Connor RS
- Subjects
- Adult, Animals, Female, Humans, Levilactobacillus brevis metabolism, Male, Mice, Mice, Inbred NOD, Mice, SCID, NAD metabolism, Oxidation-Reduction, T-Lymphocytes metabolism, Adenosine Triphosphate metabolism, Multienzyme Complexes metabolism, NADH, NADPH Oxidoreductases metabolism, Receptors, Antigen, T-Cell metabolism
- Abstract
The metabolic milieu of solid tumors provides a barrier to chimeric antigen receptor (CAR) T-cell therapies. Excessive lactate or hypoxia suppresses T-cell growth, through mechanisms including NADH buildup and the depletion of oxidized metabolites. NADH is converted into NAD
+ by the enzyme Lactobacillus brevis NADH Oxidase ( LbNOX ), which mimics the oxidative function of the electron transport chain without generating ATP. Here we determine if LbNOX promotes human CAR T-cell metabolic activity and antitumor efficacy. CAR T-cells expressing LbNOX have enhanced oxygen as well as lactate consumption and increased pyruvate production. LbNOX renders CAR T-cells resilient to lactate dehydrogenase inhibition. But in vivo in a model of mesothelioma, CAR T-cell's expressing LbNOX showed no increased antitumor efficacy over control CAR T-cells. We hypothesize that T cells in hostile environments face dual metabolic stressors of excessive NADH and insufficient ATP production. Accordingly, futile T-cell NADH oxidation by LbNOX is insufficient to promote tumor clearance.- Published
- 2021
- Full Text
- View/download PDF
29. 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
30. 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
31. Efficacy and Safety of Hydroxychloroquine vs Placebo for Pre-exposure SARS-CoV-2 Prophylaxis Among Health Care Workers: A Randomized Clinical Trial.
- Author
-
Abella BS, Jolkovsky EL, Biney BT, Uspal JE, Hyman MC, Frank I, Hensley SE, Gill S, Vogl DT, Maillard I, Babushok DV, Huang AC, Nasta SD, Walsh JC, Wiletyo EP, Gimotty PA, Milone MC, and Amaravadi RK
- Subjects
- Adult, COVID-19 epidemiology, Double-Blind Method, Female, Hospitals, Urban, Humans, Incidence, Male, Pennsylvania epidemiology, SARS-CoV-2, COVID-19 prevention & control, Cross Infection prevention & control, Cross Infection virology, Hydroxychloroquine administration & dosage, Personnel, Hospital, Pre-Exposure Prophylaxis, COVID-19 Drug Treatment
- Abstract
Importance: Health care workers (HCWs) caring for patients with coronavirus disease 2019 (COVID-19) are at risk of exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Currently, to our knowledge, there is no effective pharmacologic prophylaxis for individuals at risk., Objective: To evaluate the efficacy of hydroxychloroquine to prevent transmission of SARS-CoV-2 in hospital-based HCWs with exposure to patients with COVID-19 using a pre-exposure prophylaxis strategy., Design, Setting, and Participants: This randomized, double-blind, placebo-controlled clinical trial (the Prevention and Treatment of COVID-19 With Hydroxychloroquine Study) was conducted at 2 tertiary urban hospitals, with enrollment from April 9, 2020, to July 14, 2020; follow-up ended August 4, 2020. The trial randomized 132 full-time, hospital-based HCWs (physicians, nurses, certified nursing assistants, emergency technicians, and respiratory therapists), of whom 125 were initially asymptomatic and had negative results for SARS-CoV-2 by nasopharyngeal swab. The trial was terminated early for futility before reaching a planned enrollment of 200 participants., Interventions: Hydroxychloroquine, 600 mg, daily, or size-matched placebo taken orally for 8 weeks., Main Outcomes and Measures: The primary outcome was the incidence of SARS-CoV-2 infection as determined by a nasopharyngeal swab during the 8 weeks of treatment. Secondary outcomes included adverse effects, treatment discontinuation, presence of SARS-CoV-2 antibodies, frequency of QTc prolongation, and clinical outcomes for SARS-CoV-2-positive participants., Results: Of the 132 randomized participants (median age, 33 years [range, 20-66 years]; 91 women [69%]), 125 (94.7%) were evaluable for the primary outcome. There was no significant difference in infection rates in participants randomized to receive hydroxychloroquine compared with placebo (4 of 64 [6.3%] vs 4 of 61 [6.6%]; P > .99). Mild adverse events were more common in participants taking hydroxychloroquine compared with placebo (45% vs 26%; P = .04); rates of treatment discontinuation were similar in both arms (19% vs 16%; P = .81). The median change in QTc (baseline to 4-week evaluation) did not differ between arms (hydroxychloroquine: 4 milliseconds; 95% CI, -9 to 17; vs placebo: 3 milliseconds; 95% CI, -5 to 11; P = .98). Of the 8 participants with positive results for SARS-CoV-2 (6.4%), 6 developed viral symptoms; none required hospitalization, and all clinically recovered., Conclusions and Relevance: In this randomized clinical trial, although limited by early termination, there was no clinical benefit of hydroxychloroquine administered daily for 8 weeks as pre-exposure prophylaxis in hospital-based HCWs exposed to patients with COVID-19., Trial Registration: ClinicalTrials.gov Identifier: NCT04329923.
- Published
- 2021
- Full Text
- View/download PDF
32. Adoptive T cell immunotherapy for medullary thyroid carcinoma targeting GDNF family receptor alpha 4.
- Author
-
Bhoj VG, Li L, Parvathaneni K, Zhang Z, Kacir S, Arhontoulis D, Zhou K, McGettigan-Croce B, Nunez-Cruz S, Gulendran G, Boesteanu AC, Johnson L, Feldman MD, Radaelli E, Mansfield K, Nasrallah M, Goydel RS, Peng H, Rader C, Milone MC, and Siegel DL
- Abstract
Metastatic medullary thyroid cancer (MTC) is a rare but often aggressive thyroid malignancy with a 5-year survival rate of less than 40% and few effective therapeutic options. Adoptive T cell immunotherapy using chimeric antigen receptor (CAR)-modified T cells (CAR Ts) is showing encouraging results in the treatment of cancer, but development is challenged by the availability of suitable target antigens. We identified glial-derived neurotrophic factor (GDNF) family receptor alpha 4 (GFRα4) as a putative antigen target for CAR-based therapy of MTC. We show that GFRα4 is highly expressed in MTC, in parafollicular cells within the thyroid from which MTC originates, and in normal thymus. We isolated two single-chain variable fragments (scFvs) targeting GFRα4 isoforms a and b by antibody phage display. CARs bearing the CD3ζ and the CD137 costimulatory domains were constructed using these GFRα4-specific scFvs. GFRα4-specific CAR Ts trigger antigen-dependent cytotoxicity and cytokine production in vitro , and they are able to eliminate tumors derived from the MTC TT cell line in an immunodeficient mouse xenograft model of MTC. These data demonstrate the feasibility of targeting GFRα4 by CAR T and support this antigen as a promising target for adoptive T cell immunotherapy and other antibody-based therapies for MTC., Competing Interests: D.L.S., V.G.B., C.R., R.S.G., and M.C.M. have filed a patent application on technology presented in this manuscript., (© 2021.)
- Published
- 2021
- Full Text
- View/download PDF
33. Pharmacology of Chimeric Antigen Receptor-Modified T Cells.
- Author
-
Song EZ and Milone MC
- Subjects
- B-Cell Maturation Antigen, Humans, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes metabolism, Tissue Distribution, Multiple Myeloma, Receptors, Chimeric Antigen metabolism
- Abstract
Cell-based immunotherapies using T cells that are engineered to express a chimeric antigen receptor (CAR-T cells) are an effective treatment option for several B cell malignancies. Compared with most drugs, CAR-T cell products are highly complex, as each cell product is composed of a heterogeneous mixture of millions of cells. The biodistribution and kinetics of CAR-T cells, following administration, are unique given the ability of T cells to actively migrate as well as replicate within the patient. CAR-T cell therapies also have multiple mechanisms of action that contribute to both their antitumor activity and their toxicity. This review provides an overview of the unique pharmacology of CAR-T cells, with a focus on CD19-targeting and B cell maturation antigen (BCMA)-targeting CAR-T cells.
- Published
- 2021
- Full Text
- View/download PDF
34. Itacitinib (INCB039110), a JAK1 Inhibitor, Reduces Cytokines Associated with Cytokine Release Syndrome Induced by CAR T-cell Therapy.
- Author
-
Huarte E, O'Connor RS, Peel MT, Nunez-Cruz S, Leferovich J, Juvekar A, Yang YO, Truong L, Huang T, Naim A, Milone MC, and Smith PA
- Subjects
- Animals, Apoptosis, Cell Proliferation, Cytokine Release Syndrome etiology, Cytokine Release Syndrome pathology, Female, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, SCID, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Azetidines pharmacology, Cytokine Release Syndrome drug therapy, Cytokines antagonists & inhibitors, Immunotherapy, Adoptive adverse effects, Isonicotinic Acids pharmacology, Janus Kinase 1 antagonists & inhibitors, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Purpose: T cells engineered to express a chimeric antigen receptor (CAR) are a promising cancer immunotherapy. Such targeted therapies have shown long-term relapse-free survival in patients with B-cell leukemia and lymphoma. However, cytokine release syndrome (CRS) represents a serious, potentially life-threatening side effect often associated with CAR T-cell therapy. CRS manifests as a rapid (hyper)immune reaction driven by excessive inflammatory cytokine release, including IFNγ and IL6., Experimental Design: Many cytokines implicated in CRS are known to signal through the JAK-STAT pathway. Here we study the effect of blocking JAK pathway signaling on CAR T-cell proliferation, antitumor activity, and cytokine levels in in vitro and in vivo models., Results: We report that itacitinib, a potent, selective JAK1 inhibitor, was able to significantly and dose-dependently reduce levels of multiple cytokines implicated in CRS in several in vitro and in vivo models. Importantly, we also report that at clinically relevant doses that mimic human JAK1 pharmacologic inhibition, itacitinib did not significantly inhibit proliferation or antitumor killing capacity of three different human CAR T-cell constructs (GD2, EGFR, and CD19). Finally, in an in vivo model, antitumor activity of CD19-CAR T cells adoptively transferred into CD19
+ tumor-bearing immunodeficient animals was unabated by oral itacitinib treatment., Conclusions: Together, these data suggest that itacitinib has potential as a prophylactic agent for the prevention of CAR T cell-induced CRS, and a phase II clinical trial of itacitinib for prevention of CRS induced by CAR T-cell therapy has been initiated (NCT04071366)., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
35. Antigen-specific B cell depletion for precision therapy of mucosal pemphigus vulgaris.
- Author
-
Lee J, Lundgren DK, Mao X, Manfredo-Vieira S, Nunez-Cruz S, Williams EF, Assenmacher CA, Radaelli E, Oh S, Wang B, Ellebrecht CT, Fraietta JA, Milone MC, and Payne AS
- Subjects
- Adult, Animals, Autoantibodies immunology, B-Lymphocytes pathology, Desmoglein 3 genetics, Desmoglein 3 immunology, Disease Models, Animal, Female, Humans, Interferon-gamma immunology, Isoantigens genetics, Isoantigens immunology, Male, Mice, Mice, Inbred NOD, Mice, SCID, Pemphigus genetics, Pemphigus immunology, Pemphigus pathology, Adoptive Transfer, B-Lymphocytes immunology, Lymphocyte Depletion, Pemphigus therapy, Precision Medicine
- Abstract
Desmoglein 3 chimeric autoantibody receptor T cells (DSG3-CAART) expressing the pemphigus vulgaris (PV) autoantigen DSG3 fused to CD137-CD3ζ signaling domains, represent a precision cellular immunotherapy approach for antigen-specific B cell depletion. Here, we present definitive preclinical studies enabling a first-in-human trial of DSG3-CAART for mucosal PV. DSG3-CAART specifically lysed human anti-DSG3 B cells from PV patients and demonstrated activity consistent with a threshold dose in vivo, resulting in decreased target cell burden, decreased serum and tissue-bound autoantibodies, and increased DSG3-CAART engraftment. In a PV active immune model with physiologic anti-DSG3 IgG levels, DSG3-CAART inhibited antibody responses against pathogenic DSG3 epitopes and autoantibody binding to epithelial tissues, leading to clinical and histologic resolution of blisters. DSG3 autoantibodies stimulated DSG3-CAART IFN-γ secretion and homotypic clustering, consistent with an activated phenotype. Toxicology screens using primary human cells and high-throughput membrane proteome arrays did not identify off-target cytotoxic interactions. These preclinical data guided the trial design for DSG3-CAART and may help inform CAART preclinical development for other antibody-mediated diseases.
- Published
- 2020
- Full Text
- View/download PDF
36. Stable HLA antibodies following sustained CD19+ cell depletion implicate a long-lived plasma cell source.
- Author
-
Zhang Z, Schuster SJ, Lacey SF, Milone MC, Monos D, and Bhoj VG
- Subjects
- Antibodies, Antigens, CD19, Plasma Cells
- Published
- 2020
- Full Text
- View/download PDF
37. Ligand-Induced Degradation of a CAR Permits Reversible Remote Control of CAR T Cell Activity In Vitro and In Vivo.
- Author
-
Richman SA, Wang LC, Moon EK, Khire UR, Albelda SM, and Milone MC
- Published
- 2020
- Full Text
- View/download PDF
38. Enhancing Chimeric Antigen Receptor T Cell Anti-tumor Function through Advanced Media Design.
- Author
-
Ghassemi S, Martinez-Becerra FJ, Master AM, Richman SA, Heo D, Leferovich J, Tu Y, García-Cañaveras JC, Ayari A, Lu Y, Wang A, Rabinowitz JD, Milone MC, June CH, and O'Connor RS
- Abstract
Effective chimeric antigen receptor (CAR)-T cell therapy is dependent on optimal cell culture methods conducive to the activation and expansion of T cells ex vivo , as well as infection with CAR. Media formulations used in CAR-T cell manufacturing have not been optimized for gene delivery, cell expansion, and overall potency. Bioactive components and derivatives that support the generation of functionally-competent T cell progeny with long-lasting persistence are largely undefined. Current media formulations rely on fetal bovine serum (FBS) or human serum (HS), which suffer from a lack of consistency or supply issues. We recognize that components of blood cellular fractions that are absent in serum may have therapeutic value. Here we investigate whether a concentrated growth factor extract, purified from human transfusion grade whole blood fractions, and marketed as PhysiologixTM xeno-free (XF) hGFC (Phx), supports CAR-T cell expansion and function. We show that Phx supports T cell proliferation in clinical and research-grade media. We also show that Phx treatment enhances lentiviral-mediated gene expression across a wide range of multiplicity of infections (MOIs). We compared the ability of anti-GD-2 CAR-T cells expanded ex vivo in medium conditioned with either Phx or HS to clear tumor burden in a human xenograft model of neuroblastoma. We show that T cells expanded in Phx have superior engraftment and potency in vivo , as well as CAR-induced cytolytic activity in vitro . Metabolomic profiling revealed several factors unique to Phx that may have relevance for CAR-T cell preclinical discovery, process development, and manufacturing. In particular, we show that carnosine, a biogenic amine modestly enriched in Phx relative to HS, enhances lentiviral gene delivery in activated T cells. By limiting extracellular acidification, carnosine enhances the metabolic fitness of T cells, shifting their metabolic profile from an acidic, stressed state toward an oxidative, energetic state. These findings are very informative regarding potential derivatives to include in medium customized for gene delivery and overall potency for T cell adoptive immunotherapies., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
39. 4-1BB costimulation promotes CAR T cell survival through noncanonical NF-κB signaling.
- Author
-
Philipson BI, O'Connor RS, May MJ, June CH, Albelda SM, and Milone MC
- Subjects
- Cell Line, Humans, Receptors, Chimeric Antigen genetics, Signal Transduction genetics, Tumor Necrosis Factor Receptor Superfamily, Member 9 genetics, NF-kappa B immunology, Receptors, Chimeric Antigen immunology, Signal Transduction immunology, T-Lymphocytes immunology, Tumor Necrosis Factor Receptor Superfamily, Member 9 immunology
- Abstract
Clinical response to chimeric antigen receptor (CAR) T cell therapy is correlated with CAR T cell persistence, especially for CAR T cells that target CD19
+ hematologic malignancies. 4-1BB-costimulated CAR (BBζ) T cells exhibit longer persistence after adoptive transfer than do CD28-costimulated CAR (28ζ) T cells. 4-1BB signaling improves T cell persistence even in the context of 28ζ CAR activation, which indicates distinct prosurvival signals mediated by the 4-1BB cytoplasmic domain. To specifically study signal transduction by CARs, we developed a cell-free, ligand-based activation and ex vivo culture system for CD19-specific CAR T cells. We observed greater ex vivo survival and subsequent expansion of BBζ CAR T cells when compared to 28ζ CAR T cells. We showed that only BBζ CARs activated noncanonical nuclear factor κB (ncNF-κB) signaling in T cells basally and that the anti-CD19 BBζ CAR further enhanced ncNF-κB signaling after ligand engagement. Reducing ncNF-κB signaling reduced the expansion and survival of anti-CD19 BBζ T cells and was associated with a substantial increase in the abundance of the most pro-apoptotic isoforms of Bim. Although our findings do not exclude the importance of other signaling differences between BBζ and 28ζ CARs, they demonstrate the necessary and nonredundant role of ncNF-κB signaling in promoting the survival of BBζ CAR T cells, which likely underlies the engraftment persistence observed with this CAR design., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
40. Imaging CAR T Cell Trafficking with eDHFR as a PET Reporter Gene.
- Author
-
Sellmyer MA, Richman SA, Lohith K, Hou C, Weng CC, Mach RH, O'Connor RS, Milone MC, and Farwell MD
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Female, Fluorine Radioisotopes, Gangliosides metabolism, HCT116 Cells, Healthy Volunteers, Heterografts diagnostic imaging, Humans, Interleukin Receptor Common gamma Subunit genetics, Mice, Mice, Inbred NOD, Mice, Knockout, Mice, Nude, Mice, SCID, Spleen diagnostic imaging, Spleen metabolism, Tetrahydrofolate Dehydrogenase metabolism, Trimethoprim, CD8-Positive T-Lymphocytes immunology, Escherichia coli enzymology, Genes, Reporter, Positron Emission Tomography Computed Tomography methods, Receptors, Chimeric Antigen metabolism, Tetrahydrofolate Dehydrogenase genetics
- Abstract
Cell-based therapeutics have considerable promise across diverse medical specialties; however, reliable human imaging of the distribution and trafficking of genetically engineered cells remains a challenge. We developed positron emission tomography (PET) probes based on the small-molecule antibiotic trimethoprim (TMP) that can be used to image the expression of the Escherichia coli dihydrofolate reductase enzyme (eDHFR) and tested the ability of [
18 F]-TMP, a fluorine-18 probe, to image primary human chimeric antigen receptor (CAR) T cells expressing the PET reporter gene eDHFR, yellow fluorescent protein (YFP), and Renilla luciferase (rLuc). Engineered T cells showed an approximately 50-fold increased bioluminescent imaging signal and 10-fold increased [18 F]-TMP uptake compared to controls in vitro. eDHFR-expressing anti-GD2 CAR T cells were then injected into mice bearing control GD2- and GD2+ tumors. PET/computed tomography (CT) images acquired on days 7 and 13 demonstrated early residency of CAR T cells in the spleen followed by on-target redistribution to the GD2+ tumors. This was corroborated by autoradiography and anti-human CD8 immunohistochemistry. We found a high sensitivity of detection for identifying tumor-infiltrating CD8 CAR T cells, ∼11,000 cells per mm3 . These data suggest that the [18 F]-TMP/eDHFR PET pair offers important advantages that could better allow investigators to monitor immune cell trafficking to tumors in patients., (Copyright © 2019 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
41. Testing the Specificity of Compounds Designed to Inhibit CPT1A in T Cells.
- Author
-
O'Connor RS and Milone MC
- Subjects
- Carnitine O-Palmitoyltransferase metabolism, Epoxy Compounds pharmacology, Humans, Immunoblotting, Lymphocyte Activation drug effects, T-Lymphocytes drug effects, Carnitine O-Palmitoyltransferase antagonists & inhibitors, Drug Evaluation, Preclinical methods, Enzyme Inhibitors pharmacology, T-Lymphocytes enzymology
- Abstract
In response to antigen and costimulation, T cells undergo a series of metabolic transitions that fulfill the biosynthetic demands of clonal expansion, differentiation, and effector function. Following antigen clearance, the oxidation of long-chain fatty acids (LCFAO) has been implicated in the transition from effector to central memory T cells. However, studies demonstrating a role for LCFAO in memory T-cell development have largely relied on the use of etomoxir (ETO), a small molecule inhibitor of the long-chain fatty acid transporter CPT1A. Understanding how the depletion of nutrients including LCFA that might occur in tumor microenvironments affects T-cell proliferation, differentiation, and function has important implications for tumor immunotherapy. Here, we combine the analysis of posttranscriptional gene silencing with extracellular flux assays to determine if etomoxir exerts nonspecific effects on oxidative metabolism. The off-target effects of ETO that we describe highlight the challenges of using pharmacologic inhibitors in loss-of-function approaches in T cells.
- Published
- 2020
- Full Text
- View/download PDF
42. Manufacturing Chimeric Antigen Receptor (CAR) T Cells for Adoptive Immunotherapy.
- Author
-
Ghassemi S and Milone MC
- Subjects
- Animals, Cell Differentiation immunology, Cell Line, Tumor, Humans, T-Lymphocytes cytology, Immunotherapy, Adoptive methods, Receptors, Antigen, T-Cell immunology, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology
- Abstract
Adoptive immunotherapy holds promise for the treatment of cancer and infectious disease. We describe a simple approach to transduce primary human T cells with chimeric antigen receptor (CAR) and expand their progeny ex vivo. We include assays to measure CAR expression as well as differentiation, proliferative capacity and cytolytic activity. We describe assays to measure effector cytokine production and inflammatory cytokine secretion in CAR T cells. Our approach provides a reliable and comprehensive method to culture CAR T cells for preclinical models of adoptive immunotherapy.
- Published
- 2019
- Full Text
- View/download PDF
43. Commentary.
- Author
-
Sheng F and Milone MC
- Subjects
- Adult, Female, Humans, Electrocardiography, Syncope
- Published
- 2019
- Full Text
- View/download PDF
44. T-cell phenotypes associated with effective CAR T-cell therapy in postinduction vs relapsed multiple myeloma.
- Author
-
Garfall AL, Dancy EK, Cohen AD, Hwang WT, Fraietta JA, Davis MM, Levine BL, Siegel DL, Stadtmauer EA, Vogl DT, Waxman A, Rapoport AP, Milone MC, June CH, and Melenhorst JJ
- Subjects
- Adult, Aged, Humans, Middle Aged, Phenotype, Multiple Myeloma genetics, Receptors, Chimeric Antigen metabolism
- Published
- 2019
- Full Text
- View/download PDF
45. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer.
- Author
-
Feins S, Kong W, Williams EF, Milone MC, and Fraietta JA
- Subjects
- Antigens, CD19 immunology, Antigens, CD19 therapeutic use, Biomarkers, Humans, Immune Tolerance, Immunotherapy, Adoptive methods, Neoplasms therapy, Receptors, Chimeric Antigen therapeutic use
- Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a major advancement in personalized cancer treatment. In this strategy, a patient's own T cells are genetically engineered to express a synthetic receptor that binds a tumor antigen. CAR T cells are then expanded for clinical use and infused back into the patient's body to attack and destroy chemotherapy-resistant cancer. Dramatic clinical responses and high rates of complete remission have been observed in the setting of CAR T-cell therapy of B-cell malignancies. This resulted in two recent FDA approvals of CAR T cells directed against the CD19 protein for treatment of acute lymphoblastic leukemia and diffuse large B-cell lymphoma. Thus, CAR T cells are arguably one of the first successful examples of synthetic biology and personalized cellular cancer therapy to become commercially available. In this review, we introduce the concept of using CAR T cells to break immunological tolerance to tumors, highlight several challenges in the field, discuss the utility of biomarkers in the context of predicting clinical responses, and offer prospects for developing next-generation CAR T cell-based approaches that will improve outcome., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
46. Immunotherapy Using Chimeric Antigen Receptor-Engineered T Cells: A Novel Cellular Therapy with Important Implications for the Clinical Laboratory.
- Author
-
Thibodeaux SR and Milone MC
- Subjects
- Cell Transplantation adverse effects, Cell Transplantation methods, Humans, Immunotherapy, Adoptive adverse effects, Neoplasms immunology, Receptors, Chimeric Antigen genetics, Immunotherapy, Adoptive methods, Neoplasms therapy, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology
- Abstract
Background: We have entered a new era of cancer therapy, with a number of immune-based therapies already used clinically as a standard of care. Adoptive cellular immunotherapy using T cells genetically modified with chimeric antigen receptors (CAR-T cells) represents a novel therapeutic approach. CAR-T cells have produced clinical responses in B-cell malignancies that are otherwise refractory to conventional therapies. Two CAR-T cell therapies obtained regulatory approval in 2017, with many more of these therapies under clinical development., Content: This review focuses on the current state of adoptive cellular immunotherapy, specifically CAR-T cells, in the clinic and how this therapy differs from traditional small molecule and biologic therapies. Areas in which the clinical laboratory is affected by these novel therapies are discussed. Opportunities for the clinical laboratory to help guide these therapies are also highlighted., Summary: The clinical laboratory will play an integral role in the care of patients undergoing adoptive cellular therapy with engineered T cells. There are many ways that this new therapeutic approach affects the clinical laboratory, and the clinical laboratory will likely play a critical role in managing patients that are treated with CAR-T cell therapy., (© 2018 American Association for Clinical Chemistry.)
- Published
- 2019
- Full Text
- View/download PDF
47. B cell maturation antigen-specific CAR T cells are clinically active in multiple myeloma.
- Author
-
Cohen AD, Garfall AL, Stadtmauer EA, Melenhorst JJ, Lacey SF, Lancaster E, Vogl DT, Weiss BM, Dengel K, Nelson A, Plesa G, Chen F, Davis MM, Hwang WT, Young RM, Brogdon JL, Isaacs R, Pruteanu-Malinici I, Siegel DL, Levine BL, June CH, and Milone MC
- Subjects
- Adult, Aged, Autografts, B-Cell Maturation Antigen immunology, Cyclophosphamide adverse effects, Disease-Free Survival, Female, Humans, Male, Middle Aged, Multiple Myeloma genetics, Multiple Myeloma immunology, Multiple Myeloma mortality, Neoplasm Proteins genetics, Neoplasm Proteins immunology, Survival Rate, T-Lymphocytes immunology, T-Lymphocytes pathology, T-Lymphocytes transplantation, Transduction, Genetic, Cyclophosphamide administration & dosage, Immunotherapy, Adoptive, Lymphocyte Depletion, Multiple Myeloma therapy, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen immunology
- Abstract
Background: Chimeric antigen receptor (CAR) T cells are a promising therapy for hematologic malignancies. B-cell maturation antigen (BCMA) is a rational target in multiple myeloma (MM)., Methods: We conducted a phase I study of autologous T cells lentivirally-transduced with a fully-human, BCMA-specific CAR containing CD3ζ and 4-1BB signaling domains (CART-BCMA), in subjects with relapsed/refractory MM. Twenty-five subjects were treated in 3 cohorts: 1) 1-5 x 108 CART-BCMA cells alone; 2) Cyclophosphamide (Cy) 1.5 g/m2 + 1-5 x 107 CART-BCMA cells; and 3) Cy 1.5 g/m2 + 1-5 x 108 CART-BCMA cells. No pre-specified BCMA expression level was required., Results: CART-BCMA cells were manufactured and expanded in all subjects. Toxicities included cytokine release syndrome and neurotoxicity, which were grade 3-4 in 8 (32%) and 3 (12%) subjects, respectively, and reversible. One subject died at day 24 from candidemia and progressive myeloma, following treatment for severe CRS and encephalopathy. Responses (based on treated subjects) were seen in 4/9 (44%) in cohort 1, 1/5 (20%) in cohort 2, and 7/11 (64%) in cohort 3, including 5 partial, 5 very good partial, and 2 complete responses, 3 of which were ongoing at 11, 14, and 32 months. Decreased BCMA expression on residual MM cells was noted in responders; expression increased at progression in most. Responses and CART-BCMA expansion were associated with CD4:CD8 T cell ratio and frequency of CD45RO-CD27+CD8+ T cells in the pre-manufacturing leukapheresis product., Conclusion: CART-BCMA infusions with or without lymphodepleting chemotherapy are clinically active in heavily-pretreated MM patients., Trial Registration: NCT02546167., Funding: University of Pennsylvania-Novartis Alliance and NIH.
- Published
- 2019
- Full Text
- View/download PDF
48. Anti-CD19 CAR T cells with high-dose melphalan and autologous stem cell transplantation for refractory multiple myeloma.
- Author
-
Garfall AL, Stadtmauer EA, Hwang WT, Lacey SF, Melenhorst JJ, Krevvata M, Carroll MP, Matsui WH, Wang Q, Dhodapkar MV, Dhodapkar K, Das R, Vogl DT, Weiss BM, Cohen AD, Mangan PA, Ayers EC, Nunez-Cruz S, Kulikovskaya I, Davis MM, Lamontagne A, Dengel K, Kerr ND, Young RM, Siegel DL, Levine BL, Milone MC, Maus MV, and June CH
- Published
- 2019
- Full Text
- View/download PDF
49. Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells.
- Author
-
Ghassemi S, Nunez-Cruz S, O'Connor RS, Fraietta JA, Patel PR, Scholler J, Barrett DM, Lundh SM, Davis MM, Bedoya F, Zhang C, Leferovich J, Lacey SF, Levine BL, Grupp SA, June CH, Melenhorst JJ, and Milone MC
- Subjects
- Animals, Antigens, CD19 genetics, Antigens, CD19 immunology, Antigens, Neoplasm immunology, Cell Differentiation immunology, Cell Line, Tumor, Cytotoxicity, Immunologic, Humans, Lymphocyte Activation, Mice, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology, Time Factors, Xenograft Model Antitumor Assays, Cell Culture Techniques, Immunotherapy, Adoptive, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
The success of chimeric antigen receptor (CAR)-mediated immunotherapy in acute lymphoblastic leukemia (ALL) highlights the potential of T-cell therapies with directed cytotoxicity against specific tumor antigens. The efficacy of CAR T-cell therapy depends on the engraftment and persistence of T cells following adoptive transfer. Most protocols for T-cell engineering routinely expand T cells ex vivo for 9 to 14 days. Because the potential for engraftment and persistence is related to the state of T-cell differentiation, we hypothesized that reducing the duration of ex vivo culture would limit differentiation and enhance the efficacy of CAR T-cell therapy. We demonstrated that T cells with a CAR-targeting CD19 (CART19) exhibited less differentiation and enhanced effector function in vitro when harvested from cultures at earlier (day 3 or 5) compared with later (day 9) timepoints. We then compared the therapeutic potential of early versus late harvested CART19 in a murine xenograft model of ALL and showed that the antileukemic activity inversely correlated with ex vivo culture time: day 3 harvested cells showed robust tumor control despite using a 6-fold lower dose of CART19, whereas day 9 cells failed to control leukemia at limited cell doses. We also demonstrated the feasibility of an abbreviated culture in a large-scale current good manufacturing practice-compliant process. Limiting the interval between T-cell isolation and CAR treatment is critical for patients with rapidly progressing disease. Generating CAR T cells in less time also improves potency, which is central to the effectiveness of these therapies. Cancer Immunol Res; 6(9); 1100-9. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
50. Clinical use of lentiviral vectors.
- Author
-
Milone MC and O'Doherty U
- Subjects
- Animals, Clinical Studies as Topic, Genetic Engineering methods, Genetic Vectors biosynthesis, Humans, Transduction, Genetic, Gene Transfer Techniques, Genetic Therapy methods, Genetic Vectors genetics, Lentivirus genetics
- Abstract
Viral vectors provide an efficient means for modification of eukaryotic cells, and their use is now commonplace in academic laboratories and industry for both research and clinical gene therapy applications. Lentiviral vectors, derived from the human immunodeficiency virus, have been extensively investigated and optimized over the past two decades. Third-generation, self-inactivating lentiviral vectors have recently been used in multiple clinical trials to introduce genes into hematopoietic stem cells to correct primary immunodeficiencies and hemoglobinopathies. These vectors have also been used to introduce genes into mature T cells to generate immunity to cancer through the delivery of chimeric antigen receptors (CARs) or cloned T-cell receptors. CAR T-cell therapies engineered using lentiviral vectors have demonstrated noteworthy clinical success in patients with B-cell malignancies leading to regulatory approval of the first genetically engineered cellular therapy using lentiviral vectors. In this review, we discuss several aspects of lentiviral vectors that will be of interest to clinicians, including an overview of lentiviral vector development, the current uses of viral vectors as therapy for primary immunodeficiencies and cancers, large-scale manufacturing of lentiviral vectors, and long-term follow-up of patients treated with gene therapy products.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.