117 results on '"Siegel DL"'
Search Results
2. Expression and characterization of recombinant anti-Rh(D) antibodies on filamentous phage: a model system for isolating human red blood cell antibodies by repertoire cloning
- Author
-
Siegel, DL, primary and Silberstein, LE, additional
- Published
- 1994
- Full Text
- View/download PDF
3. A molecular approach for isolating high-affinity Fab fragments that are useful in blood group serology.
- Author
-
Czerwinski M, Krop-Watorek A, Siegel DL, Spitalnik SL, Czerwinski, M, Krop-Watorek, A, Siegel, D L, and Spitalnik, S L
- Published
- 1999
4. Cytokine-mediated CAR T therapy resistance in AML.
- Author
-
Bhagwat AS, Torres L, Shestova O, Shestov M, Mellors PW, Fisher HR, Farooki SN, Frost BF, Loken MR, Gaymon AL, Frazee D, Rogal W, Frey N, Hexner EO, Luger SM, Loren AW, Martin ME, McCurdy SR, Perl AE, Stadtmauer EA, Brogdon JL, Fraietta JA, Hwang WT, Siegel DL, Plesa G, Aplenc R, Porter DL, June CH, and Gill SI
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Cytokine Release Syndrome immunology, Drug Resistance, Neoplasm immunology, Pilot Projects, T-Lymphocytes immunology, Cytokines metabolism, Cytokines immunology, Immunotherapy, Adoptive methods, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute drug therapy, Receptors, Chimeric Antigen immunology
- Abstract
Acute myeloid leukemia (AML) is a rapidly progressive malignancy without effective therapies for refractory disease. So far, chimeric antigen receptor (CAR) T cell therapy in AML has not recapitulated the efficacy seen in B cell malignancies. Here we report a pilot study of autologous anti-CD123 CAR T cells in 12 adults with relapsed or refractory AML. CAR T cells targeting CD123
+ cells were successfully manufactured in 90.4% of runs. Cytokine release syndrome was observed in 10 of 12 infused individuals (83.3%, 90% confidence interval 0.5-0.97). Three individuals achieved clinical response (25%, 90% confidence interval 0.07-0.53). We found that myeloid-supporting cytokines are secreted during cell therapy and support AML blast survival via kinase signaling, leading to CAR T cell exhaustion. The prosurvival effect of therapy-induced cytokines presents a unique resistance mechanism in AML that is distinct from any observed in B cell malignancies. Our findings suggest that autologous CART manufacturing is feasible in AML, but treatment is associated with high rates of cytokine release syndrome and relatively poor clinical efficacy. Combining CAR T cell therapies with cytokine signaling inhibitors could enhance immunotherapy efficacy in AML and achieve improved outcomes (ClinicalTrials.gov identifier: NCT03766126 )., Competing Interests: Competing interests: D.L.P. declares funding from the National Marrow Donor Program; membership on an entity’s Board of Directors or advisory committees of Kite/Gilead, Janssen, Genentech, DeCart, Sana Biotechnology, Verismo and Novartis; is a current equity holder of the American Society for Transplantation and Cellular Therapy and Verismo; declares honoraria for Incyte; and has patents and royalties in Tmunity and Wiley and Sons Publishing. J.A.F. is a member of the scientific advisory boards of Cartography Bio and Shennon Biotechnologies and has patents, royalties and other intellectual property. M.R.L. is an employee of Hematoloics, Inc. J.L.B. is an employee of Novartis. C.H.J. is an inventor of patents related to CAR therapy products and may be eligible to receive a select portion of royalties paid from Kite to the University of Pennsylvania. C.H.J. is a scientific cofounder and holds equity in Capstan Therapeutics, Dispatch Biotherapeutics and Bluewhale Bio. C.H.J. serves on the board of AC Immune and is a scientific advisor to BlueSphere Bio, Cabaletta, Carisma, Cartography, Cellares, Cellcarta, Celldex, Danaher, Decheng, ImmuneSensor, Kite, Poseida, Verismo, Viracta and WIRB-Copernicus group. S.I.G. has patents related to CAR therapy with royalties paid from Novartis to the University of Pennsylvania. S.I.G. is a scientific cofounder and holds equity in Interius Biotherapeutics and Carisma Therapeutics. S.I.G. is a scientific advisor to Carisma, Cartography, Currus, Interius, Kite, NKILT and Mission Bio. The other authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
5. CCR5-edited CD4+ T cells augment HIV-specific immunity to enable post-rebound control of HIV replication.
- Author
-
Tebas P, Jadlowsky JK, Shaw PA, Tian L, Esparza E, Brennan AL, Kim S, Naing SY, Richardson MW, Vogel AN, Maldini CR, Kong H, Liu X, Lacey SF, Bauer AM, Mampe F, Richman LP, Lee G, Ando D, Levine BL, Porter DL, Zhao Y, Siegel DL, Bar KJ, June CH, and Riley JL
- Published
- 2024
- Full Text
- View/download PDF
6. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results.
- Author
-
Bagley SJ, Logun M, Fraietta JA, Wang X, Desai AS, Bagley LJ, Nabavizadeh A, Jarocha D, Martins R, Maloney E, Lledo L, Stein C, Marshall A, Leskowitz R, Jadlowsky JK, Christensen S, Oner BS, Plesa G, Brennan A, Gonzalez V, Chen F, Sun Y, Gladney W, Barrett D, Nasrallah MP, Hwang WT, Ming GL, Song H, Siegel DL, June CH, Hexner EO, Binder ZA, and O'Rourke DM
- Subjects
- Humans, Middle Aged, Male, Female, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local pathology, Adult, Aged, Brain Neoplasms immunology, Brain Neoplasms therapy, Brain Neoplasms pathology, Injections, Spinal, Maximum Tolerated Dose, Glioblastoma therapy, Glioblastoma immunology, Glioblastoma diagnostic imaging, Glioblastoma pathology, Interleukin-13 Receptor alpha2 Subunit immunology, Receptors, Chimeric Antigen immunology, ErbB Receptors, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods
- Abstract
Recurrent glioblastoma (rGBM) remains a major unmet medical need, with a median overall survival of less than 1 year. Here we report the first six patients with rGBM treated in a phase 1 trial of intrathecally delivered bivalent chimeric antigen receptor (CAR) T cells targeting epidermal growth factor receptor (EGFR) and interleukin-13 receptor alpha 2 (IL13Rα2). The study's primary endpoints were safety and determination of the maximum tolerated dose. Secondary endpoints reported in this interim analysis include the frequency of manufacturing failures and objective radiographic response (ORR) according to modified Response Assessment in Neuro-Oncology criteria. All six patients had progressive, multifocal disease at the time of treatment. In both dose level 1 (1 ×10
7 cells; n = 3) and dose level 2 (2.5 × 107 cells; n = 3), administration of CART-EGFR-IL13Rα2 cells was associated with early-onset neurotoxicity, most consistent with immune effector cell-associated neurotoxicity syndrome (ICANS), and managed with high-dose dexamethasone and anakinra (anti-IL1R). One patient in dose level 2 experienced a dose-limiting toxicity (grade 3 anorexia, generalized muscle weakness and fatigue). Reductions in enhancement and tumor size at early magnetic resonance imaging timepoints were observed in all six patients; however, none met criteria for ORR. In exploratory endpoint analyses, substantial CAR T cell abundance and cytokine release in the cerebrospinal fluid were detected in all six patients. Taken together, these first-in-human data demonstrate the preliminary safety and bioactivity of CART-EGFR-IL13Rα2 cells in rGBM. An encouraging early efficacy signal was also detected and requires confirmation with additional patients and longer follow-up time. ClinicalTrials.gov identifier: NCT05168423 ., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
7. Author Correction: Potent suppression of neuroendocrine tumors and gastrointestinal cancers by CDH17CAR T cells without toxicity to normal tissues.
- Author
-
Feng Z, He X, Zhang X, Wu Y, Xing B, Knowles A, Shan Q, Miller S, Hojnacki T, Ma J, Katona BW, Gade TPF, Siegel DL, Schrader J, Metz DC, June CH, and Hua X
- Published
- 2024
- Full Text
- View/download PDF
8. Red cell exchange for rapid leukoreduction in adults with hyperleukocytosis and leukostasis.
- Author
-
Mack EA, Dougher MC, Ginda AM, Cahill C, Murter M, Schell K, Tanhehco YC, Bhoj VG, Fesnak AD, Siegel DL, Kambayashi T, Aqui NA, and O'Doherty U
- Subjects
- Adult, Humans, Acute Disease, Leukapheresis, Leukocytosis therapy, Leukostasis therapy, Leukemia, Myeloid, Acute therapy, Leukemia, Monocytic, Acute therapy
- Abstract
Abstract: We show that red cell exchange (RCE) treats hyperleukocytosis in acute leukemia. RCE provided similar leukoreduction to standard therapeutic leukoreduction and could be superior in patients with severe anemia or monocytic leukemias or when requiring rapid treatment., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
9. Type I Interferon Signaling via the EGR2 Transcriptional Regulator Potentiates CAR T Cell-Intrinsic Dysfunction.
- Author
-
Jung IY, Bartoszek RL, Rech AJ, Collins SM, Ooi SK, Williams EF, Hopkins CR, Narayan V, Haas NB, Frey NV, Hexner EO, Siegel DL, Plesa G, Porter DL, Cantu A, Everett JK, Guedan S, Berger SL, Bushman FD, Herbst F, and Fraietta JA
- Subjects
- Humans, T-Lymphocytes, Immunotherapy, Adoptive, Signal Transduction, Early Growth Response Protein 2 genetics, Early Growth Response Protein 2 metabolism, Neoplasms genetics, Neoplasms therapy, Hematologic Neoplasms metabolism
- Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promise in treating hematologic cancers, but resistance is common and efficacy is limited in solid tumors. We found that CAR T cells autonomously propagate epigenetically programmed type I interferon signaling through chronic stimulation, which hampers antitumor function. EGR2 transcriptional regulator knockout not only blocks this type I interferon-mediated inhibitory program but also independently expands early memory CAR T cells with improved efficacy against liquid and solid tumors. The protective effect of EGR2 deletion in CAR T cells against chronic antigen-induced exhaustion can be overridden by interferon-β exposure, suggesting that EGR2 ablation suppresses dysfunction by inhibiting type I interferon signaling. Finally, a refined EGR2 gene signature is a biomarker for type I interferon-associated CAR T cell failure and shorter patient survival. These findings connect prolonged CAR T cell activation with deleterious immunoinflammatory signaling and point to an EGR2-type I interferon axis as a therapeutically amenable biological system., Significance: To improve CAR T cell therapy outcomes, modulating molecular determinants of CAR T cell-intrinsic resistance is crucial. Editing the gene encoding the EGR2 transcriptional regulator renders CAR T cells impervious to type I interferon pathway-induced dysfunction and improves memory differentiation, thereby addressing major barriers to progress for this emerging class of cancer immunotherapies. This article is highlighted in the In This Issue feature, p. 1501., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
10. Tissue-resident memory CAR T cells with stem-like characteristics display enhanced efficacy against solid and liquid tumors.
- Author
-
Jung IY, Noguera-Ortega E, Bartoszek R, Collins SM, Williams E, Davis M, Jadlowsky JK, Plesa G, Siegel DL, Chew A, Levine BL, Berger SL, Moon EK, Albelda SM, and Fraietta JA
- Subjects
- Humans, Immunotherapy, Adoptive methods, Cytokines metabolism, Immunotherapy, Receptors, Chimeric Antigen metabolism, Neoplasms therapy
- Abstract
Chimeric antigen receptor (CAR) T cells demonstrate remarkable success in treating hematological malignancies, but their effectiveness in non-hematopoietic cancers remains limited. This study proposes enhancing CAR T cell function and localization in solid tumors by modifying the epigenome governing tissue-residency adaptation and early memory differentiation. We identify that a key factor in human tissue-resident memory CAR T cell (CAR-T
RM ) formation is activation in the presence of the pleotropic cytokine, transforming growth factor β (TGF-β), which enforces a core program of both "stemness" and sustained tissue residency by mediating chromatin remodeling and concurrent transcriptional changes. This approach leads to a practical and clinically actionable in vitro production method for engineering peripheral blood T cells into a large number of "stem-like" CAR-TRM cells resistant to tumor-associated dysfunction, possessing an enhanced ability to accumulate in situ and rapidly eliminate cancer cells for more effective immunotherapy., Competing Interests: Declaration of interests I.Y.J., D.L.S., M.M.D., A.C., B.L.L., E.K.M., S.M.A., and J.A.F. hold patents and intellectual property related to T cell-based cancer immunotherapy and have received royalties. M.M.D. has obtained research funding from Tmunity Therapeutics and is a member of the Scientific Advisory Board for Cellares Corporation. A.C. is a co-founder and has equity in Tmunity Therapeutics. B.L.L. serves as a consultant for Terumo and GSK; is on the Scientific Advisory Boards for Akron, Avectas, Immuneel, Immusoft, In8bio, Ori Biotech, Oxford Biomedica, and Vycellix; and holds equity as a co-founder in both Tmunity Therapeutics and Capstan Therapeutics. S.M.A. is a co-founder and equity holder in Capstan Therapeutics. J.A.F. receives research funding from Tmunity Therapeutics and Danaher Corporation, consults for Retro Biosciences, and is a member of the Scientific Advisory Boards for Cartography Bio and Shennon Biotechnologies Inc., (Published by Elsevier Inc.)- Published
- 2023
- Full Text
- View/download PDF
11. Mechanisms of inhibition of human monoclonal antibodies in immune thrombotic thrombocytopenic purpura.
- Author
-
Halkidis K, Meng C, Liu S, Mayne L, Siegel DL, and Zheng XL
- Subjects
- Humans, Antibodies, Monoclonal, von Willebrand Factor metabolism, ADAM Proteins chemistry, ADAM Proteins metabolism, ADAMTS13 Protein, Autoantibodies, Purpura, Thrombotic Thrombocytopenic, Thrombosis, Purpura, Thrombocytopenic, Idiopathic
- Abstract
Antibody binding to a plasma metalloprotease, a disintegrin and metalloproteinase with thrombospondin type 1 repeats 13 (ADAMTS13), is necessary for the development of immune thrombotic thrombocytopenic purpura (iTTP). Inhibition of ADAMTS13-mediated von Willebrand factor (VWF) cleavage by such antibodies clearly plays a role in the pathophysiology of the disease, although the mechanisms by which they inhibit ADAMTS13 enzymatic function are not fully understood. At least some immunoglobulin G-type antibodies appear to affect the conformational accessibility of ADAMTS13 domains involved in both substrate recognition and inhibitory antibody binding. We used single-chain fragments of the variable region previously identified via phage display from patients with iTTP to explore the mechanisms of action of inhibitory human monoclonal antibodies. Using recombinant full-length ADAMTS13, truncated ADAMTS13 variants, and native ADAMTS13 in normal human plasma, we found that, regardless of the conditions tested, all 3 inhibitory monoclonal antibodies tested affected enzyme turnover rate much more than substrate recognition of VWF. Hydrogen-to-deuterium exchange plus mass spectrometry experiments with each of these inhibitory antibodies demonstrated that residues in the active site of the catalytic domain of ADAMTS13 are differentially exposed to solvent in the presence and absence of monoclonal antibody binding. These results support the hypothesis that inhibition of ADAMTS13 in iTTP may not necessarily occur because the antibodies directly prevent VWF binding, but instead because of allosteric effects that impair VWF cleavage, likely by affecting the conformation of the catalytic center in the protease domain of ADAMTS13. Our findings provide novel insight into the mechanism of autoantibody-mediated inhibition of ADAMTS13 and pathogenesis of iTTP., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
12. 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
13. Development and pharmacokinetic assessment of a fully canine anti-PD-1 monoclonal antibody for comparative translational research in dogs with spontaneous tumors.
- Author
-
Yoshimoto S, Chester N, Xiong A, Radaelli E, Wang H, Brillantes M, Gulendran G, Glassman P, Siegel DL, and Mason NJ
- Subjects
- Humans, Dogs, Animals, Programmed Cell Death 1 Receptor, Antibodies, Monoclonal therapeutic use, Immune Checkpoint Inhibitors, B7-H1 Antigen, Translational Research, Biomedical, Melanoma
- Abstract
PD-1 checkpoint inhibitors have revolutionized the treatment of patients with different cancer histologies including melanoma, renal cell carcinoma, and non-small cell lung carcinoma. However, only a subset of patients show a dramatic clinical response to treatment. Despite intense biomarker discovery efforts, no single robust, prognostic correlation has emerged as a valid outcome predictor. Immune competent, pet dogs develop spontaneous tumors that share similar features to human cancers including chromosome aberrations, molecular subtypes, immune signatures, tumor heterogeneity, metastatic behavior, and chemotherapeutic response. As such, they represent a valuable parallel patient population in which to investigate predictive biomarkers of checkpoint inhibition. However, the lack of a validated, non-immunogenic, canine anti-PD-1 antibody for pre-clinical use hinders this comparative approach and prevents potential clinical benefits of PD-1 blockade being realized in the veterinary clinic. To address this, fully canine single-chain variable fragments (scFvs) that bind canine (c)PD-1 were isolated from a comprehensive canine scFv phage display library. Lead candidates were identified that bound with high affinity to cPD-1 and inhibited its interaction with canine PD-L1 (cPD-L1). The lead scFv candidate re-formatted into a fully canine IgG
D reversed the inhibitory effects of cPD-1:cPD-L1 interaction on canine chimeric antigen receptor (CAR) T cell function. In vivo administration showed no toxicity and revealed favorable pharmacokinetics for a reasonable dosing schedule. These results pave the way for clinical trials with anti-cPD-1 in canine cancer patients to investigate predictive biomarkers and combination regimens to inform human clinical trials and bring a promising checkpoint inhibitor into the veterinary armamentarium.- Published
- 2023
- Full Text
- View/download PDF
14. 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
15. A Keratinocyte-Tethered Biologic Enables Location-Precise Treatment in Mouse Vitiligo.
- Author
-
Hsueh YC, Wang Y, Riding RL, Catalano DE, Lu YJ, Richmond JM, Siegel DL, Rusckowski M, Stanley JR, and Harris JE
- Subjects
- Mice, Animals, Tissue Distribution, Keratinocytes metabolism, Skin pathology, Vitiligo drug therapy, Biological Products therapeutic use
- Abstract
Despite the central role of IFN-γ in vitiligo pathogenesis, systemic IFN-γ neutralization is an impractical treatment option owing to strong immunosuppression. However, most patients with vitiligo present with <20% affected body surface area, which provides an opportunity for localized treatments that avoid systemic side effects. After identifying keratinocytes as key cells that amplify IFN-γ signaling during vitiligo, we hypothesized that tethering an IFN-γ‒neutralizing antibody to keratinocytes would limit anti‒IFN-γ effects on the treated skin for the localized treatment. To that end, we developed a bispecific antibody capable of blocking IFN-γ signaling while binding to desmoglein expressed by keratinocytes. We characterized the effect of the bispecific antibody in vitro, ex vivo, and in a mouse model of vitiligo. Single-photon emission computed tomography/computed tomography biodistribution and serum assays after local footpad injection revealed that the bispecific antibody had improved skin retention, faster elimination from the blood, and less systemic IFN-γ inhibition than the nontethered version. Furthermore, the bispecific antibody conferred localized protection almost exclusively to the treated footpad during vitiligo, which was not possible by local injection of the nontethered anti‒IFN-γ antibody. Thus, keratinocyte tethering proved effective while significantly diminishing the off-tissue effects of IFN-γ blockade, offering a safer treatment strategy for localized skin diseases, including vitiligo., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. BLIMP1 and NR4A3 transcription factors reciprocally regulate antitumor CAR T cell stemness and exhaustion.
- Author
-
Jung IY, Narayan V, McDonald S, Rech AJ, Bartoszek R, Hong G, Davis MM, Xu J, Boesteanu AC, Barber-Rotenberg JS, Plesa G, Lacey SF, Jadlowsky JK, Siegel DL, Hammill DM, Cho-Park PF, Berger SL, Haas NB, and Fraietta JA
- Subjects
- Male, Humans, Mice, Animals, Transcription Factors genetics, Transcription Factors metabolism, CD8-Positive T-Lymphocytes, Immunotherapy, Adoptive methods, Hepatitis A Virus Cellular Receptor 2 genetics, Hepatitis A Virus Cellular Receptor 2 metabolism, Cell Line, Tumor, DNA-Binding Proteins metabolism, Receptors, Thyroid Hormone metabolism, Nerve Tissue Proteins metabolism, Neoplasms pathology, Receptors, Steroid metabolism
- Abstract
Chimeric antigen receptor (CAR) T cells have not induced meaningful clinical responses in solid tumors. Loss of T cell stemness, poor expansion capacity, and exhaustion during prolonged tumor antigen exposure are major causes of CAR T cell therapeutic resistance. Single-cell RNA-sequencing analysis of CAR T cells from a first-in-human trial in metastatic prostate cancer identified two independently validated cell states associated with antitumor potency or lack of efficacy. Low expression of PRDM1 , encoding the BLIMP1 transcription factor, defined highly potent TCF7 [encoding T cell factor 1 (TCF1)]-expressing CD8
+ CAR T cells, whereas enrichment of HAVCR2 [encoding T cell immunoglobulin and mucin-domain containing-3 (TIM-3)]-expressing CD8+ T cells with elevated PRDM1 was associated with poor outcomes. PRDM1 knockout promoted TCF7 -dependent CAR T cell stemness and proliferation, resulting in marginally enhanced leukemia control in mice. However, in the setting of PRDM1 deficiency, a negative epigenetic feedback program of nuclear factor of activated T cells (NFAT)-driven T cell dysfunction was identified. This program was characterized by compensatory up-regulation of NR4A3 and other genes encoding exhaustion-related transcription factors that hampered T cell effector function in solid tumors. Dual knockout of PRDM1 and NR4A3 skewed CAR T cell phenotypes away from TIM-3+ CD8+ and toward TCF1+ CD8+ to counter exhaustion of tumor-infiltrating CAR T cells and improve antitumor responses, effects that were not achieved with PRDM1 and NR4A3 single knockout alone. These data underscore dual targeting of PRDM1 and NR4A3 as a promising approach to advance adoptive cell immuno-oncotherapy.- Published
- 2022
- Full Text
- View/download PDF
17. 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
18. National Blood Foundation 2021 Research and Development summit: Discovery, innovation, and challenges in advancing blood and biotherapies.
- Author
-
Holmberg JA, Henry SM, Burnouf T, Devine D, Marschner S, Boothby TC, Burger SR, Chou ST, Custer B, Blumberg N, Siegel DL, and Spitalnik SL
- Subjects
- Humans, Research, Biological Therapy
- Published
- 2022
- Full Text
- View/download PDF
19. Targeted In Vivo Loading of Red Blood Cells Markedly Prolongs Nanocarrier Circulation.
- Author
-
Glassman PM, Villa CH, Marcos-Contreras OA, Hood ED, Walsh LR, Greineder CF, Myerson JW, Shuvaeva T, Puentes L, Brenner JS, Siegel DL, and Muzykantov VR
- Subjects
- Animals, Drug Delivery Systems, Erythrocytes, Mice, Polymers, Liposomes pharmacokinetics, Polyethylene Glycols pharmacokinetics
- Abstract
Engineering drug delivery systems for prolonged pharmacokinetics (PK) has been an ongoing pursuit for nearly 50 years. The gold standard for PK enhancement is the coating of nanoparticles with polymers, namely polyethylene glycol (PEGylation), which has been applied in several clinically used products. In the present work, we utilize the longest circulating and most abundant component of blood─the erythrocyte─to improve the PK behavior of liposomes. Antibody-mediated coupling of liposomes to erythrocytes was tested in vitro to identify a loading dose that did not adversely impact the carrier cells. Injection of erythrocyte targeting liposomes into mice resulted in a ∼2-fold improvement in the area under the blood concentration versus time profile versus PEGylated liposomes and a redistribution from the plasma into the cellular fraction of blood. These results suggest that in vivo targeting of erythrocytes is a viable strategy to improve liposome PK relative to current, clinically viable strategies.
- Published
- 2022
- Full Text
- View/download PDF
20. Potent suppression of neuroendocrine tumors and gastrointestinal cancers by CDH17CAR T cells without toxicity to normal tissues.
- Author
-
Feng Z, He X, Zhang X, Wu Y, Xing B, Knowles A, Shan Q, Miller S, Hojnacki T, Ma J, Katona BW, Gade TPF, Siegel DL, Schrader J, Metz DC, June CH, and Hua X
- Subjects
- Animals, Humans, Mice, T-Lymphocytes, Xenograft Model Antitumor Assays, Gastrointestinal Neoplasms therapy, Neuroendocrine Tumors therapy, Receptors, Chimeric Antigen
- Abstract
Gastrointestinal cancers (GICs) and neuroendocrine tumors (NETs) are often refractory to therapy after metastasis. Adoptive cell therapy using chimeric antigen receptor (CAR) T cells, though remarkably efficacious for treating leukemia, is yet to be developed for solid tumors such as GICs and NETs. Here we isolated a llama-derived nanobody, VHH1, and found that it bound cell surface adhesion protein CDH17 upregulated in GICs and NETs. VHH1-CAR T cells (CDH17CARTs) killed both human and mouse tumor cells in a CDH17-dependent manner. CDH17CARTs eradicated CDH17-expressing NETs and gastric, pancreatic and colorectal cancers in either tumor xenograft or autochthonous mouse models. Notably, CDH17CARTs do not attack normal intestinal epithelial cells, which also express CDH17, to cause toxicity, likely because CDH17 is localized only at the tight junction between normal intestinal epithelial cells. Thus, CDH17 represents a class of previously unappreciated tumor-associated antigens that is 'masked' in healthy tissues from attack by CAR T cells for developing safer cancer immunotherapy., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
21. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial.
- Author
-
Narayan V, Barber-Rotenberg JS, Jung IY, Lacey SF, Rech AJ, Davis MM, Hwang WT, Lal P, Carpenter EL, Maude SL, Plesa G, Vapiwala N, Chew A, Moniak M, Sebro RA, Farwell MD, Marshall A, Gilmore J, Lledo L, Dengel K, Church SE, Hether TD, Xu J, Gohil M, Buckingham TH, Yee SS, Gonzalez VE, Kulikovskaya I, Chen F, Tian L, Tien K, Gladney W, Nobles CL, Raymond HE, Hexner EO, Siegel DL, Bushman FD, June CH, Fraietta JA, and Haas NB
- Subjects
- Humans, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Male, Prostate-Specific Antigen metabolism, T-Lymphocytes, Transforming Growth Factor beta metabolism, Tumor Microenvironment, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Chimeric Antigen
- Abstract
Chimeric antigen receptor (CAR) T cells have demonstrated promising efficacy, particularly in hematologic malignancies. One challenge regarding CAR T cells in solid tumors is the immunosuppressive tumor microenvironment (TME), characterized by high levels of multiple inhibitory factors, including transforming growth factor (TGF)-β. We report results from an in-human phase 1 trial of castration-resistant, prostate cancer-directed CAR T cells armored with a dominant-negative TGF-β receptor (NCT03089203). Primary endpoints were safety and feasibility, while secondary objectives included assessment of CAR T cell distribution, bioactivity and disease response. All prespecified endpoints were met. Eighteen patients enrolled, and 13 subjects received therapy across four dose levels. Five of the 13 patients developed grade ≥2 cytokine release syndrome (CRS), including one patient who experienced a marked clonal CAR T cell expansion, >98% reduction in prostate-specific antigen (PSA) and death following grade 4 CRS with concurrent sepsis. Acute increases in inflammatory cytokines correlated with manageable high-grade CRS events. Three additional patients achieved a PSA reduction of ≥30%, with CAR T cell failure accompanied by upregulation of multiple TME-localized inhibitory molecules following adoptive cell transfer. CAR T cell kinetics revealed expansion in blood and tumor trafficking. Thus, clinical application of TGF-β-resistant CAR T cells is feasible and generally safe. Future studies should use superior multipronged approaches against the TME to improve outcomes., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
22. 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
23. A randomized controlled study of convalescent plasma for individuals hospitalized with COVID-19 pneumonia.
- Author
-
Bar KJ, Shaw PA, Choi GH, Aqui N, Fesnak A, Yang JB, Soto-Calderon H, Grajales L, Starr J, Andronov M, Mastellone M, Amonu C, Feret G, DeMarshall M, Buchanan M, Caturla M, Gordon J, Wanicur A, Monroy MA, Mampe F, Lindemuth E, Gouma S, Mullin AM, Barilla H, Pronina A, Irwin L, Thomas R, Eichinger RA, Demuth F, Luning Prak ET, Pascual JL, Short WR, Elovitz MA, Baron J, Meyer NJ, Degnan KO, Frank I, Hensley SE, Siegel DL, and Tebas P
- Subjects
- Adult, Aged, Antibodies, Viral, Female, Hospitalization, Humans, Immune Tolerance, Immunization, Passive methods, Immunosuppression Therapy, Incidence, Male, Middle Aged, Oxygen therapeutic use, RNA, Viral, Respiration, Artificial, Risk Factors, Treatment Outcome, COVID-19 Serotherapy, COVID-19 therapy, Pneumonia, Viral therapy, SARS-CoV-2
- Abstract
BackgroundAntibody-based strategies for COVID-19 have shown promise in prevention and treatment of early disease. COVID-19 convalescent plasma (CCP) has been widely used but results from randomized trials supporting its benefit in hospitalized patients with pneumonia are limited. Here, we assess the efficacy of CCP in severely ill, hospitalized adults with COVID-19 pneumonia.MethodsWe performed a randomized control trial (PennCCP2), with 80 adults hospitalized with COVID-19 pneumonia, comparing up to 2 units of locally sourced CCP plus standard care versus standard care alone. The primary efficacy endpoint was comparison of a clinical severity score. Key secondary outcomes include 14- and 28-day mortality, 14- and 28-day maximum 8-point WHO ordinal score (WHO8) score, duration of supplemental oxygenation or mechanical ventilation, respiratory SARS-CoV-2 RNA, and anti-SARS-CoV-2 antibodies.ResultsEighty hospitalized adults with confirmed COVID-19 pneumonia were enrolled at median day 6 of symptoms and day 1 of hospitalization; 60% were anti-SARS-CoV-2 antibody seronegative. Participants had a median of 3 comorbidities, including risk factors for severe COVID-19 and immunosuppression. CCP treatment was safe and conferred significant benefit by clinical severity score (median [MED] and interquartile range [IQR] 10 [5.5-30] vs. 7 [2.75-12.25], P = 0.037) and 28-day mortality (n = 10, 26% vs. n = 2, 5%; P = 0.013). All other prespecified outcome measures showed weak evidence toward benefit of CCP.ConclusionTwo units of locally sourced CCP administered early in hospitalization to majority seronegative participants conferred a significant benefit in clinical severity score and 28-day mortality. Results suggest CCP may benefit select populations, especially those with comorbidities who are treated early.Trial RegistrationClinicalTrials.gov NCT04397757.FundingUniversity of Pennsylvania.
- Published
- 2021
- Full Text
- View/download PDF
24. Red blood cells: The metamorphosis of a neglected carrier into the natural mothership for artificial nanocarriers.
- Author
-
Glassman PM, Hood ED, Ferguson LT, Zhao Z, Siegel DL, Mitragotri S, Brenner JS, and Muzykantov VR
- Subjects
- Drug Carriers chemistry, Drug Delivery Systems, Humans, Lipids chemistry, Liposomes chemistry, Erythrocytes chemistry, Nanoparticles chemistry
- Abstract
Drug delivery research pursues many types of carriers including proteins and other macromolecules, natural and synthetic polymeric structures, nanocarriers of diverse compositions and cells. In particular, liposomes and lipid nanoparticles represent arguably the most advanced and popular human-made nanocarriers, already in multiple clinical applications. On the other hand, red blood cells (RBCs) represent attractive natural carriers for the vascular route, featuring at least two distinct compartments for loading pharmacological cargoes, namely inner space enclosed by the plasma membrane and the outer surface of this membrane. Historically, studies of liposomal drug delivery systems (DDS) astronomically outnumbered and surpassed the RBC-based DDS. Nevertheless, these two types of carriers have different profile of advantages and disadvantages. Recent studies showed that RBC-based drug carriers indeed may feature unique pharmacokinetic and biodistribution characteristics favorably changing benefit/risk ratio of some cargo agents. Furthermore, RBC carriage cardinally alters behavior and effect of nanocarriers in the bloodstream, so called RBC hitchhiking (RBC-HH). This article represents an attempt for the comparative analysis of liposomal vs RBC drug delivery, culminating with design of hybrid DDSs enabling mutual collaborative advantages such as RBC-HH and camouflaging nanoparticles by RBC membrane. Finally, we discuss the key current challenges faced by these and other RBC-based DDSs including the issue of potential unintended and adverse effect and contingency measures to ameliorate this and other concerns., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
25. Pathologically stiff erythrocytes impede contraction of blood clots: Reply to comment.
- Author
-
Tutwiler V, Litvinov RI, Protopopova A, Nagaswami C, Villa C, Woods E, Abdulmalik O, Siegel DL, Russell JE, Muzykantov VR, Lam WA, Myers DR, and Weisel JW
- Subjects
- Erythrocytes, Humans, Thrombosis
- Published
- 2021
- Full Text
- View/download PDF
26. Humanized CD19-Targeted Chimeric Antigen Receptor (CAR) T Cells in CAR-Naive and CAR-Exposed Children and Young Adults With Relapsed or Refractory Acute Lymphoblastic Leukemia.
- Author
-
Myers RM, Li Y, Barz Leahy A, Barrett DM, Teachey DT, Callahan C, Fasano CC, Rheingold SR, DiNofia A, Wray L, Aplenc R, Baniewicz D, Liu H, Shaw PA, Pequignot E, Getz KD, Brogdon JL, Fesnak AD, Siegel DL, Davis MM, Bartoszek C, Lacey SF, Hexner EO, Chew A, Wertheim GB, Levine BL, June CH, Grupp SA, and Maude SL
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Female, Humans, Male, Pilot Projects, Young Adult, Antigens, CD19 metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Receptors, Antigen, T-Cell metabolism, Receptors, Chimeric Antigen metabolism
- Abstract
Purpose: CD19-targeted chimeric antigen receptor (CAR)-modified T cells demonstrate unprecedented responses in B-cell acute lymphoblastic leukemia (B-ALL); however, relapse remains a substantial challenge. Short CAR T-cell persistence contributes to this risk; therefore, strategies to improve persistence are needed., Methods: We conducted a pilot clinical trial of a humanized CD19 CAR T-cell product (huCART19) in children and young adults with relapsed or refractory B-ALL (n = 72) or B-lymphoblastic lymphoma (n = 2), treated in two cohorts: with (retreatment, n = 33) or without (CAR-naive, n = 41) prior CAR exposure. Patients were monitored for toxicity, response, and persistence of huCART19., Results: Seventy-four patients 1-29 years of age received huCART19. Cytokine release syndrome developed in 62 (84%) patients and was grade 4 in five (6.8%). Neurologic toxicities were reported in 29 (39%), three (4%) grade 3 or 4, and fully resolved in all cases. The overall response rate at 1 month after infusion was 98% (100% in B-ALL) in the CAR-naive cohort and 64% in the retreatment cohort. At 6 months, the probability of losing huCART19 persistence was 27% (95% CI, 14 to 41) for CAR-naive and 48% (95% CI, 30 to 64) for retreatment patients, whereas the incidence of B-cell recovery was 15% (95% CI, 6 to 28) and 58% (95% CI, 33 to 77), respectively. Relapse-free survival at 12 and 24 months, respectively, was 84% (95% CI, 72 to 97) and 74% (95% CI, 60 to 90) in CAR-naive and 74% (95% CI, 56 to 97) and 58% (95% CI, 37 to 90) in retreatment cohorts., Conclusion: HuCART19 achieved durable remissions with long-term persistence in children and young adults with relapsed or refractory B-ALL, including after failure of prior CAR T-cell therapy., Competing Interests: David M. BarrettTravel, Accommodations, Expenses: Therakos David T. TeacheyConsulting or Advisory Role: SobiResearch Funding: Novartis, Beam Therapeutics, NeoImmuneTech Colleen CallahanConsulting or Advisory Role: NovartisSpeakers' Bureau: Peerview Susan R. RheingoldEmployment: OptiNoseStock and Other Ownership Interests: OptiNoseConsulting or Advisory Role: PfizerResearch Funding: Pfizer Richard AplencExpert Testimony: Vorys Pamela A. ShawPatents, Royalties, Other Intellectual Property: I am part of a patent owned by UPenn and currently licensed to Novartis for an algorithm that predicts severe cytokine release syndrome for the CART 19 therapy. I receive 10% of the licensing fees Edward PequignotPatents, Royalties, Other Intellectual Property: As part of Penn's role in the FDA-approval of CAR-T therapy, and for my part as an employee of Penn involved in their research in CAR-T, I have received royalties of approximately $300 in US dollars over the past 2 years Jennifer L. BrogdonEmployment: Novartis Institutes for BioMedical ResearchStock and Other Ownership Interests: Novartis Institutes for BioMedical Research Donald L. SiegelResearch Funding: Tmunity Therapeutics IncPatents, Royalties, Other Intellectual Property: A patent, owned by the Trustees of the University of Pennsylvania, on which I am listed as an inventor is licensed to Alexion Pharmaceuticals. I receive royalties as stipulated in the University of Pennsylvania Faculty HandbookExpert Testimony: Regeneron Megan M. DavisLeadership: Cellares CorporationConsulting or Advisory Role: Tmunity Therapeutics IncResearch Funding: Tmunity Therapeutics IncPatents, Royalties, Other Intellectual Property: Novartis Institutes for Biomedical Research—royalties and milestones for patents/knowhow, Tmunity Therapeutics—royalties andmilestones from patents/knowhow Simon F. LaceyConsulting or Advisory Role: Gilead SciencesResearch Funding: Tmunity Therapeutics Inc, Cabaletta Bio, Novartis Institutes for BioMedical ResearchPatents, Royalties, Other Intellectual Property: Patents and IP related to CTL019 (Kymriah) assigned by the University of Pennsylvania to Novartis Elizabeth O. HexnerConsulting or Advisory Role: Blueprint Medicines, ABIM Subspecialty BoardResearch Funding: Blueprint Medicines, Tmunity Therapeutics Inc Gerald B. WertheimEmployment: Johnson & JohnsonStock and Other Ownership Interests: Johnson & Johnson Bruce L. LevineStock and Other Ownership Interests: Tmunity Therapeutics IncHonoraria: Novartis, TerumoConsulting or Advisory Role: Avectas, Ori Biotech, Vycellix, Immuneel Therapeutics, In8bio, Patheon/ThermoFisher Viral Vector ServicesResearch Funding: Tmunity Therapeutics IncPatents, Royalties, Other Intellectual Property: Intellectual property and patents in the field of cell and gene therapyTravel, Accommodations, Expenses: Avectas, Terumo Carl H. JuneLeadership: AC ImmuneStock and Other Ownership Interests: Celldex, Tmunity Therapeutics Inc, Cabaletta Bio, Carisma Therapeutics, DeCART Therapeutics, Bluesphere Bio, Cellares, ZIOPHARM Oncology, Decheng Capital, Posieda Therapeutics, VerismaHonoraria: PfizerConsulting or Advisory Role: Celldex, Viracta Therapeutics, Cabaletta Bio, Carisma Therapeutics, Kiadis Pharma, WIRB-Copernicus Group, Janssen OncologyResearch Funding: Novartis, Tmunity Therapeutics IncPatents, Royalties, Other Intellectual Property: IP licensed to Novartis; Royalties paid to University of Pennsylvania, Office of Naval Research; IP and patent royalties, IP licensed to Tmunity Stephan A. GruppConsulting or Advisory Role: Novartis, Jazz Pharmaceuticals, Janssen, Cellular Biomedicine Group, TCR2 Therapeutics, Humanigen, Roche, Adaptimmune, Alimera Sciences, Cabaletta Bio, CRISPR Therapeutics/VertexResearch Funding: Novartis, Kite/Gilead, Servier, Jazz Pharmaceuticals, VertexPatents, Royalties, Other Intellectual Property: UPenn Toxicity management patentExpert Testimony: Juno Therapeutics Shannon L. MaudeConsulting or Advisory Role: Novartis, Wugen IncResearch Funding: NovartisTravel, Accommodations, Expenses: Novartis, Kite, a Gilead company, Wugen IncNo other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
27. A human monoclonal antibody against the distal carboxyl terminus of ADAMTS-13 modulates its susceptibility to an inhibitor in thrombotic thrombocytopenic purpura.
- Author
-
Halkidis K, Siegel DL, and Zheng XL
- Subjects
- ADAMTS13 Protein, Antibodies, Monoclonal, Autoantibodies, Humans, Thrombospondin 1, Purpura, Thrombotic Thrombocytopenic
- Abstract
Background: Immune thrombotic thrombocytopenic purpura (iTTP) is a potentially fatal thrombotic microangiopathy, resulting from a severe deficiency of plasma ADAMTS-13 (A Disintegrin And Metalloproteinase with ThromboSpondin type 1 motif, member 13) activity. IgG-type autoantibodies are primarily responsible for the inhibition of plasma ADAMTS-13 activity. However, the mechanism underlying autoantibody-mediated inhibition is not fully understood., Objective: The purpose of the present study is to determine the role of IgG autoantibodies against various carboxyl-terminal domains of ADAMTS-13 in regulating ADAMTS-13 activity and its inhibition., Method: Various human monoclonal antibodies isolated by phage display, recombinant protein expression and purification, and biochemical analyses were employed for the study., Results: Our results demonstrate for the first time that a human monoclonal antibody fragment, the single chain fragment of the variable region (scFv) isolated from a patient with acute iTTP that binds the distal carboxyl-terminus of ADAMTS-13, is able to activate ADAMTS-13 and increase the proteolytic cleavage of a FRETS-VWF73 substrate; moreover, binding of such a human monoclonal antibody against the carboxyl-terminus of ADAMTS-13 to plasma ADAMTS-13 appears to modulate inhibition by another human monoclonal antibody (i.e., scFv4-20), also isolated from an iTTP patient, that targets the spacer domain of ADAMTS-13. These results provide new insights into our understanding of the pathogenesis of iTTP., (© 2021 International Society on Thrombosis and Haemostasis.)
- Published
- 2021
- Full Text
- View/download PDF
28. CCR5-edited CD4+ T cells augment HIV-specific immunity to enable post-rebound control of HIV replication.
- Author
-
Tebas P, Jadlowsky JK, Shaw PA, Tian L, Esparza E, Brennan AL, Kim S, Naing SY, Richardson MW, Vogel AN, Maldini CR, Kong H, Liu X, Lacey SF, Bauer AM, Mampe F, Richman LP, Lee G, Ando D, Levine BL, Porter DL, Zhao Y, Siegel DL, Bar KJ, June CH, and Riley JL
- Subjects
- Adult, CD8-Positive T-Lymphocytes immunology, Female, Humans, Male, Middle Aged, Viral Load genetics, Viral Load immunology, Virus Replication drug effects, Virus Replication genetics, Anti-Retroviral Agents administration & dosage, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes transplantation, Gene Editing, HIV Infections genetics, HIV Infections immunology, HIV Infections therapy, HIV-1 physiology, Lymphocyte Transfusion, Receptors, CCR5 genetics, Receptors, CCR5 immunology, Virus Replication immunology
- Abstract
BackgroundWe conducted a phase I clinical trial that infused CCR5 gene-edited CD4+ T cells to determine how these T cells can better enable HIV cure strategies.MethodsThe aim of trial was to develop RNA-based approaches to deliver zinc finger nuclease (ZFN), evaluate the effect of CCR5 gene-edited CD4+ T cells on the HIV-specific T cell response, test the ability of infused CCR5 gene-edited T cells to delay viral rebound during analytical treatment interruption, and determine whether individuals heterozygous for CCR5 Δ32 preferentially benefit. We enrolled 14 individuals living with HIV whose viral load was well controlled by antiretroviral therapy (ART). We measured the time to viral rebound after ART withdrawal, the persistence of CCR5-edited CD4+ T cells, and whether infusion of 10 billion CCR5-edited CD4+ T cells augmented the HIV-specific immune response.ResultsInfusion of the CD4+ T cells was well tolerated, with no serious adverse events. We observed a modest delay in the time to viral rebound relative to historical controls; however, 3 of the 14 individuals, 2 of whom were heterozygous for CCR5 Δ32, showed post-viral rebound control of viremia, before ultimately losing control of viral replication. Interestingly, only these individuals had substantial restoration of HIV-specific CD8+ T cell responses. We observed immune escape for 1 of these reinvigorated responses at viral recrudescence, illustrating a direct link between viral control and enhanced CD8+ T cell responses.ConclusionThese findings demonstrate how CCR5 gene-edited CD4+ T cell infusion could aid HIV cure strategies by augmenting preexisting HIV-specific immune responses.REGISTRATIONClinicalTrials.gov NCT02388594.FundingNIH funding (R01AI104400, UM1AI126620, U19AI149680, T32AI007632) was provided by the National Institute of Allergy and Infectious Diseases (NIAID), the National Institute on Drug Abuse (NIDA), the National Institute of Mental Health (NIMH), and the National Institute of Neurological Disorders and Stroke (NINDS). Sangamo Therapeutics also provided funding for these studies.
- Published
- 2021
- Full Text
- View/download PDF
29. 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
30. 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
31. Development of a fully canine anti-canine CTLA4 monoclonal antibody for comparative translational research in dogs with spontaneous tumors.
- Author
-
Mason NJ, Chester N, Xiong A, Rotolo A, Wu Y, Yoshimoto S, Glassman P, Gulendran G, and Siegel DL
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, CTLA-4 Antigen, Dogs, Humans, Mice, Translational Research, Biomedical, Lung Neoplasms, Melanoma
- Abstract
The immune checkpoint inhibitor (ICI) ipilimumab has revolutionized the treatment of patients with different cancer histologies, including melanoma, renal cell carcinoma, and non-small cell lung carcinoma. However, only a subset of patients shows dramatic clinical responses to treatment. Despite intense biomarker discovery efforts linked to clinical trials using CTLA4 checkpoint blockade, no single prognostic correlate has emerged as a valid predictor of outcome. Client-owned, immune competent, pet dogs develop spontaneous tumors that exhibit similar features to human cancers, including shared chromosome aberrations, molecular subtypes, immune signatures, tumor heterogeneity, metastatic behavior, and response to chemotherapy. As such, they represent a valuable parallel patient population in which to investigate novel predictive biomarkers and rational therapeutic ICI combinations. However, the lack of validated, non-immunogenic, canine ICIs for preclinical use hinders this comparative approach. To address this, fully canine single-chain variable fragments (scFvs) that bind canine CTLA4 were isolated from a comprehensive canine scFv phage display library. A lead candidate for clinical development was selected based on its subnanomolar binding affinity to canine CTLA4 and its ability to prevent CTLA4 binding to CD80/CD86 and promote T cell proliferation and effector function. In vivo mouse studies revealed pharmacokinetics similar to isotype control IgG with no evidence of short-term adverse effects. This work paves the way for in vivo analysis of the first fully canine, anti-canine CTLA4 antibody to promote anti-tumor immunity in dogs with immune-responsive cancers and provide an important comparative tool to investigate correlative biomarkers of response and mechanisms of resistance to CTLA4 checkpoint inhibition.
- Published
- 2021
- Full Text
- View/download PDF
32. 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
33. Synthetic nucleic acid antibody prophylaxis confers rapid and durable protective immunity against Zika virus challenge.
- Author
-
Choi H, Kudchodkar SB, Reuschel EL, Asija K, Borole P, Agarwal S, Van Gorder L, Reed CC, Gulendran G, Ramos S, Broderick KE, Kim JJ, Ugen KE, Kobinger G, Siegel DL, Weiner DB, and Muthumani K
- Subjects
- Animals, Antibodies, Neutralizing, Antibodies, Viral, Mice, Nucleic Acids, Viral Vaccines, Zika Virus, Zika Virus Infection prevention & control
- Abstract
Significant concerns have arisen over the past 3 y from the increased global spread of the mosquito-borne flavivirus, Zika. Accompanying this spread has been an increase in cases of the devastating birth defect microcephaly as well as of Guillain-Barré syndrome in adults in many affected countries. Currently there is no vaccine or therapy for this infection; however, we sought to develop a combination approach that provides more rapid and durable protection than traditional vaccination alone. A novel immune-based prophylaxis/therapy strategy entailing the facilitated delivery of a synthetic DNA consensus prME vaccine along with DNA-encoded anti-ZIKV envelope monoclonal antibodies (dMAb) were developed and evaluated for antiviral efficacy. This immediate and persistent protection strategy confers the ability to overcome shortcomings inherent with conventional active vaccination or passive immunotherapy. A collection of novel dMAbs were developed which were potent against ZIKV and could be expressed in serum within 24-48 h of in vivo administration. The DNA vaccine, from a previous development, was potent after adaptive immunity was developed, protecting against infection, brain and testes pathology in relevant mouse challenge models and in an NHP challenge. Delivery of potent dMAbs protected mice from the same murine viral challenge within days of delivery. Combined injection of dMAb and the DNA vaccine afforded rapid and long-lived protection in this challenge model, providing an important demonstration of the advantage of this synergistic approach to pandemic outbreaks.
- Published
- 2020
- Full Text
- View/download PDF
34. Bispecific and split CAR T cells targeting CD13 and TIM3 eradicate acute myeloid leukemia.
- Author
-
He X, Feng Z, Ma J, Ling S, Cao Y, Gurung B, Wu Y, Katona BW, O'Dwyer KP, Siegel DL, June CH, and Hua X
- Subjects
- Animals, CD13 Antigens, Hepatitis A Virus Cellular Receptor 2, Humans, Immunotherapy, Adoptive, Mice, T-Lymphocytes, Leukemia, Myeloid, Acute, Receptors, Antigen, T-Cell
- Abstract
Chimeric antigen receptor (CAR) T cells have radically improved the treatment of B cell-derived malignancies by targeting CD19. The success has not yet expanded to treat acute myeloid leukemia (AML). We developed a Sequentially Tumor-Selected Antibody and Antigen Retrieval (STAR) system to rapidly isolate multiple nanobodies (Nbs) that preferentially bind AML cells and empower CAR T cells with anti-AML efficacy. STAR-isolated Nb157 specifically bound CD13, which is highly expressed in AML cells, and CD13 CAR T cells potently eliminated AML in vitro and in vivo. CAR T cells bispecific for CD13 and TIM3, which are upregulated in AML leukemia stem cells, eradicated patient-derived AML, with much reduced toxicity to human bone marrow stem cells and peripheral myeloid cells in mouse models, highlighting a promising approach for developing effective AML CAR T cell therapy., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
35. CRISPR-engineered T cells in patients with refractory cancer.
- Author
-
Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, Mangan PA, Kulikovskaya I, Gupta M, Chen F, Tian L, Gonzalez VE, Xu J, Jung IY, Melenhorst JJ, Plesa G, Shea J, Matlawski T, Cervini A, Gaymon AL, Desjardins S, Lamontagne A, Salas-Mckee J, Fesnak A, Siegel DL, Levine BL, Jadlowsky JK, Young RM, Chew A, Hwang WT, Hexner EO, Carreno BM, Nobles CL, Bushman FD, Parker KR, Qi Y, Satpathy AT, Chang HY, Zhao Y, Lacey SF, and June CH
- Subjects
- Aged, CRISPR-Associated Protein 9, Cell Engineering, Female, Humans, Male, Middle Aged, Programmed Cell Death 1 Receptor genetics, Transgenes, Adoptive Transfer, CRISPR-Cas Systems, Gene Editing, Receptors, Antigen, T-Cell, alpha-beta genetics, T-Lymphocytes immunology, T-Lymphocytes transplantation
- Abstract
CRISPR-Cas9 gene editing provides a powerful tool to enhance the natural ability of human T cells to fight cancer. We report a first-in-human phase 1 clinical trial to test the safety and feasibility of multiplex CRISPR-Cas9 editing to engineer T cells in three patients with refractory cancer. Two genes encoding the endogenous T cell receptor (TCR) chains, TCRα ( TRAC ) and TCRβ ( TRBC ), were deleted in T cells to reduce TCR mispairing and to enhance the expression of a synthetic, cancer-specific TCR transgene (NY-ESO-1). Removal of a third gene encoding programmed cell death protein 1 (PD-1; PDCD1 ), was performed to improve antitumor immunity. Adoptive transfer of engineered T cells into patients resulted in durable engraftment with edits at all three genomic loci. Although chromosomal translocations were detected, the frequency decreased over time. Modified T cells persisted for up to 9 months, suggesting that immunogenicity is minimal under these conditions and demonstrating the feasibility of CRISPR gene editing for cancer immunotherapy., (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
36. CD19-targeting CAR T cell immunotherapy outcomes correlate with genomic modification by vector integration.
- Author
-
Nobles CL, Sherrill-Mix S, Everett JK, Reddy S, Fraietta JA, Porter DL, Frey N, Gill SI, Grupp SA, Maude SL, Siegel DL, Levine BL, June CH, Lacey SF, Melenhorst JJ, and Bushman FD
- Subjects
- Antigens, CD19 genetics, Female, Humans, Male, Antigens, CD19 immunology, Genetic Vectors, Immunotherapy, Adoptive, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology
- Abstract
Chimeric antigen receptor-engineered T cells targeting CD19 (CART19) provide an effective treatment for pediatric acute lymphoblastic leukemia but are less effective for chronic lymphocytic leukemia (CLL), focusing attention on improving efficacy. CART19 harbor an engineered receptor, which is delivered through lentiviral vector integration, thereby marking cell lineages and modifying the cellular genome by insertional mutagenesis. We recently reported that vector integration within the host TET2 gene was associated with CLL remission. Here, we investigated clonal population structure and therapeutic outcomes in another 39 patients by high-throughput sequencing of vector-integration sites. Genes at integration sites enriched in responders were commonly found in cell-signaling and chromatin modification pathways, suggesting that insertional mutagenesis in these genes promoted therapeutic T cell proliferation. We also developed a multivariate model based on integration-site distributions and found that data from preinfusion products forecasted response in CLL successfully in discovery and validation cohorts and, in day 28 samples, reported responders to CLL therapy with high accuracy. These data clarify how insertional mutagenesis can modulate cell proliferation in CART19 therapy and how data on integration-site distributions can be linked to treatment outcomes.
- Published
- 2020
- Full Text
- View/download PDF
37. CAR T cell viability release testing and clinical outcomes: is there a lower limit?
- Author
-
Chong EA, Levine BL, Grupp SA, Davis MM, Siegel DL, Maude SL, Gladney WL, Frey NV, Porter DL, Hwang WT, Chong ER, June CH, and Schuster SJ
- Subjects
- Humans, Lymphoma, Large B-Cell, Diffuse mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Receptors, Chimeric Antigen, Treatment Outcome, Cell Survival, Immunotherapy, Adoptive standards, Lymphoma, Large B-Cell, Diffuse therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Antigen, T-Cell therapeutic use, T-Lymphocytes
- Published
- 2019
- Full Text
- View/download PDF
38. 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
39. Bullous pemphigoid autoantibody-mediated complement fixation is abolished by the low-molecular-weight heparin tinzaparin sodium.
- Author
-
Gutjahr A, Heck F, Emtenani S, Hammers AK, Hundt JE, Muck P, Siegel DL, Schmidt E, Stanley JR, Zillikens D, and Hammers CM
- Subjects
- Autoantibodies blood, Autoantibodies immunology, Autoantigens immunology, Complement Activation immunology, Complement C5a immunology, Complement Fixation Tests, Dystonin immunology, Humans, Non-Fibrillar Collagens immunology, Off-Label Use, Pemphigoid, Bullous blood, Pemphigoid, Bullous drug therapy, Skin immunology, Skin pathology, Tinzaparin therapeutic use, Collagen Type XVII, Autoantibodies metabolism, Complement Activation drug effects, Complement C5a metabolism, Pemphigoid, Bullous immunology, Tinzaparin pharmacology
- Published
- 2019
- Full Text
- View/download PDF
40. 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
41. 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
42. 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
- Subjects
- Animals, Female, Interleukin-6 metabolism, Male, Mice, STAT3 Transcription Factor metabolism, Transcription, Genetic, Treatment Outcome, Antigens, CD19 metabolism, Immunotherapy, Adoptive, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Receptors, Chimeric Antigen metabolism
- Abstract
Tolerance to self-antigens prevents the elimination of cancer by the immune system
1,2 . We used synthetic chimeric antigen receptors (CARs) to overcome immunological tolerance and mediate tumor rejection in patients with chronic lymphocytic leukemia (CLL). Remission was induced in a subset of subjects, but most did not respond. Comprehensive assessment of patient-derived CAR T cells to identify mechanisms of therapeutic success and failure has not been explored. We performed genomic, phenotypic and functional evaluations to identify determinants of response. Transcriptomic profiling revealed that CAR T cells from complete-responding patients with CLL were enriched in memory-related genes, including IL-6/STAT3 signatures, whereas T cells from nonresponders upregulated programs involved in effector differentiation, glycolysis, exhaustion and apoptosis. Sustained remission was associated with an elevated frequency of CD27+ CD45RO- CD8+ T cells before CAR T cell generation, and these lymphocytes possessed memory-like characteristics. Highly functional CAR T cells from patients produced STAT3-related cytokines, and serum IL-6 correlated with CAR T cell expansion. IL-6/STAT3 blockade diminished CAR T cell proliferation. Furthermore, a mechanistically relevant population of CD27+ PD-1- CD8+ CAR T cells expressing high levels of the IL-6 receptor predicts therapeutic response and is responsible for tumor control. These findings uncover new features of CAR T cell biology and underscore the potential of using pretreatment biomarkers of response to advance immunotherapies.- Published
- 2018
- Full Text
- View/download PDF
43. 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
- Subjects
- Aged, B-Cell Maturation Antigen immunology, Combined Modality Therapy methods, Female, Hematopoietic Stem Cell Transplantation methods, Humans, Immunity, Cellular drug effects, Male, Melphalan administration & dosage, Middle Aged, Multiple Myeloma immunology, Myeloablative Agonists therapeutic use, Receptors, Antigen, T-Cell administration & dosage, Receptors, Antigen, T-Cell immunology, SOXB1 Transcription Factors immunology, T-Lymphocytes drug effects, T-Lymphocytes immunology, Transplantation, Autologous, Treatment Outcome, Antigens, CD19 immunology, Immunotherapy, Adoptive methods, Melphalan therapeutic use, Multiple Myeloma drug therapy, Receptors, Antigen, T-Cell therapeutic use
- Abstract
Background: Multiple myeloma is usually fatal due to serial relapses that become progressively refractory to therapy. CD19 is typically absent on the dominant multiple myeloma cell population but may be present on minor subsets with unique myeloma-propagating properties. To target myeloma-propagating cells, we clinically evaluated autologous T cells transduced with a chimeric antigen receptor (CAR) against CD19 (CTL019)., Methods: Subjects received CTL019 following salvage high-dose melphalan and autologous stem cell transplantation (ASCT). All subjects had relapsed/refractory multiple myeloma and had previously undergone ASCT with less than 1 year progression-free survival (PFS)., Results: ASCT + CTL019 was safe and feasible, with most toxicity attributable to ASCT and no severe cytokine release syndrome. Two of 10 subjects exhibited significantly longer PFS after ASCT + CTL019 compared with prior ASCT (479 vs. 181 days; 249 vs. 127 days). Correlates of favorable clinical outcome included peak CTL019 frequency in bone marrow and emergence of humoral and cellular immune responses against the stem-cell antigen Sox2. Ex vivo treatment of primary myeloma samples with a combination of CTL019 and CAR T cells against the plasma cell antigen BCMA reliably inhibited myeloma colony formation in vitro, whereas treatment with either CAR alone inhibited colony formation inconsistently., Conclusion: CTL019 may improve duration of response to standard multiple myeloma therapies by targeting and precipitating secondary immune responses against myeloma-propagating cells., Trial Registration: Clinicaltrials.gov identifier NCT02135406., Funding: Novartis, NIH, Conquer Cancer Foundation.
- Published
- 2018
- Full Text
- View/download PDF
44. Biocompatible coupling of therapeutic fusion proteins to human erythrocytes.
- Author
-
Villa CH, Pan DC, Johnston IH, Greineder CF, Walsh LR, Hood ED, Cines DB, Poncz M, Siegel DL, and Muzykantov VR
- Subjects
- Animals, Anion Exchange Protein 1, Erythrocyte immunology, Humans, Inflammation drug therapy, Macaca, Mice, Recombinant Fusion Proteins immunology, Recombinant Fusion Proteins therapeutic use, Rh-Hr Blood-Group System immunology, Single-Chain Antibodies administration & dosage, Single-Chain Antibodies genetics, Single-Chain Antibodies immunology, Thrombomodulin administration & dosage, Thrombomodulin genetics, Thrombosis pathology, Drug Delivery Systems methods, Erythrocytes immunology, Recombinant Fusion Proteins administration & dosage, Thrombosis drug therapy
- Abstract
Carriage of drugs by red blood cells (RBCs) modulates pharmacokinetics, pharmacodynamics, and immunogenicity. However, optimal targets for attaching therapeutics to human RBCs and adverse effects have not been studied. We engineered nonhuman-primate single-chain antibody fragments (scFvs) directed to human RBCs and fused scFvs with human thrombomodulin (hTM) as a representative biotherapeutic cargo (hTM-scFv). Binding fusions to RBCs on band 3/glycophorin A (GPA; Wright b [Wr
b ] epitope) and RhCE (Rh17/Hr0 epitope) similarly endowed RBCs with hTM activity, but differed in their effects on RBC physiology. scFv and hTM-scFv targeted to band 3/GPA increased membrane rigidity and sensitized RBCs to hemolysis induced by mechanical stress, while reducing sensitivity to hypo-osmotic hemolysis. Similar properties were seen for other ligands bound to GPA and band 3 on human and murine RBCs. In contrast, binding of scFv or hTM-scFv to RhCE did not alter deformability or sensitivity to mechanical and osmotic stress at similar copy numbers bound per RBCs. Contrasting responses were also seen for immunoglobulin G antibodies against band 3, GPA, and RhCE. RBC-bound hTM-scFv generated activated protein C (APC) in the presence of thrombin, but RhCE-targeted hTM-scFv demonstrated greater APC generation per bound copy. Both Wrb - and RhCE-targeted fusion proteins inhibited fibrin deposition induced by tumor necrosis factor-α in an endothelialized microfluidic model using human whole blood. RhCE-bound hTM-scFv more effectively reduced platelet and leukocyte adhesion, whereas anti-Wrb scFv appeared to promote platelet adhesion. These data provide a translational framework for the development of engineered affinity ligands to safely couple therapeutics to human RBCs., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
45. Targeting thrombomodulin to circulating red blood cells augments its protective effects in models of endotoxemia and ischemia-reperfusion injury.
- Author
-
Carnemolla R, Villa CH, Greineder CF, Zaitsev S, Patel KR, Kowalska MA, Atochin DN, Cines DB, Siegel DL, Esmon CT, and Muzykantov VR
- Subjects
- Animals, Inflammation drug therapy, Male, Membrane Fusion Proteins, Mice, Mice, Inbred C57BL, Thrombomodulin chemistry, Endotoxemia prevention & control, Erythrocytes metabolism, Reperfusion Injury prevention & control, Thrombomodulin administration & dosage, Thrombomodulin therapeutic use
- Abstract
Endothelial thrombomodulin (TM) regulates coagulation and inflammation via several mechanisms, including production of activated protein C (APC). Recombinant APC and soluble fragments of TM (sTM) have been tested in settings associated with insufficiency of the endogenous TM/APC pathway, such as sepsis. We previously designed a fusion protein of TM [single-chain variable fragment antibody (scFv)/TM] targeted to red blood cells (RBCs) to improve pharmacokinetics and antithrombotic effects without increasing bleeding. Here, scFv/TM was studied in mouse models of systemic inflammation and ischemia-reperfusion injury. Injected concomitantly with or before endotoxin, scFv/TM provided more potent protection against liver injury and release of pathological mediators than sTM, showing similar efficacy at up to 50-fold lower doses. scFv/TM provided protection when injected after endotoxin, whereas sTM did not, and augmented APC production by thrombin ∼50-fold more than sTM. However, scFv/TM injected after endotoxin did not reduce thrombin/antithrombin complexes; nor did antibodies that block APC anticoagulant activity suppress the prophylactic anti-inflammatory effect of scFv/TM. Therefore, similar to endogenous TM, RBC-anchored scFv/TM activates several protective pathways. Finally, scFv/TM was more effective at reducing cerebral infarct volume and alleviated neurological deficits than sTM after cerebral ischemia/reperfusion injury. These results indicate that RBC-targeted scFv/TM exerts multifaceted cytoprotective effects and may find utility in systemic and focal inflammatory and ischemic disorders.-Carnemolla, R., Villa, C. H., Greineder, C. F., Zaitseva, S., Patel, K. R., Kowalska, M. A., Atochin, D. N., Cines, D. B., Siegel, D. L., Esmon, C. T., Muzykantov, V. R. Targeting thrombomodulin to circulating red blood cells augments its protective effects in models of endotoxemia and ischemia-reperfusion injury., (© FASEB.)
- Published
- 2017
- Full Text
- View/download PDF
46. Proteomic Analysis of Pemphigus Autoantibodies Indicates a Larger, More Diverse, and More Dynamic Repertoire than Determined by B Cell Genetics.
- Author
-
Chen J, Zheng Q, Hammers CM, Ellebrecht CT, Mukherjee EM, Tang HY, Lin C, Yuan H, Pan M, Langenhan J, Komorowski L, Siegel DL, Payne AS, and Stanley JR
- Subjects
- Amino Acid Sequence, Cell Surface Display Techniques, Chromatography, Liquid, Clone Cells, Complementarity Determining Regions genetics, Desmogleins metabolism, Humans, Mutation genetics, Pemphigus blood, Single-Chain Antibodies chemistry, Single-Chain Antibodies metabolism, Tandem Mass Spectrometry, Autoantibodies immunology, B-Lymphocytes immunology, Pemphigus genetics, Pemphigus immunology, Proteomics methods
- Abstract
In autoantibody-mediated diseases such as pemphigus, serum antibodies lead to disease. Genetic analysis of B cells has allowed characterization of antibody repertoires in such diseases but would be complemented by proteomic analysis of serum autoantibodies. Here, we show using proteomic analysis that the serum autoantibody repertoire in pemphigus is much more polyclonal than that found by genetic studies of B cells. In addition, many B cells encode pemphigus autoantibodies that are not secreted into the serum. Heavy chain variable gene usage of serum autoantibodies is not shared among patients, implying targeting of the coded proteins will not be a useful therapeutic strategy. Analysis of autoantibodies in individual patients over several years indicates that many antibody clones persist but the proportion of each changes. These studies indicate a dynamic and diverse autoantibody response not revealed by genetic studies and explain why similar overall autoantibody titers may give variable disease activity., Competing Interests: JL and LK are employees of the Euroimmun AG, a company that develops, produces and manufactures immunoassays for the detection of disease-associated antibodies. The other authors have nothing to disclose., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. Erythrocytes as Carriers for Drug Delivery in Blood Transfusion and Beyond.
- Author
-
Villa CH, Cines DB, Siegel DL, and Muzykantov V
- Subjects
- Humans, Blood Transfusion methods, Drug Carriers therapeutic use, Drug Delivery Systems methods, Erythrocytes physiology
- Abstract
Red blood cells (RBCs) are innate carriers that can also be engineered to improve the pharmacokinetics and pharmacodynamics of many drugs, particularly biotherapeutics. Successful loading of drugs, both internally and on the external surface of RBCs, has been demonstrated for many drugs including anti-inflammatory, antimicrobial, and antithrombotic agents. Methods for internal loading of drugs within RBCs are now entering clinical use. Although internal loading can result in membrane disruption that may compromise biocompatibility, surface loading using either affinity or chemical ligands offers a diverse set of approaches for the production of RBC drug carriers. A wide range of surface determinants is potentially available for this approach, although there remains a need to characterize the effects of coupling agents to these surface proteins. Somewhat surprisingly, recent data also suggest that red cell-mediated delivery may confer tolerogenic immune effects. Questions remaining before widespread application of these technologies include determining the optimal loading protocol, source of RBCs, and production logistics, as well as addressing regulatory hurdles. Red blood cell drug carriers, after many decades of progress, are now poised to enter the clinic and broaden the potential application of RBCs in blood transfusion., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Modulation of natural IgM autoantibodies to oxidative stress-related neo-epitopes on apoptotic cells in newborns of mothers with anti-Ro autoimmunity.
- Author
-
Grönwall C, Clancy RM, Getu L, Lloyd KA, Siegel DL, Reed JH, Buyon JP, and Silverman GJ
- Subjects
- Adult, Antibodies, Anti-Idiotypic blood, Autoantibodies blood, Autoantigens immunology, Cohort Studies, Enzyme-Linked Immunosorbent Assay, Epitopes chemistry, Female, Fetal Blood immunology, Humans, Immunoglobulin G immunology, Infant, Newborn, Malondialdehyde adverse effects, Malondialdehyde chemistry, Malondialdehyde immunology, Mothers, Phosphorylcholine adverse effects, Phosphorylcholine blood, Pregnancy, Pregnancy Complications, Ribonucleoproteins chemistry, Apoptosis immunology, Autoantibodies immunology, Epitopes immunology, Fetus immunology, Immunoglobulin M immunology, Maternal-Fetal Exchange immunology, Oxidative Stress immunology, Ribonucleoproteins immunology
- Abstract
At birth, the human immune system already contains substantial levels of polymeric IgM, that include autoantibodies to neo-epitopes on apoptotic cells (ACs) that are proposed to play homeostatic and anti-inflammatory roles. Yet the biologic origins and developmental regulation of these naturally arising antibodies remain poorly understood. Herein, we report that levels of IgM-antibodies to malondialdehyde (MDA) protein adducts, a common type of in vivo generated oxidative stress-related neoepitope, directly correlate with the relative binding of neonatal-IgM to ACs. Levels of IgM to phosphorylcholine (PC), a natural antibody prevalent in adults, were relatively scant in cord blood, while there was significantly greater relative representation of IgM anti-MDA antibodies in newborns compared to adults. To investigate the potential interrelationships between neonatal IgM with pathogenic IgG-autoantibodies, we studied 103 newborns born to autoimmune mothers with IgG anti-Ro (i.e., 70 with neonatal lupus and 33 without neonatal lupus). In these subjects the mean levels of IgM anti-Ro60 were significantly higher than in the newborns from non-autoimmune mothers. In contrast, levels of IgM anti-MDA in IgG anti-Ro exposed neonates were significantly lower than in neonates from non-autoimmune mothers. The presence or absence of neonatal lupus did not appear to influence the total levels of IgM in the anti-Ro exposed newborns. Taken together, our studies provide evidence that the immune development of the natural IgM-repertoire may be affected, and become imprinted by, the transfer of maternal IgG into the fetus., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
49. ADAMTS13 autoantibodies cloned from patients with acquired thrombotic thrombocytopenic purpura: 1. Structural and functional characterization in vitro.
- Author
-
Ostertag EM, Kacir S, Thiboutot M, Gulendran G, Zheng XL, Cines DB, and Siegel DL
- Subjects
- Adult, Antibody Specificity, Cell Surface Display Techniques, Child, Cloning, Molecular, Cross Reactions immunology, Epitope Mapping, Humans, Immunoglobulin G blood, Middle Aged, ADAMTS13 Protein immunology, Autoantibodies isolation & purification, Purpura, Thrombotic Thrombocytopenic immunology
- Abstract
Background: Acquired thrombotic thrombocytopenia purpura (TTP) is a life-threatening illness caused by autoantibodies that decrease the activity of ADAMTS13, the von Willebrand factor-cleaving protease. Despite efficacy of plasma exchange, mortality remains high and relapse is common. Improved therapies may come from understanding the diversity of pathogenic autoantibodies on a molecular or genetic level. Cloning comprehensive repertoires of patient autoantibodies can provide the necessary tools for studying immunobiology of disease and developing animal models., Study Design and Methods: Anti-ADAMTS13 antibodies were cloned from four patients with acquired TTP using phage display and characterized with respect to genetic origin, inhibition of ADAMTS13 proteolytic activity, and epitope specificity. Anti-idiotypic antisera raised to a subset of autoantibodies enabled comparison of their relatedness to each other and to polyclonal immunoglobulin (Ig)G in patient plasma., Results: Fifty-one unique antibodies were isolated comprising epitope specificities resembling the diversity found in circulating patient IgG. Antibodies directed both to the amino terminal domains and to those requiring the ADAMTS13 cysteine-rich/spacer region for binding inhibited proteolytic activity, while those solely targeting carboxy-terminal domains were noninhibitory. Anti-idiotypic antisera raised to a subset of antibody clones crossreacted with and reduced the inhibitory activity of polyclonal IgG from a set of unrelated patients., Conclusions: Anti-ADAMTS13 autoantibodies isolated by repertoire cloning display the diversity of epitope specificities found in patient plasma and provide tools for developing animal models of acquired TTP. Shared idiotypes of inhibitory clones with circulating IgG from multiple patients suggest common features of pathogenic autoantibodies that could be exploited for developing more targeted therapies., (© 2016 AABB.)
- Published
- 2016
- Full Text
- View/download PDF
50. ADAMTS13 autoantibodies cloned from patients with acquired thrombotic thrombocytopenic purpura: 2. Pathogenicity in an animal model.
- Author
-
Ostertag EM, Bdeir K, Kacir S, Thiboutot M, Gulendran G, Yunk L, Hayes VM, Motto DG, Poncz M, Zheng XL, Cines DB, and Siegel DL
- Subjects
- ADAMTS13 Protein immunology, Animals, Cloning, Molecular, DNA, Complementary administration & dosage, Humans, Mice, Models, Animal, Molecular Targeted Therapy methods, Single-Chain Antibodies genetics, Single-Chain Antibodies toxicity, von Willebrand Factor metabolism, ADAMTS13 Protein antagonists & inhibitors, Autoantibodies genetics, Purpura, Thrombotic Thrombocytopenic immunology, Purpura, Thrombotic Thrombocytopenic therapy
- Abstract
Background: Acquired thrombotic thrombocytopenic purpura (TTP) is a potentially fatal disease in which ultralarge von Willebrand factor (UL-VWF) multimers accumulate as a result of autoantibody inhibition of the VWF protease, ADAMTS13. Current treatment is not specifically directed at the responsible autoantibodies and in some cases is ineffective or of transient benefit. More rational, reliable, and durable therapies are needed, and a human autoantibody-mediated animal model would be useful for their development. Previously, TTP patient anti-ADAMTS13 single-chain variable-region fragments (scFv's) were cloned that inhibited ADAMTS13 proteolytic activity in vitro and expressed features in common with inhibitory immunoglobulin G in patient plasma. Here, pathogenicity of these scFv's is explored in vivo by transfecting mice with inhibitory antibody cDNA., Study Design and Methods: Hydrodynamic tail vein injection of naked DNA encoding human anti-ADAMTS13 scFv was used to create sustained ADAMTS13 inhibition in mice. Accumulation of UL-VWF multimers was measured and formation of platelet (PLT) thrombi after focal or systemic vascular injury was examined., Results: Transfected mice expressed physiological plasma levels of human scFv and developed sustained ADAMTS13 inhibition and accumulation of unprocessed UL-VWF multimers. Induced focal endothelial injury generated PLT thrombi extending well beyond the site of initial injury, and systemic endothelial injury induced thrombocytopenia, schistocyte formation, PLT thrombi, and death., Conclusions: These results demonstrate for the first time the ability of human recombinant monovalent anti-ADAMTS13 antibody fragments to recapitulate key pathologic features of untreated acquired TTP in vivo, validating their clinical significance and providing an animal model for testing novel targeted therapeutic approaches., (© 2016 AABB.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.