1
|
Ramasamy K, Vij R, Kuter D, Cella D, Durie BGM, Abonour R, Rifkin RM, Ailawadhi S, Lee HC, Cowan AJ, Ho C, Dhanasiri S, Fish S, Yu E, Dhamane AD, Fang J, Marshall TS, Samuel A, Liu L, Katz J, Gu T, Jagannath S. Real-World Treatment Patterns and Clinical Outcomes in Patients With Multiple Myeloma Previously Treated With Lenalidomide and an Anti-CD38 Monoclonal Antibody. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:337-348.e2. [PMID: 39799046 DOI: 10.1016/j.clml.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND This analysis explored real-world characteristics, treatment patterns and clinical outcomes in patients with relapsed or refractory multiple myeloma (RRMM) previously treated with lenalidomide and an anti-CD38 monoclonal antibody (mAb) and requiring subsequent treatment. MATERIALS AND METHODS The PREAMBLE and Connect MM prospective registries of patients with multiple myeloma (MM), and the US nationwide Flatiron Health electronic health record-derived de-identified database were analysed. MM-specific treatment patterns (prior/index therapies) and outcomes (progression-free survival [PFS]/overall survival [OS]) were assessed. RESULTS This analysis included: PREAMBLE n = 215; Connect MM n = 232; Flatiron Health n = 845. Median age at index was 69.0 years, median 3 prior lines of therapy; > 50% male. The most common index regimens accounted < 15% of treatments (most common PREAMBLE, Connect MM: carfilzomib±dexamethasone; Flatiron Health: pomalidomide+daratumumab+dexamethasone); most patients received classes that they had previously; ≥ 93% were triple-class exposed (immunomodulatory drug, proteasome inhibitor, anti-CD38 mAb). In PREAMBLE, Connect MM and Flatiron Health, respectively: 80.9%, 68.1% and 77.2% were lenalidomide- and anti-CD38 mAb-refractory; 69.3%, 67.2% and 71.1% were triple-class refractory (TCR); median PFS: 5.2 (95% CI 3.7-6.7), 4.4 (3.5-5.6) and 5.3 months (4.8-6.0); median OS: 19.3 (15.8-26.1), 14.2 (11.0-16.9) and 23.1 months (19.0-28.6). PFS and OS were shorter in lenalidomide- and anti-CD38 mAb-refractory patients versus those who were not refractory to both. A similar pattern was observed for TCR patients versus non-TCR patients. CONCLUSION There is no uniform standard of care for patients with RRMM with prior exposure to lenalidomide and anti-CD38 mAbs. Survival outcomes are poor, with a need for effective treatments for these patients.
Collapse
Affiliation(s)
- Karthik Ramasamy
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford Translational Myeloma Centre, NDORMS, University of Oxford, Oxford, UK.
| | - Ravi Vij
- Siteman Cancer Center, Washington University, St. Louis, MO
| | - David Kuter
- Hematology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - David Cella
- Department of Medical Social Sciences and Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | - Rafat Abonour
- Indiana University School of Medicine, Indianapolis, IN
| | - Robert M Rifkin
- Rocky Mountain Cancer Centers/US Oncology Research, Denver, CO
| | | | - Hans C Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Andrew J Cowan
- University of Washington / Fred Hutchinson Cancer Center, Seattle, WA
| | - Carrie Ho
- University of Washington / Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | | | | | | | | | | | | | | | - Tao Gu
- Bristol Myers Squibb, Princeton, NJ
| | | |
Collapse
|
2
|
Khan S, Bergstrom DJ, Côté J, Kotb R, LeBlanc R, Louzada ML, Mian HS, Othman I, Colasurdo G, Visram A. First Line Treatment of Newly Diagnosed Transplant Eligible Multiple Myeloma Recommendations From a Canadian Consensus Guideline Consortium. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e151-e172. [PMID: 39567294 DOI: 10.1016/j.clml.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
The availability of effective therapies for multiple myeloma (MM) has sparked debate on the role of first line autologous stem cell transplantation (ASCT), particularly in standard-risk patients. However, treatment for individuals with high-risk disease continues to display suboptimal outcomes. With novel therapies used earlier, practice is changing rapidly in the field of MM. Presently, quadruplet induction therapy incorporating an anti-CD38 monoclonal antibody to a proteasome inhibitor and an immunomodulatory drug prior to ASCT followed by maintenance therapy stands as the foremost strategy for attaining deep and sustained responses in transplant eligible MM (TEMM). This Canadian Consensus Guideline Consortium (CGC) proposes consensus recommendations for the first line treatment of TEMM. To address the needs of physicians and people diagnosed with MM, this document focuses on ASCT eligibility, induction therapy, mobilization and collection, conditioning, consolidation, and maintenance therapy, as well as, high-risk populations, management of adverse events, assessment of treatment response, and monitoring for disease relapse. The CGC will periodically review the recommendations herein and update as necessary.
Collapse
Affiliation(s)
- Sahar Khan
- Windsor Regional Hospital, University of Western Ontario, Windsor, Ontario, Canada.
| | - Debra J Bergstrom
- Division of Hematology, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Julie Côté
- Centre Hospitalier Universitaire de Québec, Quebec, Quebec, Canada
| | - Rami Kotb
- Department of Medical Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Richard LeBlanc
- Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, Quebec, Canada
| | - Martha L Louzada
- London Health Sciences Centre, Western University, London, Ontario, Canada
| | - Hira S Mian
- Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ibraheem Othman
- Allan Blair Cancer Centre, University of Saskatchewan, Regina, Saskatchewan, Canada
| | | | - Alissa Visram
- The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
3
|
Anwer F, Lan T, Dolph M, Moradian H, Slaff S, Shih YH, Tang D. Survival trends using DPd vs. other triplets in early RRMM patients: a population-adjusted indirect treatment comparison. Future Oncol 2025; 21:221-230. [PMID: 39611661 PMCID: PMC11792785 DOI: 10.1080/14796694.2024.2426443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024] Open
Abstract
AIMS Limited head-to-head data exist for daratumumab plus pomalidomide and dexamethasone (DPd) and non-pomalidomide-containing triplet regimens to treat relapsed/refractory multiple myeloma (RRMM). This study conducted population-adjusted indirect comparisons of overall survival (OS) for DPd vs. daratumumab, carfilzomib, and dexamethasone (DKd) and daratumumab, bortezomib, and dexamethasone (DVd). MATERIALS & METHODS A systematic literature review was performed via searches of databases and relevant conference proceedings. Both simulated treatment comparison (STC) and matching-adjusted indirect comparison (MAIC) were used to adjust for between-trial differences. RESULTS Seven randomized controlled trials were identified, five of which were subsequently excluded from the indirect treatment comparison during feasibility assessment. A consistent OS benefit was observed for DPd vs. DKd and DVd for patients with RRMM, using both STC and MAIC methods. CONCLUSIONS The findings of this study support the use of DPd over DKd and DVd for the treatment of patients with early RRMM.
Collapse
Affiliation(s)
- Faiz Anwer
- Hematology, Oncology, Multiple Myeloma, Blood and Marrow Transplant Program, Taussig Cancer Center, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | | | | | | | | | - Derek Tang
- Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
4
|
Ward MB, Jones AB, Krenciute G. Therapeutic advantage of combinatorial chimeric antigen receptor T cell and chemotherapies. Pharmacol Rev 2025; 77:100011. [PMID: 39952691 DOI: 10.1124/pharmrev.124.001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/28/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapies have transformed outcomes for many patients with hematological malignancies. However, some patients do not respond to CAR T cell treatment, and adapting CAR T cells for treatment of solid and brain tumors has been met with many challenges, including a hostile tumor microenvironment and poor CAR T cell persistence. Thus, it is unlikely that CAR T cell therapy alone will be sufficient for consistent, complete tumor clearance across patients with cancer. Combinatorial therapies of CAR T cells and chemotherapeutics are a promising approach for overcoming this because chemotherapeutics could augment CAR T cells for improved antitumor activity or work in tandem with CAR T cells to clear tumors. Herein, we review efforts toward achieving successful CAR T cell and chemical drug combination therapies. We focus on combination therapies with approved chemotherapeutics because these will be more easily translated to the clinic but also review nonapproved chemotherapeutics and drug screens designed to reveal promising new CAR T cell and chemical drug combinations. Overall, this review highlights the promise of CAR T cell and chemotherapy combinations with a specific focus on how combinatorial therapy overcomes challenges faced by either monotherapy and supports the potential of this therapeutic strategy to improve outcomes for patients with cancer. SIGNIFICANCE STATEMENT: Improving currently available CAR T cell products via combinatorial therapy with chemotherapeutics has the potential to drastically expand the types of cancers and number of patients that could benefit from these therapies when neither alone has been sufficient to achieve tumor clearance. Herein, we provide a thorough review of the current efforts toward studying CAR T and chemotherapy combinatorial therapies and offer perspectives on optimal ways to identify new and effective combinations moving forward.
Collapse
Affiliation(s)
- Meghan B Ward
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Amber B Jones
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
5
|
Ohlstrom D, Walker ZJ, Pandey A, Davis LN, Engel KL, Pan Z, Forsberg PA, Mark TM, Gillen AE, Sherbenou DW. Single-Cell RNA Sequencing before and after Light Chain Escape Reveals Intrapatient Multiple Myeloma Subpopulations with Divergent Osteolytic Gene Expression. CANCER RESEARCH COMMUNICATIONS 2025; 5:106-118. [PMID: 39699274 PMCID: PMC11737298 DOI: 10.1158/2767-9764.crc-24-0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/09/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
SIGNIFICANCE scRNA-seq was used to study a patient with high-risk multiple myeloma featuring LCE. LCE was rooted in a transcriptomic subpopulation that corresponded to a genetic subclone and established novel links between LCE and LAMP5 overexpression to osteolysis and prognosis, validated in RNA-seq databases.
Collapse
Affiliation(s)
- Denis Ohlstrom
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Biomedical Sciences and Biotechnology, Graduate School, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zachary J. Walker
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Abhishek Pandey
- Hematology-Oncology Fellowship Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lorraine N. Davis
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Krysta L. Engel
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zenggang Pan
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Peter A. Forsberg
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Tomer M. Mark
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Austin E. Gillen
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel W. Sherbenou
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
6
|
Bahlis NJ, Samaras C, Reece D, Sebag M, Matous J, Berdeja JG, Shustik J, Schiller GJ, Ganguly S, Song K, Seet CS, Acosta-Rivera M, Bar M, Quick D, Fonseca G, Liu H, Gentili C, Singh P, Siegel D. Pomalidomide/Daratumumab/Dexamethasone in Relapsed or Refractory Multiple Myeloma: Final Overall Survival From MM-014. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:852-862. [PMID: 39237427 DOI: 10.1016/j.clml.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Patients with relapsed or refractory multiple myeloma (RRMM) who have exhausted lenalidomide benefits require improved therapies. The 3-cohort phase 2 MM-014 trial (NCT01946477) explored pomalidomide in early lines of treatment for lenalidomide-exposed RRMM. In cohort B, pomalidomide plus daratumumab and dexamethasone (DPd) showed promising efficacy (median follow-up 28.4 months), as previously reported. Here, we report final overall survival (OS) in cohort B. METHODS Patients aged ≥ 18 years were treated in 28-day cycles: pomalidomide 4 mg orally daily from days 1 to 21; daratumumab 16 mg/kg intravenously on days 1, 8, 15, and 22 (cycles 1-2), days 1 and 15 (cycles 3-6), and day 1 (cycle ≥ 7); and dexamethasone 40 mg (age ≤ 75 years) or 20 mg (age > 75 years) orally on days 1, 8, 15, and 22. The primary endpoint was ORR. OS and safety were secondary endpoints. RESULTS Among 112 patients enrolled, 85 (75.9%) had lenalidomide-refractory disease and 27 (24.1%) had lenalidomide-relapsed disease. At a median follow-up of 41.9 months (range, 0.4-73.1), median OS was 56.7 months (95% confidence interval, 46.5-not reached). Treatment-emergent adverse events related to, and leading to discontinuation of, pomalidomide, dexamethasone, or daratumumab occurred in 7 (6.3%), 9 (8.0%), and 6 (5.4%) patients, respectively. CONCLUSION With long-term follow-up, our results show favorable OS with DPd. The safety profile was consistent with previous reports, with no new safety signals identified. IMiD agent-based therapy can still be considered in patients with RRMM who experience progressive disease on or after lenalidomide.
Collapse
Affiliation(s)
| | | | - Donna Reece
- Princess Margaret Hospital, Toronto, ON, Canada
| | - Michael Sebag
- McGill University Health Centre, Montreal, QC, Canada
| | - Jeffrey Matous
- Colorado Blood Cancer Institute, Sarah Cannon Research Institute, Denver, CO
| | | | | | | | | | - Kevin Song
- Vancouver General Hospital, Vancouver, BC, Canada
| | | | | | | | - Donald Quick
- Joe Arrington Cancer Research and Treatment Center, Lubbock, TX
| | | | | | | | | | | |
Collapse
|
7
|
Yu Y, Tian W, Grauffel C, Lin W, Hsieh M, Wu P, Lee H, Peng C, Lin P, Chu H, Lim C, Chang TW. An Antibody-Drug Conjugate for Multiple Myeloma Prepared by Multi-Arm Linkers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307852. [PMID: 38477561 PMCID: PMC11132082 DOI: 10.1002/advs.202307852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/31/2024] [Indexed: 03/14/2024]
Abstract
First-line treatment of multiple myeloma, a prevalent blood cancer lacking a cure, using anti-CD38 daratumumab antibody and lenalidomide is often inadequate due to relapse and severe side effects. To enhance drug safety and efficacy, an antibody-drug conjugate, TE-1146, comprising six lenalidomide drug molecules site-specifically conjugated to a reconfigured daratumumab to deliver cytotoxic lenalidomide to tumor cells is developed. TE-1146 is prepared using the HighDAR platform, which employs i) a maleimide-containing "multi-arm linker" to conjugate multiple drug molecules creating a drug bundle, and ii) a designed peptide with a Zn2+-binding cysteine at the C-termini of a reconfigured daratumumab for site-specific drug bundle conjugation. It is shown that TE-1146 remains intact and effectively enters CD38-expressing tumor cells, releasing lenalidomide, leading to enhanced cell-killing effects compared to lenalidomide/daratumumab alone or their combination. This reveals the remarkable potency of lenalidomide once internalized by myeloma cells. TE-1146 precisely delivers lenalidomide to target CD38-overexpressing tumor cells. In contrast, lenalidomide without daratumumab cannot easily enter cells, whereas daratumumab without lenalidomide relies on Fc-dependent effector functions to kill tumor cells.
Collapse
Affiliation(s)
- Yueh‐Hsiang Yu
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Wei‐Ting Tian
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | | | - Wei‐Chen Lin
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Ming‐Yu Hsieh
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Pei‐Wen Wu
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Hui‐Ju Lee
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Chi‐Jiun Peng
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Pei‐Hsuan Lin
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Hsing‐Mao Chu
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| | - Carmay Lim
- Institute of Biomedical SciencesAcademia SinicaAcademia Rd.Taipei115Taiwan
| | - Tse Wen Chang
- Immunwork, Inc.Academia Rd., Sec. 1, NangangTaipei115Taiwan
| |
Collapse
|
8
|
Kunyu L, Shuping S, Chang S, Yiyue C, Qinyu X, Ting Z, Bin W. An Updated Comprehensive Pharmacovigilance Study of Drug-Induced Thrombocytopenia Based on FDA Adverse Event Reporting System Data. J Clin Pharmacol 2024; 64:478-489. [PMID: 38041205 DOI: 10.1002/jcph.2389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/18/2023] [Indexed: 12/03/2023]
Abstract
Drug-induced thrombocytopenia (DIT) deserves both clinical and research attention for the serious clinical consequences and high prevalence of the condition. The current study aimed to perform a comprehensive pharmacovigilance analysis of DIT reported in the US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) database, with a particular focus on drugs associated with thrombocytopenia events. A disproportionality analysis of DIT was conducted using reports submitted to FARES from January 2004 to December 2022. Both the information component (IC) and reporting odds ratio (ROR) algorithms were applied to identify an association between target drugs and DIT events. A total of 15,940,383 cases were gathered in FAERS, 168,657 of which were related to DIT events. The top 50 drugs ranked by number of cases and ranked by signal strength were documented. The top 5 drugs ranked by number of cases were lenalidomide (10,601 cases), niraparib (3726 cases), ruxolitinib (3624 cases), eltrombopag (3483 cases), and heparin (3478 cases). The top 5 drugs ranked by signal strength were danaparoid (ROR 37.61, 95%CI 30.46-46.45), eptifibatide (ROR 34.75, 95%CI 30.65-39.4), inotersen (ROR 34.00, 95%CI 29.47-39.23), niraparib (ROR 30.53, 95%CI 29.42-31.69), and heparin (ROR 28.84, 95%CI 27.76-29.97). The top 3 involved drug groups were protein kinase inhibitors, antimetabolites, and monoclonal antibodies and antibody-drug conjugates. The current comprehensive pharmacovigilance study identified more drugs associated with thrombocytopenia. Although the mechanisms of DIT have been elucidated for some drugs, others still require further investigation.
Collapse
Affiliation(s)
- Li Kunyu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shi Shuping
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Su Chang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Cao Yiyue
- School of Mathematics, Sichuan University, Chengdu, China
| | - Xiong Qinyu
- School of Mathematics, Sichuan University, Chengdu, China
| | - Zhang Ting
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Wu Bin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Weisel K, Dimopoulos MA, Beksac M, Leleu X, Richter J, Heeg B, Patel S, Majer I, McFadden I, Mikhael J. Carfilzomib, daratumumab, and dexamethasone (KdD) vs. lenalidomide-sparing pomalidomide-containing triplet regimens for relapsed/refractory multiple myeloma: an indirect treatment comparison. Leuk Lymphoma 2024; 65:481-492. [PMID: 38345269 DOI: 10.1080/10428194.2023.2300051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/23/2023] [Indexed: 02/24/2024]
Abstract
Nearly all patients with multiple myeloma eventually relapse or become refractory to treatment. Lenalidomide is increasingly administered in the frontline until disease progression or intolerance to therapy, resulting in the need for highly effective, lenalidomide-sparing options. In this study, carfilzomib plus daratumumab and dexamethasone were evaluated against lenalidomide-sparing, pomalidomide-containing triplets using matching-adjusted indirect comparison in the absence of head-to-head data. The analyses utilized long-term follow-up data from the CANDOR study (NCT03158688). Treatment with carfilzomib, daratumumab, and dexamethasone resulted in significantly longer progression-free survival (hazard ratio 0.60 [95% confidence interval: 0.37, 0.88])vs. pomalidomide plus bortezomib and dexamethasone, and numerically longer progression-free survival (hazard ratio 0.77 [95% confidence interval: 0.50, 1.08]) vs. daratumumab plus pomalidomide and dexamethasone in patients with relapsed/refractory multiple myeloma and previous lenalidomide exposure, the majority of whom were lenalidomide refractory. Carfilzomib plus daratumumab and dexamethasone offers a highly effective, lenalidomide-sparing treatment option for this population.
Collapse
Affiliation(s)
- Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Xavier Leleu
- CHU de Poitiers - La Miletrie, INSERM CIC 1402, Poitiers, France
| | - Joshua Richter
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Bart Heeg
- Cytel Inc, Rotterdam, The Netherlands
| | | | | | | | - Joseph Mikhael
- Translational Genomics Research Institute (TGen), City of Hope Cancer Center, Phoenix, AZ, USA
| |
Collapse
|
10
|
Oriol A, Dimopoulos M, Schjesvold F, Beksac M, Facon T, Dhanasiri S, Guo S, Mu Y, Hong K, Gentili C, Galli M, Yagci M, Larocca A, Richardson P, Weisel K. Pomalidomide, Bortezomib, and Dexamethasone in Lenalidomide-Pretreated Multiple Myeloma: A Subanalysis of OPTIMISMM by Frailty and Bortezomib Dose Adjustment. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:165-176.e4. [PMID: 38072743 DOI: 10.1016/j.clml.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/06/2023] [Accepted: 10/26/2023] [Indexed: 03/01/2024]
Abstract
INTRODUCTION A proportion of patients with multiple myeloma (MM) are older and/or have comorbidities, requiring dose adjustments. Data from OPTIMISMM (NCT01734928) supported the use of pomalidomide, bortezomib, and dexamethasone (PVd) for treating relapsed/refractory MM. This subanalysis of OPTIMISMM assessed outcome by frailty and/or bortezomib dose adjustment. METHODS Patient frailty (nonfrail vs. frail) was classified using age, Charlson Comorbidity Index, and Eastern Cooperative Oncology Group performance status. Data from patients requiring a bortezomib dose reduction, interruption, and/or withdrawal during PVd treatment were assessed. RESULTS Among 559 patients, 93 of 281 (33.1%) and 93 of 278 (33.5%) patients who received PVd and bortezomib and dexamethasone (Vd), respectively, were frail. Overall response rate (ORR) and median progression-free survival (PFS) were higher in nonfrail vs. frail with PVd treatment (ORR, 82.8% vs. 79.6%; PFS, 14.7 vs. 9.7 months); significantly higher than with Vd regardless of frailty. Grade ≥ 3 treatment-emergent adverse events (TEAEs) were higher with PVd vs. Vd, regardless of frailty. Discontinuations of PVd were lower in nonfrail vs. frail patients (19.2% vs. 30.1%); the median duration of treatment was similar (DoT; 8.8 vs. 8.9 months, respectively). Patients who received PVd with a bortezomib dose adjustment (n = 240) had a longer median DoT (9.3 vs. 4.5 months) and PFS (12.1 vs. 8.4 months) vs. those without. CONCLUSION Frail patients treated with PVd demonstrated a higher ORR and a longer PFS and DoT vs. Vd, despite a higher frequency of grade ≥ 3 TEAEs leading to pomalidomide, bortezomib, and/or dexamethasone discontinuation. Therefore, PVd treatment may improve patient outcomes, regardless of frailty.
Collapse
Affiliation(s)
- Albert Oriol
- Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Barcelona, Spain.
| | | | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital and KG Jebsen Center for B Cell Malignancies, University of Oslo, Oslo, Norway
| | - Meral Beksac
- Ankara Üniversitesi Tip Fakültes, Ankara, Turkey
| | | | - Sujith Dhanasiri
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | | | | | | | | | | | - Munci Yagci
- Gazi University Medical Faculty, Ankara, Turkey
| | | | | | - Katja Weisel
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Abduh MS. An overview of multiple myeloma: A monoclonal plasma cell malignancy's diagnosis, management, and treatment modalities. Saudi J Biol Sci 2024; 31:103920. [PMID: 38283805 PMCID: PMC10818257 DOI: 10.1016/j.sjbs.2023.103920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Multiple Myeloma (MM) is a plasma cell cancer with high mortality and morbidity rates. Its incidence rate has increased by 143% since 1975. Adipokines, cytokines, chemokines, and genetic variations influence the development and progression of MM. Chromosomal translocations cause mutations associated with MM. The pathogenesis of MM is complicated by novel issues like miRNAs, RANKL, Wnt/DKK1, Wnt, and OPG. Conventional diagnosis methods include bone marrow biopsy, sPEP or uPEP, sIFE and uIFE, and sFLC assay, along with advanced techniques such as FISH, SNPA, and gene expression technologies. A novel therapeutic strategy has been developed recently. Chemotherapy, hematopoietic stem cell transplantation, and a variety of drug classes in combination are used to treat patients with high-risk diseases. Alkylating agents, PIs, and IMiDs have all been developed as effective treatment options for MM in recent years. This review overviews the current recommendations for managing MGUS, SMM, MM, SP and NSMM and discusses practices in diagnosing and treating MM.
Collapse
Affiliation(s)
- Maisa Siddiq Abduh
- Immune Responses in Different Diseases Research Group, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Fu XH, Guan RY, Huang Z, Li Y, Lu G, Mou WW, Du J. From Multiple Myeloma to Acute Myeloid Leukemia: A Case Report of a 61-year-old Woman after 8 Years of Chemotherapy and Immunotherapy. Recent Pat Anticancer Drug Discov 2024; 19:396-401. [PMID: 38214323 DOI: 10.2174/1574892818666230619093300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/28/2023] [Accepted: 05/17/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND As the second most prevalent hematologic malignancy, multiple myeloma (MM) affects plasma cells and is characterized by chromosomal abnormalities, particularly involving the immunoglobulin heavy chain switch region. MM represents a biologically and clinically heterogeneous hematological malignancy that serves as a clonal evolution model, exhibiting clonal heterogeneity throughout all stages from monoclonal gammopathy undetermined significance (MGUS) and smoldering multiple myeloma (SMM) to MM. Although significant progress has been made in the treatment of MM, leading to improved patient outcomes, concerns are arising regarding disease relapse due to the presence and selection of pre-existing resistant clones or selective pressure during therapy. CASE PRESENTATION We present a case of multiple myeloma (MM) in a female patient, who underwent an 8-year course of treatment, including chemotherapy, immunomodulators, hematopoietic stem cell transplantation, CD38 monoclonal antibody, and chimeric antigen receptor T-cell (CAR-T), and was recently diagnosed with concurrent progressive MM and acute myeloid leukemia (AML). This patient has witnessed the evolution of MM treatment paradigms. CONCLUSION In this course, disease relapses occurred twice, one of which was manifested by a light chain escape (LCE). Moreover, through the course of the disease in this patient, we review the process of clonal evolution that may be relevant.
Collapse
Affiliation(s)
- Xue-Hang Fu
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Rong-Yan Guan
- Department of Hematology, Aviation General Hospital, Beijing, 100012, China
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yun Li
- Department of Hematology, Aviation General Hospital, Beijing, 100012, China
| | - Guang Lu
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, 257099, China
| | - Wei-Wei Mou
- Department of Pediatrics, Shengli Oilfield Central Hospital, Dongying, 257034, China
| | - Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
13
|
Liu Y, Li B, Chen X, Xiong H, Huang C. The effect of immunomodulatory drugs on bone metabolism of patients with multiple myeloma. Expert Rev Hematol 2024; 17:47-54. [PMID: 38319240 DOI: 10.1080/17474086.2024.2316090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/05/2024] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Immunomodulatory drugs (IMiDs) are widely used in the management of newly diagnosed and relapsed/refractory multiple myeloma patients. These agents show their potential effect on myeloma bone disease (MBD), including inhibition of osteoclasts activity and effects on osteoblasts differentiation. It is unclear whether these effects are direct, which may have an impact on bone formation markers when combined with proteasome inhibitors. AREAS COVERED This review summarizes the available evidence on the role of IMiDs in microenvironment regulation and their potential effects on bone metabolism. The literature search methodology consisted of searching PubMed for basic and clinical trials using medical subject terms. Included articles were screened and evaluated by the coauthors of this review. EXPERT OPINION As a therapeutic option, IMiDs directly affect preosteoblast/osteoclast differentiation. The combination of proteasome inhibitors may counteract the short-term up-regulation of osteogenic activity markers, and therefore intravenous zoledronic acid is recommended, however, obtaining a more significant myeloma response will have a long-term positive impact on myeloma bone disease.
Collapse
Affiliation(s)
- Yang Liu
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Li
- Department of Intensive Care Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaomin Chen
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Xiong
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunlan Huang
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
14
|
Perez de Acha O, Reiman L, Jayabalan DS, Walker ZJ, Bosma G, Keller AL, Parzych SE, Abbott D, Idler BM, Ribadeneyra D, Niesvizky R, Forsberg PA, Mark TM, Sherbenou DW. CD38 antibody re-treatment in daratumumab-refractory multiple myeloma after time on other therapies. Blood Adv 2023; 7:6430-6440. [PMID: 37648670 PMCID: PMC10598487 DOI: 10.1182/bloodadvances.2023010162] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/03/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Monoclonal antibodies targeting CD38 are important for treatment of both newly diagnosed and relapsed multiple myeloma (MM). Daratumumab and isatuximab are anti-CD38 antibodies with the US Food and Drugs Administration approval in multiple different combinations. Despite good initial efficacy, patients inevitably develop drug resistance. Whether patients can be effectively re-treated with these antibodies in subsequent lines of therapy is unclear. Thus far, studies have mostly been limited to clinical retrospectives with short washout periods. To answer whether patients regain sensitivity after longer washouts, we used ex vivo sensitivity testing to isolate the anti-CD38 antibody-specific cytotoxicity in samples obtained from patients who had been exposed to and then off daratumumab for up to 53 months. MM cells from patients who had been off daratumumab for >1 year showed greater sensitivity than those with <1 year, although they still were less sensitive than those who were daratumumab naïve. CD38 expression on MM cells gradually recovered, although, again, not to the level of anti-CD38 antibody-naïve patients. Interestingly, low MM CD38 explained only 45% of cases identified to have daratumumab resistance. With clinical follow-up, we found ex vivo sensitivity predicted subsequent clinical response but CD38 overexpression did not. Patients clinically re-treated with anti-CD38 antibodies had <6 months of clinical benefit, but 1 patient who was daratumumab exposed but not refractory achieved complete response lasting 13 months. We conclude that transient efficacy can be achieved by waiting 1 year before CD38 antibody rechallenge, but this approach may be best used as a bridge to, or after, chimeric antigen receptor T-cell therapy.
Collapse
Affiliation(s)
- Olivia Perez de Acha
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Lauren Reiman
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - David S. Jayabalan
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York-Presbyterian, New York City, NY
| | - Zachary J. Walker
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Grace Bosma
- Department of Biostatistics and Informatics, Center for Innovative Design and Analysis, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Alana L. Keller
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Sarah E. Parzych
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Diana Abbott
- Department of Biostatistics and Informatics, Center for Innovative Design and Analysis, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Beau M. Idler
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Drew Ribadeneyra
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York-Presbyterian, New York City, NY
| | - Ruben Niesvizky
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York-Presbyterian, New York City, NY
| | - Peter A. Forsberg
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tomer M. Mark
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Daniel W. Sherbenou
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
15
|
Osada N, Kikuchi J, Iha H, Yasui H, Ikeda S, Takahashi N, Furukawa Y. c-FOS is an integral component of the IKZF1 transactivator complex and mediates lenalidomide resistance in multiple myeloma. Clin Transl Med 2023; 13:e1364. [PMID: 37581569 PMCID: PMC10426395 DOI: 10.1002/ctm2.1364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/28/2023] [Accepted: 08/06/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND The immunomodulatory drug lenalidomide, which is now widely used for the treatment of multiple myeloma (MM), exerts pharmacological action through the ubiquitin-dependent degradation of IKZF1 and subsequent down-regulation of interferon regulatory factor 4 (IRF4), a critical factor for the survival of MM cells. IKZF1 acts principally as a tumour suppressor via transcriptional repression of oncogenes in normal lymphoid lineages. In contrast, IKZF1 activates IRF4 and other oncogenes in MM cells, suggesting the involvement of unknown co-factors in switching the IKZF1 complex from a transcriptional repressor to an activator. The transactivating components of the IKZF1 complex might promote lenalidomide resistance by residing on regulatory regions of the IRF4 gene to maintain its transcription after IKZF1 degradation. METHODS To identify unknown components of the IKZF1 complex, we analyzed the genome-wide binding of IKZF1 in MM cells using chromatin immunoprecipitation-sequencing (ChIP-seq) and screened for the co-occupancy of IKZF1 with other DNA-binding factors on the myeloma genome using the ChIP-Atlas platform. RESULTS We found that c-FOS, a member of the activator protein-1 (AP-1) family, is an integral component of the IKZF1 complex and is primarily responsible for the activator function of the complex in MM cells. The genome-wide screening revealed the co-occupancy of c-FOS with IKZF1 on the regulatory regions of IKZF1-target genes, including IRF4 and SLAMF7, in MM cells but not normal bone marrow progenitors, pre-B cells or mature T-lymphocytes. c-FOS and IKZF1 bound to the same consensus sequence as the IKZF1 complex through direct protein-protein interactions. The complex also includes c-JUN and IKZF3 but not IRF4. Treatment of MM cells with short-hairpin RNA against FOS or a selective AP-1 inhibitor significantly enhanced the anti-MM activity of lenalidomide in vitro and in two murine MM models. Furthermore, an AP-1 inhibitor mitigated the lenalidomide resistance of MM cells. CONCLUSIONS C-FOS determines lenalidomide sensitivity and mediates drug resistance in MM cells as a co-factor of IKZF1 and thus, could be a novel therapeutic target for further improvement of the prognosis of MM patients.
Collapse
Affiliation(s)
- Naoki Osada
- Division of Stem Cell RegulationCenter for Molecular MedicineJichi Medical UniversityTochigiJapan
| | - Jiro Kikuchi
- Division of Stem Cell RegulationCenter for Molecular MedicineJichi Medical UniversityTochigiJapan
| | - Hidekatsu Iha
- Division of PathophysiologyThe Research Center for GLOBAL and LOCAL Infectious Diseases (RCGLID)Oita UniversityOitaJapan
| | - Hiroshi Yasui
- Division of Hematology and Oncology, Department of Internal MedicineSt. Marianna University School of MedicineKanagawaJapan
- Project Division of Innovative Diagnostics Technology Platform, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Sho Ikeda
- Department of HematologyNephrology and RheumatologyAkita University Graduate School of MedicineAkitaJapan
| | - Naoto Takahashi
- Department of HematologyNephrology and RheumatologyAkita University Graduate School of MedicineAkitaJapan
| | - Yusuke Furukawa
- Division of Stem Cell RegulationCenter for Molecular MedicineJichi Medical UniversityTochigiJapan
- Center for Medical EducationTeikyo University of ScienceTokyoJapan
| |
Collapse
|
16
|
Wang X, Wang X, Zhao Y, Qi Z. LY103, a pomalidomide derivative, alleviates taxol resistance in NSCLC via energy metabolism crosstalk and tumor microenvironment intervention. Bioorg Chem 2023; 136:106558. [PMID: 37105001 DOI: 10.1016/j.bioorg.2023.106558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
In this study, we identified HIF 1α as a potential target for reversing taxol resistance in lung cancer by combining bioinformatics analysis with pharmacological analysis. Furthermore, pomalidomide derivative LY103 was also be synthesized by introducing an isatin analogue into the amino terminal ofpomalidomide, and it has a broad antitumor spectrum and showed excellent activity against A549/Taxol cells (IC50 = 6.33 ± 0.51 μM). The results of molecular docking showed that not only LY103 was inclined to bind to HIF 1α stably, it could also form multiple hydrogen bonds with VAL376, ASP256, ILE454, and GLU455 of HIF 1α even was reduced to LY103-NH2 by nitroreductase, which was further stabilized the complex formed by them, thereby inhibiting the activity of HIF 1α. LY103 was able to significantly induce DNA damage and inhibit angiogenesis. Concurrently, LY103 activated the immune response, reduced the expression of cytokines TNF-α, IL-6, and IL-1β, thus might be inhibit the proliferation and metastasis of tumor cells. Pharmacological analysis proved that LY103 led to cell apoptosis through the mitochondrial pathway, and its combination with taxol significantly promoted this process. In general, the consumption of glutathione, the crosstalk of energy metabolism, and the improvement of the tumor microenvironment caused by LY103 eventually led to the decrease of ABCC1 protein expression and the drug resistance was reversed. The rational design of LY103 provided a basis for the application of nitro compounds in the treatment of hypoxic tumors and the reversal of taxol resistance.
Collapse
Affiliation(s)
- Xing Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xiaohan Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yongfei Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Zhengjian Qi
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
17
|
Guinn BA, Schuler PJ, Schrezenmeier H, Hofmann S, Weiss J, Bulach C, Götz M, Greiner J. A Combination of the Immunotherapeutic Drug Anti-Programmed Death 1 with Lenalidomide Enhances Specific T Cell Immune Responses against Acute Myeloid Leukemia Cells. Int J Mol Sci 2023; 24:ijms24119285. [PMID: 37298237 DOI: 10.3390/ijms24119285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Immune checkpoint inhibitors can block inhibitory molecules on the surface of T cells, switching them from an exhausted to an active state. One of these inhibitory immune checkpoints, programmed cell death protein 1 (PD-1) is expressed on T cell subpopulations in acute myeloid leukemia (AML). PD-1 expression has been shown to increase with AML progression following allo-haematopoeitic stem cell transplantation, and therapy with hypomethylating agents. We have previously shown that anti-PD-1 can enhance the response of leukemia-associated antigen (LAA)-specific T cells against AML cells as well as leukemic stem and leukemic progenitor cells (LSC/LPCs) ex vivo. In concurrence, blocking of PD-1 with antibodies such as nivolumab has been shown to enhance response rates post-chemotherapy and stem cell transplant. The immune modulating drug lenalidomide has been shown to promote anti-tumour immunity including anti-inflammatory, anti-proliferative, pro-apoptotic and anti-angiogenicity. The effects of lenalidomide are distinct from chemotherapy, hypomethylating agents or kinase inhibitors, making lenalidomide an attractive agent for use in AML and in combination with existing active agents. To determine whether anti-PD-1 (nivolumab) and lenalidomide alone or in combination could enhance LAA-specific T cell immune responses, we used colony-forming immune and ELISpot assays. Combinations of immunotherapeutic approaches are believed to increase antigen-specific immune responses against leukemic cells including LPC/LSCs. In this study we used a combination of LAA-peptides with the immune checkpoint inhibitor anti-PD-1 and lenalidomide to enhance the killing of LSC/LPCs ex vivo. Our data offer a novel insight into how we could improve AML patient responses to treatment in future clinical studies.
Collapse
Affiliation(s)
- Barbara-Ann Guinn
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | - Patrick J Schuler
- Department of Otorhinolaryngology, University Hospital Ulm, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm and German Red Cross, 89073 Ulm, Germany
| | - Susanne Hofmann
- Department of Internal Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Johanna Weiss
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany
| | - Christiane Bulach
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany
| | - Marlies Götz
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany
| | - Jochen Greiner
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany
- Department of Internal Medicine, Diakonie Hospital Stuttgart, 70176 Stuttgart, Germany
| |
Collapse
|
18
|
Beksac M, Seval GC, Koyun D, Topcuoglu P, Yuksel MK, Gurman G, Ilhan O. Generic Lenalidomide Rivelime Versus Brand-name Revlimid® in the Treatment of Relapsed/Refractory Multiple Myeloma: A Retrospective Single-center Experience on Efficacy, Safety and Survival Outcome. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e164-e170. [PMID: 36610852 DOI: 10.1016/j.clml.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND This study aimed to compare use of original brand-name lenalidomide (Revlimid®) vs. generic equivalent (Rivelime®) in terms of efficacy, safety and survival outcome in patients with relapsed/refractory multiple myeloma (RRMM) PATIENTS AND METHODS: A total of 184 patients RRMM (median age: 62 years, 60.9% were males) who received singlet, doublet or triplet lenalidomide-containing regimens including either Revlimid® (n=74) or Rivelime® (n=110) were included in this study. Treatment response was based on evaluation of objective response to treatment (ORR) including the sum of patients who achieved partial response (PR), very good partial responses (VGPR) or complete response (CR) to therapy. Progression-free survival (PFS), overall survival (OS) and safety data were also recorded. RESULTS Revlimid® and Rivelime® groups were similar in terms of ORR (54.1 vs. 60.0%), CR (22.5 vs. 28.8%), VGPR (55.0 vs. 50.0%) and PR (22.5 vs. 21.2%) rates. Median (SE) PFS time were similar between Rivelime® vs. Revlimid® treated patients who were in the 2nd line (30.3(3.8) vs. 22.7(7.0) months, p=0.827) or 3rd line of therapy (38.1(12.1) vs. 20.1(0.9) months, p=0.147) at lenalidomide initiation. Two groups also had similar OS rate (83.8 vs. 73.6%) and OS time (mean 122.3 vs. 123.5 months). Side effects were manageable in both groups. CONCLUSION In conclusion, replacing Revlimid® with its generic version Rivelime® in singlet, doublet or triplet lenalidomide containing RRMM regimens seems not to compromise the efficacy of treatment, and to yield a similarly improved response rates and survival outcome and no additional toxic effects, enabling a long-term therapy.
Collapse
Affiliation(s)
- Meral Beksac
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey.
| | | | - Derya Koyun
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Pervin Topcuoglu
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Meltem-Kurt Yuksel
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Gunhan Gurman
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Osman Ilhan
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
19
|
Guo H, Yang J, Wang H, Liu X, Liu Y, Zhou K. Reshaping the tumor microenvironment: The versatility of immunomodulatory drugs in B-cell neoplasms. Front Immunol 2022; 13:1017990. [PMID: 36311747 PMCID: PMC9596992 DOI: 10.3389/fimmu.2022.1017990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) such as thalidomide, lenalidomide and pomalidomide are antitumor compounds that have direct tumoricidal activity and indirect effects mediated by multiple types of immune cells in the tumor microenvironment (TME). IMiDs have shown remarkable therapeutic efficacy in a set of B-cell neoplasms including multiple myeloma, B-cell lymphomas and chronic lymphocytic leukemia. More recently, the advent of immunotherapy has revolutionized the treatment of these B-cell neoplasms. However, the success of immunotherapy is restrained by immunosuppressive signals and dysfunctional immune cells in the TME. Due to the pleiotropic immunobiological properties, IMiDs have shown to generate synergetic effects in preclinical models when combined with monoclonal antibodies, immune checkpoint inhibitors or CAR-T cell therapy, some of which were successfully translated to the clinic and lead to improved responses for both first-line and relapsed/refractory settings. Mechanistically, despite cereblon (CRBN), an E3 ubiquitin ligase, is considered as considered as the major molecular target responsible for the antineoplastic activities of IMiDs, the exact mechanisms of action for IMiDs-based TME re-education remain largely unknown. This review presents an overview of IMiDs in regulation of immune cell function and their utilization in potentiating efficacy of immunotherapies across multiple types of B-cell neoplasms.
Collapse
Affiliation(s)
| | | | | | | | | | - Keshu Zhou
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
20
|
Li B, Hu M, Chen C, Yin H, Deng Y, Li H, Zhang J, He L. Synthesis and antitumor activity of a series of novel N-aryl-5-(2,2,2-trifluoroethoxy)-1,5-dihydro-2H-pyrrol-2-ones derivatives. Bioorg Med Chem Lett 2022; 73:128919. [PMID: 35931243 DOI: 10.1016/j.bmcl.2022.128919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022]
Abstract
With the help of the establishment of novel reaction methodology, a series of N-Aryl-5-(2,2,2-trifluoroethoxy)-1,5-dihydro-2H-pyrrol-2-one conjugates were designed and synthesized in 2-4 steps, and subsequent anticancer activity of these compounds was evaluated. Preliminary results showed that these compounds have moderate to potent activities against human acute leukemia cells K562, human lung cancer A549, human breast cancer MDA-MB-231, and human cervical cancer HeLa cancer cell lines. Among them, compounds 2d and 2k were the most potent against K562 cell line with IC50 values of 0.07 and 0.52 µM, respectively, and the toxicity of 2d to the normal of hepatocytes (LO2) cell line was low (the survival rate 81 %). Flow cytometry analysis showed that 2d arrested K562 cells in the G2/M phase potently, even much better than Combretastatin A4 (CA4). In addition, the results demonstrated the involvement of the caspase-dependent or independent pathways of apoptosis, evidenced by the upregulation of FADD, pro-caspase 3, cleaved-caspase 3, HTRA2/Omi, SMAC/Diablo and the ratio of Bax/Bcl-2.The biological effects founding of 2d in this work point to prospective uses against acute leukemia.
Collapse
Affiliation(s)
- Bin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mingli Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chen Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Honglu Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yan Deng
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Haibo Li
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jing Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University.
| | - Ling He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
21
|
Yang Y, Chen R, Gong Y, Yang W, Li K, Fan W, Gou S, Gao P, He T, Cai K. Double-drug loading upconversion nanoparticles for monitoring and therapy of a MYC/BCL6-positive double-hit diffuse large B-cell lymphoma. Biomaterials 2022; 287:121607. [PMID: 35696785 DOI: 10.1016/j.biomaterials.2022.121607] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 05/14/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a systemic hematological malignancy. Herein, through whole exome sequencing (WES), we found that DLBCL genome changes and expression characteristics are associated with various immune cells. Lenalidomide (Len) is a leading candidate for the immunomodulatory treatment of multiple myeloma in the clinic. Inspired by lenalidomide as an immunomodulatory drug for the treatment of multiple myeloma, we constructed a multifunctional nanoplatform with therapeutic and imaging properties for DLBCL by co-loading lenalidomide and dexamethasone (Dex) with upconversion nanoparticles using a GSH-sensitive linker (named as UCNPs-Len-Dex). In vitro cell experiments proved that the UCNPs-Len-Dex had good biocompatibility and obvious antitumor efficacy. UCNPs-Len-Dex also exhibited excellent anti-tumor efficacy and imaging properties in vivo. RNA sequencing showed that UCNPs-Len-Dex targeted and activated the E3 ligase of CRBN, resulting in IKZF1/3 degradation, which inhibited MYC/BCL6-positive DLBCL and maintained the stability of the immune microenvironment. Therefore, this study provided a new monitoring and therapeutic synergetic strategy for DLBCL.
Collapse
Affiliation(s)
- Yulu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Rui Chen
- Department of Pathology, Chongqing Cancer Institute/Hospital, Chongqing, 400030, China
| | - Yi Gong
- Department of Phase I Clinical Trial Ward, Chongqing Cancer Institute/Hospital, Chongqing, 400030, China.
| | - Weihu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Ke Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wuzhe Fan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shuangquan Gou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Pengfei Gao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tingting He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
22
|
Rinaldi I, Muthalib A, Edina BC, Wiyono L, Winston K. Role of Anti-B-Cell Maturation Antigen (BCMA) in the Management of Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14143507. [PMID: 35884566 PMCID: PMC9317279 DOI: 10.3390/cancers14143507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Multiple myeloma is the most prevalent hematological cancer, and further treatments for this disease are required. Despite progress in the development of treatment regimens, multiple myeloma is still an incurable disease because of its poor response to therapy and high rate of resistance to treatment. However, anti-BCMA (B-cell maturation antigen) has shown promise in the treatment of multiple myeloma, and it may have the potential to be a new first-line treatment for patients. Thus, in this review, we objectively discussed the treatment potential of anti-BCMA, its mechanisms, and its future clinical implications for multiple myeloma patients. Abstract Over the past few decades, treatment options have become more advanced for multiple myeloma (MM), one of the most prevalent hematological cancers; however, multiple myeloma remains an incurable disease due to its poor response to therapy and high rates of resistance, which cause relapsed/refractory or multiple myeloma. Researchers have described anti-BCMA (B-cell maturation antigen) as a promising treatment regimen that targets the BCMA biomarker in the affected plasma cells. BCMA is a protein that is specifically expressed in plasma-cell neoplasms by using several mechanisms, such as CAR T cells (Chimeric Antigen Receptor T cells), antibody-drug conjugates, and bispecific T-cell engagers, thus allowing for a rapid response in the treatment of resistant or relapsed/refractory multiple myeloma patients. Anti-BCMA treatment is novel and specific in its mechanisms of action, with noninferior complete responses, higher overall survival rates, and fewer reported adverse events compared to other currently available treatment of MM. In this review, we compared anti-BCMA mechanisms with those of previously available therapies, such as those using immunomodulators and proteasome inhibitors, and discussed the advantages of using anti-BCMA as a potential first-line treatment for multiple myeloma patients.
Collapse
Affiliation(s)
- Ikhwan Rinaldi
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Ciptomangunkusumo General Hospital—Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
- Correspondence: ; Tel.: +62-811-1001-758
| | - Abdul Muthalib
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Ciptomangunkusumo General Hospital—Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Brenda Cristie Edina
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia; (B.C.E.); (L.W.); (K.W.)
| | - Lowilius Wiyono
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia; (B.C.E.); (L.W.); (K.W.)
| | - Kevin Winston
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia; (B.C.E.); (L.W.); (K.W.)
- Hospital Medicine, Bhakti Medicare Hospital, Cicurug 43359, Indonesia
| |
Collapse
|
23
|
Weisel K, Nooka AK, Terpos E, Spencer A, Goldschmidt H, Dirnberger F, DeCosta L, Yusuf A, Kumar S. Carfilzomib 56 mg/m 2 twice-weekly in combination with dexamethasone and daratumumab (KdD) versus daratumumab in combination with bortezomib and dexamethasone (DVd): a matching-adjusted indirect treatment comparison. Leuk Lymphoma 2022; 63:1887-1896. [DOI: 10.1080/10428194.2022.2047962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Katja Weisel
- Department of Oncology and Hematology, University Medical Center of Hamburg-Eppendorf, Hamburg, Germany
| | - Ajay K. Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Andrew Spencer
- Australian Centre for Blood Diseases, Monash University, Malignant Haematology & Stem Cell Transplantation, Alfred Hospital and Department of Clinical Haematology, Monash University, Melbourne, Australia
| | - Hartmut Goldschmidt
- Internal Medicine V and National Center of Tumor Diseases, University Clinic Heidelberg, Heidelberg, Germany
| | | | - Lucy DeCosta
- Global Biostatistical Science, Amgen Ltd., Uxbridge, UK
| | - Akeem Yusuf
- Global Medical Affairs, Amgen Inc., Thousand Oaks, CA, USA
| | - Shaji Kumar
- Department of Internal Medicine, Division of Hematology, Mayo Clinic Rochester, Rochester, MN, USA
| |
Collapse
|
24
|
Invasive Orbital Squamous Cell Carcinoma in a Patient with Multiple Myeloma. Case Rep Ophthalmol Med 2022; 2022:8585692. [PMID: 35815062 PMCID: PMC9259366 DOI: 10.1155/2022/8585692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/03/2022] [Accepted: 06/18/2022] [Indexed: 11/18/2022] Open
Abstract
Background Orbital squamous cell carcinoma (SCC) is a rare entity. It is often a result of local invasion of SCC originating from the skin, nasopharynx, nasal cavity, paranasal sinuses, conjunctiva, lacrimal glands, or sac or less commonly occurs through hematogenous metastasis. Herein, we report a patient with orbital SCC with a history of multiple myeloma (MM). Case presentation. A 45-year-old woman with a history of MM in the past two years presented to our clinic complaining of gradual right eye proptosis for six months. The relative afferent pupillary defect was detected in the right eye on her examination. Ocular movements of the right eye were limited in all directions. Orbital magnetic resonance imaging demonstrated an infiltrative mass in the right orbit extended from the anterior to the orbital apex and the optic canal. The patient underwent debulking, and a histopathology examination revealed SCC results. No other secondary site was found to be the origin of the tumor. Result The patient underwent chemotherapy and subsequent radiotherapy. To our knowledge, this is the first report of concomitant MM and primary orbital SCC.
Collapse
|
25
|
Gutowska A, McKinnon K, Sarkis S, Doster MN, Bissa M, Moles R, Stamos JD, Rahman MA, Washington-Parks R, Davis D, Yarchoan R, Franchini G, Pise-Masison CA. Transient Viral Activation in Human T Cell Leukemia Virus Type 1-Infected Macaques Treated With Pomalidomide. Front Med (Lausanne) 2022; 9:897264. [PMID: 35602479 PMCID: PMC9119179 DOI: 10.3389/fmed.2022.897264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/11/2022] [Indexed: 12/31/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) persists in the host despite a vigorous immune response that includes cytotoxic T cells (CTL) and natural killer (NK) cells, suggesting the virus has developed effective mechanisms to counteract host immune surveillance. We recently showed that in vitro treatment of HTLV-1-infected cells with the drug pomalidomide (Pom) increases surface expression of MHC-I, ICAM-1, and B7-2, and significantly increases the susceptibility of HTLV-1-infected cells to NK and CTL killing, which is dependent on viral orf-I expression. We reasoned that by restoring cell surface expression of these molecules, Pom treatment has the potential to reduce virus burden by rendering infected cells susceptible to NK and CTL killing. We used the rhesus macaque model to determine if Pom treatment of infected individuals activates the host immune system and allows recognition and clearance of HTLV-1-infected cells. We administered Pom (0.2 mg/kg) orally to four HTLV-1-infected macaques over a 24 day period and collected blood, urine, and bone marrow samples throughout the study. Pom treatment caused immune activation in all four animals and a marked increase in proliferating CD4+, CD8+, and NK cells as measured by Ki-67+ cells. Activation markers HLA-DR, CD11b, and CD69 also increased during treatment. While we detected an increased frequency of cells with a memory CD8+ phenotype, we also found an increased frequency of cells with a Treg-like phenotype. Concomitant with immune activation, the frequency of detection of viral DNA and the HTLV-1-specific humoral response increased as well. In 3 of 4 animals, Pom treatment resulted in increased antibodies to HTLV-1 antigens as measured by western blot and p24Gag ELISA. Consistent with Pom inducing immune and HTLV-1 activation, we measured elevated leukotrienes LTB4 and LTE4 in the urine of all animals. Despite an increase in plasma LTB4, no significant changes in plasma cytokine/chemokine levels were detected. In all cases, however, cellular populations, LTB4, and LTE4 decreased to baseline or lower levels 2 weeks after cessation of treatment. These results indicated that Pom treatment induces a transient HTLV-1-specific immune activation in infected individuals, but also suggest Pom may not be effective as a single-agent therapeutic.
Collapse
Affiliation(s)
- Anna Gutowska
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Białystok, Białystok, Poland
| | - Katherine McKinnon
- Vaccine Branch Flow Cytometry Core, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sarkis Sarkis
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Melvin N. Doster
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Massimiliano Bissa
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Ramona Moles
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - James D. Stamos
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Mohammad Arif Rahman
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Robyn Washington-Parks
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - David Davis
- HIV and AIDS Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Robert Yarchoan
- HIV and AIDS Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Cynthia A. Pise-Masison
- Animal Models and Retroviral Vaccine Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
- *Correspondence: Cynthia A. Pise-Masison,
| |
Collapse
|
26
|
Choi T, Kang Y. Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma. Pharmacol Ther 2022; 232:108007. [PMID: 34582835 PMCID: PMC8930424 DOI: 10.1016/j.pharmthera.2021.108007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022]
Abstract
Although treatment outcomes of multiple myeloma patients have improved significantly during the last two decades, myeloma is still an incurable disease. There are newly emerging immunotherapies to treat multiple myeloma including monoclonal antibodies, antibody-drug conjugate, bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapy. Impressive response rate and clinical efficacy in heavily pretreated myeloma patients led to the FDA approval of the first myeloma CAR-T therapy in March 2021. Among many different targets for myeloma CAR-T therapies, B Cell Maturation Antigen (BCMA) has been the most successful target so far, but other targets which can be used either for single-target or dual-target CAR-T's are actively being explored. Clinical efficacy and safety of current myeloma CAR-T therapies will be presented here. Potential mechanisms leading to resistance include clearance of CAR-T cells, antigenic escape, and immunosuppressive tumor microenvironment. Novel strategies to enhance myeloma CAR-T will also be described. In this article, we provide a comprehensive review of the current data and the future directions of myeloma CAR-T therapies.
Collapse
Affiliation(s)
- Taewoong Choi
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
27
|
Richardson PG, Schjesvold F, Weisel K, Moreau P, Anderson LD, White D, Rodriguez-Otero P, Sonneveld P, Engelhardt M, Jenner M, Corso A, Dürig J, Pavic M, Salomo M, Beksac M, Oriol A, Lindsay J, Liberati AM, Galli M, Robak P, Larocca A, Yagci M, Vural F, Kanate AS, Jiang R, Grote L, Peluso T, Dimopoulos M. Pomalidomide, bortezomib, and dexamethasone at first relapse in lenalidomide-pretreated myeloma: A subanalysis of OPTIMISMM by clinical characteristics. Eur J Haematol 2021; 108:73-83. [PMID: 34496096 PMCID: PMC9293199 DOI: 10.1111/ejh.13706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 01/17/2023]
Abstract
Objective We evaluated the efficacy and safety of pomalidomide, bortezomib, and dexamethasone (PVd) vs bortezomib and dexamethasone (Vd) by age, renal function, and high‐risk cytogenetic abnormalities in lenalidomide‐pretreated patients with multiple myeloma at first relapse. Methods OPTIMISMM was a phase 3, multicenter, open‐label, randomized study (NCT01734928; N = 559). The primary endpoint was progression‐free survival (PFS). Results Overall, 226 patients had received one prior line of therapy. PVd significantly prolonged PFS vs Vd in patients aged ≤65 years (median, 22.0 vs 13.1 months; P = .0258) and >65 years (median, 17.6 vs 9.9 months; P = .0369). Median PFS in patients with renal impairment (RI; creatinine clearance <60 mL/min) was 15.1 months with PVd vs 9.5 months with Vd (hazard ratio [HR], 0.67 [95% CI, 0.34‐1.34]). In patients without RI, median PFS was 22.0 vs 13.1 months (HR, 0.45 [95% CI, 0.27‐0.76]). In patients with high‐risk cytogenetics, median PFS was 14.7 vs 9.9 months (HR, 0.39 [95% CI, 0.13‐1.17]). PVd significantly improved overall response rate vs Vd in all subgroups. The safety profile of PVd was consistent with previous reports. Conclusions These findings confirmed the benefits of PVd at first relapse, including in patients with poor prognostic factors.
Collapse
Affiliation(s)
- Paul G Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Fredrik Schjesvold
- Oslo Myeloma Center at Oslo University Hospital, KG Jebsen Center for B-Cell Malignancies, University of Oslo, Oslo, Norway
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Larry D Anderson
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Darrell White
- Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| | | | | | | | | | | | - Jan Dürig
- University Hospital Essen, Essen, Germany
| | - Michel Pavic
- Centre Hospitalier Universitaire De Sherbrooke (CHUS), Centre de Recherche Clinique Etienne-Le Bel (CRCELB) Hopital Fleurimont, Sherbrooke, QC, Canada
| | - Morten Salomo
- Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Albert Oriol
- Institut Català d'Oncologia I Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Jindriska Lindsay
- East Kent Hospitals University NHS Foundation Trust, Kent and Canterbury Hospital, Canterbury, UK
| | | | - Monica Galli
- Ospedale Papa Giovanni XXIII, U.O. di Ematologia, Ispedali Riuniti di Bergamo, Bergamo, Italy
| | | | | | - Munci Yagci
- Gazi University Medical Faculty, Ankara, Turkey
| | | | | | - Ruiyun Jiang
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Lara Grote
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Teresa Peluso
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | | |
Collapse
|
28
|
Barilà G, Pavan L, Vedovato S, Berno T, Lo Schirico M, Arangio Febbo M, Teramo A, Calabretto G, Vicenzetto C, Gasparini VR, Fregnani A, Manni S, Trimarco V, Carraro S, Facco M, Piazza F, Semenzato G, Zambello R. Treatment Induced Cytotoxic T-Cell Modulation in Multiple Myeloma Patients. Front Oncol 2021; 11:682658. [PMID: 34211851 PMCID: PMC8239308 DOI: 10.3389/fonc.2021.682658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/10/2021] [Indexed: 11/15/2022] Open
Abstract
The biology of plasma cell dyscrasias (PCD) involves both genetic and immune-related factors. Since genetic lesions are necessary but not sufficient for Multiple Myeloma (MM) evolution, several authors hypothesized that immune dysfunction involving both B and T cell counterparts plays a key role in the pathogenesis of the disease. The aim of this study is to evaluate the impact of cornerstone treatments for Multiple Myeloma into immune system shaping. A large series of 976 bone marrow samples from 735 patients affected by PCD was studied by flow analysis to identify discrete immune subsets. Treated MM samples displayed a reduction of CD4+ cells (p<0.0001) and an increase of CD8+ (p<0.0001), CD8+/DR+ (p<0.0001) and CD3+/CD57+ (p<0.0001) cells. Although these findings were to some extent demonstrated also following bortezomib treatment, a more pronounced cytotoxic polarization was shown after exposure to autologous stem cell transplantation (ASCT) and Lenalidomide (Len) treatment. As a matter of fact, samples of patients who received ASCT (n=110) and Len (n=118) were characterized, towards untreated patients (n=138 and n=130, respectively), by higher levels of CD8+ (p<0.0001 and p<0.0001, respectively), CD8+/DR+ (p=0.0252 and p=0.0001, respectively) and CD3+/CD57+ cells (p<0.0001 and p=0.0006, respectively) and lower levels of CD4+ lymphocytes (p<0.0001 and p=0.0005, respectively). We demonstrated that active MM patients are characterized by a relevant T cell modulation and that most of these changes are therapy-related. Current Myeloma treatments, notably ASCT and Len treatments, polarize immune system towards a dominant cytotoxic response, likely contributing to the anti-Myeloma effect of these regimens.
Collapse
Affiliation(s)
- Gregorio Barilà
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Laura Pavan
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Susanna Vedovato
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Tamara Berno
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Mariella Lo Schirico
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Massimiliano Arangio Febbo
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Antonella Teramo
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Giulia Calabretto
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Cristina Vicenzetto
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Vanessa Rebecca Gasparini
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Anna Fregnani
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Sabrina Manni
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Valentina Trimarco
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Samuela Carraro
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Monica Facco
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| | - Renato Zambello
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padua University School of Medicine, Padova, Italy
| |
Collapse
|
29
|
Ancidoni A, Bacigalupo I, Remoli G, Lacorte E, Piscopo P, Sarti G, Corbo M, Vanacore N, Canevelli M. Anticancer drugs repurposed for Alzheimer's disease: a systematic review. ALZHEIMERS RESEARCH & THERAPY 2021; 13:96. [PMID: 33952306 PMCID: PMC8101105 DOI: 10.1186/s13195-021-00831-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/19/2021] [Indexed: 12/28/2022]
Abstract
Background The relationship between cancer and dementia is triggering growing research interest. Several preclinical studies have provided the biological rationale for the repurposing of specific anticancer agents in Alzheimer’s disease (AD), and a growing number of research protocols are testing their efficacy and safety/tolerability in patients with AD. Methods The aim of the present systematic review was to provide an overview on the repurposing of approved anticancer drugs in clinical trials for AD by considering both ongoing and completed research protocols in all phases. In parallel, a systematic literature review was conducted on PubMed, ISI Web, and the Cochrane Library to identify published clinical studies on repurposed anticancer agents in AD. Results Based on a structured search on the ClinicalTrials.gov and the EudraCT databases, we identified 13 clinical trials testing 11 different approved anticancer agents (five tyrosine kinase inhibitors, two retinoid X receptor agonists, two immunomodulatory agents, one histone deacetylase inhibitor, and one monoclonal antibody) in the AD continuum. The systematic literature search led to the identification of five published studies (one phase I, three phase II, and one phase IIb/III) reporting the effects of antitumoral treatments in patients with mild cognitive impairment or AD dementia. The clinical findings and the methodological characteristics of these studies are described and discussed. Conclusion Anticancer agents are triggering growing interest in the context of repurposed therapies in AD. Several clinical trials are underway, and data are expected to be available in the near future. To date, data emerging from published clinical studies are controversial. The promising results emerging from preclinical studies and identified research protocols should be confirmed and extended by larger, adequately designed, and high-quality clinical trials.
Collapse
Affiliation(s)
- Antonio Ancidoni
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Via Giano della Bella 34, 00162, Rome, Italy.
| | - Ilaria Bacigalupo
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Via Giano della Bella 34, 00162, Rome, Italy
| | - Giulia Remoli
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Via Giano della Bella 34, 00162, Rome, Italy
| | - Eleonora Lacorte
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Via Giano della Bella 34, 00162, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Giulia Sarti
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Via Dezza 48, 20144, Milan, Italy
| | - Nicola Vanacore
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Via Giano della Bella 34, 00162, Rome, Italy
| | - Marco Canevelli
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Via Giano della Bella 34, 00162, Rome, Italy.,Department of Human Neuroscience, Sapienza University, Rome, Italy
| |
Collapse
|
30
|
Emerging Therapeutic Strategies to Overcome Drug Resistance in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13071686. [PMID: 33918370 PMCID: PMC8038312 DOI: 10.3390/cancers13071686] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Multiple myeloma is a deadly blood cancer, but fortunately drug development has substantially prolonged the lifespan of patients to average more than a decade after diagnosis with optimal therapy. As a result, the population of patients living with multiple myeloma has grown considerably. Through its course, patients suffer repeated relapses for which they require new lines of treatment. Currently, the key drug classes for treatment are immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies. The goal of this review is to summarize the understanding of the problem of resistance to these drugs, which is ultimately responsible for patient fatality. In addition, we will focus on how new agents that are promising in clinical trials overcome resistance. Abstract Multiple myeloma is a malignant plasma cell neoplasm that remains incurable and is ultimately fatal when patients acquire multi-drug resistance. Thus, advancing our understanding of the mechanisms behind drug resistance in multi-relapsed patients is critical for developing better strategies to extend their lifespan. Here, we review the understanding of resistance to the three key drug classes approved for multiple myeloma treatment: immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies. We consider how the complex, heterogenous biology of multiple myeloma may influence the acquisition of drug resistance and reflect on the gaps in knowledge where additional research is needed to improve our treatment approaches. Fortunately, many agents are currently being evaluated preclinically and in clinical trials that have the potential to overcome or delay drug resistance, including next-generation immunomodulatory drugs and proteasome inhibitors, novel small molecule drugs, chimeric antigen receptor T cells, antibody-drug conjugates, and bispecific antibodies. For each class, we discuss the potential of these strategies to overcome resistance through modifying agents within each class or new classes without cross-resistance to currently available drugs.
Collapse
|
31
|
Bolaman AZ, Turgutkaya A, Sahip B, Selim C, Eroğlu Küçükerdiler H, Ertop Ş, Sargın G, Yavaşoğlu İ. Original Versus Generic Lenalidomide in Patients with Relapsed/Refractory Multiple Myeloma: Comparison of Efficacy and Adverse Events. Turk J Haematol 2021; 38:41-48. [PMID: 33342205 PMCID: PMC7927438 DOI: 10.4274/tjh.galenos.2020.2020.0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Objective: Lenalidomide is an effective immunomodulatory derivative drug used in the treatment of multiple myeloma (MM). It is available in original and generic forms in Turkey, but there is no clinical study that has compared the effectiveness and adverse events (AEs) of the generic and original forms of lenalidomide. We compared the effectivity and AEs of generic and original lenalidomide in patients with relapsed/refractory MM (RRMM). Materials and Methods: Patients with RRMM using original or generic lenalidomide were evaluated retrospectively. Overall response (OR), complete response (CR), very good partial response (VGPR), partial response (PR), stable disease, and progressive disease rates and hematologic and nonhematologic AEs were evaluated in these RRMM patients. The results were described as numbers, frequencies, and percentages and were analyzed using PASW 19.0 for Windows with chi-square and Fisher exact tests. Results: The number of patients using original lenalidomide was 55 and the number of patients using generic lenalidomide was 43. The OR rate was 67.2% for patients using original lenalidomide and 60.4% for those on generic lenalidomide. CR and VGPR rates were 14.5% and 45.4% in the original group while the CR and VGPR rates were 20.9% and 18.6%, respectively, in patients using generic lenalidomide. Hematologic AEs were similar in the two groups while some nonhematologic AEs were less common in the original lenalidomide group than the generic group. Only pyrexia as a grade 3-4 AE was more common in the original lenalidomide than the generic lenalidomide group. Conclusion: This study showed that the generic form of lenalidomide has similar efficacy with the original form of lenalidomide in the treatment of RRMM. The AEs of original lenalidomide were generally fewer than those of generic lenalidomide. Further studies involving a larger number of patients with RRMM would be useful for comparing the efficacy and AEs of original and generic lenalidomide.
Collapse
Affiliation(s)
- Ali Zahit Bolaman
- Aydın Adnan Menderes University Faculty of Medicine, Department of Hematology, Aydın, Turkey
| | - Atakan Turgutkaya
- Aydın Adnan Menderes University Faculty of Medicine, Department of Hematology, Aydın, Turkey
| | - Birsen Sahip
- Zonguldak Bülent Ecevit University Faculty of Medicine, Division of Hematology, Zonguldak, Turkey
| | - Cem Selim
- Aydın Adnan Menderes University Faculty of Medicine, Department of Hematology, Aydın, Turkey
| | | | - Şehmus Ertop
- Zonguldak Bülent Ecevit University Faculty of Medicine, Division of Hematology, Zonguldak, Turkey
| | - Gökhan Sargın
- Aydın Adnan Menderes University Faculty of Medicine, Department of Immunology-Rheumatology, Aydın, Turkey
| | - İrfan Yavaşoğlu
- Aydın Adnan Menderes University Faculty of Medicine, Department of Hematology, Aydın, Turkey
| |
Collapse
|
32
|
Innao V, Rizzo V, Allegra AG, Musolino C, Allegra A. Promising Anti-Mitochondrial Agents for Overcoming Acquired Drug Resistance in Multiple Myeloma. Cells 2021; 10:439. [PMID: 33669515 PMCID: PMC7922387 DOI: 10.3390/cells10020439] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable tumor due to the high rate of relapse that still occurs. Acquired drug resistance represents the most challenging obstacle to the extension of survival and several studies have been conducted to understand the mechanisms of this phenomenon. Mitochondrial pathways have been extensively investigated, demonstrating that cancer cells become resistant to drugs by reprogramming their metabolic assessment. MM cells acquire resistance to proteasome inhibitors (PIs), activating protection programs, such as a reduction in oxidative stress, down-regulating pro-apoptotic, and up-regulating anti-apoptotic signals. Knowledge of the mechanisms through which tumor cells escape control of the immune system and acquire resistance to drugs has led to the creation of new compounds that can restore the response by leading to cell death. In this scenario, based on all literature data available, our review represents the first collection of anti-mitochondrial compounds able to overcome drug resistance in MM. Caspase-independent mechanisms, mainly based on increased oxidative stress, result from 2-methoxyestradiol, Artesunate, ascorbic acid, Dihydroartemisinin, Evodiamine, b-AP15, VLX1570, Erw-ASNase, and TAK-242. Other agents restore PIs' efficacy through caspase-dependent tools, such as CDDO-Im, NOXA-inhibitors, FTY720, GCS-100, LBH589, a derivative of ellipticine, AT-101, KD5170, SMAC-mimetics, glutaminase-1 (GLS1)-inhibitors, and thenoyltrifluoroacetone. Each of these substances improved the efficacy rates when employed in combination with the most frequently used antimyeloma drugs.
Collapse
Affiliation(s)
- Vanessa Innao
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (V.I.); (A.G.A.); (C.M.)
| | - Vincenzo Rizzo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Andrea Gaetano Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (V.I.); (A.G.A.); (C.M.)
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (V.I.); (A.G.A.); (C.M.)
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (V.I.); (A.G.A.); (C.M.)
| |
Collapse
|
33
|
Tan TD, Hong YC, Li SS, Yu JT, Sung YC, Wang PN, Teng CLJ. Lenalidomide with dexamethasone to multiple myeloma patients relapsing from bortezomib-based induction therapies: A prospective, observational study. CHINESE J PHYSIOL 2020; 63:211-217. [PMID: 33109787 DOI: 10.4103/cjp.cjp_68_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Lenalidomide with dexamethasone (Len/Dex) is considered to be an effective and well-tolerated regimen to treat multiple myeloma (MM) patients relapsing after bortezomib induction therapy. With the increase in novel agents targeting refractory and relapsed MM, the identification of clinical or laboratory variables that can predict the appropriate candidates of Len/Dex is essential. To address this question, we prospectively assessed 38 adult MM patients who received bortezomib-based induction therapy and were administered Len/Dex for their first relapse. These 38 patients were stratified into the symptomatic relapse group (n = 10) and biological relapse group (n = 28) according to the disease status when Len/Dex was initiated. The overall response rate in the symptomatic group and biological relapse group was 70.0% (7/10) and 60.7% (17/28), respectively (P = 0.964). These two groups harbored a comparable median Len/Dex treatment duration (139 vs. 225 days; P = 0.876) and progression-free survival 2 (PFS2) (501 vs. 1289 days; P = 0.410). Multivariate analyses failed to show that treating biological relapse (hazard ratio [HR]: 1.29; 95% confidence interval [CI]: 0.43-3.88; P = 0.648), PFS with bortezomib-based induction therapies ≥18 months (HR: 1.79; 95% CI: 0.64-5.01; P = 0.266), autologous hematopoietic stem cell transplantation (HR: 2.18; 95% CI: 0.56-8.55; P = 0.262), and high-risk cytogenetics (HR: 0.85; 95% CI: 0.18-3.93; P = 0.835) were attributed to depth of Len/Dex treatment. In conclusion, whether MM patients treated by Len/Dex for biological relapse would have a better outcome than those prescribed for symptomatic relapse remains inconclusive. Treating significant biological relapse and symptomatic relapse remains the current consensus.
Collapse
Affiliation(s)
- Tran-Der Tan
- Hematology and Medical Oncology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Ying-Chung Hong
- Division of Hematology and Oncology, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Sin-Syue Li
- Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital; College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jui-Ting Yu
- Division of Hematology/Medical Oncology, Department of Medicine, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Yung-Chuan Sung
- Division of Hematology/Oncology, Department of Medicine, Cathay General Hospital, Taipei; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Po-Nan Wang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chieh-Lin Jerry Teng
- Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung; Department of Life Science, Tunghai University; School of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| |
Collapse
|
34
|
Sherbenou DW, Su Y, Behrens CR, Aftab BT, Perez de Acha O, Murnane M, Bearrows SC, Hann BC, Wolf JL, Martin TG, Liu B. Potent Activity of an Anti-ICAM1 Antibody-Drug Conjugate against Multiple Myeloma. Clin Cancer Res 2020; 26:6028-6038. [PMID: 32917735 PMCID: PMC7669584 DOI: 10.1158/1078-0432.ccr-20-0400] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/15/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE New therapies have changed the outlook for patients with multiple myeloma, but novel agents are needed for patients who are refractory or relapsed on currently approved drug classes. Novel targets other than CD38 and BCMA are needed for new immunotherapy development, as resistance to daratumumab and emerging anti-BCMA approaches appears inevitable. One potential target of interest in myeloma is ICAM1. Naked anti-ICAM1 antibodies were active in preclinical models of myeloma and safe in patients, but showed limited clinical efficacy. Here, we sought to achieve improved targeting of multiple myeloma with an anti-ICAM1 antibody-drug conjugate (ADC). EXPERIMENTAL DESIGN Our anti-ICAM1 human mAb was conjugated to an auristatin derivative, and tested against multiple myeloma cell lines in vitro, orthotopic xenografts in vivo, and patient samples ex vivo. The expression of ICAM1 was also measured by quantitative flow cytometry in patients spanning from diagnosis to the daratumumab-refractory state. RESULTS The anti-ICAM1 ADC displayed potent anti-myeloma cytotoxicity in vitro and in vivo. In addition, we have verified that ICAM1 is highly expressed on myeloma cells and shown that its expression is further accentuated by the presence of bone marrow microenvironmental factors. In primary samples, ICAM1 is differentially overexpressed on multiple myeloma cells compared with normal cells, including daratumumab-refractory patients with decreased CD38. In addition, ICAM1-ADC showed selective cytotoxicity in multiple myeloma primary samples. CONCLUSIONS We propose that anti-ICAM1 ADC should be further studied for toxicity, and if safe, tested for clinical efficacy in patients with relapsed or refractory multiple myeloma.
Collapse
Affiliation(s)
- Daniel W Sherbenou
- Department of Medicine, University of California at San Francisco, California
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Yang Su
- Department of Anesthesia, University of California at San Francisco, California
| | | | - Blake T Aftab
- Department of Medicine, University of California at San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Olivia Perez de Acha
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Megan Murnane
- Department of Medicine, University of California at San Francisco, California
| | - Shelby C Bearrows
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Byron C Hann
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jeffery L Wolf
- Department of Medicine, University of California at San Francisco, California
| | - Thomas G Martin
- Department of Medicine, University of California at San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California at San Francisco, California.
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| |
Collapse
|
35
|
Anti-tumor activities of the new oral pan-RAF inhibitor, TAK-580, used as monotherapy or in combination with novel agents in multiple myeloma. Oncotarget 2020; 11:3984-3997. [PMID: 33216827 PMCID: PMC7646837 DOI: 10.18632/oncotarget.27775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 09/29/2020] [Indexed: 12/21/2022] Open
Abstract
Many RAS pathway inhibitors, including pan-RAF inhibitors, have shown significant anti-tumor activities in both solid and hematological tumors. The pan-RAF inhibitor, TAK-580, is a representative of the novel RAF inhibitors that act by disrupting RAF homo- or heterodimerization. In this study, we examined the anti-tumor effects of TAK-580 used as monotherapy or in combination with bortezomib, lenalidomide, or other novel agents in multiple myeloma (MM) cells in vitro. TAK-580 monotherapy potently targeted proteins in the RAS-RAF-MEK-ERK signaling pathway and induced potent cytotoxicity and apoptosis in MM cell lines and myeloma cells from patients with newly diagnosed and relapsed and/or refractory MM, compared with a representative RAF inhibitor, dabrafenib. Normal donor peripheral blood B lymphocytes and cord blood CD34-positive cells were not affected. Importantly, TAK-580 significantly inhibited phospho-FOXO3 and induced upregulation of BimL and BimS in a dose-dependent manner, finally leading to apoptosis in MM cells. Moreover, TAK-580 enhanced bortezomib-induced cytotoxicity and apoptosis in MM cells via the FOXO3-Bim axis and the terminal unfolded protein response. Importantly, TAK-580 also enhanced lenalidomide-induced cytotoxicity and apoptosis in MM cells. Taken together, our results provide the rationale for TAK-580 monotherapy and/or treatment in combination with novel agents to improve outcomes in patients with MM.
Collapse
|
36
|
Walker ZJ, Idler BM, Davis LN, Stevens BM, VanWyngarden MJ, Ohlstrom D, Bearrows SC, Hammes A, Smith CA, Jordan CT, Mark TM, Forsberg PA, Sherbenou DW. Exploiting Protein Translation Dependence in Multiple Myeloma with Omacetaxine-Based Therapy. Clin Cancer Res 2020; 27:819-830. [PMID: 33109736 DOI: 10.1158/1078-0432.ccr-20-2246] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/18/2020] [Accepted: 10/22/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE The prognosis of patients with multiple myeloma who are resistant to proteasome inhibitors, immunomodulatory drugs (IMiD), and daratumumab is extremely poor. Even B-cell maturation antigen-specific chimeric antigen receptor T-cell therapies provide only a temporary benefit before patients succumb to their disease. In this article, we interrogate the unique sensitivity of multiple myeloma cells to the alternative strategy of blocking protein translation with omacetaxine. EXPERIMENTAL DESIGN We determined protein translation levels (n = 17) and sensitivity to omacetaxine (n = 51) of primary multiple myeloma patient samples. Synergy was evaluated between omacetaxine and IMiDs in vitro, ex vivo, and in vivo. Underlying mechanism was investigated via proteomic analysis. RESULTS Almost universally, primary patient multiple myeloma cells exhibit >2.5-fold increased rates of protein translation compared with normal marrow cells. Ex vivo treatment with omacetaxine resulted in >50% reduction in viable multiple myeloma cells. In this cohort, high levels of translation serve as a biomarker for patient multiple myeloma cell sensitivity to omacetaxine. Unexpectedly, omacetaxine demonstrated synergy with IMiDs in multiple myeloma cell lines in vitro. In addition, in an IMiD-resistant relapsed patient sample, omacetaxine/IMiD combination treatment resensitized the multiple myeloma cells to the IMiD. Proteomic analysis found that the omacetaxine/IMiD combination treatment produced a double-hit on the IRF4/c-MYC pathway, which is critical to multiple myeloma survival. CONCLUSIONS Overall, protein translation inhibitors represent a potential new drug class for myeloma treatment and provide a rationale for conducting clinical trials with omacetaxine alone and in combination with IMiDs for patients with relapsed/refractory multiple myeloma.
Collapse
Affiliation(s)
- Zachary J Walker
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Beau M Idler
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lorraine N Davis
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brett M Stevens
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael J VanWyngarden
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Denis Ohlstrom
- Biomedical Sciences and Biotechnology, Graduate School, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shelby C Bearrows
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Andrew Hammes
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Clayton A Smith
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Tomer M Mark
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Peter A Forsberg
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel W Sherbenou
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado. .,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
37
|
Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity. Eur J Med Chem 2020; 209:112912. [PMID: 33328101 DOI: 10.1016/j.ejmech.2020.112912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022]
Abstract
Lenalidomide and its analogs are well-known for treating multiple myeloma. In this work, designed sulfide-modified lenalidomide and pomalidomide were synthesized and evaluated. The anti-proliferative activity against MM.1S cell line of 3ak (IC50 = 79 nM) was similar to lenalidomide (IC50 = 81 nM). Compared to benzylic thioether substituted lenalidomide 3a, the half-live (T1/2) of 4-F-phenyl-thioether analogs 3ak in human liver microsomes was promoted from 3 min to 416.7 min. The corresponding metabolic factor of 3ak was increased from 2.8% to 79.5%, which was slightly lower than lenalidomide (91.5%). Moreover, the IKZF1 degradation of 3y and 3ak was well related with corresponding IC50 values, which suggested the IKZF1 degradation efficiency is correlated to the responses of MM1. S cells. Furthermore, the oral administration of compounds 3y and 3ak at dosages of 60 mg/kg could delay tumor growth in female CB-17 SCID mice. This research helped to prompt the stability of thioether lenalidomide analogs, which paved the way for developing better molecules for treating multiple myeloma.
Collapse
|
38
|
Abstract
Despite considerable advances in treatment approaches in the past two decades, multiple myeloma remains an incurable disease. Treatments for myeloma continue to evolve with many emerging immunotherapies. The first immunotherapy used to treat hematologic cancers, including multiple myeloma, was an allogeneic stem cell transplant. In the mid-2000s, immunomodulatory drugs thalidomide, lenalidomide, and subsequently pomalidomide were proven to be effective in multiple myeloma and substantially improved survival. The next wave of immunotherapies for multiple myeloma included the monoclonal antibodies daratumumab and elotuzumab, which were approved by the Food and Drug Administration in 2015. Subsequently, a variety of immunotherapies have been developed for multiple myeloma, including chimeric antigen receptor T cells, bispecific antibodies, antibody drug conjugates, and checkpoint inhibitors. Many of these emerging treatments target the B cell maturation antigen, which is expressed on plasma cells, although several other novel receptors are also being studied. This review summarizes the evidence of these various immunotherapies, their mechanism of action, and data from clinical trials regarding the treatments' safety and efficacy.
Collapse
Affiliation(s)
- Urvi A Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| |
Collapse
|
39
|
Overview of the Side-Effects of FDA- and/or EMA-Approved Targeted Therapies for the Treatment of Hematological Malignancies. J Clin Med 2020; 9:jcm9092903. [PMID: 32911829 PMCID: PMC7565707 DOI: 10.3390/jcm9092903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
In the last decade there has been tremendous effort in offering better therapeutic management strategies to patients with hematologic malignancies. These efforts have ranged from biological to clinical approaches and resulted in the rapid development of new approaches. The main “problem” that comes with the high influx of newly approved drugs, which not only influences hematologists that frequently work with these drugs but also affects other healthcare professionals that work with hematologists in patient management, including intensive care unit (ICU) physicians, is they have to keep up within their specialty and, in addition, with the side-effects that can occur when encountering hematology-specific therapies. Nonetheless, there are few people that have an in-depth understanding of a specialty outside theirs. Thus, this manuscript offers an overview of the most common side-effects caused by therapies used in hematology nowadays, or that are currently being investigated in clinical trials, with the purpose to serve as an aid to other specialties. Nevertheless, because of the high amount of information on this subject, each chapter will offer an overview of the side-effects of a drug class with each reference of the section being intended as further reading.
Collapse
|
40
|
Pomalidomide, bortezomib, and dexamethasone for multiple myeloma previously treated with lenalidomide (OPTIMISMM): outcomes by prior treatment at first relapse. Leukemia 2020; 35:1722-1731. [PMID: 32895455 PMCID: PMC8179841 DOI: 10.1038/s41375-020-01021-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/14/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022]
Abstract
In the phase 3 OPTIMISMM trial, pomalidomide, bortezomib, and dexamethasone (PVd) demonstrated superior efficacy vs bortezomib and dexamethasone (Vd) in patients with relapsed or refractory multiple myeloma previously treated with lenalidomide, including those refractory to lenalidomide. This analysis evaluated outcomes in patients at first relapse (N = 226) by lenalidomide-refractory status, prior bortezomib exposure, and prior stem cell transplant (SCT). Second-line PVd significantly improved PFS vs Vd in lenalidomide-refractory (17.8 vs 9.5 months; P = 0.0276) and lenalidomide-nonrefractory patients (22.0 vs 12.0 months; P = 0.0491), patients with prior bortezomib (17.8 vs 12.0 months; P = 0.0068), and patients with (22.0 vs 13.8 months; P = 0.0241) or without (16.5 vs 9.5 months; P = 0.0454) prior SCT. In patients without prior bortezomib, median PFS was 20.7 vs 9.5 months (P = 0.1055). Significant improvement in overall response rate was also observed with PVd vs Vd in lenalidomide-refractory (85.9% vs 50.8%; P < 0.001) and lenalidomide-nonrefractory (95.7% vs 60.0%; P < 0.001) patients, with similar results regardless of prior bortezomib or SCT. No new safety signals were observed. These data demonstrate the benefit of PVd at first relapse, including immediately after upfront lenalidomide treatment failure and other common first-line treatments.
Collapse
|
41
|
Poh C, Keegan T, Rosenberg AS. Second primary malignancies in multiple myeloma: A review. Blood Rev 2020; 46:100757. [PMID: 32972803 DOI: 10.1016/j.blre.2020.100757] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/24/2020] [Accepted: 08/27/2020] [Indexed: 12/16/2022]
Abstract
As survival times of multiple myeloma (MM) patients continue to improve, second primary malignancies (SPM) have become an increasingly relevant long-term risk among MM survivors. Population studies since the 1950s have consistently observed an increased incidence of hematologic SPMs, specifically acute leukemia, among MM survivors. Prolonged treatment with alkylators, especially melphalan, was associated with an increased hematologic SPM risk; likewise, autologous stem cell transplantation appeared to minimally increase SPM risk. Immunomodulatory drugs, specifically lenalidomide, was associated with an increased SPM incidence, although most studies concluded that the benefits of therapy outweighed any risks of SPM. Newer anti-myeloma therapy such as proteasome inhibitors and monoclonal antibodies did not appear to increase SPM risk although robust long-term follow-up is lacking. This review discusses current understanding regarding SPMs among survivors of MM, how different host-, disease- and treatment-related factors contribute to SPM incidence and highlights emerging screening guidelines and prognosis for SPMs.
Collapse
Affiliation(s)
- Christina Poh
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA; University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA.
| | - Theresa Keegan
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Aaron Seth Rosenberg
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| |
Collapse
|
42
|
Rauch DA, Olson SL, Harding JC, Sundaramoorthi H, Kim Y, Zhou T, MacLeod AR, Challen G, Ratner L. Interferon regulatory factor 4 as a therapeutic target in adult T-cell leukemia lymphoma. Retrovirology 2020; 17:27. [PMID: 32859220 PMCID: PMC7456374 DOI: 10.1186/s12977-020-00535-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022] Open
Abstract
Background Adult T-cell leukemia lymphoma (ATLL) is a chemotherapy-resistant malignancy with a median survival of less than one year that will afflict between one hundred thousand and one million individuals worldwide who are currently infected with human T-cell leukemia virus type 1. Recurrent somatic mutations in host genes have exposed the T-cell receptor pathway through nuclear factor κB to interferon regulatory factor 4 (IRF4) as an essential driver for this malignancy. We sought to determine if IRF4 represents a therapeutic target for ATLL and to identify downstream effectors and biomarkers of IRF4 signaling in vivo. Results ATLL cell lines, particularly Tax viral oncoprotein-negative cell lines, that most closely resemble ATLL in humans, were sensitive to dose- and time-dependent inhibition by a next-generation class of IRF4 antisense oligonucleotides (ASOs) that employ constrained ethyl residues that mediate RNase H-dependent RNA degradation. ATLL cell lines were also sensitive to lenalidomide, which repressed IRF4 expression. Both ASOs and lenalidomide inhibited ATLL proliferation in vitro and in vivo. To identify biomarkers of IRF4-mediated CD4 + T-cell expansion in vivo, transcriptomic analysis identified several genes that encode key regulators of ATLL, including interleukin 2 receptor subunits α and β, KIT ligand, cytotoxic T-lymphocyte-associated protein 4, and thymocyte selection-associated high mobility group protein TOX 2. Conclusions These data support the pursuit of IRF4 as a therapeutic target in ATLL with the use of either ASOs or lenalidomide.
Collapse
Affiliation(s)
- Daniel A Rauch
- Division of Oncology, Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Box 8069, St Louis, MO, 63110, USA
| | - Sydney L Olson
- Division of Oncology, Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Box 8069, St Louis, MO, 63110, USA
| | - John C Harding
- Division of Oncology, Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Box 8069, St Louis, MO, 63110, USA
| | - Hemalatha Sundaramoorthi
- Division of Oncology, Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Box 8069, St Louis, MO, 63110, USA
| | | | | | | | - Grant Challen
- Division of Oncology, Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Box 8069, St Louis, MO, 63110, USA
| | - Lee Ratner
- Division of Oncology, Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Box 8069, St Louis, MO, 63110, USA.
| |
Collapse
|
43
|
Gamberi B, Berthou C, Hernandez M, Semenzato G, Tholouli E, Hájek R, Caers J, Dimopoulos M, Minnema MC, Andreasson B, Parreira J, Crotty G, Remes K, Kueenburg E, Rosettani B, Di Micco A, Peters S, Bacon P, Blau IW. A Noninterventional, Observational, European Post-Authorization Safety Study of Patients With Relapsed/Refractory Multiple Myeloma Treated With Lenalidomide. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e629-e644. [PMID: 32605897 DOI: 10.1016/j.clml.2020.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Lenalidomide plus dexamethasone is effective and well tolerated in relapsed/refractory multiple myeloma (RRMM). In this observational, noninterventional European post-authorization safety study, the safety profile of lenalidomide plus dexamethasone was investigated and compared with that of other agents in the treatment of RRMM in a real-world setting. PATIENTS AND METHODS Patients had received ≥ 1 prior antimyeloma therapy; prior lenalidomide was excluded. Treatment was per investigator's routine practice. Adverse events were analyzed by incidence rates per 100 person-years to account for differences in observation length and treatment duration. RESULTS In total, 2150 patients initiated lenalidomide, and 1479 initiated any other antimyeloma therapy, predominately bortezomib (80.3%), which was primarily administered intravenously (74.3%). The incidence rate of neuropathy was lower with lenalidomide (10.5) than with bortezomib (78.9) or thalidomide (38.7). Lenalidomide also had a lower incidence rate of infections (68.7) versus bortezomib (95.9) and thalidomide (76.0). Conversely, the incidence rate of neutropenia was higher with lenalidomide (38.0) than with bortezomib (18.2) or thalidomide (25.7). The incidence rates of thrombocytopenia were 24.4, 40.4, and 14.4 with lenalidomide, bortezomib, and thalidomide, respectively. CONCLUSION No new safety signals for lenalidomide were identified in this study, which is the largest prospective real-world European study of lenalidomide in patients with RRMM to date. These results confirm that the safety profile of lenalidomide plus dexamethasone in RRMM in a real-world setting is comparable to that reported in clinical trials.
Collapse
Affiliation(s)
- Barbara Gamberi
- Department of Hematology, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| | - Christian Berthou
- Centre Hospitalier Régional Universitaire, Hôpital Auguste Morvan, Brest, France
| | - Miguel Hernandez
- Hemotherapy Service, Hospital Universitario de Canarias, Tenerife, Spain
| | | | - Eleni Tholouli
- Department of Haematology, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Roman Hájek
- Department of Clinic Subjects, University Hospital Ostrava and Faculty of Medicine Ostrava, Ostrava, Czech Republic
| | - Jo Caers
- Department of Hematology, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - Meletios Dimopoulos
- National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Joana Parreira
- Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa and Instiuto Português de Oncologia, Francisco Gentil, Lisboa, Portugal
| | - Gerard Crotty
- Department of Haematology, Midland Regional Hospital, Tullamore, Ireland
| | - Kari Remes
- Department of Internal Medicine, Turku University Hospital, Turku, Finland
| | - Elisabeth Kueenburg
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Barbara Rosettani
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Antonia Di Micco
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Sarah Peters
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Pamela Bacon
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Igor Wolfgang Blau
- Department of Internal Medicine III, Charité Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
44
|
Armando RG, Gómez DLM, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. Int J Oncol 2020; 56:651-684. [PMID: 32124955 PMCID: PMC7010222 DOI: 10.3892/ijo.2020.4966] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
Collapse
Affiliation(s)
- Romina Gabriela Armando
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Diego Luis Mengual Gómez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Daniel Eduardo Gomez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Autologous stem cell transplantation has been the standard of care in myeloma treatment for many years, but the availability of newer antimyeloma drugs and the emerging data from chimeric antigen receptor (CAR) T-cell clinical studies make us question the relevance of it. The purpose of this review is to go over recent data and to reassess the current status of autologous stem cell transplantation as a standard of care. RECENT FINDINGS Autologous stem cell transplantation can be safely performed for elderly patients and there is no absolute age limit. Recent data on BEAM (Carmustine, Etoposide, Cytarabine, and Melphalan), Busulfan/Melphalan, and Carmustine/Melphalan conditioning when compared with Melphalan showed favorable survival outcomes with manageable toxicities although we need to see data from randomized, multicenter studies. Posttransplant maintenance and consolidation can maximize the benefit of transplant by prolonging progression-free survival. Current B-cell maturation antigen CAR T-cell therapy showed remarkably high response rates, but didn't seem to provide durable response yet. SUMMARY Recent advances in myeloma therapy and autologous stem cell transplantation are described. Although we've seen many new developments including CAR T-cell therapies, autologous stem cell transplantation remains as the standard of care. However, it may be replaced by or combined with newer therapies in the future.
Collapse
|
46
|
Direct potentiation of NK cell cytotoxicity by 8-azaguanine with potential antineoplastic activity. Int Immunopharmacol 2018; 67:152-159. [PMID: 30551032 DOI: 10.1016/j.intimp.2018.12.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/01/2018] [Accepted: 12/07/2018] [Indexed: 12/22/2022]
Abstract
This study identified 8-azaguanine (8-AG) as a novel immunomodulatory drug (IMiD) through a high-throughput screen of the Preswick Chemical Library in a model of human NK cell cytotoxicity against blood cancer cells. 8-AG, originally developed as an antineoplastic agent, significantly increased the cytotoxicity of NK cells and was superior in this activity to previously known IMiDs, such as fluoxetine and amphotericin B, identified from the same library. IFN-γ expression was also slightly increased by 8-AG. Mechanistically, 8-AG increased conjugate formation between NK and target cells and subsequent cytolytic granule polarization, but not calcium mobilization, regulation of activating receptors, or expression of perforin or granzyme B. Thus, the antineoplastic activity of 8-AG should be re-evaluated in light of this novel potentiating effect on NK cells.
Collapse
|
47
|
Robak P, Drozdz I, Szemraj J, Robak T. Drug resistance in multiple myeloma. Cancer Treat Rev 2018; 70:199-208. [DOI: 10.1016/j.ctrv.2018.09.001] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/05/2018] [Accepted: 09/01/2018] [Indexed: 02/07/2023]
|