65 results on '"Garfall AL"'
Search Results
2. B06: SUBCUTANEOUS TALQUETAMAB IN COMBINATION WITH DARATUMUMAB IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM): PHASE 1B RESULTS
- Author
-
Mateos, M, primary, Hari, P, additional, Bahlis, N, additional, Chari, A, additional, van de Donk, NWCJ, additional, Dholaria, B, additional, Garfall, AL, additional, Goldschmidt, H, additional, Kortüm, KM, additional, Krishnan, A, additional, Martin, T, additional, Morillo, D, additional, Oriol, A, additional, Reece, D, additional, Rodriguez, C, additional, Rodríguez-Otero, P, additional, San-Miguel, JF, additional, Usmani, SZ, additional, Verona, R, additional, Lin, SXW, additional, Prior, TJ, additional, Wade, M, additional, Weiss, B, additional, Goldberg, JD, additional, and Askari, E, additional
- Published
- 2022
- Full Text
- View/download PDF
3. Updated phase 1 results of teclistamab, a B-cell maturation antigen (BCMA) x CD3 bispecific antibody, in relapsed/refractory multiple myeloma (MM)
- Author
-
Krishnan, AY, Garfall, AL, Mateos, MV, van de Donk, NWCJ, Nahi, H, San-Miguel, JF, Oriol, A, Rosinol, L, Chari, A, Bhutani, M, Karlin, L, Benboubker, L, Pei, LX, Verona, R, Girgis, S, Stephenson, T, Goldberg, JD, Banerjee, A, and Usmani, SZ
- Published
- 2021
4. Efficacy and safety of teclistamab in patients with relapsed/refractory multiple myeloma after BCMA-targeting therapies.
- Author
-
Touzeau C, Krishnan AY, Moreau P, Perrot A, Usmani SZ, Manier S, Cavo M, Martinez Chamorro C, Nooka AK, Martin TG, Karlin L, Leleu X, Bahlis NJ, Besemer B, Pei L, Stein S, Wang Lin SX, Trancucci D, Verona RI, Girgis S, Miao X, Uhlar CM, Chastain K, and Garfall AL
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Adult, Immunotherapy, Adoptive adverse effects, Treatment Outcome, Aged, 80 and over, Multiple Myeloma drug therapy, Multiple Myeloma mortality, Multiple Myeloma therapy, B-Cell Maturation Antigen antagonists & inhibitors, B-Cell Maturation Antigen immunology, Antibodies, Bispecific therapeutic use, Antibodies, Bispecific adverse effects, Antibodies, Bispecific administration & dosage
- Abstract
Abstract: Teclistamab is a B-cell maturation antigen (BCMA)-directed bispecific antibody approved for the treatment of patients with triple-class exposed relapsed/refractory multiple myeloma (R/RMM). In the phase 1/2 MajesTEC-1 study, a cohort of patients who had prior BCMA-targeted therapy (antibody-drug conjugate [ADC] or chimeric antigen receptor T-cell [CAR-T] therapy) was enrolled to explore teclistamab in patients previously exposed to anti-BCMA treatment. At a median follow-up of 28.0 months (range, 0.7-31.1), 40 patients with prior BCMA-targeted therapy had received subcutaneous 1.5 mg/kg weekly teclistamab. The median prior lines of treatment was 6 (range, 3-14). Prior anti-BCMA therapy included ADC (n = 29), CAR-T (n = 15), or both (n = 4). The overall response rate was 52.5%; 47.5% of patients achieved very good partial response or better, and 30.0% achieved complete response or better. The median duration of response was 14.8 months, the median progression-free survival was 4.5 months, and the median overall survival was 15.5 months. The most common treatment-emergent adverse events (TEAEs) were neutropenia, infections, cytokine release syndrome, and anemia; cytopenias and infections were the most common grade ≥3 TEAEs. Infections occurred in 28 patients (70.0%; maximum grade 3/4, n = 13 [32.5%]; grade 5, n = 4 [10%]). Before starting teclistamab, baseline BCMA expression and immune characteristics were unaffected by prior anti-BCMA treatment. The MajesTEC-1 trial cohort C results demonstrate favorable efficacy and safety of teclistamab in patients with heavily pretreated R/RMM and prior anti-BCMA treatment. This trial was registered at www.ClinicalTrials.gov as #NCT03145181 and #NCT04557098., (© 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
5. T Cell-Redirecting Bispecific Antibodies in Multiple Myeloma: Optimal Dosing Schedule and Duration of Treatment.
- Author
-
van de Donk NWCJ, Rasche L, Sidana S, Zweegman S, and Garfall AL
- Subjects
- Humans, T-Lymphocytes immunology, Drug Administration Schedule, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological pharmacology, Multiple Myeloma immunology, Multiple Myeloma therapy, Multiple Myeloma drug therapy, Antibodies, Bispecific therapeutic use, Antibodies, Bispecific administration & dosage
- Abstract
T cell-redirecting bispecific antibodies (BsAb) induce significant responses in heavily pretreated multiple myeloma. BsAbs are currently administered in a dose-dense manner until disease progression. However, continuous therapy is associated with safety concerns, including a high risk of infections and high costs. In addition, chronic exposure to BsAbs, and thus long-term T-cell stimulation, induces T-cell exhaustion, which may contribute to relapse. There is increasing evidence that the strategy of induction treatment followed by maintenance with longer intervals between BsAb doses, or limited treatment duration with cessation of therapy in patients who achieve deep remission, improves the balance between toxicity and efficacy. Significance: There is increasing evidence that after initial debulking, less-frequent BsAb administration mitigates T-cell exhaustion and minimizes the potential for chronic or cumulative toxicity while maintaining durable clinical responses. In addition, specific patient subsets may experience an extended treatment-free period following fixed-duration treatment. Fixed-duration treatment may, therefore, decrease cumulative toxicities and the burden on patients and healthcare systems., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
6. Impact of COVID-19 on outcomes with teclistamab in patients with relapsed/refractory multiple myeloma in the phase 1/2 MajesTEC-1 study.
- Author
-
van de Donk NWCJ, Bahlis N, Costa LJ, Mateos MV, Nooka AK, Perrot A, Garfall AL, Thaman P, Qi K, Uhlar C, Chastain K, Doyle M, and Usmani SZ
- Published
- 2024
- Full Text
- View/download PDF
7. Bendamustine as Lymphodepletion for Brexucabtagene Autoleucel Therapy of Mantle Cell Lymphoma.
- Author
-
Chong EA, Chong ER, Therwhanger D, Nasta SD, Landsburg DJ, Barta SK, Svoboda J, Gerson JN, Ghilardi G, Paruzzo L, Fraietta JA, Weber E, Stefano N, Porter DL, Frey NV, Garfall AL, Ruella M, and Schuster SJ
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Retrospective Studies, Vidarabine analogs & derivatives, Vidarabine therapeutic use, Vidarabine administration & dosage, Adult, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating administration & dosage, Antigens, CD19 immunology, Treatment Outcome, Bendamustine Hydrochloride therapeutic use, Bendamustine Hydrochloride administration & dosage, Lymphoma, Mantle-Cell drug therapy, Immunotherapy, Adoptive methods
- Abstract
Brexucabtagene autoleucel (brexu-cel) is an autologous CD19-directed chimeric antigen receptor (CAR) T-cell therapy approved for treatment of relapsed/refractory mantle cell lymphoma (MCL). During a fludarabine shortage, we used bendamustine as an alternative to standard cyclophosphamide/fludarabine (cy/flu) lymphodepletion (LD) prior to brexu-cel. We assessed MCL patient outcomes as well as CAR T-cell expansion and persistence after brexu-cel following bendamustine or cy/flu LD at our center. This was a retrospective single institution study that utilized prospectively banked blood and tissue samples. Clinical efficacy was assessed by 2014 Lugano guidelines. CAR T-cell expansion and persistence in peripheral blood were assessed on day 7 and at ≥month 6 for patients with available samples. Seventeen patients received bendamustine and 5 received cy/flu. For the bendamustine cohort, 14 (82%) received bridging therapy and 4 (24%) had CNS involvement. Fifteen patients (88%) developed CRS with 4 (24%) ≥grade 3 events. Six (35%) patients developed ICANS with 4 (24%) events ≥grade 3. No patient had ≥grade 3 cytopenias at day 90. Best objective (BOR) and complete response (CRR) rates were 82% and 65%, respectively. At 24.5 months median follow-up, 12-month progression-free survival (PFS) was 45%, 24-month PFS was 25%, and median duration of response was 19 months. Median OS was not reached. BOR was 25% (1/4) for patients with CNS involvement. CAR transgene expansion after bendamustine LD was observed on day 7 in all (4/4) patients tested and persisted at ≥6 months (2/2), regardless of response. Bendamustine LD before brexu-cel for MCL is feasible and safe with a lower frequency and shorter duration of cytopenias than reported for cy/flu. Both CAR T-cell expansion and persistence were observed after bendamustine LD. Outcomes appear comparable to the real world outcomes reported with cy/flu LD., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
8. Human IL-6 fosters long-term engraftment of patient-derived disease-driving myeloma cells in immunodeficient mice.
- Author
-
Hasanali ZS, Garfall AL, Burzenski L, Shultz LD, Tang Y, Kadu S, Sheppard NC, Liu W, Dopkin D, Vogl DT, Cohen AD, Waxman AJ, Susanibar-Adaniya SP, Carroll M, Stadtmauer EA, and Allman D
- Subjects
- Animals, Female, Humans, Male, Mice, Bortezomib pharmacology, Bortezomib therapeutic use, Disease Models, Animal, Mice, Transgenic, Monoclonal Gammopathy of Undetermined Significance immunology, Monoclonal Gammopathy of Undetermined Significance pathology, Plasma Cells immunology, Interleukin-6 metabolism, Multiple Myeloma immunology, Multiple Myeloma pathology
- Abstract
Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned human IL-6-transgenic (hIL-6-transgenic) NSG (NSG+hIL6) mice reliably support the engraftment of malignant and premalignant human plasma cells, including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and postrelapse myeloma, plasma cell leukemia, and amyloid light chain amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single-cell RNA sequencing showed nonmalignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma-engrafted mice given CAR T cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient-derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.
- Published
- 2024
- Full Text
- View/download PDF
9. Efficacy and safety of bendamustine for lymphodepletion before lisocabtagene maraleucel.
- Author
-
Ghilardi G, Paruzzo L, Patel V, Svoboda J, Chong ER, Fardella E, Chong EA, Gabrielli G, Nasta SD, Landsburg DJ, Carter J, Pajarillo R, Barta SK, White G, Weber E, Napier E, Porter DL, Garfall AL, Schuster SJ, and Ruella M
- Subjects
- Humans, Middle Aged, Male, Female, Aged, Retrospective Studies, Adult, Lymphoma, Large B-Cell, Diffuse drug therapy, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Biological Products therapeutic use, Biological Products adverse effects, Aged, 80 and over, Treatment Outcome, Bendamustine Hydrochloride therapeutic use, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects
- Abstract
Bendamustine has been retrospectively shown to be an effective and safe lymphodepletion regimen prior to the anti-CD19 chimeric antigen receptor T cell (CART) products tisagenlecleucel and axicabtagene ciloleucel, as well as the anti-BCMA CART products idecabtagene vicleucel and ciltacabtagene autoleucel. However, bendamustine as lymphodepletion prior to lisocabtagene maraleucel (liso-cel), a 4-1BB co-stimulated, fixed CD4:CD8 ratio anti-CD19 CART product, has not been described yet. Thus, we studied a cohort of sequentially-treated patients with large B-cell lymphomas who received bendamustine lymphodepletion before liso-cel at the University of Pennsylvania between 5/2021 and 12/2023 (n = 31). Patients were evaluated for toxicities and responses. Of note, 7 patients (22.6%) would have dnot met the inclusion criteria for the registrational liso-cel clinical trials, mostly due to older age. Overall and complete response rates were 76.9% and 73.1%, respectively. At a median follow-up of 6.3 months, the 6-month progression-free and overall survival were 59.9% and 91.1%, respectively. Rates of cytokine-release syndrome (CRS) and neurotoxicity (ICANS) of any grade were 9.7% and 9.7%, respectively, with no grade ≥ 3 events. No infections were reported during the first 30 days following liso-cel infusion. Neutropenia ≥ grade 3 was observed in 29.0% of patients; thrombocytopenia ≥ grade 3 occurred in 9.7%. In conclusion, bendamustine lymphodepletion before liso-cel appears to be a strategy that can drive tumor responses while ensuring a mild toxicity profile., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. CAR T-Cell Immunotherapy in Minority Patients with Lymphoma.
- Author
-
Ghilardi G, Williamson S, Pajarillo R, Paruzzo L, Chen L, Grady C, Doucette A, Nemecek E, Gabrielli G, Barta SK, Svoboda J, Chong EA, Yelton R, Nasta SD, Landsburg DJ, Ugwuanyi O, Chen AI, Schachter L, White G, Ballard HJ, Weber E, Curley C, Porter DL, Garfall AL, Hwang WT, Guerra CE, Maziarz RT, Schuster SJ, and Ruella M
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Minority Groups statistics & numerical data, Receptors, Chimeric Antigen immunology, Antigens, CD19 immunology, Antigens, CD19 therapeutic use, Immunotherapy, Adoptive adverse effects
- Abstract
Background: Administration of anti-CD19 chimeric antigen receptor T-cell (CART19) immunotherapy for large B-cell lymphomas (LBCLs), a subset of non-Hodgkin lymphoma (NHL), involves high costs and access to specialized tertiary care centers. We investigated whether minority health populations (MHPs) have equal access to CART19 and whether their outcomes are similar to those of non-MHPs., Methods: We analyzed the prevalence and clinical outcomes of patients treated with commercial CART19 at two geographically and socioeconomically different institutions: the Abramson Cancer Center (ACC, Philadelphia, Pennsylvania) and the Knight Cancer Institute (KCI, Portland, Oregon)., Results: In the ACC catchment area, 8956 patients were diagnosed with NHL between 2015 and 2019 (latest available data from the state registry), including 17.9% MHPs. In the ACC, between 2018 and 2022 (CART became available in 2018), 1492 patients with LBCL were treated, and 194 received CART19. The proportion of MHPs was 15.7% for the entire LBCL cohort but only 6.7% for the CART19 cohort. During the same time, in the KCI catchment area, 4568 patients were diagnosed with NHL, including 4.2% MHPs. In the KCI, 396 patients with LBCL were treated, and 47 received CART19. The proportion of MHPs was 6.6% for the entire LBCL cohort and 4.2% for the CART19 cohort. The 3-month response, survival, and toxicities after CART19 infusion showed similar results, although the number of patients who were treated was limited., Conclusions: This study shows that the access of MHPs to tertiary centers for LBCL care was preserved but appeared reduced for commercial CART19 immunotherapy. Although clinical outcomes of MHPs seemed similar to those of non-MHPs, the small sample size precludes drawing firm conclusions. Further studies are needed. (Funded by the Laffey McHugh Foundation and others.).
- Published
- 2024
- Full Text
- View/download PDF
11. Chimeric Antigen Receptor T Cells in the Treatment of Multiple Myeloma.
- Author
-
Hasanali ZS, Razzo B, Susanibar-Adaniya SP, Garfall AL, Stadtmauer EA, and Cohen AD
- Subjects
- Humans, Immunotherapy, Adoptive, B-Cell Maturation Antigen, T-Lymphocytes, Multiple Myeloma therapy, Multiple Myeloma pathology, Receptors, Chimeric Antigen genetics
- Abstract
Chimeric antigen receptor T cells (CARTs) represent another powerful way to leverage the immune system to fight malignancy. Indeed, in multiple myeloma, the high response rate and duration of response to B cell maturation antigen-targeted therapies in later lines of disease has led to 2 Food and Drug Administration (FDA) drug approvals and opened the door to the development of this drug class. This review aims to provide an update on the 2 FDA-approved products, summarize the data for the most promising next-generation multiple myeloma CARTs, and outline current challenges in the field and potential solutions., Competing Interests: Disclosure Z.S. Hasanali, B. Razzo and S.P. Susanibar-Adaniya have nothing to disclose. A.L. Garfall: Consulting: BMS, Janssen, Novartis, GSK, Legend; Research funding: Novartis, Switzerland, Janssen, United States, Tmunity, CRISPR therapeutics; Patents in CAR T cell therapy: Novartis. E.A. Stadtmauer: Consultant for Janssen and BMS and grant funding from Sorrento and Abbvie. A.D. Cohen: Consulting: Janssen, GSK, BMS/Celgene, Genentech/Roche, Pfizer, Abbvie, Arcellx, Ichnos, Takeda; Research Funding: Novartis, GSK, Genentech, United States; Patents in CAR T cell therapy: Novartis., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. T cell lymphoma and secondary primary malignancy risk after commercial CAR T cell therapy.
- Author
-
Ghilardi G, Fraietta JA, Gerson JN, Van Deerlin VM, Morrissette JJD, Caponetti GC, Paruzzo L, Harris JC, Chong EA, Susanibar Adaniya SP, Svoboda J, Nasta SD, Ugwuanyi OH, Landsburg DJ, Fardella E, Waxman AJ, Chong ER, Patel V, Pajarillo R, Kulikovskaya I, Lieberman DB, Cohen AD, Levine BL, Stadtmauer EA, Frey NV, Vogl DT, Hexner EO, Barta SK, Porter DL, Garfall AL, Schuster SJ, June CH, and Ruella M
- Subjects
- Humans, Immunotherapy, Adoptive adverse effects, Receptors, Antigen, T-Cell genetics, Antigens, CD19, Receptors, Chimeric Antigen genetics, Lymphoma, B-Cell, Lymphoma, T-Cell, Hematologic Neoplasms, Lung Neoplasms
- Abstract
We report a T cell lymphoma (TCL) occurring 3 months after anti-CD19 chimeric antigen receptor (CAR) T cell immunotherapy for non-Hodgkin B cell lymphoma. The TCL was diagnosed from a thoracic lymph node upon surgery for lung cancer. The TCL exhibited CD8
+ cytotoxic phenotype and a JAK3 variant, while the CAR transgene was very low. The T cell clone was identified at low levels in the blood before CAR T infusion and in lung cancer. To assess the overall risk of secondary primary malignancy after commercial CAR T (CD19, BCMA), we analyzed 449 patients treated at the University of Pennsylvania. At a median follow-up of 10.3 months, 16 patients (3.6%) had a secondary primary malignancy. The median onset time was 26.4 and 9.7 months for solid and hematological malignancies, respectively. The projected 5-year cumulative incidence is 15.2% for solid and 2.3% for hematological malignancies. Overall, one case of TCL was observed, suggesting a low risk of TCL after CAR T., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
13. Bendamustine lymphodepletion before axicabtagene ciloleucel is safe and associates with reduced inflammatory cytokines.
- Author
-
Ghilardi G, Paruzzo L, Svoboda J, Chong EA, Shestov AA, Chen L, Cohen IJ, Gabrielli G, Nasta SD, Porazzi P, Landsburg DJ, Gerson JN, Carter J, Barta SK, Yelton R, Pajarillo R, Patel V, White G, Ballard HJ, Weber E, Napier E, Chong ER, Fraietta JA, Garfall AL, Porter DL, Milone MC, O'Connor R, Schuster SJ, and Ruella M
- Subjects
- Humans, Bendamustine Hydrochloride adverse effects, CD28 Antigens, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Cyclophosphamide, Cytokines, Lymphoma, Follicular, Biological Products
- Abstract
Abstract: Lymphodepletion (LD) is an integral component of chimeric antigen receptor T-cell (CART) immunotherapies. In this study, we compared the safety and efficacy of bendamustine (Benda) to standard fludarabine/cyclophosphamide (Flu/Cy) LD before CD19-directed, CD28-costimulated CART axicabtagene ciloleucel (axi-cel) for patients with large B-cell lymphoma (LBCL) and follicular lymphoma (FL). We analyzed 59 patients diagnosed with LBCL (n = 48) and FL (n = 11) consecutively treated with axi-cel at the University of Pennsylvania. We also analyzed serum samples for cytokine levels and metabolomic changes before and after LD. Flu/Cy and Benda demonstrated similar efficacy, with complete remission rates of 51.4% and 50.0% (P = .981), respectively, and similar progression-free and overall survivals. Any-grade cytokine-release syndrome occurred in 91.9% of patients receiving Flu/Cy vs 72.7% of patients receiving Benda (P = .048); any-grade neurotoxicity after Flu/Cy occurred in 45.9% of patients and after Benda in 18.2% of patients (P = .031). In addition, Flu/Cy was associated with a higher incidence of grade ≥3 neutropenia (100% vs 54.5%; P < .001), infections (78.4% vs 27.3%; P < .001), and neutropenic fever (78.4% vs 13.6%; P < .001). These results were confirmed both in patients with LBCL and those with FL. Mechanistically, patients with Flu/Cy had a greater increase in inflammatory cytokines associated with neurotoxicity and reduced levels of metabolites critical for redox balance and biosynthesis. This study suggests that Benda LD may be a safe alternative to Flu/Cy for CD28-based CART CD19-directed immunotherapy with similar efficacy and reduced toxicities. Benda is associated with reduced levels of inflammatory cytokines and increased anabolic metabolites., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. New Biological Therapies for Multiple Myeloma.
- Author
-
Garfall AL
- Subjects
- Humans, Immunotherapy, Proteasome Inhibitors therapeutic use, Antibodies, Monoclonal therapeutic use, Biological Therapy, Multiple Myeloma drug therapy
- Abstract
Multiple myeloma is a cancer of bone marrow plasma cells that represents approximately 10% of hematologic malignancies. Though it is typically incurable, a remarkable suite of new therapies developed over the last 25 years has enabled durable disease control in most patients. This article briefly introduces the clinical features of multiple myeloma and aspects of multiple myeloma biology that modern therapies exploit. Key current and emerging treatment modalities are then reviewed, including cereblon-modulating agents, proteasome inhibitors, monoclonal antibodies, other molecularly targeted therapies (selinexor, venetoclax), chimeric antigen receptor T cells, T cell-engaging bispecific antibodies, and antibody-drug conjugates. For each modality, mechanism of action and clinical considerations are discussed. These therapies are combined and sequenced in modern treatment pathways, discussed at the conclusion of the article, which have led to substantial improvements in outcomes for multiple myeloma patients in recent years.
- Published
- 2024
- Full Text
- View/download PDF
15. Exploiting the CD200-CD200R immune checkpoint axis in multiple myeloma to enhance CAR T-cell therapy.
- Author
-
Tang Y, Liu W, Kadu S, Johnson O, Hasanali ZS, Kelly A, Shestov A, Pajarillo R, Greenblatt E, Holmes M, Wang LP, Shih N, O'Connor RS, Ruella M, Garfall AL, Allman D, Vogl DT, Cohen A, June CH, and Sheppard NC
- Subjects
- Humans, Mice, Animals, CD28 Antigens metabolism, T-Lymphocytes, B-Cell Maturation Antigen metabolism, Neoplasm Recurrence, Local metabolism, Immunotherapy, Adoptive, Multiple Myeloma metabolism
- Abstract
Abstract: Patients with multiple myeloma (MM) treated with B-cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T cells usually relapse with BCMA+ disease, indicative of CAR T-cell suppression. CD200 is an immune checkpoint that is overexpressed on aberrant plasma cells (aPCs) in MM and is an independent negative prognostic factor for survival. However, CD200 is not present on MM cell lines, a potential limitation of current preclinical models. We engineered MM cell lines to express CD200 at levels equivalent to those found on aPCs in MM and show that these are sufficient to suppress clinical-stage CAR T-cells targeting BCMA or the Tn glycoform of mucin 1 (TnMUC1), costimulated by 4-1BB and CD2, respectively. To prevent CD200-mediated suppression of CAR T cells, we compared CRISPR-Cas9-mediated knockout of the CD200 receptor (CD200RKO), to coexpression of versions of the CD200 receptor that were nonsignaling, that is, dominant negative (CD200RDN), or that leveraged the CD200 signal to provide CD28 costimulation (CD200R-CD28 switch). We found that the CD200R-CD28 switch potently enhanced the polyfunctionality of CAR T cells, and improved cytotoxicity, proliferative capacity, CAR T-cell metabolism, and performance in a chronic antigen exposure assay. CD200RDN provided modest benefits, but surprisingly, the CD200RKO was detrimental to CAR T-cell activity, adversely affecting CAR T-cell metabolism. These patterns held up in murine xenograft models of plasmacytoma, and disseminated bone marrow predominant disease. Our findings underscore the importance of CD200-mediated immune suppression in CAR T-cell therapy of MM, and highlight a promising approach to enhance such therapies by leveraging CD200 expression on aPCs to provide costimulation via a CD200R-CD28 switch., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Immunotherapy targeting B cells and long-lived plasma cells effectively eliminates pre-existing donor-specific allo-antibodies.
- Author
-
Zhang Z, Markmann C, Yu M, Agarwal D, Rostami S, Wang W, Liu C, Zhao H, Ochoa T, Parvathaneni K, Xu X, Li E, Gonzalez V, Khadka R, Hoffmann J, Knox JJ, Scholler J, Marcellus B, Allman D, Fraietta JA, Samelson-Jones B, Milone MC, Monos D, Garfall AL, Naji A, and Bhoj VG
- Subjects
- Humans, Animals, Mice, Plasma Cells, B-Cell Maturation Antigen, T-Lymphocytes, Immunotherapy, Antibodies, Receptors, Chimeric Antigen
- Abstract
Pre-existing anti-human leukocyte antigen (HLA) allo-antibodies constitute a major barrier to transplantation. Current desensitization approaches fail due to ineffective depletion of allo-specific memory B cells (Bmems) and long-lived plasma cells (LLPCs). We evaluate the efficacy of chimeric antigen receptor (CAR) T cells targeting CD19 and B cell maturation antigen (BCMA) to eliminate allo-antibodies in a skin pre-sensitized murine model of islet allo-transplantation. We find that treatment of allo-sensitized hosts with CAR T cells targeting Bmems and LLPCs eliminates donor-specific allo-antibodies (DSAs) and mitigates hyperacute rejection of subsequent islet allografts. We then assess the clinical efficacy of the CAR T therapy for desensitization in patients with multiple myeloma (MM) with pre-existing HLA allo-antibodies who were treated with the combination of CART-BCMA and CART-19 (ClinicalTrials.gov: NCT03549442) and observe clinically meaningful allo-antibody reduction. These findings provide logical rationale for clinical evaluation of CAR T-based immunotherapy in highly sensitized candidates to promote successful transplantation., Competing Interests: Declaration of interests J.A.F. is an inventor on patents in the field of T cell therapy for cancer, from which he has received royalties. J.A.F. is also a member of the scientific advisory boards of Cartography Bio. and Shennon Biotechnologies, Inc. M.C.M. is an inventor on patents pending or granted related to CAR T cell technology that are assigned to the University of Pennsylvania and licensed to Novartis Ag, from which he receives royalties; these include patents on the CAR T cell therapies described in this study (PCT/US2011/064191, PCT/US2018/061239, PCT/US2018/063255). M.C.M. is also a founder, stockholder, and co-chair of the scientific advisory boards for Cabaletta Bio and Verismo Therapeutics. A.L.G. has received grants from Novartis, grants from National Institutes of Health, and grants from the Leukemia & Lymphoma Society while this study was conducted, grants and personal fees from Janssen, personal fees from GlaxoSmithKline, personal fees from Legend Biotech, grants from CRISPR Therapeutics, grants from Tmunity Therapeutics, personal fees from Amgen, grants from the Leukemia & Lymphoma Society, and grants from National Institutes of Health outside the submitted work; in addition, A.L.G. has a patent for US15/757,123 pending and licensed to Novartis, a patent for US16/764,459 pending, and a patent for US16/768,260 pending and stock ownership in Cabaletta Bio. V.G.B. is an inventor on patents pending or granted related to CAR T cell technology that are assigned to the University of Pennsylvania and licensed to Cabaletta Bio and Tmunity, from which he receives royalties., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Options at the time of relapse after anti-BCMA therapy.
- Author
-
Razzo B, Garfall AL, and Cohen AD
- Subjects
- Humans, B-Cell Maturation Antigen, Chronic Disease, Immunotherapy, Adoptive, Neoplasm Recurrence, Local, Prospective Studies, Retrospective Studies, Recurrence, Multiple Myeloma therapy
- Abstract
B-cell maturation antigen (BCMA)-directed therapies, including antibody-drug conjugates, bispecific antibodies (BsAbs), and chimeric antigen receptor T cells (CARTs), have shown remarkable efficacy in patients with late-line myeloma with prior exposure to immunomodulatory agents, proteasome inhibitors, and anti-CD38 antibodies. However, optimal sequencing of these agents remains to be determined, and management of these patients once they relapse has become a new unmet need. Fortunately, there are multiple options with demonstrated activity after anti-BCMA therapy, including a different BCMA-directed therapy, non-BCMA-directed CARTs and BsAbs, novel non-T-cell-engaging drugs, and standard triplet/quadruplet regimens or salvage stem cell transplant. Factors to consider when choosing a next therapy after anti-BCMA therapy include patient characteristics and preferences, prior therapies and toxicities, disease biology, timing from last anti-BCMA therapy, and, in the future, BCMA expression and immune profiling. While current data are limited to retrospective studies and small prospective cohorts, the serial use of T-cell-engaging therapies looks particularly promising, especially as BCMA-directed therapies move up earlier in the myeloma treatment course and additional CARTs and BsAbs against alternative targets (eg, G protein-coupled receptor, family C, group 5, member D and Fc receptor-homolog 5) become available. Going forward, ongoing prospective studies, large real-world data sets, and better tools to interrogate antigen expression and immune cell fitness hopefully will provide further insight into how to best individualize therapy for this difficult-to-treat population., (Copyright © 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
18. Understanding Infection Risk with Anti-BCMA Bispecific Antibodies.
- Author
-
Garfall AL and Stadtmauer EA
- Subjects
- Humans, Immunotherapy, Immunization, Passive, B-Cell Maturation Antigen, Antibodies, Bispecific therapeutic use, Multiple Myeloma
- Abstract
Summary: Lancman and colleagues find that infection risk in patients treated with anti-BCMA bispecific antibodies for relapsed/refractory multiple myeloma is associated with severe immunoglobulin deficiency and may be mitigated by immunoglobulin replacement therapy. The study has implications for managing infection risk and raises questions about the optimal duration of treatment with these potent, novel immunotherapies. See related article by Lancman et al., p. 440 (4) ., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
19. Population Pharmacokinetics and Exposure-Response with Teclistamab in Patients With Relapsed/Refractory Multiple Myeloma: Results From MajesTEC-1.
- Author
-
Miao X, Wu LS, Lin SXW, Xu Y, Chen Y, Iwaki Y, Kobos R, Stephenson T, Kemmerer K, Uhlar CM, Banerjee A, Goldberg JD, Trancucci D, Apte A, Verona R, Pei L, Desai R, Hickey K, Su Y, Ouellet D, Samtani MN, Guo Y, Garfall AL, Krishnan A, Usmani SZ, Zhou H, and Girgis S
- Subjects
- Humans, Proteasome Inhibitors, Body Weight, Multiple Myeloma drug therapy, Antineoplastic Agents, Neutropenia
- Abstract
Background: Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, is approved in patients with relapsed/refractory multiple myeloma (RRMM) who have previously received an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 antibody., Objective: We report the population pharmacokinetics of teclistamab administered intravenously and subcutaneously (SC) and exposure-response relationships from the phase I/II, first-in-human, open-label, multicenter MajesTEC-1 study., Methods: Phase I of MajesTEC-1 consisted of dose escalation and expansion at the recommended phase II dose (RP2D; 1.5 mg/kg SC weekly, preceded by step-up doses of 0.06 and 0.3 mg/kg); phase II investigated the efficacy of teclistamab RP2D in patients with RRMM. Population pharmacokinetics and the impact of covariates on teclistamab systemic exposure were assessed using a 2-compartment model with first-order absorption for SC and parallel time-independent and time-dependent elimination pathways. Exposure-response analyses were conducted, including overall response rate (ORR), duration of response (DoR), progression-free survival (PFS), overall survival (OS), and the incidence of grade ≥ 3 anemia, neutropenia, lymphopenia, leukopenia, thrombocytopenia, and infection., Results: In total, 4840 measurable serum concentration samples from 338 pharmacokinetics-evaluable patients who received teclistamab were analyzed. The typical population value of time-independent and time-dependent clearance were 0.449 L/day and 0.547 L/day, respectively. The time-dependent clearance decreased rapidly to < 10% after 8 weeks of teclistamab treatment. Patients who discontinue teclistamab after the 13th dose are expected to have a 50% reduction from C
max in teclistamab concentration at a median (5th to 95th percentile) time of 15 days (7-33 days) after Tmax and a 97% reduction from Cmax in teclistamab concentration at a median time of 69 days (32-163 days) after Tmax . Body weight, multiple myeloma type (immunoglobulin G vs non-immunoglobulin G), and International Staging System (ISS) stage (II vs I and III vs I) were statistically significant covariates on teclistamab pharmacokinetics; however, these covariates had no clinically relevant effect on the efficacy of teclistamab at the RP2D. Across all doses, ORR approached a plateau at the concentration range associated with RP2D, and in patients who received the RP2D, a flat exposure-response curve was observed. No apparent relationship was observed between DoR, PFS, OS, and the incidence of grade ≥3 adverse events across the predicted exposure quartiles., Conclusion: Body weight, myeloma type, and ISS stage impacted systemic teclistamab exposure without any clinically relevant effect on efficacy. The exposure-response analyses for ORR showed a positive trend with increasing teclistamab systemic exposure, with a plateau at the RP2D, and there was no apparent exposure-response trend for safety or other efficacy endpoints. These analyses support the RP2D of teclistamab in patients with RRMM., Clinical Trial Registration: NCT03145181 (phase I, 09 May 2017); NCT04557098 (phase II, 21 September 2020)., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
20. Survival Outcomes for Patients with Relapsed/ Refractory Aggressive B Cell Lymphomas Following Receipt of High-Dose Chemotherapy/Autologous Stem Transplantation and/or Chimeric Antigen Receptor-Modified T Cells.
- Author
-
Landsburg DJ, Nasta SD, Svoboda J, Gerson JN, Schuster SJ, Barta SK, Chong EA, Difilippo H, Weber E, Cunningham K, Catania C, Garfall AL, Stadtmauer EA, Frey NV, and Porter DL
- Subjects
- Humans, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Transplantation, Autologous methods, T-Lymphocytes, Hematopoietic Stem Cell Transplantation, Receptors, Chimeric Antigen therapeutic use, Lymphoma, Large B-Cell, Diffuse therapy
- Abstract
Patients diagnosed with relapsed/refractory (R/R) diffuse large B cell lymphoma (DLBCL) or high-grade B cell lymphoma (HGBL) may achieve prolonged survival following receipt of high-dose chemotherapy/autologous stem cell transplantation (HDC/ASCT) or CD19-directed chimeric antigen receptor modified T cell therapy (CART19). Although early results from randomized clinical trials suggest that assignment to CART19 versus salvage immunochemotherapy as second-line therapy results in improved survival, analysis of a large series of patients who actually received HDC/ASCT or CART19 has yet to be performed. Such an analysis may inform future research efforts to optimize the risk stratification of R/R DLBCL/HGBL patients who are candidates for either therapy. The aim of this study was to evaluate clinicopathologic factors predictive of freedom from treatment failure (FFTF) for R/R DLBCL/HGBL patients following receipt of HDC/ASCT or CART19, and to compare patterns of treatment failure (TF) in R/R DLBCL/HGBL patients receiving HDC/ASCT and those receiving CART19. THE STUDY GROUP COMPRISED: patients age ≤75 years with R/R DLBCL/HGBL who received HDC/ASCT demonstrating partial or complete metabolic response to salvage immunochemotherapy and/or CART19 in the standard of care setting at the University of Pennsylvania between 2013 and 2021. Survival analyses were performed from the time of infusion of either HDC/ASCT or CART19, as well as at landmark time points postinfusion for patients who achieved FFTF. For 100 HDC/ASCT patients with a median follow-up of 62.7 months, the estimated 36-month FFTF and overall survival (OS) rates were 59% and 81%, respectively. For 109 CART19 patients with a median follow-up of 37.6 months, the estimated 36-month FFTF and OS rates were 24% and 48%, respectively. HDC/ASCT patients had significantly higher rates of estimated 36-month FFTF when they achieved actual FFTF at 3, 6, 12 and 24 months. Additionally, the rates of baseline characteristics predictive of TF at 36 months for either HDC/ASCT or CART19 patients were either similar to or significantly lower for CART19 patients compared to HDC/ASCT patients who achieved actual FFTF at 3, 6, 12, and 24 months. Patients with R/R DLBCL/HGBL achieving response to salvage immunochemotherapy who received HDC/ASCT had a high rate of estimated FFTF regardless of whether they harbored features predictive of resistance to salvage immunochemotherapy, which may be more durable than that of R/R DLBCL/HGBL patients receiving CART19. These findings support further investigation of disease characteristics, such as molecular features, that may predict response to salvage immunochemotherapy in patients fit for HDC/ASCT., (Copyright © 2023 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Teclistamab versus real-world physician's choice of therapy in triple-class exposed relapsed/refractory multiple myeloma.
- Author
-
Krishnan A, Nooka AK, Chari A, Garfall AL, Martin TG, Nair S, Lin X, Qi K, Londhe A, Pei L, Ammann E, Kobos R, Smit J, Parekh T, Marshall A, Slavcev M, and Usmani SZ
- Subjects
- Humans, Treatment Outcome, Dexamethasone therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Multiple Myeloma drug therapy, Antineoplastic Agents therapeutic use, Physicians
- Abstract
Aim: We compared the effectiveness of teclistamab versus real-world physician's choice of therapy (RWPC) in triple-class exposed relapsed/refractory multiple myeloma. Materials & methods: MajesTEC-1 eligibility criteria were applied to the RWPC cohort. Baseline covariate imbalances were adjusted using inverse probability of treatment weighting. Overall survival, progression-free survival and time to next treatment were compared. Results: After inverse probability of treatment weighting, baseline characteristics were similar between cohorts (teclistamab, n = 165; RWPC, n = 364 [766 observations]). Teclistamab treated patients had numerically better overall survival (hazard ratio [HR]: 0.82 [95% CI: 0.59-1.14]; p = 0.233) and significantly greater progression-free survival (HR: 0.43 [0.33-0.56]; p < 0.0001) and time to next treatment (HR: 0.36 [0.27-0.49]; p < 0.0001) versus the RWPC cohort. Conclusion: Teclistamab offered clinical benefit over RWPC in triple-class exposed relapsed/refractory multiple myeloma.
- Published
- 2023
- Full Text
- View/download PDF
22. High SOX2 expression is associated with poor survival in patients with newly diagnosed multiple myeloma.
- Author
-
Shan X, Long Q, Garfall AL, and Susanibar-Adaniya SP
- Subjects
- Humans, Disease-Free Survival, Prognosis, SOXB1 Transcription Factors, Multiple Myeloma diagnosis, Multiple Myeloma genetics, Multiple Myeloma metabolism
- Published
- 2023
- Full Text
- View/download PDF
23. Progressive multifocal leukoencephalopathy in multiple myeloma.
- Author
-
Hoeynck BW, Cohen AD, Stadtmauer EA, Susanibar-Adaniya SP, Vogl DT, Waxman AJ, Bardsley M, Le S, LaMaestra L, and Garfall AL
- Subjects
- Humans, Central Nervous System pathology, Immunosuppression Therapy adverse effects, Leukoencephalopathy, Progressive Multifocal diagnosis, Leukoencephalopathy, Progressive Multifocal etiology, Multiple Myeloma complications, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy, JC Virus physiology
- Abstract
Progressive multifocal leukoencephalopathy (PML) is a rare and often fatal demyelinating disease of the central nervous system caused by reactivation of the JC virus in the context of immune suppression such as HIV, malignancy, and certain immunomodulatory medications. PML has been reported only rarely in multiple myeloma patients, and its presenting features and natural history in this population are not well known. We describe six cases of PML among multiple myeloma patients treated at our institution between 2013 and 2022, including two that developed on or shortly after treatment with recently developed BCMA-directed immunotherapies., (© 2022 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
24. 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
25. Changes in Bone Marrow Tumor and Immune Cells Correlate with Durability of Remissions Following BCMA CAR T Therapy in Myeloma.
- Author
-
Dhodapkar KM, Cohen AD, Kaushal A, Garfall AL, Manalo RJ, Carr AR, McCachren SS, Stadtmauer EA, Lacey SF, Melenhorst JJ, June CH, Milone MC, and Dhodapkar MV
- Subjects
- Humans, B-Cell Maturation Antigen genetics, Bone Marrow pathology, Neoplasm Recurrence, Local, T-Lymphocytes immunology, Tumor Microenvironment, Receptors, Chimeric Antigen genetics, Multiple Myeloma immunology, Bone Marrow Neoplasms
- Abstract
Chimeric antigen-receptor (CAR) T cells lead to high response rates in myeloma, but most patients experience recurrent disease. We combined several high-dimensional approaches to study tumor/immune cells in the tumor microenvironment (TME) of myeloma patients pre- and post-B-cell maturation antigen (BCMA)-specific CAR T therapy. Lower diversity of pretherapy T-cell receptor (TCR) repertoire, presence of hyperexpanded clones with exhaustion phenotype, and BAFF+PD-L1+ myeloid cells in the marrow correlated with shorter progression-free survival (PFS) following CAR T therapy. In contrast, longer PFS was associated with an increased proportion of CLEC9A+ dendritic cells (DC), CD27+TCF1+ T cells with diverse T-cell receptors, and emergence of T cells expressing marrow-residence genes. Residual tumor cells at initial response express stemlike genes, and tumor recurrence was associated with the emergence of new dominant clones. These data illustrate a dynamic interplay between endogenous T, CAR T, myeloid/DC, and tumor compartments that affects the durability of response following CAR T therapy in myeloma., Significance: There is an unmet need to identify determinants of durable responses following BCMA CAR T therapy of myeloma. High-dimensional analysis of the TME was performed to identify features of immune and tumor cells that correlate with survival and suggest several strategies to improve outcomes following CAR T therapy. See related commentary by Graham and Maus, p. 478. This article is highlighted in the In This Issue feature, p. 476., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
26. Teclistamab in Relapsed or Refractory Multiple Myeloma.
- Author
-
Moreau P, Garfall AL, van de Donk NWCJ, Nahi H, San-Miguel JF, Oriol A, Nooka AK, Martin T, Rosinol L, Chari A, Karlin L, Benboubker L, Mateos MV, Bahlis N, Popat R, Besemer B, Martínez-López J, Sidana S, Delforge M, Pei L, Trancucci D, Verona R, Girgis S, Lin SXW, Olyslager Y, Jaffe M, Uhlar C, Stephenson T, Van Rampelbergh R, Banerjee A, Goldberg JD, Kobos R, Krishnan A, and Usmani SZ
- Subjects
- Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Humans, Injections, Subcutaneous, Neoplasm Recurrence, Local drug therapy, Recurrence, T-Lymphocytes drug effects, T-Lymphocytes immunology, Antibodies, Bispecific administration & dosage, Antibodies, Bispecific adverse effects, Antibodies, Bispecific therapeutic use, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Agents, Immunological therapeutic use, B-Cell Maturation Antigen antagonists & inhibitors, CD3 Complex antagonists & inhibitors, Multiple Myeloma drug therapy, Multiple Myeloma immunology, Multiple Myeloma pathology
- Abstract
Background: Teclistamab is a T-cell-redirecting bispecific antibody that targets both CD3 expressed on the surface of T cells and B-cell maturation antigen expressed on the surface of myeloma cells. In the phase 1 dose-defining portion of the study, teclistamab showed promising efficacy in patients with relapsed or refractory multiple myeloma., Methods: In this phase 1-2 study, we enrolled patients who had relapsed or refractory myeloma after at least three therapy lines, including triple-class exposure to an immunomodulatory drug, a proteasome inhibitor, and an anti-CD38 antibody. Patients received a weekly subcutaneous injection of teclistamab (at a dose of 1.5 mg per kilogram of body weight) after receiving step-up doses of 0.06 mg and 0.3 mg per kilogram. The primary end point was the overall response (partial response or better)., Results: Among 165 patients who received teclistamab, 77.6% had triple-class refractory disease (median, five previous therapy lines). With a median follow-up of 14.1 months, the overall response rate was 63.0%, with 65 patients (39.4%) having a complete response or better. A total of 44 patients (26.7%) were found to have no minimal residual disease (MRD); the MRD-negativity rate among the patients with a complete response or better was 46%. The median duration of response was 18.4 months (95% confidence interval [CI], 14.9 to not estimable). The median duration of progression-free survival was 11.3 months (95% CI, 8.8 to 17.1). Common adverse events included cytokine release syndrome (in 72.1% of the patients; grade 3, 0.6%; no grade 4), neutropenia (in 70.9%; grade 3 or 4, 64.2%), anemia (in 52.1%; grade 3 or 4, 37.0%), and thrombocytopenia (in 40.0%; grade 3 or 4, 21.2%). Infections were frequent (in 76.4%; grade 3 or 4, 44.8%). Neurotoxic events occurred in 24 patients (14.5%), including immune effector cell-associated neurotoxicity syndrome in 5 patients (3.0%; all grade 1 or 2)., Conclusions: Teclistamab resulted in a high rate of deep and durable response in patients with triple-class-exposed relapsed or refractory multiple myeloma. Cytopenias and infections were common; toxic effects that were consistent with T-cell redirection were mostly grade 1 or 2. (Funded by Janssen Research and Development; MajesTEC-1 ClinicalTrials.gov numbers, NCT03145181 and NCT04557098.)., (Copyright © 2022 Massachusetts Medical Society.)
- Published
- 2022
- Full Text
- View/download PDF
27. Femtomolar SARS-CoV-2 Antigen Detection Using the Microbubbling Digital Assay with Smartphone Readout Enables Antigen Burden Quantitation and Tracking.
- Author
-
Chen H, Li Z, Feng S, Richard-Greenblatt M, Hutson E, Andrianus S, Glaser LJ, Rodino KG, Qian J, Jayaraman D, Collman RG, Glascock A, Bushman FD, Lee JS, Cherry S, Fausto A, Weiss SR, Koo H, Corby PM, Oceguera A, O'Doherty U, Garfall AL, Vogl DT, Stadtmauer EA, and Wang P
- Subjects
- Humans, Machine Learning, SARS-CoV-2, Sensitivity and Specificity, Antigens, Viral analysis, COVID-19 diagnosis, COVID-19 Testing methods, Smartphone
- Abstract
Background: High-sensitivity severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigen assays are desirable to mitigate false negative results. Limited data are available to quantify and track SARS-CoV-2 antigen burden in respiratory samples from different populations., Methods: We developed the Microbubbling SARS-CoV-2 Antigen Assay (MSAA) with smartphone readout, with a limit of detection of 0.5 pg/mL (10.6 fmol/L) nucleocapsid antigen or 4000 copies/mL inactivated SARS-CoV-2 virus in nasopharyngeal (NP) swabs. We developed a computer vision and machine learning-based automatic microbubble image classifier to accurately identify positives and negatives and quantified and tracked antigen dynamics in intensive care unit coronavirus disease 2019 (COVID-19) inpatients and immunocompromised COVID-19 patients., Results: Compared to qualitative reverse transcription-polymerase chain reaction methods, the MSAA demonstrated a positive percentage agreement of 97% (95% CI 92%-99%) and a negative percentage agreement of 97% (95% CI 94%-100%) in a clinical validation study with 372 residual clinical NP swabs. In immunocompetent individuals, the antigen positivity rate in swabs decreased as days-after-symptom-onset increased, despite persistent nucleic acid positivity. Antigen was detected for longer and variable periods of time in immunocompromised patients with hematologic malignancies. Total microbubble volume, a quantitative marker of antigen burden, correlated inversely with cycle threshold values and days-after-symptom-onset. Viral sequence variations were detected in patients with long duration of high antigen burden., Conclusions: The MSAA enables sensitive and specific detection of acute infections and quantification and tracking of antigen burden and may serve as a screening method in longitudinal studies to identify patients who are likely experiencing active rounds of ongoing replication and warrant close viral sequence monitoring., (© American Association for Clinical Chemistry 2021. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
28. The Safety of Bridging Radiation with Anti-BCMA CAR T-Cell Therapy for Multiple Myeloma.
- Author
-
Manjunath SH, Cohen AD, Lacey SF, Davis MM, Garfall AL, Melenhorst JJ, Maxwell R, Arscott WT, Maity A, Jones JA, Plastaras JP, Stadtmauer EA, Levine BL, June CH, Milone MC, and Paydar I
- Subjects
- B-Cell Maturation Antigen, Humans, Receptors, Chimeric Antigen, Retrospective Studies, Immunotherapy, Adoptive adverse effects, Multiple Myeloma drug therapy
- Abstract
Purpose: B-cell maturation antigen (BCMA)-targeted chimeric antigen receptor (CAR) T cells (CART-BCMA) are a promising treatment for relapsed/refractory multiple myeloma (r/rMM). We evaluated the safety and feasibility of bridging radiation (RT) in subjects treated on a phase I trial of CART-BCMA., Experimental Design: Twenty-five r/rMM subjects were treated in three cohorts with two doses of CART-BCMA cells ± cyclophosphamide. We retrospectively analyzed toxicity, response, and CART manufacturing data based on RT receipt., Results: Thirteen subjects received no RT <1 year before CART infusion (Group A). Eight subjects received RT <1 year before CART infusion (Group B) with median time from RT to apheresis of 114 days (range 40-301). Four subjects received bridging-RT (Group C) with a median dose of 22 Gy and time from RT to infusion of 25 days (range 18-35). Group C had qualitatively lower rates of grade 4 (G4) hematologic toxicities (25%) versus A (61.5%) and B (62.5%). G3-4 neurotoxicity occurred in 7.7%, 25%, and 25% in Group A, B, and C, respectively. G3-4 cytokine release syndrome was observed in 38.5%, 25%, and 25% in Group A, B, and C, respectively. Partial response or better was observed in 54%, 38%, and 50% of Group A, B, and C, respectively. RT administered <1 year ( P = 0.002) and <100 days ( P = 0.069) before apheresis was associated with lower in vitro proliferation during manufacturing; however, in vivo CART-BCMA expansion appeared similar across groups., Conclusions: Bridging-RT appeared safe and feasible with CART-BCMA therapy in our r/rMM patients, though larger future studies are needed to draw definitive conclusions., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
29. Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, in patients with relapsed or refractory multiple myeloma (MajesTEC-1): a multicentre, open-label, single-arm, phase 1 study.
- Author
-
Usmani SZ, Garfall AL, van de Donk NWCJ, Nahi H, San-Miguel JF, Oriol A, Rosinol L, Chari A, Bhutani M, Karlin L, Benboubker L, Pei L, Verona R, Girgis S, Stephenson T, Elsayed Y, Infante J, Goldberg JD, Banerjee A, Mateos MV, and Krishnan A
- Subjects
- Administration, Intravenous, Aged, Antibodies, Monoclonal, Humanized administration & dosage, B-Cell Maturation Antigen immunology, Dose-Response Relationship, Drug, Female, Humans, Injections, Subcutaneous, Male, Middle Aged, Neoplasm Recurrence, Local drug therapy, T-Lymphocytes drug effects, T-Lymphocytes immunology, Treatment Outcome, Antibodies, Bispecific pharmacology, Antineoplastic Agents, Immunological therapeutic use, B-Cell Maturation Antigen therapeutic use, Multiple Myeloma drug therapy
- Abstract
Background: There is a need for novel therapies for relapsed or refractory multiple myeloma, and B-cell maturation antigen (BCMA) is a validated target. Teclistamab is a bispecific antibody that binds BCMA and CD3 to redirect T cells to multiple myeloma cells. The aim of the MajesTEC-1 study was to evaluate the safety, tolerability, and preliminary efficacy of teclistamab in patients with relapsed or refractory multiple myeloma., Methods: This open-label, single-arm, phase 1 study enrolled patients with multiple myeloma who were relapsed, refractory, or intolerant to established therapies. Teclistamab was administered intravenously (range 0·3-19·2 μg/kg [once every 2 weeks] or 19·2-720 μg/kg [once per week]) or subcutaneously (range 80-3000 μg/kg [once per week]) in different cohorts, with step-up dosing for 38·4 μg/kg or higher doses. The primary objectives were to identify the recommended phase 2 dose (part one) and characterise teclistamab safety and tolerability at the recommended phase 2 dose (part two). Safety was assessed in all patients treated with at least one dose of teclistamab. Efficacy was analysed in response-evaluable patients (ie, patients who received at least one dose of teclistamab and had at least one post-baseline response evaluation). This ongoing trial is registered with ClinicalTrials.gov, NCT03145181., Findings: Between June 8, 2017, and March 29, 2021, 219 patients were screened for study inclusion, and 157 patients (median six previous therapy lines) were enrolled and received at least one dose of teclistamab (intravenous n=84; subcutaneous n=73). 40 patients were administered the recommended phase 2 dose, identified as once per week subcutaneous administration of teclistamab at 1500 μg/kg, after 60 μg/kg and 300 μg/kg step-up doses (median follow-up 6·1 months, IQR 3·6-8·2). There were no dose-limiting toxicities at the recommended phase 2 dose in part one. In the 40 patients treated at the recommended phase 2 dose, the most common treatment-emergent adverse events were cytokine release syndrome in 28 (70%; all grade 1 or 2 events) and neutropenia in 26 (65%) patients (grade 3 or 4 in 16 [40%]). The overall response rate in response-evaluable patients treated at the recommended phase 2 dose (n=40) was 65% (95% CI 48-79); 58% achieved a very good partial response or better. At the recommended phase 2 dose, the median duration of response was not reached. 22 (85%) of 26 responders were alive and continuing treatment after 7·1 months' median follow-up (IQR 5·1-9·1). At the recommended phase 2 dose, teclistamab exposure was maintained above target exposure levels, and consistent T-cell activation was reported., Interpretation: Teclistamab is a novel treatment approach for relapsed or refractory multiple myeloma. At the recommended phase 2 dose, teclistamab showed promising efficacy, with durable responses that deepened over time, and was well tolerated, supporting further clinical development., Funding: Janssen Research & Development., Competing Interests: Declaration of interests SZU received grant support from Bristol Myers Squibb and Pharmacyclics; grant support and consulting fees from Amgen, Celgene, Sanofi, Seattle Genetics, Janssen, Takeda, SkylineDx, Merck, and GlaxoSmithKline; and consulting fees from AbbVie, Karyopharm, and Mundipharma. ALG received grant support from Novartis and CRISPR Therapeutics; grant support and consulting fees from Janssen and Tmunity Therapeutics; consulting fees from GlaxoSmithKline and Surface Oncology; and holds a pending patent (15/757,123) with royalties paid, and pending patents (16/768,260 and 16/746,459) licensed to Novartis. NWCJvdD received grant support and advisory board fees from Bristol Myers Squibb/Celgene, Janssen, Novartis, and Amgen; advisory board fees from Takeda, Servier, Bayer, Adaptive Biotechnologies, Roche, GlaxoSmithKline, and Sanofi; and grant support from Cellectis. JFS-M received consulting and advisory board fees from Amgen, Celgene, Takeda, Bristol Myers Squibb, Merck, Sharp & Dohme, Novartis, Sanofi, Janssen, Roche, AbbVie, GlaxoSmithKline, Karyopharm Therapeutics, and Secura Bio. AO received consulting fees from Amgen, Celgene, Sanofi, Janssen, and GlaxoSmithKline. LR received consulting fees from Amgen, Celgene, Sanofi, Janssen, and Takeda. AC received grant support and consulting fees from Amgen, Janssen, Celgene, Novartis Pharmaceuticals, Seattle Genetics, Millennium/Takeda; consulting fees from Bristol Myers Squibb, Karyopharm, Sanofi, Oncopeptides, Antengene, GlaxoSmithKline, Secura Bio, Shattuck Labs; and grant support from Pharmacyclics. MB received payment for speaker bureaus from Amgen, Bristol Myers Squibb, and Takeda; served as a consultant for Sanofi Genzyme; and received grant support from Janssen, MedImmune, Takeda, Prothena, Bristol Myers Squibb/Celgene, Cerecor, and Celularity. LK received consulting fees from Janssen-Cilag, Celgene, Amgen, Takeda, and GlaxoSmithKline. LP, RV, SG, TS, YE, JI, JDG, and AB are employed by Janssen. M-VM received consulting fees from Janssen-Cilag, GlaxoSmithKline, Celgene, Amgen, Regeneron, Pfizer, AbbVie, Takeda, Roche/Genentech, and Adaptive Biotechnologies. AK received consulting fees from Janssen Oncology, GlaxoSmithKline, Celgene, Adaptive Biotechnologies, Regeneron, Pfizer, Bristol-Myers Squib, Takeda, and Sutro Biopharma. HN and LB declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. CD8 + T cells contribute to survival in patients with COVID-19 and hematologic cancer.
- Author
-
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Hwee MA, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CAG, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella LA, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer NJ, DeMichele AM, Vardhana SA, Mamtani R, and Huang AC
- Subjects
- Aged, Antibodies, Viral immunology, B-Lymphocytes immunology, COVID-19 complications, COVID-19 mortality, Cohort Studies, Female, Hematologic Neoplasms complications, Humans, Immunity, Cellular immunology, Immunity, Humoral immunology, Immunophenotyping, Logistic Models, Male, Middle Aged, Multivariate Analysis, Neoplasms complications, Proportional Hazards Models, Prospective Studies, SARS-CoV-2, Survival Rate, CD8-Positive T-Lymphocytes immunology, COVID-19 immunology, Hematologic Neoplasms immunology, Neoplasms immunology
- Abstract
Patients with cancer have high mortality from coronavirus disease 2019 (COVID-19), and the immune parameters that dictate clinical outcomes remain unknown. In a cohort of 100 patients with cancer who were hospitalized for COVID-19, patients with hematologic cancer had higher mortality relative to patients with solid cancer. In two additional cohorts, flow cytometric and serologic analyses demonstrated that patients with solid cancer and patients without cancer had a similar immune phenotype during acute COVID-19, whereas patients with hematologic cancer had impairment of B cells and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific antibody responses. Despite the impaired humoral immunity and high mortality in patients with hematologic cancer who also have COVID-19, those with a greater number of CD8 T cells had improved survival, including those treated with anti-CD20 therapy. Furthermore, 77% of patients with hematologic cancer had detectable SARS-CoV-2-specific T cell responses. Thus, CD8 T cells might influence recovery from COVID-19 when humoral immunity is deficient. These observations suggest that CD8 T cell responses to vaccination might provide protection in patients with hematologic cancer even in the setting of limited humoral responses., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
31. B-cell maturation antigen chimeric antigen receptor T-cell re-expansion in a patient with myeloma following salvage programmed cell death protein 1 inhibitor-based combination therapy.
- Author
-
Bernabei L, Tian L, Garfall AL, Melenhorst JJ, Lacey SF, Stadtmauer EA, Vogl DT, Gonzalez VE, Plesa G, Young RM, Waxman A, Levine BL, June CH, Milone MC, and Cohen AD
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized administration & dosage, Combined Modality Therapy, Dexamethasone administration & dosage, Disease-Free Survival, Female, Humans, Immune Checkpoint Inhibitors administration & dosage, Lenalidomide administration & dosage, Male, Middle Aged, Survival Rate, Thalidomide administration & dosage, Thalidomide analogs & derivatives, Antineoplastic Combined Chemotherapy Protocols administration & dosage, B-Cell Maturation Antigen blood, Immunotherapy, Adoptive, Multiple Myeloma blood, Multiple Myeloma mortality, Multiple Myeloma therapy, Neoplasm Proteins blood, Programmed Cell Death 1 Receptor blood, Receptors, Chimeric Antigen
- Published
- 2021
- Full Text
- View/download PDF
32. Femtomolar SARS-CoV-2 Antigen Detection Using the Microbubbling Digital Assay with Smartphone Readout Enables Antigen Burden Quantitation and Dynamics Tracking.
- Author
-
Chen H, Li Z, Feng S, Wang A, Richard-Greenblatt M, Hutson E, Andrianus S, Glaser LJ, Rodino KG, Qian J, Jayaraman D, Collman RG, Glascock A, Bushman FD, Lee JS, Cherry S, Fausto A, Weiss SR, Koo H, Corby PM, O'Doherty U, Garfall AL, Vogl DT, Stadtmauer EA, and Wang P
- Abstract
Background: Little is known about the dynamics of SARS-CoV-2 antigen burden in respiratory samples in different patient populations at different stages of infection. Current rapid antigen tests cannot quantitate and track antigen dynamics with high sensitivity and specificity in respiratory samples., Methods: We developed and validated an ultra-sensitive SARS-CoV-2 antigen assay with smartphone readout using the Microbubbling Digital Assay previously developed by our group, which is a platform that enables highly sensitive detection and quantitation of protein biomarkers. A computer vision-based algorithm was developed for microbubble smartphone image recognition and quantitation. A machine learning-based classifier was developed to classify the smartphone images based on detected microbubbles. Using this assay, we tracked antigen dynamics in serial swab samples from COVID patients hospitalized in ICU and immunocompromised COVID patients., Results: The limit of detection (LOD) of the Microbubbling SARS-CoV-2 Antigen Assay was 0.5 pg/mL (10.6 fM) recombinant nucleocapsid (N) antigen or 4000 copies/mL inactivated SARS-CoV-2 virus in nasopharyngeal (NP) swabs, comparable to many rRT-PCR methods. The assay had high analytical specificity towards SARS-CoV-2. Compared to EUA-approved rRT-PCR methods, the Microbubbling Antigen Assay demonstrated a positive percent agreement (PPA) of 97% (95% confidence interval (CI), 92-99%) in symptomatic individuals within 7 days of symptom onset and positive SARS-CoV-2 nucleic acid results, and a negative percent agreement (NPA) of 97% (95% CI, 94-100%) in symptomatic and asymptomatic individuals with negative nucleic acid results. Antigen positivity rate in NP swabs gradually decreased as days-after-symptom-onset increased, despite persistent nucleic acid positivity of the same samples. The computer vision and machine learning-based automatic microbubble image classifier could accurately identify positives and negatives, based on microbubble counts and sizes. Total microbubble volume, a potential marker of antigen burden, correlated inversely with Ct values and days-after-symptom-onset. Antigen was detected for longer periods of time in immunocompromised patients with hematologic malignancies, compared to immunocompetent individuals. Simultaneous detectable antigens and nucleic acids may indicate the presence of replicating viruses in patients with persistent infections., Conclusions: The Microbubbling SARS-CoV-2 Antigen Assay enables sensitive and specific detection of acute infections, and quantitation and tracking of antigen dynamics in different patient populations at various stages of infection. With smartphone compatibility and automated image processing, the assay is well-positioned to be adapted for point-of-care diagnosis and to explore the clinical implications of antigen dynamics in future studies.
- Published
- 2021
- Full Text
- View/download PDF
33. CD8 T cells compensate for impaired humoral immunity in COVID-19 patients with hematologic cancer.
- Author
-
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CAG, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella L, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer N, DeMichele AM, Vardhana SA, Mamtani R, and Huang AC
- Abstract
Cancer patients have increased morbidity and mortality from Coronavirus Disease 2019 (COVID-19), but the underlying immune mechanisms are unknown. In a cohort of 100 cancer patients hospitalized for COVID-19 at the University of Pennsylvania Health System, we found that patients with hematologic cancers had a significantly higher mortality relative to patients with solid cancers after accounting for confounders including ECOG performance status and active cancer status. We performed flow cytometric and serologic analyses of 106 cancer patients and 113 non-cancer controls from two additional cohorts at Penn and Memorial Sloan Kettering Cancer Center. Patients with solid cancers exhibited an immune phenotype similar to non-cancer patients during acute COVID-19 whereas patients with hematologic cancers had significant impairment of B cells and SARS-CoV-2-specific antibody responses. High dimensional analysis of flow cytometric data revealed 5 distinct immune phenotypes. An immune phenotype characterized by CD8 T cell depletion was associated with a high viral load and the highest mortality of 71%, among all cancer patients. In contrast, despite impaired B cell responses, patients with hematologic cancers and preserved CD8 T cells had a lower viral load and mortality. These data highlight the importance of CD8 T cells in acute COVID-19, particularly in the setting of impaired humoral immunity. Further, depletion of B cells with anti-CD20 therapy resulted in almost complete abrogation of SARS-CoV-2-specific IgG and IgM antibodies, but was not associated with increased mortality compared to other hematologic cancers, when adequate CD8 T cells were present. Finally, higher CD8 T cell counts were associated with improved overall survival in patients with hematologic cancers. Thus, CD8 T cells likely compensate for deficient humoral immunity and influence clinical recovery of COVID-19. These observations have important implications for cancer and COVID-19-directed treatments, immunosuppressive therapies, and for understanding the role of B and T cells in acute COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
34. Axicabtagene ciloleucel for CD19+ plasmablastic lymphoma.
- Author
-
Raychaudhuri R, Qualtieri J, and Garfall AL
- Subjects
- Adult, Antigens, CD19 analysis, Biological Products, Fatal Outcome, Female, Humans, Immunotherapy, Adoptive, Salvage Therapy methods, Antigens, CD19 therapeutic use, Plasmablastic Lymphoma therapy
- Published
- 2020
- Full Text
- View/download PDF
35. Trispecific antibodies offer a third way forward for anticancer immunotherapy.
- Author
-
Garfall AL and June CH
- Subjects
- Immunologic Factors, Antibodies, Immunotherapy
- Published
- 2019
- Full Text
- View/download PDF
36. 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
37. Multitargeted CAR T-cell therapy in multiple myeloma.
- Author
-
Susanibar Adaniya S and Garfall AL
- Subjects
- Humans, Receptors, Antigen, T-Cell, T-Lymphocytes, Immunotherapy, Adoptive, Multiple Myeloma
- Published
- 2019
- Full Text
- View/download PDF
38. Serial treatment of relapsed/refractory multiple myeloma with different BCMA-targeting therapies.
- Author
-
Cohen AD, Garfall AL, Dogan A, Lacey SF, Martin C, Lendvai N, Vogl DT, Spear M, and Lesokhin AM
- Subjects
- Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Combined Modality Therapy adverse effects, Combined Modality Therapy methods, Drug Resistance, Neoplasm, Female, Humans, Male, Middle Aged, Multiple Myeloma diagnosis, Recurrence, Retreatment, Treatment Outcome, Antineoplastic Agents, Immunological therapeutic use, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Molecular Targeted Therapy, Multiple Myeloma immunology, Multiple Myeloma therapy
- Published
- 2019
- Full Text
- View/download PDF
39. Prospective Study of Cardiac Events During Proteasome Inhibitor Therapy for Relapsed Multiple Myeloma.
- Author
-
Cornell RF, Ky B, Weiss BM, Dahm CN, Gupta DK, Du L, Carver JR, Cohen AD, Engelhardt BG, Garfall AL, Goodman SA, Harrell SL, Kassim AA, Jadhav T, Jagasia M, Moslehi J, O'Quinn R, Savona MR, Slosky D, Smith A, Stadtmauer EA, Vogl DT, Waxman A, and Lenihan D
- Subjects
- Adult, Aged, Aged, 80 and over, Bortezomib administration & dosage, Cardiovascular Diseases chemically induced, Cardiovascular Diseases complications, Disease-Free Survival, Electrocardiography, Female, Heart Diseases complications, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Molecular Targeted Therapy, Multiple Myeloma mortality, Natriuretic Peptide, Brain analysis, Neoplasm Recurrence, Local complications, Neoplasm Recurrence, Local drug therapy, Oligopeptides administration & dosage, Prospective Studies, Proteasome Inhibitors therapeutic use, Risk Factors, Time-to-Treatment, Treatment Outcome, Troponin I analysis, Troponin T analysis, Heart Diseases chemically induced, Multiple Myeloma complications, Multiple Myeloma drug therapy, Proteasome Inhibitors adverse effects
- Abstract
Purpose: Cardiovascular adverse events (CVAEs) can occur during proteasome inhibitor (PI) therapy. We conducted a prospective, observational, multi-institutional study to define risk factors and outcomes in patients with multiple myeloma (MM) receiving PIs., Patients and Methods: Patients with relapsed MM initiating carfilzomib- or bortezomib-based therapy underwent baseline assessments and repeated assessments at regular intervals over 6 months, including cardiac biomarkers (troponin I or T, brain natriuretic peptide [BNP], and N-terminal proBNP), ECG, and echocardiography. Monitoring occurred over 18 months for development of CVAEs., Results: Of 95 patients enrolled, 65 received carfilzomib and 30 received bortezomib, with median 25 months of follow-up. Sixty-four CVAEs occurred, with 55% grade 3 or greater in severity. CVAEs occurred in 51% of patients treated with carfilzomib and 17% of those treated with bortezomib ( P = .002). Median time to first CVAE from treatment start was 31 days, and 86% occurred within the first 3 months. Patients receiving carfilzomib-based therapy with a baseline elevated BNP level higher than 100 pg/mL or N-terminal proBNP level higher than 125 pg/mL had increased risk for CVAE (odds ratio, 10.8; P < .001). Elevated natriuretic peptides occurring mid-first cycle of treatment with carfilzomib were associated with a substantially higher risk of CVAEs (odds ratio, 36.0; P < .001). Patients who experienced a CVAE had inferior progression-free survival (log-rank P = .01) and overall survival (log-rank P < .001). PI therapy was safely resumed in 89% of patients, although 41% required chemotherapy modifications., Conclusion: CVAEs are common during PI therapy for relapsed MM, especially with carfilzomib, particularly within the first 3 months of therapy. CVAEs were associated with worse overall outcomes, but usually, discontinuation of therapy was not required. Natriuretic peptides were highly predictive of CVAEs; however, validation of this finding is necessary before uniform incorporation into the routine management of patients receiving carfilzomib.
- Published
- 2019
- Full Text
- View/download PDF
40. RUNX proteins desensitize multiple myeloma to lenalidomide via protecting IKZFs from degradation.
- Author
-
Zhou N, Gutierrez-Uzquiza A, Zheng XY, Chang R, Vogl DT, Garfall AL, Bernabei L, Saraf A, Florens L, Washburn MP, Illendula A, Bushweller JH, and Busino L
- Subjects
- Adaptor Proteins, Signal Transducing, Cell Line, Tumor, Core Binding Factor alpha Subunits antagonists & inhibitors, Core Binding Factor alpha Subunits chemistry, Humans, Peptide Hydrolases physiology, Ubiquitin-Protein Ligases, Core Binding Factor alpha Subunits physiology, Ikaros Transcription Factor metabolism, Lenalidomide therapeutic use, Multiple Myeloma drug therapy
- Abstract
Ikaros family zinc finger protein 1 and 3 (IKZF1 and IKZF3) are transcription factors that promote multiple myeloma (MM) proliferation. The immunomodulatory imide drug (IMiD) lenalidomide promotes myeloma cell death via Cereblon (CRBN)-dependent ubiquitylation and proteasome-dependent degradation of IKZF1 and IKZF3. Although IMiDs have been used as first-line drugs for MM, the overall survival of refractory MM patients remains poor and demands the identification of novel agents to potentiate the therapeutic effect of IMiDs. Using an unbiased screen based on mass spectrometry, we identified the Runt-related transcription factor 1 and 3 (RUNX1 and RUNX3) as interactors of IKZF1 and IKZF3. Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation, and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Inhibition of RUNXs, via genetic ablation or a small molecule (AI-10-104), results in sensitization of myeloma cell lines and primary tumors to lenalidomide. Thus, RUNX inhibition represents a valuable therapeutic opportunity to potentiate IMiDs therapy for the treatment of multiple myeloma.
- Published
- 2019
- Full Text
- View/download PDF
41. Chimeric antigen receptor T cell immunotherapy for multiple myeloma: A review of current data and potential clinical applications.
- Author
-
Susanibar Adaniya SP, Cohen AD, and Garfall AL
- Subjects
- Combined Modality Therapy, Humans, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive trends, Immunotherapy, Adoptive methods, Multiple Myeloma therapy, Receptors, Chimeric Antigen therapeutic use
- Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder that remains incurable for most patients despite significant improvements achieved with modern therapy. Tumor evasion is a key process in the pathogenesis of MM and a compromised immune system is associated with more aggressive forms of the disease. In contrast, the emergence of myeloma-specific immune responses after both autologous and allogeneic stem cell transplantation is associated with better prognosis. Adoptive T cell therapies may improve specific anti-myeloma immunity resulting in long-lasting remissions. CAR T cell therapies for MM are at an early stage of clinical development. To date, anti-BCMA CAR T cells have shown the greatest results in early-phase clinical trials. Toxicities have included cytokine release syndrome (CRS) and neurotoxicity. Current areas of research in CAR T cell therapies include the use of gene-editing to enhance their effectiveness and safety, the integration of CAR T cells with other therapies (immunomodulatory drugs, checkpoint inhibitors) and CAR T cells to target multiple antigens., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
42. 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
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
- Published
- 2019
- Full Text
- View/download PDF
44. Acute Liver Failure Associated With Pomalidomide Therapy for Multiple Myeloma.
- Author
-
Reed-Guy L, Hoteit MA, and Garfall AL
- Subjects
- Chemical and Drug Induced Liver Injury diagnosis, Chemical and Drug Induced Liver Injury therapy, Humans, Liver Failure, Acute diagnosis, Liver Failure, Acute therapy, Liver Function Tests, Male, Middle Aged, Multiple Myeloma diagnosis, Multiple Myeloma immunology, Recovery of Function, Thalidomide adverse effects, Treatment Outcome, Antineoplastic Agents adverse effects, Chemical and Drug Induced Liver Injury etiology, Liver Failure, Acute chemically induced, Multiple Myeloma drug therapy, Thalidomide analogs & derivatives
- Published
- 2018
- Full Text
- View/download PDF
45. 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
46. Cellular and vaccine immunotherapy for multiple myeloma.
- Author
-
Garfall AL and Stadtmauer EA
- Subjects
- Animals, Humans, Receptors, Antigen, T-Cell immunology, Adoptive Transfer, Cancer Vaccines immunology, Cancer Vaccines therapeutic use, Cell- and Tissue-Based Therapy, Immunologic Factors therapeutic use, Multiple Myeloma immunology, Multiple Myeloma therapy
- Abstract
Allogeneic hematopoietic cell transplantation and donor lymphocyte infusion for multiple myeloma (MM) can induce graft-versus-myeloma immunity and long-term survivorship, but limited efficacy and associated toxicities have prevented its widespread use. Cellular immunotherapies and vaccines seek to induce more specific, reliable, and potent antimyeloma immune responses with less treatment-related risk than is possible with allogeneic transplantation. Advances in molecular biology, and basic and applied immunology, have led to promising approaches such as genetically engineered T cells with chimeric antigen receptors and T-cell receptors targeting myeloma-specific epitopes, vaccine primed ex vivo expanded autologous T cells, expanded marrow-infiltrating lymphocytes, and plasma cell/dendritic cell fusion vaccines. The addition of these emerging therapies to immunomodulatory drugs and inhibitors of programmed death-1 T-cell regulatory pathways are poised to improve outcome for our patients with myeloma., (© 2016 by The American Society of Hematology. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. Origin and Role of a Subset of Tumor-Associated Neutrophils with Antigen-Presenting Cell Features in Early-Stage Human Lung Cancer.
- Author
-
Singhal S, Bhojnagarwala PS, O'Brien S, Moon EK, Garfall AL, Rao AS, Quatromoni JG, Stephen TL, Litzky L, Deshpande C, Feldman MD, Hancock WW, Conejo-Garcia JR, Albelda SM, and Eruslanov EB
- Subjects
- Gene Expression Regulation, Neoplastic, Granulocyte-Macrophage Colony-Stimulating Factor immunology, Humans, Ikaros Transcription Factor metabolism, Interferon-gamma immunology, Neoplasm Staging, Neutrophils cytology, T-Lymphocytes, Cytotoxic immunology, Antigen-Presenting Cells immunology, Lung Neoplasms immunology, Neutrophils immunology
- Abstract
Based on studies in mouse tumor models, granulocytes appear to play a tumor-promoting role. However, there are limited data about the phenotype and function of tumor-associated neutrophils (TANs) in humans. Here, we identify a subset of TANs that exhibited characteristics of both neutrophils and antigen-presenting cells (APCs) in early-stage human lung cancer. These APC-like "hybrid neutrophils," which originate from CD11b(+)CD15(hi)CD10(-)CD16(low) immature progenitors, are able to cross-present antigens, as well as trigger and augment anti-tumor T cell responses. Interferon-γ and granulocyte-macrophage colony-stimulating factor are requisite factors in the tumor that, working through the Ikaros transcription factor, synergistically exert their APC-promoting effects on the progenitors. Overall, these data demonstrate the existence of a specialized TAN subset with anti-tumor capabilities in human cancer., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
48. Combined deletion of cathepsin protease family members reveals compensatory mechanisms in cancer.
- Author
-
Akkari L, Gocheva V, Quick ML, Kester JC, Spencer AK, Garfall AL, Bowman RL, and Joyce JA
- Subjects
- Animals, Apoptosis genetics, Carcinogenesis genetics, Carcinoma, Neuroendocrine genetics, Carcinoma, Neuroendocrine physiopathology, Disease Models, Animal, Gene Expression Regulation, Neoplastic, Gene Knockout Techniques, Macrophages enzymology, Mice, Mice, Inbred C57BL, Neoplasm Invasiveness genetics, Neovascularization, Pathologic enzymology, Neovascularization, Pathologic genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms physiopathology, Carcinoma, Neuroendocrine enzymology, Cathepsins genetics, Cathepsins metabolism, Gene Deletion, Pancreatic Neoplasms enzymology
- Abstract
Proteases are important for regulating multiple tumorigenic processes, including angiogenesis, tumor growth, and invasion. Elevated protease expression is associated with poor patient prognosis across numerous tumor types. Several multigene protease families have been implicated in cancer, including cysteine cathepsins. However, whether individual family members have unique roles or are functionally redundant remains poorly understood. Here we demonstrate stage-dependent effects of simultaneously deleting cathepsin B (CtsB) and CtsS in a murine pancreatic neuroendocrine tumor model. Early in tumorigenesis, the double knockout results in an additive reduction in angiogenic switching, whereas at late stages, several tumorigenic phenotypes are unexpectedly restored to wild-type levels. We identified CtsZ, which is predominantly supplied by tumor-associated macrophages, as the compensatory protease that regulates the acquired tumor-promoting functions of lesions deficient in both CtsB and CtsS. Thus, deletion of multiple cathepsins can lead to stage-dependent, compensatory mechanisms in the tumor microenvironment, which has potential implications for the clinical consideration of selective versus pan-family cathepsin inhibitors in cancer., (© 2016 Akkari et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2016
- Full Text
- View/download PDF
49. Chimeric Antigen Receptor T Cells in Myeloma.
- Author
-
Garfall AL, Stadtmauer EA, and June CH
- Subjects
- Female, Humans, Antigens, CD19 metabolism, Multiple Myeloma drug therapy, Receptors, Antigen, T-Cell therapeutic use
- Published
- 2016
- Full Text
- View/download PDF
50. The Promise of Chimeric Antigen Receptor Engineered T Cells in the Treatment of Hematologic Malignancies.
- Author
-
Nagle SJ, Garfall AL, and Stadtmauer EA
- Subjects
- Animals, Antigens, Neoplasm immunology, Cell- and Tissue-Based Therapy, Genetic Therapy, Hematologic Neoplasms genetics, Humans, Treatment Outcome, Hematologic Neoplasms immunology, Hematologic Neoplasms therapy, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Receptors, Antigen, T-Cell genetics, Recombinant Fusion Proteins genetics, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Relapsed and refractory hematologic malignancies have a very poor prognosis. Chimeric antigen receptor T cells are emerging as a powerful therapy in this setting. Early clinical trials of genetically modified T cells for the treatment of non-Hodgkin lymphoma, chronic lymphocytic leukemia, and acute lymphoblastic leukemia have shown high complete response rates in patients with few therapeutic options. Exploration is ongoing for other hematologic malignancies including multiple myeloma, acute myeloid leukemia, and Hodgkin lymphoma (HL). At the same time, the design and production of chimeric antigen receptor T cells are being advanced so that this therapy can be more widely utilized. Cytokine release syndrome and neurotoxicity are common, but they are treatable and fully reversible. This review will review available data as well as future developments and challenges in the field.
- 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.