37 results on '"Alexander Lesokhin"'
Search Results
2. Immunophenotypic correlates of sustained MRD negativity in patients with multiple myeloma
- Author
-
David G. Coffey, Francesco Maura, Edgar Gonzalez-Kozlova, J. Javier Diaz-Mejia, Ping Luo, Yong Zhang, Yuexin Xu, Edus H. Warren, Travis Dawson, Brian Lee, Hui Xie, Eric Smith, Amanda Ciardiello, Hearn J. Cho, Adeeb Rahman, Seunghee Kim-Schulze, Benjamin Diamond, Alexander Lesokhin, Dickran Kazandjian, Trevor J. Pugh, Damian J. Green, Sacha Gnjatic, and Ola Landgren
- Subjects
Science - Abstract
Abstract The role of the immune microenvironment in maintaining disease remission in patients with multiple myeloma (MM) is not well understood. In this study, we comprehensively profile the immune system in patients with newly diagnosed MM receiving continuous lenalidomide maintenance therapy with the aim of discovering correlates of long-term treatment response. Leveraging single-cell RNA sequencing and T cell receptor β sequencing of the peripheral blood and CyTOF mass cytometry of the bone marrow, we longitudinally characterize the immune landscape in 23 patients before and one year after lenalidomide exposure. We compare patients achieving sustained minimal residual disease (MRD) negativity to patients who never achieved or were unable to maintain MRD negativity. We observe that the composition of the immune microenvironment in both the blood and the marrow varied substantially according to both MRD negative status and history of autologous stem cell transplant, supporting the hypothesis that the immune microenvironment influences the depth and duration of treatment response.
- Published
- 2023
- Full Text
- View/download PDF
3. Bortezomib, lenalidomide and dexamethasone (VRd) vs carfilzomib, lenalidomide and dexamethasone (KRd) as induction therapy in newly diagnosed multiple myeloma
- Author
-
Carlyn Rose Tan, Andriy Derkach, David Nemirovsky, Amanda Ciardiello, Benjamin Diamond, Malin Hultcrantz, Hani Hassoun, Sham Mailankody, Urvi Shah, Kylee Maclachlan, Dhwani Patel, Oscar B. Lahoud, Heather J. Landau, David J. Chung, Gunjan L. Shah, Michael Scordo, Sergio A. Giralt, Alexander Lesokhin, Saad Z. Usmani, Ola Landgren, and Neha Korde
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Lenalidomide and dexamethasone with bortezomib (VRd) or carfilzomib (KRd) are commonly used induction regimens in the U.S. This single-center, retrospective study evaluated outcomes and safety of VRd and KRd. Primary endpoint was progression-free survival (PFS). Of 389 patients with newly diagnosed multiple myeloma, 198 received VRd and 191 received KRd. Median PFS was not reached (NR) in both groups; 5-year PFS was 56% (95%CI, 48–64%) for VRd and 67% (60–75%) for KRd (P = 0.027). Estimated 5-year EFS was 34% (95%CI, 27–42%) for VRd and 52% (45–60%) for KRd (P
- Published
- 2023
- Full Text
- View/download PDF
4. Isatuximab in combination with cemiplimab in patients with relapsed/refractory multiple myeloma: A phase 1/2 study
- Author
-
Alexander Lesokhin, Richard LeBlanc, Meletios A. Dimopoulos, Marcelo Capra, Carmelo Carlo‐Stella, Lionel Karlin, Jean‐Francois Castilloux, Peter Forsberg, Gurdeep Parmar, Axel Tosikyan, Ludek Pour, Vincent Ribrag, Rossella Ribolla, Al‐Ola Abdallah, Nadia Le Roux, Liyan Dong, Helgi van deVelde, Laurent Mayrargue, Lucie Lépine, Sandrine Macé, and Philippe Moreau
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Given the incurable nature of multiple myeloma (MM), efforts are made to improve the efficacy of anti‐CD38 monoclonal antibodies via combinations with other potentially synergistic therapies. This Phase 1/2 trial (NCT03194867) was designed to determine whether cemiplimab (anti‐PD‐1) enhances the anti‐myeloma activity of isatuximab (anti‐CD38) in patients with relapsed and refractory multiple myeloma (RRMM), to confirm the feasibility of the combination, determine its efficacy, and further evaluate its safety. Methods Patients received isatuximab 10 mg/kg once weekly for 4 weeks followed by every 2 weeks (Isa), or isatuximab 10 mg/kg plus cemiplimab 250 mg every 2 (Isa + CemiQ2W) or every 4 weeks (Isa + CemiQ4W). Results Overall, 106 patients with RRMM treated with a median of 4 prior lines were included; 25.5% had high‐risk cytogenetics, 63.2% were refractory to proteasome inhibitors and immunomodulatory agents, 26.4% were previously exposed to daratumumab, and 84.0% were refractory to their last treatment line. There were no major changes in the safety or pharmacokinetic profile of isatuximab with the addition of cemiplimab. As assessed by investigators, four patients (11.8%) in the Isa arm, nine patients (25.0%) in the Isa + CemiQ2W arm, and eight patients (22.2%) in the Isa + CemiQ4W arm were responders. Though response rates were numerically higher in cemiplimab‐containing arms, differences were not statistically significant and did not translate to improved progression‐free or overall survival after a median follow‐up of 9.99 months. Conclusion Our results suggest a marginal benefit by adding cemiplimab to isatuximab, despite demonstration of target engagement, without additional observed safety issues.
- Published
- 2023
- Full Text
- View/download PDF
5. S196: ELRANATAMAB, A B-CELL MATURATION ANTIGEN (BCMA)-CD3 BISPECIFIC ANTIBODY, FOR PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA: EXTENDED FOLLOW UP AND BIWEEKLY ADMINISTRATION FROM MAGNETISMM-3
- Author
-
Mohamad Mohty, Michael H. Tomasson, Bertrand Arnulf, Nizar J Bahlis, Paula Rodríguez-Otero, Joaquín Martinez-Lopez, Cyrille Touzeau, Hang Quach, Julien Depaus, Hisayuki Yokoyama, Salomon Manier, Noopur Raje, Marc S. Raab, Emma Searle, Eric Leip, Sharon T. Sullivan, Akos Czibere, Andrea Viqueira, and Alexander Lesokhin
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
6. S202: CARTITUDE-1 FINAL RESULTS: PHASE 1B/2 STUDY OF CILTACABTAGENE AUTOLEUCEL IN HEAVILY PRETREATED PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA
- Author
-
Nikhil Munshi, Tom Martin, Saad Z Usmani, Jesus Berdeja, Andrzej Jakubowiak, Mounzer Agha, Adam D Cohen, Abhinav Deol, Myo Htut, Alexander Lesokhin, Yi Lin, Elizabeth O’donnell, Carolyn C Jackson, Tzu-Min Yeh, Arnob Banerjee, Enrique Zudaire, Deepu Madduri, Christopher Delcorral, Lida Pacaud, and Sundar Jagannath
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
7. P880: EFFICACY AND SAFETY OF ELRANATAMAB BY AGE AND FRAILTY IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM): A SUBGROUP ANALYSIS FROM MAGNETISMM-3
- Author
-
Xavier Leleu, Noopur Raje, Alexander Lesokhin, Mohamad Mohty, Ajay Nooka, Eric Leip, Sharon T. Sullivan, Andrea Viqueira, and Salomon Manier
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
8. P870: EFFICACY AND SAFETY OF ELRANATAMAB IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA AND PRIOR B-CELL MATURATION ANTIGEN (BCMA)-DIRECTED THERAPIES: A POOLED ANALYSIS FROM MAGNETISMM STUDIES
- Author
-
Salomon Manier, Alexander Lesokhin, Mohamad Mohty, Ruben Niesvizky, Christopher Maisel, Bertrand Arnulf, Sarah M. Larson, Asya Nina Varshavsky-Yanovsky, Xavier Leleu, Lionel Karlin, David H. Vesole, Nizar J Bahlis, Carlos Fernandez de Larrea, Noopur Raje, Eric Leip, Sharon T. Sullivan, Mohamed Elmeliegy, Andrea Viqueira, and Ajay Nooka
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
9. P930: ELRANATAMAB MONOTHERAPY OR IN COMBINATION WITH DARATUMUMAB VS DARATUMUMAB + POMALIDOMIDE + DEXAMETHASONE FOR PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA: PHASE 3 MAGNETISMM-5 STUDY, PART 2
- Author
-
Xavier Leleu, Shinsuke Iida, C. Ola Landgren, Alexander Lesokhin, Eric Leip, Arthur Kudla, Gregory Finn, and Lugui Qiu
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
10. P962: COMMERCIAL TECLISTAMAB IN ANTI-BCMA THERAPY EXPOSED RELAPSED REFRACTORY MULTIPLE MYELOMA PATIENTS: THE MSKCC EXPERIENCE.
- Author
-
Ross Firestone, Tala Shekarkhand, Dhwani Patel, Malin Hultcrantz, Alexander Lesokhin, Sham Mailankody, Hani Hassoun, Carlyn Tan, Urvi Shah, Neha Korde, Kylee Maclachlan, Heather Landau, David Chung, Gunjan Shah, Sergio Giralt, Oscar Lahoud, and Saad Usmani
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
11. Whole-genome sequencing reveals progressive versus stable myeloma precursor conditions as two distinct entities
- Author
-
Bénedith Oben, Guy Froyen, Kylee H. Maclachlan, Daniel Leongamornlert, Federico Abascal, Binbin Zheng-Lin, Venkata Yellapantula, Andriy Derkach, Ellen Geerdens, Benjamin T. Diamond, Ingrid Arijs, Brigitte Maes, Kimberly Vanhees, Malin Hultcrantz, Elisabet E. Manasanch, Dickran Kazandjian, Alexander Lesokhin, Ahmet Dogan, Yanming Zhang, Aneta Mikulasova, Brian Walker, Gareth Morgan, Peter J. Campbell, Ola Landgren, Jean-Luc Rummens, Niccolò Bolli, and Francesco Maura
- Subjects
Science - Abstract
The factors that are associated with myeloma precursor condition progression are not well understood. Here the authors find that monoclonal gammopathies of undetermined significance and smoldering myelomas that did not progress to multiple myelomas have a distinct genomic profile and emerge later in the patient’s life.
- Published
- 2021
- Full Text
- View/download PDF
12. Diabetes mellitus and risk of plasma cell and lymphoproliferative disorders in 94,579 cases and 368,348 matched controls
- Author
-
Urvi A. Shah, Sæmundur Rögnvaldsson, Andriy Derkach, Magnus Björkholm, Ingemar Turesson, Yael David, Malin Hultcrantz, Carlyn Tan, Hani Hassoun, Neha Korde, Alexander Lesokhin, Sham Mailankody, Sigurður Yngvi Kristinsson, and C. Ola Landgren
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2021
- Full Text
- View/download PDF
13. First-in-human phase I/Ib open-label dose-escalation study of GWN323 (anti-GITR) as a single agent and in combination with spartalizumab (anti-PD-1) in patients with advanced solid tumors and lymphomas
- Author
-
Ravit Geva, Jennifer Mataraza, Osama Rahma, Jason John Luke, Philippe L Bedard, Sarina A Piha-Paul, Angad Singh, Tira J Tan, Darren WT Lim, Cinta Hierro, Toshikiko Doi, Alexander Lesokhin, Javier Otero, Lisa Nardi, Alexandros Xyrafas, and Xinhui Chen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background GWN323 is an IgG1 monoclonal antibody (mAb) against the glucocorticoid-induced tumor necrosis factor receptor-related protein. This first-in-human, open-label phase I/Ib study aimed to investigate the safety and tolerability and to identify the recommended doses of GWN323 with/without spartalizumab, an anti-programmed cell death receptor-1 agent, for future studies. Pharmacokinetics, preliminary efficacy and efficacy biomarkers were also assessed.Methods Patients (aged ≥18 years) with advanced/metastatic solid tumors with Eastern Cooperative Oncology Group performance status of ≤2 were included. GWN323 (10–1500 mg) or GWN323+spartalizumab (GWN323 10–750 mg+spartalizumab 100–300 mg) were administered intravenously at various dose levels and schedules during the dose-escalation phase. Dose-limiting toxicities (DLTs) were assessed during the first 21 days in a single-agent arm and 42 days in a combination arm. Adverse events (AEs) were graded per National Cancer Institute-Common Toxicity Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors V.1.1.Results Overall, 92 patients (single-agent, n=39; combination, n=53) were included. The maximum administered doses (MADs) in the single-agent and combination arms were GWN323 1500 mg every 3 weeks (q3w) and GWN323 750 mg+spartalizumab 300 mg q3w, respectively. No DLTs were observed with single-agent treatment. Three DLTs (6%, all grade ≥3) were noted with combination treatment: blood creatine phosphokinase increase, respiratory failure and small intestinal obstruction. Serious AEs were reported in 30.8% and 34.0%, and drug-related AEs were reported in 82.1% and 77.4% of patients with single-agent and combination treatments, respectively. Disease was stable in 7 patients and progressed in 26 patients with single-agent treatment. In combination arm patients, 1 had complete response (endometrial cancer); 3, partial response (rectal cancer, adenocarcinoma of colon and melanoma); 14, stable disease; and 27, disease progression. GWN323 exhibited a pharmacokinetic profile typical of mAbs with a dose-dependent increase in the pharmacokinetic exposure. Inconsistent decreases in regulatory T cells and increases in CD8+ T cells were observed in the combination arm. Gene expression analyses showed no significant effect of GWN323 on interferon-γ or natural killer-cell signatures.Conclusions GWN323, as a single agent and in combination, was well tolerated in patients with relapsed/refractory solid tumors. The MAD was 1500 mg q3w for single-agent and GWN323 750 mg+spartalizumab 300 mg q3w for combination treatments. Minimal single-agent activity and modest clinical benefit were observed with the spartalizumab combination.Trial registration number NCT02740270.
- Published
- 2021
- Full Text
- View/download PDF
14. Ciltacabtagene Autoleucel, an Anti–B-cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE-1 2-Year Follow-Up
- Author
-
Thomas Martin, Saad Z. Usmani, Jesus G. Berdeja, Mounzer Agha, Adam D. Cohen, Parameswaran Hari, David Avigan, Abhinav Deol, Myo Htut, Alexander Lesokhin, Nikhil C. Munshi, Elizabeth O'Donnell, A. Keith Stewart, Jordan M. Schecter, Jenna D. Goldberg, Carolyn C. Jackson, Tzu-Min Yeh, Arnob Banerjee, Alicia Allred, Enrique Zudaire, William Deraedt, Yunsi Olyslager, Changwei Zhou, Lida Pacaud, Deepu Madduri, Andrzej Jakubowiak, Yi Lin, and Sundar Jagannath
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE CARTITUDE-1, a phase Ib/II study evaluating the safety and efficacy of ciltacabtagene autoleucel (cilta-cel) in heavily pretreated patients with relapsed/refractory multiple myeloma, yielded early, deep, and durable responses at 12 months. Here, we present updated results 2 years after last patient in (median follow-up [MFU] approximately 28 months), including analyses of high-risk patient subgroups. METHODS Eligible patients had relapsed/refractory multiple myeloma, had received ≥ 3 prior lines of therapy or were double refractory to a proteasome inhibitor and immunomodulatory drug and had received prior proteasome inhibitor, immunomodulatory drug, and anti-CD38 therapy. Patients received a single cilta-cel infusion 5-7 days after lymphodepletion. Responses were assessed by an independent review committee. RESULTS At a MFU of 27.7 months (N = 97), the overall response rate was 97.9% (95% CI, 92.7 to 99.7); 82.5% (95% CI, 73.4 to 89.4) of patients achieved a stringent complete response. Median duration of response was not estimable. Median progression-free survival (PFS) and overall survival (OS) were not reached; 27-month PFS and OS rates were 54.9% (95% CI, 44.0 to 64.6) and 70.4% (95% CI, 60.1 to 78.6), respectively. Overall response rates were high across all subgroups (95.1%-100%). Duration of response, PFS, and/or OS were shorter in patients with high-risk cytogenetics, International Staging System stage III, high tumor burden, or plasmacytomas. The safety profile was manageable with no new cilta-cel–related cytokine release syndrome and one new case of parkinsonism (day 914 after cilta-cel) since the last report. CONCLUSION At approximately 28 months MFU, patients treated with cilta-cel maintained deep and durable responses, observed in both standard and high-risk subgroups. The risk/benefit profile of cilta-cel remained favorable with longer follow-up.
- Published
- 2023
15. Interventions and outcomes of patients with multiple myeloma receiving salvage therapy after BCMA-directed CAR T therapy
- Author
-
Oliver Van Oekelen, Karthik Nath, Tarek H. Mouhieddine, Tasmin Farzana, Adolfo Aleman, David T. Melnekoff, Yogita Ghodke-Puranik, Gunjan L. Shah, Alexander Lesokhin, Sergio Giralt, Santiago Thibaud, Adriana Rossi, Cesar Rodriguez, Larysa Sanchez, Joshua Richter, Shambavi Richard, Hearn J. Cho, Ajai Chari, Saad Z. Usmani, Sundar Jagannath, Urvi A. Shah, Sham Mailankody, and Samir Parekh
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
B-cell maturation antigen (BCMA)–directed chimeric antigen receptor T-cell (CAR T) therapy has demonstrated remarkable efficacy in patients with relapsed/refractory multiple myeloma, and now there are two US Food and Drug Administration–approved BCMA-directed CAR T products. However, despite high initial response rates, most patients eventually relapse. The outcomes of patients with disease recurrence after BCMA-directed CAR T have not been comprehensively studied, and such an analysis would help define optimal treatment strategies. We analyzed the salvage treatments and outcomes of 79 patients with multiple myeloma from two academic institutions, who had progression of disease after treatment with BCMA-directed CAR T. A total of 237 post–CAR T salvage treatment lines were used, and patients received a median of 2 (range, 1-10) treatment lines. The median overall survival from the date of relapse post-CAR T therapy was 17.9 months (95% confidence interval [CI], 14.0 non-estimable). The overall response rate to the first salvage regimen was 43.4%, with a median progression-free survival of 3.5 months (CI, 2.5-4.6). Thirty-five patients (44.3%) received a T-cell–engaging therapy (bispecific antibody or subsequent CAR T) as salvage treatment. The overall survival in patients who received subsequent T-cell–engaging therapy was not reached after a median follow up of 21.3 months. Patients with multiple myeloma who relapse after BCMA-directed CAR T have a limited prognosis but can be potentially treated with multiple lines of salvage therapy. T-cell–engaging therapies appear to maintain pronounced clinical activity in this setting.
- Published
- 2023
16. GPRC5D-Targeted CAR T Cells for Myeloma
- Author
-
Sham Mailankody, Sean M. Devlin, Jonathan Landa, Karthik Nath, Claudia Diamonte, Elizabeth J. Carstens, Douglas Russo, Romany Auclair, Lisa Fitzgerald, Briana Cadzin, Xiuyan Wang, Devanjan Sikder, Brigitte Senechal, Vladimir P. Bermudez, Terence J. Purdon, Kinga Hosszu, Devin P. McAvoy, Tasmin Farzana, Elena Mead, Jessica A. Wilcox, Bianca D. Santomasso, Gunjan L. Shah, Urvi A. Shah, Neha Korde, Alexander Lesokhin, Carlyn R. Tan, Malin Hultcrantz, Hani Hassoun, Mikhail Roshal, Filiz Sen, Ahmet Dogan, Ola Landgren, Sergio A. Giralt, Jae H. Park, Saad Z. Usmani, Isabelle Rivière, Renier J. Brentjens, and Eric L. Smith
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Humans ,General Medicine ,B-Cell Maturation Antigen ,Neoplasm Recurrence, Local ,Cytokine Release Syndrome ,Multiple Myeloma ,Immunotherapy, Adoptive ,Article ,Receptors, G-Protein-Coupled - Abstract
BACKGROUND: B-cell maturation antigen (BCMA)–directed chimeric antigen receptor (CAR) T-cell therapies have generated responses in patients with advanced myeloma, but relapses are common. G protein–coupled receptor, class C, group 5, member D (GPRC5D) has been identified as an immunotherapeutic target in multiple myeloma. Preclinical studies have shown the efficacy of GPRC5D-targeted CAR T cells, including activity in a BCMA antigen escape model. METHODS: In this phase 1 dose-escalation study, we administered a GPRC5D-targeted CAR T-cell therapy (MCARH109) at four dose levels to patients with heavily pretreated multiple myeloma, including patients with relapse after BCMA CAR T-cell therapy. RESULTS: A total of 17 patients were enrolled and received MCARH109 therapy. The maximum tolerated dose was identified at 150×10(6) CAR T cells. At the 450×10(6) CAR T-cell dose, 1 patient had grade 4 cytokine release syndrome and immune effector cell–associated neurotoxicity syndrome (ICANS), and 2 patients had a grade 3 cerebellar disorder of unclear cause. No cerebellar disorder, ICANS of any grade, or cytokine release syndrome of grade 3 or higher occurred in the 12 patients who received doses of 25×10(6) to 150×10(6) cells. A response was reported in 71% of the patients in the entire cohort and in 58% of those who received doses of 25×10(6) to 150×10(6) cells. The patients who had a response included those who had received previous BCMA therapies; responses were observed in 7 of 10 such patients in the entire cohort and in 3 of 6 such patients who received 25×10(6) to 150×10(6) cells. CONCLUSIONS: The results of this study of a GPRC5D-targeted CAR T-cell therapy (MCARH109) confirm that GPRC5D is an active immunotherapeutic target in multiple myeloma. (Funded by Juno Therapeutics/Bristol Myers Squibb; ClinicalTrials.gov number, NCT04555551.)
- Published
- 2022
17. Maintenance Therapy Cessation for Sustained MRD Negative Multiple Myeloma Patients
- Author
-
Neha Korde, Benjamin Diamond, Miranda Burge, Hani Hassoun, Heather Landau, Malin Hultcrantz, Sham Mailankody, Kylee H Maclachlan, Urvi A Shah, Andriy Derkach, Saad Usmani, Ola Landgren, and Alexander Lesokhin
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
18. MM-181 CARTITUDE-1: Two-Year Post Last Patient in (LPI) Results From the Phase 1b/2 Study of Ciltacabtagene Autoleucel (Cilta-Cel), a B-Cell Maturation Antigen (BCMA)-Directed Chimeric Antigen Receptor T (CAR-T) Cell Therapy, in Patients With Relapsed/Refractory Multiple Myeloma (RRMM)
- Author
-
Saad Z Usmani, Thomas Martin, Jesus G Berdeja, Andrzej Jakubowiak, Mounzer Agha, Adam D Cohen, Abhinav Deol, Myo Htut, Alexander Lesokhin, Nikhil C Munshi, Elizabeth O'Donnell, Carolyn C Jackson, Tzu-min Yeh, Arnob Banerjee, Enrique Zudaire, Deepu Madduri, Changwei Zhou, Lida Pacaud, Yi Lin, and Sundar Jagannath
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
19. Chemotherapy Signatures Map Evolution of Therapy-Related Myeloid Neoplasms
- Author
-
Benjamin Diamond, Bachisio Ziccheddu, Kylee Maclachlan, Justin Taylor, Eileen Boyle, Juan Arrango Ossa, Jacob Jahn, Maurizio Affer, Tulasigeri M. Totiger, David Coffey, Justin Watts, Sydney X Lu, Niccolò Bolli, Kelly Bolton, Jae H. Park, Heather Landau, Karuna Ganesh, Andrew McPherson, Mikkael A. Sekeres, Alexander Lesokhin, David Chung, Yanming Zhang, Caleb Ho, Mikhail Roshal, Jeffrey Tyner, Stephen Nimer, Elli Papaemmanuil, Saad Usmani, Gareth Morgan, Ola Landgren, and Francesco Maura
- Abstract
Patients treated with cytotoxic therapies, including autologous stem cell transplantation, are at risk for developing therapy-related myeloid neoplasms1, 2. Pre-leukemic clones (i.e., clonal hematopoiesis) are detectable years before the development of these aggressive malignancies3-5, though the genomic events leading to transformation and expansion are not well-defined. Here, leveraging distinctive chemotherapy-associated mutational signatures6-12 from whole-genome sequencing data and targeted sequencing of pre-chemotherapy samples, we reconstruct the evolutionary life-history of 39 therapy-related myeloid malignancies. A dichotomy is revealed, in which neoplasms with evidence of chemotherapy-induced mutagenesis from platinum and melphalan are relatively hypermutated and enriched for complex structural variants (i.e., chromothripsis), while neoplasms with alternative exposures bear a similar profile to de novo acute myeloid leukemia. Using chemotherapy-associated mutational signatures as a temporal barcode in each patient’s life, we estimate that several complex events and genomic drivers are acquired after chemotherapy exposure. In the case of treatment with high-dose melphalan and autologous stem cell transplantation, we demonstrate that the procedure allows clonal hematopoiesis to escape chemotherapy exposure entirely, and to be reinfused to expand to malignancy. This information reveals a novel mode of malignant progression for therapy-related malignancies that is not reliant on direct mutagenesis or even exposure to chemotherapy, itself, and prompts further investigation into leukemia-permissive effects of cytotoxic drugs.
- Published
- 2022
20. Association of Patient Activity Bio-Profiles with Health-Related Quality of Life: A Prospective Trial Using Mobile Wearables in Newly Diagnosed Multiple Myeloma Patients
- Author
-
Neha Korde, Elizabeth Tavitian, Donna Mastey, Joseph Lengfellner, Gil Hevroni, Andrew Zarski, Meghan Salcedo, Sham Mailankody, Hani Hassoun, Eric L. Smith, Malin Hultcrantz, Urvi Shah, Carlyn Rose Tan, Benjamin Diamond, Gunjan Shah, Michael Scordo, Oscar B. Lahoud, David J. Chung, Heather J. Landau, Sergio A. Giralt, Andriy Derkach, Thomas Atkinson, Paul Sabbatini, Francesca Konig, Saad Usmani, Ola Landgren, and Alexander Lesokhin
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
21. P-176: Assessment of renal outcome following therapy in monoclonal immunoglobulin deposition disease: a retrospective study of 34 patients highlighting the need for consensus criteria
- Author
-
Matthew J. Pianko, Timothy Tiutan, Andriy Derkach, Insara Jaffer-Sathick, Adriana Rossi, Steven Salvatore, Heather Landau, Oscar Lahoud, Neha Korde, Sham Mailankody, Alexander Lesokhin, Malin Hultcrantz, Urvi Shah, Carlyn Tan, David Chung, Gunjan Shah, Edgar Jaimes, Saad Usmani, Sergio Giralt, and Hani Hassoun
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
22. P-170: Evaluating serum free light chain ratio as a biomarker for multiple myeloma
- Author
-
Theresia Akhlaghi, Kylee Maclachlan, Neha Korde, Sham Mailankody, Alexander Lesokhin, Hani Hassoun, Sydney Lu, Dhwani Patel, Urvi Shah, Carlyn Tan, Andriy Derkach, Oscar Lahoud, Heather Landau, Gunjan Shah, Michael Scordo, David Chung, Sergio Giralt, Saad Usmani, Ola Landgren, and Malin Hultcrantz
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
23. P-194: Plinabulin after autologous hematopoietic cell transplant to decrease duration of neutropenia and improve quality of life peri-transplant
- Author
-
Gunjan Shah, Leah Shulman, Danielle Hanley, David Chung, Ambika Datta, Prima Dhar, Gaurav Gupta, Hani Hassoun, Elizabeth Hoover, Malin Hultcrantz, Neha Korde, Oscar Lahoud, Heather Landau, Alexander Lesokhin, Sham Mailankody, Michael Scordo, Urvi Shah, Carlyn Tan, Saad Usmani, Ramon Mohanlal, and Sergio Giralt
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
24. P-190: Bone remineralization of lytic lesions in newly diagnosed multiple myeloma (NDMM) patients treated with carfilzomib, lenalidomide, dexamethasone +/- daratumumab induction regimen
- Author
-
Mina Meseha, Murad Abusamra, Azeez Farooki, Andriy Derkach, Kylee Maclachlan, Malin Hultcrantz, Hani Hassoun, Urvi Shah, Sydney Lu, Sham Mailankody, Dhwani Patel, Oscar Lahoud, Gunjan Shah, Michael Scordo, David Chung, Heather Landau, Alexander Lesokhin, Neha Korde, Sergio Giralt, Saad Usmani, and Carlyn Tan
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
25. P-046: Maintenance therapy cessation for patients with three-year sustained MRD negative remissions: initial results from a phase II study
- Author
-
Neha Korde, Benjamin Diamond, Miranda Burge, Hani Hassoun, Heather Landau, Malin Hultcrantz, Sham Mailankody, Carlyn Tan, Urvi Shah, Kylee Maclachlan, Oscar Lahoud, Saad Usmani, Ola Landgren, and Alexander Lesokhin
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
26. Ixazomib and dexamethasone in high risk smoldering multiple myeloma: a clinical and correlative pilot study
- Author
-
Sham Mailankody, Meghan Salcedo, Elizabet Tavitian, Miranda Burge, Neha Korde, Hani Hassoun, Alexander Lesokhin, Oscar Lahoud, Eric Smith, Malin Hultcrantz, Carlyn Tan, Urvi Shah, Sean Devlin, and Ola Landgren
- Subjects
Smoldering Multiple Myeloma ,Boron Compounds ,Cancer Research ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Pilot Projects ,Hematology ,Multiple Myeloma ,Dexamethasone - Published
- 2022
27. Poster: MM-181 CARTITUDE-1: Two-Year Post Last Patient in (LPI) Results From the Phase 1b/2 Study of Ciltacabtagene Autoleucel (Cilta-Cel), a B-Cell Maturation Antigen (BCMA)-Directed Chimeric Antigen Receptor T (CAR-T) Cell Therapy, in Patients With Relapsed/Refractory Multiple Myeloma (RRMM)
- Author
-
Saad Z Usmani, Thomas Martin, Jesus G Berdeja, Andrzej Jakubowiak, Mounzer Agha, Adam D Cohen, Abhinav Deol, Myo Htut, Alexander Lesokhin, Nikhil C Munshi, Elizabeth O'Donnell, Carolyn C Jackson, Tzu-min Yeh, Arnob Banerjee, Enrique Zudaire, Deepu Madduri, Changwei Zhou, Lida Pacaud, Yi Lin, and Sundar Jagannath
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
28. P-171: African American patients with smoldering multiple myeloma may have a lower risk of progression compared to white patients
- Author
-
Theresia Akhlaghi, Kylee Maclachlan, Neha Korde, Sham Mailankody, Alexander Lesokhin, Hani Hassoun, Sydney Lu, Dhwani Patel, Urvi Shah, Carlyn Tan, Andriy Derkach, Oscar Lahoud, Heather Landau, Gunjan Shah, Michael Scordo, David Chung, Sergio Giralt, Saad Usmani, Ola Landgren, and Malin Hultcrantz
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
29. P-178: A pilot plant based dietary intervention in MGUS and SMM is feasible and associated with reduction in insulin and leptin levels
- Author
-
Urvi Shah, Francesca Castro, Aishwarya Anuraj, Jenna Blaslov, Peter Adintori, Andriy Derkach, Michael Pollak, Kylee Maclachlan, Sham Mailankody, Neha Korde, Carlyn Tan, Malin Hultcrantz, Hani Hassoun, Gunjan Shah, Michael Scordo, Oscar Lahoud, David Chung, Heather Landau, Anita D’Souza, Ola Landgren, Sergio Giralt, Saad Usmani, Neil Iyengar, Marcel van den Brink, and Alexander Lesokhin
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
30. OAB-014: Chemotherapy signatures reveal the evolutionary history of post-melphalan myeloid neoplasm
- Author
-
Benjamin Diamond, Bachisio Ziccheddu, Kylee Maclachlan, Justin Taylor, Eileen Boyle, David Coffey, Sydney Lu, Niccolo Bolli, Kelly Bolton, Jae Park, Heather Landau, Andrew McPherson, Mikkael Sekeres, Alexander Lesokhin, David Chung, Yanming Zhang, Caleb Ho, Mikhail Roshal, Jeffrey Tyner, Stephen Nimer, Elli Papaemmanuil, Saad Usmani, Gareth Morgan, Ola Landgren, and Francesco Maura
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
31. Abstract 5747: Chemotherapy-related mutational signatures reveal the origins of therapy-related myeloid neoplasms
- Author
-
Benjamin Diamond, Bachisio Ziccheddu, Eileen M. Boyle, Kylee Maclachlan, Justin Taylor, Justin M. Watts, Sydney X. Lu, David G. Coffey, Niccolo Bolli, Elli Papaemmanuil, Kelly Bolton, Jae H. Park, Heather Landau, Karuna Ganesh, Mikkael A. Sekeres, Stephen Nimer, David J. Chung, Caleb H. Ho, Mikhail Roshal, Alexander Lesokhin, Gareth Morgan, Ola Landgren, and Francesco Maura
- Subjects
Cancer Research ,Oncology - Abstract
Patients treated with chemotherapy (CT) and/or autologous stem-cell transplantation (ASCT) are at risk for therapy-related myeloid neoplasms (tMN). Certain cytotoxic agents introduce mutations within distinct trinucleotide contexts resulting in a unique barcode for each exposed cell. We leveraged mutational signatures to investigate the role of CT in the genomic landscape of tMN with respect to antecedent clonal hematopoiesis (CH). We analyzed 32 tMN and 2 tALL from 33 patients and interrogated for copy number abnormalities (CNA), structural variants (SV), single nucleotide variants (SNV), and mutational signatures. For 7 patients with tMN post-melphalan/ASCT, we investigated antecedent CH using targeted sequencing on pre-melphalan samples, including autograft products. CH variants that became clonal in tumor were seen in 5/7 pre-melphalan/ASCT samples (TP53, RUNX1, NCOR1, NF1, CREBBP, DNMT3A, and PPM1D). Complex SV were seen in 7 tMNs; including chromothripsis in 6 (19.4%). In 4 cases, chromothripsis involved chromosome 19 with hyper-amplification of the SMARCA4 locus (≥5 copies). Mutational signature analysis revealed 6 known single base substitution (SBS) signatures in tMN including melphalan (SBS-MM1) and platinum signatures (SBS31, SBS35, and E-SBS37). TMNs with CT signatures had higher mutation burden than those without (p = 0.004). 17 patients with exposure to agents other than melphalan/platinum did not have increased mutational burden with respect to de novo AML (TCGA; NEJM, 2013). All patients with prior platinum exposure (including tALL, n=9) had platinum SBS signatures while only 2 of 7 patients with prior melphalan/ASCT had a melphalan signature (SBS-MM1). Detection of CT signatures in bulk sequencing relies on one cell, with its barcode of mutations, to expand to clonal dominance. Given pre-existent CH, including in 3/3 autograft products, absence of a CT signature despite melphalan exposure implies progression by a clone that escaped CT exposure with stem-cell collection and reinfusion. Conversely, all platinum-exposed tAML had signature evidence of exposure confirming existence of CH prior to exposure and supporting post-CT single-cell expansion. TMNs from 3 patients exposed to sequential platinum and melphalan/ASCT had platinum but not melphalan signatures confirming single-cell expansion of the pre-tMN CH clone post-platinum but with escape from exposure to melphalan via leukapheresis. Chromothripsis events bore only non-duplicated CT-induced mutations, indicative of acquisition prior to, and not directly caused by, CT exposure. These disparities suggest that ASCT provides a mechanism for CH clones to escape CT and re-engraft with transplant. Coupled with driver events accrued prior to CT, this suggest that CT-induced mutagenesis may be less important than other factors, such as CT-induced immunosuppression, in the expansion of pre-TMN CH clones. Citation Format: Benjamin Diamond, Bachisio Ziccheddu, Eileen M. Boyle, Kylee Maclachlan, Justin Taylor, Justin M. Watts, Sydney X. Lu, David G. Coffey, Niccolo Bolli, Elli Papaemmanuil, Kelly Bolton, Jae H. Park, Heather Landau, Karuna Ganesh, Mikkael A. Sekeres, Stephen Nimer, David J. Chung, Caleb H. Ho, Mikhail Roshal, Alexander Lesokhin, Gareth Morgan, Ola Landgren, Francesco Maura. Chemotherapy-related mutational signatures reveal the origins of therapy-related myeloid neoplasms [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5747.
- Published
- 2022
32. CD47-Blocker TTI-622 Shows Single-Agent Activity in Patients with Advanced Relapsed or Refractory Lymphoma: Update from the Ongoing First-in-Human Dose Escalation Study
- Author
-
Krish Patel, Jeffrey A. Zonder, Dahlia Sano, Michael Maris, Alexander Lesokhin, Gottfried von Keudell, Catherine Lai, Rod Ramchandren, Tina Catalano, Gloria H. Y. Lin, Bob Uger, Penka S. Petrova, Naomi Molloy, Ingmar Bruns, and Swaminathan P Iyer
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Background CD47 is an innate immune checkpoint that binds signal regulatory protein alpha (SIRPα) and delivers a "don't eat me" signal to suppress macrophage phagocytosis. Overexpression of CD47 serves as a mechanism of immune surveillance evasion, and is associated with poor prognosis in both hematologic and solid malignancies. TTI-622 is a fusion protein consisting of the CD47-binding domain of human SIRPα linked to the Fc region of human IgG4. It is designed to enhance phagocytosis and antitumor activity by preventing CD47 from delivering its inhibitory signal as well as generating a moderate pro-phagocytic signal via IgG4 Fc. Importantly, unlike many CD47-blocking agents, TTI-622 does not bind to human red blood cells. Methods This is an ongoing multicenter, Phase 1a/1b dose-escalation and expansion trial of TTI-622 (NCT03530683). Based on a modified 3+3 scheme, dose escalation proceeded through 8 dose levels ranging from 0.05 to 18 mg/kg weekly dosing in patients with refractory or relapsed (r/r) lymphomas. Testing of every two and three week schedules of single agent TTI-622 at doses up to 24 mg/kg is ongoing. Safety monitoring includes clinical laboratories and assessments of adverse events (AEs) based on CTCAE v 4.03. Results As of April 12, 2021, 42 patients (18M/24F) with a median age of 67 years (range 24-86) have received dose levels of 0.05−18 mg/kg weekly TTI-622. Lymphoma histologies enrolled include diffuse large B-cell lymphoma (DLBCL; n=19), cutaneous T-cell lymphoma - mycosis fungoides (CTCL-MF; n=6), peripheral T-cell lymphoma (PTCL; n=6), Hodgkin lymphoma (HL; n=5), and follicular lymphoma (FL; n=4). Additional histologies with single patients only accounted for the remaining 2 patients. Disease stages at entry have been III or IV in 38 patients and median prior systemic therapies of 3 (range 1-9) have been received, including CAR-T and HSCT in 9 and 10 patients, respectively. One additional patient with Erdheim-Chester disease initially diagnosed as lymphoma is included in the safety analysis only. Treatment-related AEs have occurred in 20 (47%) patients; most AEs have been Grade 1 or 2 and reversible. The most frequent treatment-related AEs include thrombocytopenia (n=9, 21%), neutropenia (n=5, 12%), and anemia and fatigue (n=4 each, 9%). Related adverse events of Grade ≥ 3 intensity include thrombocytopenia (n=2, 5%; 1 each Grade 3 & 4), neutropenia (n=4, 9%; all Grade 3), and anemia (n=1, 2%; Grade 3). These Grade 3 and 4 hematologic events occurred in dose levels ranging from 0.8 to 18 mg/kg without apparent dose relationship. Mild to moderate platelet decreases seen in the majority of patients generally occurred on dosing days and recovered quickly to baseline levels. Preliminary PK results suggest dose-dependent increases in exposure following both single and repeated TTI-622 infusions. A total of 27 patients were response-evaluable at dose levels ≥0.8 mg/kg at the time of the data-cut. Objective responses have been achieved in 9 patients, including 2 complete responses (CR) - 1 in DLBCL (0.8 mg/kg) and 1 in CTCL (18 mg/kg) - and 7 partial responses (PR) - 2 in CTCL (both 8 mg/kg), 2 in PTCL (2 and 12 mg/kg), 2 in DLBCL (4 and 18 mg/kg), and 1 in FL (8 mg/kg). Initial responses were achieved at the first disease assessment (week 8) in all 9 responders. In the dose range of 0.8-18 mg/kg, objective responses occurred in 33% (9/27) of response-evaluable patients. At the time of the data cut-off 4 responders were active on treatment; both patients with CR, ongoing 22+ months (0.8 mg/kg) and 12 weeks on an every three week dosing schedule (18 mg/kg). Two patients with PR were ongoing 18 weeks (12 mg/kg) and 11 weeks (18 mg/kg). Conclusions Results to date in patients with r/r lymphomas indicate that weekly doses of single agent TTI-622 up to 18 mg/kg are well tolerated. TTI-622 has shown activity in r/r lymphoma with objective responses across a broad dose range (0.8-18 mg/kg), in highly pre-treated patients, across multiple histologies, and with onset of action consistently observed at the first response assessment at Week 8. The study is ongoing evaluating single agent TTI-622 in every two and three week schedules. Updated data including additional patients will be presented at the conference. Based on these results demonstrating promising single-agent activity as well as good tolerability, TTI-622 is currently being investigated in combination regimens in a range of hematologic malignancies. Disclosures Patel: Xencor: Research Funding; Velos Bio: Research Funding; Trillium Therapeutics: Research Funding; TG Therapeutics: Consultancy, Speakers Bureau; Sunesis Pharmaceuticals: Research Funding; Pharmacyclics/Janssen: Consultancy, Research Funding, Speakers Bureau; Morphosys: Consultancy; Millenium/Takeda: Research Funding; MEI Pharma: Consultancy, Research Funding; Kite: Consultancy, Research Funding, Speakers Bureau; Juno Pharmaceuticals: Consultancy; Genentech/Roche: Consultancy, Research Funding, Speakers Bureau; Fate Therapeutics: Research Funding; Curis, Inc: Research Funding; Celgene: Consultancy, Research Funding, Speakers Bureau; Bristol Myers Squibb: Consultancy, Research Funding, Speakers Bureau; BeiGene: Consultancy; AstraZeneca: Consultancy, Research Funding, Speakers Bureau; Aptevo Therapeutics: Research Funding; Abbvie: Consultancy. Zonder: Amgen: Consultancy; BMS: Consultancy, Research Funding; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy; Caelum Biosciences: Consultancy; Intellia: Consultancy; Regeneron: Consultancy; Alnylam: Consultancy. Lesokhin: pfizer: Consultancy, Research Funding; Genetech: Research Funding; Janssen: Honoraria, Research Funding; Iteos: Consultancy; bristol myers squibb: Research Funding; Serametrix, Inc: Patents & Royalties; Trillium Therapeutics: Consultancy; Behringer Ingelheim: Honoraria. von Keudell: Merck: Consultancy, Honoraria; Pharmacyclics: Consultancy, Honoraria; Merck: Research Funding; Incyte: Consultancy, Honoraria; AbbVie: Research Funding; BMS: Research Funding; Janssen: Research Funding. Lai: Macrogenics: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas: Speakers Bureau; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees; Agios: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz Pharma: Speakers Bureau; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees. Ramchandren: curis: Research Funding; seattle genetics: Consultancy, Research Funding; MERCK: Consultancy, Research Funding; pharmacyclics: Consultancy, Research Funding; BMS: Consultancy; Trillium: Research Funding. Catalano: Trillium Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Lin: Trillium Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Uger: Trillium Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Petrova: Trillium Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Molloy: Trillium Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Bruns: Trillium Therapeutics: Current Employment, Current holder of stock options in a privately-held company.
- Published
- 2021
33. Genomic and Immune Signatures Predict Sustained MRD Negativity in Newly Diagnosed Multiple Myeloma Patients Treated with Daratumumab, Carfilzomib, Lenalidomide, and Dexamethasone (D-KRd)
- Author
-
Francesco Maura, Eileen M Boyle, Benjamin Diamond, Patrick Blaney, Hussein Ghamlouch, Bachisio Ziccheddu, Yubao Wang, Kylee H Maclachlan, James E. Hoffman, Hani Hassoun, Sham Mailankody, Urvi A Shah, Carlyn Tan, Malin Hultcrantz, Michael Scordo, Dickran Kazandjian, Gunjan L Shah, Heather J Landau, David J. Chung, Sergio Giralt, Benedetto Bruno, Yanming Zhang, Arnaldo A Arbini, Ahmet Dogan, Alexander Lesokhin, Faith E Davies, Neha Korde, Gareth J Morgan, and Ola Landgren
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry ,health care economics and organizations - Abstract
Introduction: Treatment combinations involving CD38 targeted monoclonal antibodies have significantly prolonged the median duration and depth of response in myeloma (MM), reflected in minimal residual disease-negativity (MRD-) rates of over 70% in newly diagnosed patients (Landgren et al. JAMA Onc 2021). Key to improving our understanding of treatment failures is the combined use of single cell analysis of the microenvironment with genome wide assessment of tumor genetics to decipher the mechanisms of disease resistance. Methods: We isolated malignant plasma cells from bone marrow (BM) samples using CD138+magnetic or flow (CD38, CD138, and CD45) sorting from 60 newly diagnosed MM patients treated with KRd with daratumumab (D-KRd n=46; NCT03290950) and without daratumumab combination therapy (KRd, n=14; NCT01402284). Fifty-five baseline samples underwent whole genome sequencing (WGS), median coverage of 80x using somatic DNA as a normal comparator. The BM cellular content of 22 patients (44 samples-5 failed) treated with D-KRd (10 MRD+ and 12 MRD-) underwent 5'single cell RNA-sequencing with an additional capture of the TCR and surface protein markers (CITEseq) to interrogate the single cell composition of the immune microenvironment at baseline (T1, n=20) and at the end of induction (T2, n=19). Paired (T1/T2) single-cell data were obtained in 17 patients and paired WGS and single cell data (T1) were available in 15 patients. MRD-, sustained MRD- (defined as two MRD- results, the first at the end of the induction (T2) and the subsequent at the first year of follow-up (T3)) and progression/loss of MRD- were used as clinical endpoints for this study. Results: After a median follow up of 29 months, 36 (54%) patients achieved MRD-; 34 (51%) had sustained MRD- >1 year after completion of combination therapy. Overall, 10 (15%) patients had clinical progression and two conversions from MRD- to MRD+. A comprehensive catalogue of MM-genomic events associated with these three clinical endpoints was defined. Deletion (del) 13, biallelic loss CYLD, del XBP1, del 20q13.12 (CD40), and 8q gains were associated with MRD+ and failure to achieve sustained MRD-. Presence of del RPL5 and multiple chromothripsis events significantly correlated with early progression and loss of MRD-. Interestingly, structural variants (SV) involving IKFZ3 were seen in all three negative clinical endpoints (p We interrogated the BM microenvironment at baseline and correlated its composition with the tumor genomic architecture. Across 15 evaluable patients, del XBP1 were associated with fewer memory B-cells (p=0.03), naïve B-cell (p=0.01) and dendritic cells (p=0.03) compared to the wild type. Also, low dendritic cell at baseline cases were observed in patients with del 20q13.12 (CD40) (p=0.03). Interestingly, low level of plasmacytoid dendritic cells at baseline was associated with failure to achieve MRD- and sustained MRD-. Patients with 6p24 amplification showed a reduced number of CD8 effectors 1 and 2 (p When comparing baseline (T1) and end of induction (T2), significant differences were seen between sustained MRD+ and MRD-. We identified significantly depleted NK, and naïve and memory B-cell after D-KRd MRD- patients had significantly more CD14+ monocytes both at T1 and T2 than their MRD+ counterparts (Fig. 1). Differential expression suggests that inflammatory response genes including IL1B are upregulated in the absence of sustained MRD- whereas genes implicated in IL2, IL6, and IFNα response as well as adipocyte differentiation are associated with sustained MRD response. Conclusion: We show, for the first time, evidence of complex interplay between MM tumor genetics and the microenvironment in the context of D-KRd treated patients. Our results highlight the importance of genomic-based mechanisms in the persistence of disease (IKZF3, XBP1) as well as heterogeneity in the composition of the BM microenvironment, with the monocytes pointing towards the importance of inflammation. Figure 1 Figure 1. Disclosures Maura: OncLive: Honoraria; Medscape: Consultancy, Honoraria. Hassoun: Celgene, Takeda, Janssen: Research Funding. Mailankody: Jansen Oncology: Research Funding; Allogene Therapeutics: Research Funding; Bristol Myers Squibb/Juno: Research Funding; Legend Biotech: Consultancy; Takeda Oncology: Research Funding; Fate Therapeutics: Research Funding; Physician Education Resource: Honoraria; Plexus Communications: Honoraria; Evicore: Consultancy. Hultcrantz: Intellisphere LLC: Consultancy; GlaxoSmithKline: Membership on an entity's Board of Directors or advisory committees, Research Funding; Curio Science LLC: Consultancy; Amgen: Research Funding; Daiichi Sankyo: Research Funding. Scordo: Omeros Corporation: Consultancy; i3 Health: Other: Speaker; Kite - A Gilead Company: Membership on an entity's Board of Directors or advisory committees; Angiocrine Bioscience: Consultancy, Research Funding; McKinsey & Company: Consultancy. Kazandjian: Arcellx: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees. Shah: Janssen Pharmaceutica: Research Funding; Amgen: Research Funding. Landau: Takeda, Janssen, Caelum Biosciences, Celgene, Pfizer, Genzyme: Membership on an entity's Board of Directors or advisory committees; Takeda: Research Funding; Genzyme: Honoraria. Giralt: SANOFI: Membership on an entity's Board of Directors or advisory committees; PFIZER: Membership on an entity's Board of Directors or advisory committees; GSK: Membership on an entity's Board of Directors or advisory committees; JAZZ: Membership on an entity's Board of Directors or advisory committees; JANSENN: Membership on an entity's Board of Directors or advisory committees; AMGEN: Membership on an entity's Board of Directors or advisory committees; Actinnum: Membership on an entity's Board of Directors or advisory committees; CELGENE: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees. Dogan: Physicians' Education Resource: Honoraria; Seattle Genetics: Consultancy; EUSA Pharma: Consultancy; Roche: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Peer View: Honoraria. Lesokhin: Behringer Ingelheim: Honoraria; pfizer: Consultancy, Research Funding; Janssen: Honoraria, Research Funding; Iteos: Consultancy; Genetech: Research Funding; bristol myers squibb: Research Funding; Trillium Therapeutics: Consultancy; Serametrix, Inc: Patents & Royalties. Davies: Janssen: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Roche: Consultancy, Honoraria. Korde: Amgen: Research Funding; Medimmune: Membership on an entity's Board of Directors or advisory committees. Morgan: BMS: Membership on an entity's Board of Directors or advisory committees; Jansen: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Oncopeptides: Membership on an entity's Board of Directors or advisory committees. Landgren: Janssen: Other: IDMC; Celgene: Research Funding; Amgen: Honoraria; Janssen: Research Funding; Janssen: Honoraria; Amgen: Research Funding; Takeda: Other: IDMC; GSK: Honoraria.
- Published
- 2021
34. Cevostamab Monotherapy Continues to Show Clinically Meaningful Activity and Manageable Safety in Patients with Heavily Pre-Treated Relapsed/Refractory Multiple Myeloma (RRMM): Updated Results from an Ongoing Phase I Study
- Author
-
Suzanne Trudel, Adam D Cohen, Amrita Y. Krishnan, Rafael Fonseca, Andrew Spencer, Jesús G. Berdeja, Alexander Lesokhin, Peter A Forsberg, Jacob P. Laubach, Luciano J. Costa, Paula Rodriguez-Otero, Rayan Kaedbey, Joshua Richter, Maria-Victoria Mateos, Sheeba K Thomas, Chihunt Wong, Mengsong Li, Voleak Choeurng, Anjali Vaze, Divya Samineni, Teiko Sumiyoshi, James Cooper, and Simon J Harrison
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Background: Fc receptor-homolog 5 (FcRH5) is a type I membrane protein that is expressed exclusively in the B-cell lineage, and at a higher level on myeloma cells than on normal B cells. Cevostamab is a FcRH5xCD3 bispecific antibody (BsAb) that facilitates T cell-directed killing of myeloma cells. Initial data from the dose-escalation phase of the ongoing Phase I study (NCT03275103) of cevostamab monotherapy in patients (pts) with heavily pre-treated RRMM demonstrated promising activity and manageable safety, along with near ubiquitous FcRH5 expression on myeloma cells (Cohen et al. ASH 2020; Sumiyoshi et al. EHA 2021). We present updated safety and efficacy data from a larger cohort of pts, including results comparing Cycle (C) 1 single step-up (SS) and double step-up (DS) dosing for the mitigation of cytokine release syndrome (CRS). Methods: Participants have RRMM for which no established therapy is available or appropriate. Cevostamab (intravenous infusion) is administered in 21-day cycles. In the SS cohorts, the step dose (0.05-3.6mg) is given on C1 Day (D) 1 and the target dose (0.15-198mg) on C1D8. In the DS cohorts, the step doses are given on C1D1 (0.3-1.2mg) and C1D8 (3.6mg), and the target dose (60-160mg) on C1D15. In both regimens, the target dose is given on D1 of subsequent cycles. Cevostamab is continued for a total of 17 cycles, unless progressive disease or unacceptable toxicity occurs. CRS is reported using ASTCT criteria (Lee et al. Biol Blood Marrow Transplant 2019). Results: At data cut-off (18 May 2021), 160 pts had been enrolled (median age: 64 years, range: 33-82 years; male: 58.1%); 21.3% of pts had extramedullary disease. Median number of prior lines of therapy was 6 (range: 2-18). Most pts (85.0%) were triple-class refractory (PI, IMiD, anti-CD38 antibody). 28 pts (17.5%) had received ≥1 prior CAR-T, 13 pts (8.1%) ≥1 prior BsAb, 27 pts (16.9%) ≥1 prior antibody-drug conjugate (ADC), and 54 pts (33.8%) ≥1 prior anti-BCMA targeting agent. Median follow-up in exposed pts was 6.1 months. Almost all had ≥1 adverse event (Table). The most common was CRS (128/160 pts [80.0%]; Grade [Gr] 1: 42.5%; Gr 2: 36.3%; Gr 3: 1.3%). Immune effector cell-associated neurotoxicity syndrome (ICANS) associated with CRS was observed in 21 pts (13.1%) and in 34/211 (16.1%) CRS events (Gr 1: 8.5%; Gr 2: 6.2%; Gr 3: 1.4%). Most CRS events occurred in C1 (87.2%), arose within 24 hours of cevostamab administration (70.5%), and resolved within 48 hours of onset (83.4%). In the pts with CRS, tocilizumab was used for CRS management in 43.8% and steroids in 25.8% (both agents: 18.0%). In SS dose-escalation (68 pts), 3.6mg was chosen as the most effective C1D1 SS dose for limiting CRS in C1, with no target dose-dependent increase in the rate or severity of CRS observed after the C1D8 administration. Likewise, in DS dose-escalation (30 pts), 0.3/3.6mg was identified as the preferred C1D1/C1D8 DS dose for limiting CRS in C1. Notably, the overall rate of CRS was lower in the pts who received the 0.3/3.6mg/target DS regimen than in those who received the 3.6mg/target SS regimen (77.3% [34/44] vs 88.2% [75/85], respectively). The rate of ICANS associated with CRS was also lower in the 0.3/3.6mg/target DS cohort than in the 3.6mg/target SS cohort (4.5% [2/44] vs 21.2% [18/85], respectively). At data cut-off, 158/160 pts were efficacy evaluable. In dose-escalation, responses were observed at the 20-198mg target dose levels, and data suggested a target dose-dependent increase in clinical efficacy. Median time to response was 29 days (range: 20-179 days). Two dose-expansion cohorts were opened: ORR was higher at the 160mg dose level (54.5%, 24/44 pts) than at the 90mg dose level (36.7%, 22/60). At target dose levels >90mg, ORRs in pts with prior exposure to CAR-Ts, BsAbs, ADCs, and anti-BCMA targeting agents were 44.4% (4/9 pts), 33.3% (3/9), 50.0% (7/14), and 36.4% (8/22) respectively. Median follow-up among all responders (n=61) was 8.1 months; estimated median duration of response was 15.6 months (95% CI: 6.4, 21.6). Conclusions: Cevostamab monotherapy continues to show clinically meaningful activity in a large cohort of pts with heavily pre-treated RRMM, with a target dose-dependent increase in ORR, but no increase in CRS rate. Responses appear durable, and are observed in pts with prior exposure to CAR-Ts, BsAbs, and ADCs. Compared with SS dosing, DS dosing at the 0.3/3.6mg level appears to be associated with a trend for an improved C1 safety profile. Figure 1 Figure 1. Disclosures Trudel: Amgen: Honoraria, Research Funding; GlaxoSmithKline: Consultancy, Honoraria, Research Funding; Roche: Consultancy; Genentech: Research Funding; Pfizer: Honoraria, Research Funding; BMS/Celgene: Consultancy, Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Sanofi: Honoraria. Cohen: BMS/Celgene: Consultancy; GlaxoSmithKline: Consultancy, Research Funding; Oncopeptides: Consultancy; Novartis: Research Funding; Genentech/Roche: Consultancy; AstraZeneca: Consultancy; Janssen: Consultancy; Takeda: Consultancy. Krishnan: MAGENTA: Consultancy; BMS: Consultancy, Current equity holder in publicly-traded company, Speakers Bureau; JANSSEN: Consultancy, Research Funding; City of Hope Cancer Center: Current Employment; REGENERON: Consultancy; SANOFI: Consultancy; GSK: Consultancy; Amgen: Speakers Bureau. Fonseca: Kite: Consultancy; Juno: Consultancy; Merck: Consultancy; Sanofi: Consultancy; Pharmacyclics: Consultancy; Novartis: Consultancy; OncoTracker: Consultancy, Membership on an entity's Board of Directors or advisory committees; Aduro: Consultancy; Caris Life Sciences: Membership on an entity's Board of Directors or advisory committees; Patent: Prognosticaton of myeloma via FISH: Patents & Royalties; AbbVie: Consultancy; GSK: Consultancy; Scientific Advisory Board: Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; Bayer: Consultancy; Amgen: Consultancy; Mayo Clinic in Arizona: Current Employment; Celgene: Consultancy; BMS: Consultancy; Takeda: Consultancy; Janssen: Consultancy. Spencer: Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees. Berdeja: Bluebird bio, BMS, Celgene, CRISPR Therapeutics, Janssen, Kite Pharma, Legend Biotech, SecuraBio, Takeda: Consultancy; Abbvie, Acetylon, Amgen: Research Funding; EMD Sorono, Genentech: Research Funding; Celularity, CRISPR Therapeutics: Research Funding; GSK, Ichnos Sciences, Incyte: Research Funding; Lilly, Novartis: Research Funding; Poseida, Sanofi, Teva: Research Funding. Lesokhin: Serametrix, Inc: Patents & Royalties; Behringer Ingelheim: Honoraria; Genetech: Research Funding; Iteos: Consultancy; Janssen: Honoraria, Research Funding; pfizer: Consultancy, Research Funding; bristol myers squibb: Research Funding; Trillium Therapeutics: Consultancy. Forsberg: University of Colorado: Current Employment; Karyopharm, Sanofi, Genentech: Research Funding. Costa: Karyopharm: Consultancy, Honoraria; BMS: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria, Speakers Bureau; Amgen: Consultancy, Honoraria, Research Funding, Speakers Bureau. Rodriguez-Otero: Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene-BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; GSK: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy; Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees; Kite: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria; Regeneron: Honoraria; Clínica Universidad de Navarra: Current Employment. Kaedbey: Takeda, Sanofi: Honoraria; Celgene/BMS, Janssen: Honoraria; Royal Victoria Hospital Lakeshore Hospital: Ended employment in the past 24 months; Jewish General Hospital - McGill University: Current Employment. Richter: Janssen, Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Adaptive biotechnologies: Speakers Bureau; BMS, Karyopharm, Antengene: Membership on an entity's Board of Directors or advisory committees; Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai: Current Employment. Mateos: Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene - Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Regeneron: Honoraria, Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologies: Honoraria, Membership on an entity's Board of Directors or advisory committees; Sea-Gen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Oncopeptides: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; AbbVie: Honoraria; Bluebird bio: Honoraria; GSK: Honoraria; Oncopeptides: Honoraria. Thomas: Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Genentech: Research Funding; Acerta Pharma: Research Funding; X4 Pharma: Research Funding; Ascentage Pharma: Research Funding; BeiGene: Membership on an entity's Board of Directors or advisory committees; BMS: Research Funding. Wong: Genentech: Current Employment; CTMX, UBX, BMRN: Current equity holder in publicly-traded company. Li: Genentech/Roche: Current Employment, Current equity holder in publicly-traded company. Choeurng: Genentech: Current Employment, Current equity holder in publicly-traded company. Vaze: Roche/Genentech: Current Employment, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Samineni: Genentech: Current Employment, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Sumiyoshi: Genentech: Current Employment, Current equity holder in publicly-traded company, Divested equity in a private or publicly-traded company in the past 24 months. Cooper: Genentech: Current Employment; Roche: Current holder of individual stocks in a privately-held company. Harrison: Haemalogix: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Roche/Genentech: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Janssen Cilag: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene/ Juno/ BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; GSK: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Eusa: Consultancy, Honoraria, Speakers Bureau; Terumo BCT: Consultancy, Honoraria. OffLabel Disclosure: Cevostamab is a FcRH5xCD3 bispecific antibody that facilitates T cell-directed killing of myeloma cells. Cevostamab is an investigational agent.
- Published
- 2021
35. Poster: MM-379: MagnetisMM-1: A Study of Elranatamab (PF-06863135), a B-Cell Maturation Antigen (BCMA)-Targeted, CD3-Engaging Bispecific Antibody, for Patients with Relapsed or Refractory Multiple Myeloma (MM)
- Author
-
Moshe Levy, Nizar Bahlis, Noopur Raje, Caitlin Costello, Bhagirathbhai Dholaria, Melhem Solh, Michael Tomasson, Harman Dube, Michael Damore, Hoi Kei Lon, Cynthia Basu, Athanasia Skoura, Edward Chan, Suzanne Trudel, Andrzej Jakubowiak, Michael Chu, Cristina Gasparetto, Andrew Dalovisio, Michael Sebag, and Alexander Lesokhin
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2021
36. Role of AID in the temporal pattern of acquisition of driver mutations in multiple myeloma
- Author
-
Francesco, Maura, Even H, Rustad, Venkata, Yellapantula, Marta, Łuksza, David, Hoyos, Kylee H, Maclachlan, Benjamin T, Diamond, Benjamin D, Greenbaum, Gareth, Morgan, Alexander, Lesokhin, Elli, Papaemmanuil, and Ola, Landgren
- Subjects
Clonal Evolution ,Whole Genome Sequencing ,Cytidine Deaminase ,DNA Mutational Analysis ,Mutation ,Biomarkers, Tumor ,Humans ,Multiple Myeloma - Published
- 2019
37. Nivolumab Plus Ipilimumab in Patients With Advanced Melanoma: Updated Survival, Response, and Safety Data in a Phase I Dose-Escalation Study.
- Author
-
Callahan MK, Kluger H, Postow MA, Segal NH, Lesokhin A, Atkins MB, Kirkwood JM, Krishnan S, Bhore R, Horak C, Wolchok JD, and Sznol M
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Drug Administration Schedule, Female, Humans, Ipilimumab adverse effects, Male, Melanoma mortality, Melanoma secondary, Middle Aged, Nivolumab adverse effects, Skin Neoplasms mortality, Skin Neoplasms pathology, Time Factors, Treatment Outcome, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Ipilimumab administration & dosage, Melanoma drug therapy, Nivolumab administration & dosage, Skin Neoplasms drug therapy
- Abstract
Purpose The clinical activity observed in a phase I dose-escalation study of concurrent therapy with nivolumab (NIVO) and ipilimumab (IPI) in patients with previously treated or untreated advanced melanoma led to subsequent clinical development, including randomized trials. Here, we report long-term follow-up data from study CA209-004, including 3-year overall survival (OS). Patients and Methods Concurrent cohorts 1, 2, 2a, and 3 received escalating doses of NIVO plus IPI once every 3 weeks for four doses, followed by NIVO once every 3 weeks for four doses, then NIVO plus IPI once every 12 weeks for eight doses. An expansion cohort (cohort 8) received concurrent NIVO 1 mg/kg plus IPI 3 mg/kg once every 3 weeks for four doses, followed by NIVO 3 mg/kg once every 2 weeks, which is the dose and schedule used in phase II and III studies and now approved for patients with unresectable or metastatic melanoma. Results Among all concurrent cohorts (N = 94) at a follow-up of 30.3 to 55.0 months, the 3-year OS rate was 63% and median OS had not been reached. Objective response rate by modified WHO criteria was 42%, and median duration of response was 22.3 months. Incidence of grade 3 and 4 treatment-related adverse events was 59%. The most common grade 3 and 4 treatment-related adverse events were increases in lipase (15%), alanine aminotransferase (12%), and aspartate aminotransferase (11%). One treatment-related death (1.1%) occurred in a patient who had multiorgan failure 70 days after the last dose of NIVO plus IPI. Conclusion This is the longest follow-up for NIVO plus IPI combination therapy in patients with advanced melanoma. The 3-year OS rate of 63% is the highest observed for this patient population and provides additional evidence for the durable clinical activity of immune checkpoint inhibitors in the treatment of advanced melanoma.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.