101
|
Mo CC, Jagannath S, Chari A, Nooka AK, Lonial S, Siegel D, Biran N, Gasparetto C, Bahlis NJ, Richardson P. Selinexor for the treatment of patients with previously treated multiple myeloma. Expert Rev Hematol 2021; 14:697-706. [PMID: 33985401 DOI: 10.1080/17474086.2021.1923473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is an increasingly treatable but still incurable hematologic malignancy. Prognosis has improved significantly over recent years, although further advances remain urgently needed, especially for patients with heavily pre-treated and resistant disease for whom there are limited options. Selinexor is a first-in-class, oral, selective inhibitor of nuclear export (SINE) compound that triggers apoptosis in malignant cells by inducing nuclear retention of oncogene messenger RNAs (mRNAs) and reactivation of tumor suppressor proteins (TSPs). In clinical studies of patients with relapsed and/or refractory MM, selinexor has demonstrated both manageable toxicity and encouraging efficacy. AREAS COVERED This review will provide an overview of the mechanism of action of selinexor as well as the efficacy and safety data from clinical studies using selinexor for the treatment of multiple myeloma. EXPERT OPINION Long-term outcomes for patients with MM will continue to improve due to numerous recent and imminent therapeutic advances, although critical areas of unmet need remain. Oral selinexor is likely to contribute to the meeting of these needs and the further advancement of MM therapy in a meaningful way.
Collapse
Affiliation(s)
- Clifton C Mo
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sundar Jagannath
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ajai Chari
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - David Siegel
- John Theurer Cancer Center, Hackensack University, Hackensack, NJ, USA
| | - Noa Biran
- John Theurer Cancer Center, Hackensack University, Hackensack, NJ, USA
| | | | - Nizar J Bahlis
- Charbonneau Cancer Research Institute, Calgary, AB, Canada
| | - Paul Richardson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
102
|
Nelson R, Mathews R, Palesi C, Carter J, Szymanski J, Uehlinger J, Weinberg R, Paroder M. Mysterious clumping in a cell therapy product. Transfusion 2021; 61:2232-2233. [PMID: 34273121 DOI: 10.1111/trf.16569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/27/2021] [Accepted: 05/21/2021] [Indexed: 12/26/2022]
Affiliation(s)
- Randin Nelson
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Carlo Palesi
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jamal Carter
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| | - James Szymanski
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| | - Joan Uehlinger
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Monika Paroder
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
103
|
Straka C, Salwender H, Knop S, Vogel M, Müller J, Metzner B, Langer C, Sayer H, Jung W, Dürk HA, Bassermann F, Gramatzki M, Rösler W, Wolf HH, Brugger W, Engelhardt M, Fischer T, Liebisch P, Einsele H. Full or intensity-reduced high-dose melphalan and single or double autologous stem cell transplant with or without bortezomib consolidation in patients with newly diagnosed multiple myeloma. Eur J Haematol 2021; 107:529-542. [PMID: 34270825 DOI: 10.1111/ejh.13690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 01/18/2023]
Abstract
OBJECTIVE A post hoc subgroup analysis of two phase III trials (NCT00416273, NCT00416208) was carried out to investigate the influence of 100/140 and 200 mg/m² melphalan as well as single/double autologous stem cell transplantation (ASCT) on progression-free survival (PFS). Additionally, the effect of bortezomib consolidation on PFS was analyzed. METHODS Following induction therapy and high-dose melphalan with subsequent ASCT, patients with newly diagnosed multiple myeloma (NDMM) were randomized 1:1 to either four 35-day cycles of bortezomib consolidation (1.6 mg/m² IV on days 1, 8, 15, 22) or observation. RESULTS Of the 340 patients included in this analysis, 13.5% received 1 × MEL100/140, 22.9% 2 × MEL100/140, 31.2% 1 × MEL200, and 32.4% 2 × MEL200. With higher cumulative melphalan dose, PFS improved (P = .0085). PFS curves of patients treated with 2 × MEL100/140 and 1 × MEL200 were very similar. The superior dose effect of MEL200 over MEL100/140 was non-existent in the bortezomib consolidation arm but pronounced in the observation arm (P = .0015). Similarly, double ASCT was only beneficial in patients without bortezomib consolidation (P = .0569). CONCLUSIONS Full dose melphalan and double transplantation seem advantageous only as long as patients are not receiving bortezomib consolidation afterwards.
Collapse
Affiliation(s)
| | - Hans Salwender
- Asklepios Tumorzentrum, AK Altona und AK St. Georg, Hamburg, Germany
| | - Stefan Knop
- Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany
| | | | | | - Bernd Metzner
- Universitätsklinik für Innere Medizin II, Oldenburg, Germany
| | - Christian Langer
- Universitätsklinikum Ulm, Klinik für Innere Medizin II, Ulm, Germany
| | - Herbert Sayer
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Jena, Germany
| | - Wolfram Jung
- Universitätsmedizin Göttingen, Göttingen, Germany
| | | | - Florian Bassermann
- Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | | | - Wolf Rösler
- Universitätsklinikum Erlangen, Erlangen, Germany
| | | | | | | | | | - Peter Liebisch
- Universitätsklinikum Ulm, Klinik für Innere Medizin II, Ulm, Germany
| | - Hermann Einsele
- Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany
| |
Collapse
|
104
|
Initial Therapeutic Approaches to Patients with Multiple Myeloma. Adv Ther 2021; 38:3694-3711. [PMID: 34145483 DOI: 10.1007/s12325-021-01824-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022]
Abstract
Multiple Myeloma (MM) is part of a spectrum of plasma cell disorders that may result in end organ damage. MM is subclassified into high and standard risk based on cytogenetic and laboratory markers. The treatment of newly diagnosed multiple myeloma is constantly changing with the advent of novel therapies. Recent advances in therapies have resulted in longer time to remission and overall survival. the introduction of targeted therapy with monoclonal antibodies such as Daratumumab has improved stringent complete response to 39%. In this review, we outline the current approach to diagnosis, prognosis, and management of newly diagnosed multiple myeloma in both transplant eligible and ineligible patients.
Collapse
|
105
|
Singh N, Singh A, Besser SA, Lang RM, Mor-Avi V, Kosuri S, Bishop MR, DeCara JM. Echocardiographic predictors of new-onset atrial arrhythmias in patients undergoing hematopoietic stem cell transplantation. Int J Cardiol 2021; 339:225-231. [PMID: 34174337 DOI: 10.1016/j.ijcard.2021.06.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Atrial arrhythmias following hematopoietic stem cell transplantation (HSCT) have been associated with increased length of stay, need for intensive care, and increased mortality within one-year post-transplant. We sought to identify echocardiographic parameters that may predict the development of new atrial arrhythmias post-HSCT. METHODS We performed a retrospective chart review of 753 consecutive patients who underwent HSCT at the University of Chicago from January 2015 through December 2019. Patients with baseline echocardiogram within 6 months prior to transplantation were included. Those with prior transplants, history of atrial arrhythmias, or unavailable echocardiographic images were excluded, resulting in 187 patients included for final analysis. Baseline clinical and demographic variables, as well as echocardiographic parameters, were compared between patients who developed new atrial arrhythmias post-HSCT versus those who did not. RESULTS Of the 187 patients included for analysis, 25 (13%) developed new atrial arrhythmias, with 13 of these occurring within 30 days of transplantation. Despite no significant difference in left atrial (LA) end-systolic volume between those with and without new arrhythmia following HSCT (OR 1.04; 95% CI 0.91-1.09, p = 0.233), univariable analysis demonstrated that patients who developed atrial arrhythmias had reduced LA function, as reflected by lower LA emptying fraction (OR 0.94; 95% CI 0.91-0.98, p = 0.003) and lower LA reservoir strain (OR 0.95; 95% CI 0.92-0.99, p = 0.009). CONCLUSIONS Echocardiographic indices of LA function, namely LA emptying fraction and LA reservoir strain, can identify patients at risk for developing new atrial arrhythmias post-HSCT, prior to the development of morphologic changes in the LA.
Collapse
Affiliation(s)
- Nikhil Singh
- Section of Cardiology, Department of Medicine, UChicago Medicine, Chicago, IL 60636, USA
| | - Amita Singh
- Section of Cardiology, Department of Medicine, UChicago Medicine, Chicago, IL 60636, USA
| | - Stephanie A Besser
- Section of Cardiology, Department of Medicine, UChicago Medicine, Chicago, IL 60636, USA
| | - Roberto M Lang
- Section of Cardiology, Department of Medicine, UChicago Medicine, Chicago, IL 60636, USA
| | - Victor Mor-Avi
- Section of Cardiology, Department of Medicine, UChicago Medicine, Chicago, IL 60636, USA
| | - Satyajit Kosuri
- Section of Hematology/Oncology, Department of Medicine, UChicago Medicine, Chicago, IL 60637, USA
| | - Michael R Bishop
- Section of Hematology/Oncology, Department of Medicine, UChicago Medicine, Chicago, IL 60637, USA
| | - Jeanne M DeCara
- Section of Cardiology, Department of Medicine, UChicago Medicine, Chicago, IL 60636, USA.
| |
Collapse
|
106
|
Heider M, Grass S, Dill V, Rämisch S, Winter C, Verbeek M, Götze KS, Bassermann F, Jilg S. Cyclophosphamide plus etoposide is a safe and effective mobilization regimen in patients with multiple myeloma. Transfus Apher Sci 2021; 60:103197. [PMID: 34175195 DOI: 10.1016/j.transci.2021.103197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 11/29/2022]
Abstract
High-dose chemotherapy followed by autologous stem cell transplantation is a major component in the treatment of patients with multiple myeloma. As a prerequisite, the successful collection of a sufficient number of viable peripheral blood hematopoietic CD34+ cells is critical. A common standard protocol for mobilization is currently not defined and critically discussed especially in German-speaking Europe. In times of the Covid-19 pandemic, safe and effective strategies have to be chosen to minimize hospitalization times and severe courses. In this single-center retrospective analysis, safety and efficacy of cyclophosphamide plus etoposide (CE) and growth-factor support (n = 33) was compared to cyclophosphamide mono treatment and growth-factor support (n = 49) in 82 patients with multiple myeloma at first diagnosis. CE was superior to cyclophosphamide mono with a significantly higher number of collected CD34+ cells (15.46 × 106 CD34+ cells/kg vs. 9.92 × 106 CD34+ cells/kg), significantly faster engraftment of granulocytes after stem cell transplantation (day 10.5 vs. day 11.6), shorter duration of the inpatient stay (17.47 days vs. 19.16 days) and significantly less transfusions (8.82 % vs. 30.61 % patients receiving transfusions). The safety profile was comparable in both groups and in line with published data. We conclude that CE is a safe and highly effective mobilization protocol in patients with multiple myeloma at first diagnosis and appears to be superior to the commonly used cyclophosphamide mono regimen.
Collapse
Affiliation(s)
- Michael Heider
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany; Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Sandra Grass
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany
| | - Veronika Dill
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany
| | - Stephanie Rämisch
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany
| | - Christof Winter
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Mareike Verbeek
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany
| | - Katharina S Götze
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK), 69120 Heidelberg, Germany
| | - Florian Bassermann
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany; Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technical University of Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK), 69120 Heidelberg, Germany.
| | - Stefanie Jilg
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Germany.
| |
Collapse
|
107
|
Wang F, Younis M, Luo Y, Zhang L, Yuan L. Iguratimod-encapsulating PLGA-NPs induce human multiple myeloma cell death via reactive oxygen species and Caspase-dependent signalling. Int Immunopharmacol 2021; 95:107532. [PMID: 33756230 DOI: 10.1016/j.intimp.2021.107532] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/28/2021] [Accepted: 02/22/2021] [Indexed: 11/26/2022]
Abstract
Human multiple myeloma (MM) is a currently incurable haematopoietic malignancies. Our research investigate the anti-tumour effect of iguratimod (IGU) encapsulated in poly(lactic-co-glycolic acid) PLGA nanoparticles (IGU-PLGA-NPs) on MM cells in vitro and in vivo. A significant inhibitory effect of IGU-PLGA-NPs on MM cancer cells and MM CSCs was demonstrated by the Cell Counting Kit-8 (CCK-8) assay. Treatment with IGU-PLGA-NPs induced significant cell cycle arrest at G1 in MM cells and reduced tumour colony formation in MM CSCs. Mechanistically, IGU-PLGA-NPs increase apoptosis in MM cells by activating Caspase-dependent signalling pathway to increase the levels of bax, cytochrome c (cyt-c), caspase-9 and caspase-3 proteins. Moreover, IGU-PLGA-NPs effectively increase ROS production assayed using a DCFH-DA fluorescent probe in MM cells. The data indicate that IGU-PLGA-NPs induce a significant reduction in the tumour volume and a marked increase in the survival rate in a mouse model of multiple myeloma. Overall, our findings indicate that IGU-PLGA-NPs are a potential therapeutic strategy that may contribute to the therapy of MM and elimination of MM CSCs in future clinical trials.
Collapse
Affiliation(s)
- Faming Wang
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Muhammad Younis
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao Road, Nanjing 210009, China; Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Yao Luo
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Le Zhang
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Liudi Yuan
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao Road, Nanjing 210009, China; Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.
| |
Collapse
|
108
|
Membranous Glomerulopathy After Autologous Hematopoietic Stem Cell Transplant in a Patient With Multiple Myeloma. Kidney Int Rep 2021; 6:2243-2245. [PMID: 34386678 PMCID: PMC8343790 DOI: 10.1016/j.ekir.2021.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/24/2021] [Indexed: 11/26/2022] Open
|
109
|
Nasr F, Ghoche AA, Diab S, Nasr L, Ammanouil E, Riachy C, Hallit S, Chahine G. Lebanese Real-World Experience in Treating Multiple Myeloma: A Multicenter Retrospective Study. Leuk Res Rep 2021; 15:100252. [PMID: 34141564 PMCID: PMC8188546 DOI: 10.1016/j.lrr.2021.100252] [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: 10/13/2020] [Revised: 03/29/2021] [Accepted: 05/23/2021] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE The present retrospective multicenter study aims at documenting characteristics of multiple myeloma (MM) patients and the effect of autologous stem cell transplant (ASCT) on survival. METHODS A total of 134 adult patients initiating any new MM therapy from January 2002 till December 2019 were included. Enrollment was stratified by disease subtype, induction protocol and transplant status. The characteristics and survival outcomes were recorded. RESULTS Mean age at diagnosis was 61.91 ± 10.83 years, with 62.7% male patients. Regarding the prognostic MM International Staging System (ISS), stage 3 was the most common at diagnosis with 50.8% of patients followed by stage 1 (25.4%) and stage 2 (23.8%). Maintenance treatment was given in 88.5% of the patients. 24.6% patients were transplanted, 41% were not and the remaining were unknown or still in induction. 86.1% of patients were alive at data cut off. A significantly higher mean progression free survival (PFS) was found in transplant patients (p=0.016). Using cox regression, creatinine >2 mg/dl (HR3.78) and hypercalcemia >11 mg/dl (HR=6.48) were significantly associated with a shorter PFS1. A significantly shorter overall survival (OS) was associated with hypercalcemia (HR=6.58), as well as male gender though not statistically significant in the latter. Difference in survival distributions by treatment was not statistically significant (bortezomib thalidomide dexamethasone (VTD) (p=0.211), bortezomib cyclophosphamide dexamethasone (VCD) (p=0.111) or bortezomib Revlimid dexamethasone (VRD) (p=0.312)). The interaction between ISS stage on diagnosis and transplant was not significantly associated with the overall survival. CONCLUSION The results of our retrospective study are in conformity with international data emphasizing the role of transplant in the treatment algorithm of newly diagnosed transplant-eligible multiple myeloma patients.
Collapse
Affiliation(s)
- Fadi Nasr
- Hotel Dieu de France University Hospital, Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
- Mount Lebanon university Hospital, oncology and hematology department, Hazmieh, Lebanon
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
| | - Ahmad Al Ghoche
- Mount Lebanon university Hospital, oncology and hematology department, Hazmieh, Lebanon
| | - Saada Diab
- Mount Lebanon university Hospital, oncology and hematology department, Hazmieh, Lebanon
| | - Lewis Nasr
- Hotel Dieu de France University Hospital, Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | - Emmanuel Ammanouil
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
| | - Christelle Riachy
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
| | - Souheil Hallit
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
| | - Georges Chahine
- Hotel Dieu de France University Hospital, Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
| |
Collapse
|
110
|
Koomen DC, Meads MB, Magaletti DM, Guingab-Cagmat JD, Oliveira PS, Fang B, Liu M, Welsh EA, Meke LE, Jiang Z, Hampton OA, Tungesvik A, De Avila G, Alugubelli RR, Nishihori T, Silva AS, Eschrich SA, Garrett TJ, Koomen JM, Shain KH. Metabolic Changes Are Associated with Melphalan Resistance in Multiple Myeloma. J Proteome Res 2021; 20:3134-3149. [PMID: 34014671 DOI: 10.1021/acs.jproteome.1c00022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple myeloma is an incurable hematological malignancy that impacts tens of thousands of people every year in the United States. Treatment for eligible patients involves induction, consolidation with stem cell rescue, and maintenance. High-dose therapy with a DNA alkylating agent, melphalan, remains the primary drug for consolidation therapy in conjunction with autologous stem-cell transplantation; as such, melphalan resistance remains a relevant clinical challenge. Here, we describe a proteometabolomic approach to examine mechanisms of acquired melphalan resistance in two cell line models. Drug metabolism, steady-state metabolomics, activity-based protein profiling (ABPP, data available at PRIDE: PXD019725), acute-treatment metabolomics, and western blot analyses have allowed us to further elucidate metabolic processes associated with melphalan resistance. Proteometabolomic data indicate that drug-resistant cells have higher levels of pentose phosphate pathway metabolites. Purine, pyrimidine, and glutathione metabolisms were commonly altered, and cell-line-specific changes in metabolite levels were observed, which could be linked to the differences in steady-state metabolism of naïve cells. Inhibition of selected enzymes in purine synthesis and pentose phosphate pathways was evaluated to determine their potential to improve melphalan's efficacy. The clinical relevance of these proteometabolomic leads was confirmed by comparison of tumor cell transcriptomes from newly diagnosed MM patients and patients with relapsed disease after treatment with high-dose melphalan and autologous stem-cell transplantation. The observation of common and cell-line-specific changes in metabolite levels suggests that omic approaches will be needed to fully examine melphalan resistance in patient specimens and define personalized strategies to optimize the use of high-dose melphalan.
Collapse
Affiliation(s)
- David C Koomen
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Mark B Meads
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Dario M Magaletti
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Joy D Guingab-Cagmat
- University of Florida College of Medicine, Gainesville, Florida 32610, United States
| | - Paula S Oliveira
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Bin Fang
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Min Liu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Eric A Welsh
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Laurel E Meke
- University of Florida College of Medicine, Gainesville, Florida 32610, United States
| | | | | | - Alexandre Tungesvik
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Gabriel De Avila
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | | | - Taiga Nishihori
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Ariosto S Silva
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Steven A Eschrich
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Timothy J Garrett
- University of Florida College of Medicine, Gainesville, Florida 32610, United States
| | - John M Koomen
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Kenneth H Shain
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| |
Collapse
|
111
|
Liang EC, Muffly LS, Shiraz P, Shizuru JA, Johnston L, Arai S, Frank MJ, Weng WK, Lowsky R, Rezvani A, Meyer EH, Negrin R, Miklos DB, Sidana S. Use of Backup Stem Cells for Stem Cell Boost and Second Transplant in Patients with Multiple Myeloma Undergoing Autologous Stem Cell Transplantation. Transplant Cell Ther 2021; 27:405.e1-405.e6. [PMID: 33775587 PMCID: PMC8113075 DOI: 10.1016/j.jtct.2021.02.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/10/2021] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
Autologous hematopoietic stem cell transplantation (ASCT) is a standard treatment for multiple myeloma (MM). Consensus guidelines recommend collecting sufficient stem cells in case there is a need for stem cell boost for delayed/poor engraftment or for future second ASCT. However, collecting and storing backup stem cells in all patients requires significant resources and cost, and the rates of backup stem cell utilization are not well studied. We sought to examine the utilization of backup stem cells (BSCs) in patients with MM undergoing ASCT. Patients with MM aged ≥18 years old who underwent first ASCT at our institution from January 2010 through December 2015 and collected sufficient stem cells for at least 2 transplants were included in this single-center retrospective study. This timeframe was selected to allow for adequate follow-up. A total of 393 patients were included. The median age was 58 years (range, 25-73). After a median follow-up of 6 years, the median progression-free survival (PFS) of the cohort was 3 years. Sixty-one percent (n = 240) of patients progressed or relapsed. Chemotherapy-based mobilization was used in almost all patients (98%). The median total CD34+ cells collected was 18.2 × 106/kg (range, 3.4-112.4). A median of 5.7 × 106 CD34+ cells/kg (range, 1.8-41.9) was infused during the first ASCT, and a median of 10.1 × 106 CD34+ cells/kg (range, 1.5-104.5) was cryopreserved for future use. Of the patients, 6.9% (n = 27) used backup stem cells, with 2.3% (n = 10) using them for stem cell boost, 4.6% (n = 18) for a second salvage ASCT, including 1 patient for both stem cell boost and second ASCT. Rates of backup stem cell use among patients aged <60, 60-69, and ≥70 years were 7.8%, 5.7%, and 5.9%, respectively. There was a trend toward higher rates of backup stem cell use for second ASCT in patients who were younger, had suboptimal disease control at time of first ASCT, and longer PFS. The median dose of stem cell boost given was 5.6 × 106 CD34+ cells/kg (range, 1.9-20). The median time from stem cell boost to neutrophil, hemoglobin, and platelet engraftment was 4 (range, 2-11), 15 (range, 4-34), and 12 (range, 0-34) days, respectively. Lower CD34+ dose and older age at time of ASCT predicted need for stem cell boost. With new salvage therapies for relapsed MM, the rates of second ASCT are very low. The low rates of use suggest that institutional policies regarding universal BSC collection and long-term storage should be reassessed and individualized. However, need for stem cell boost in 2.3% of patients may present a challenge to that.
Collapse
Affiliation(s)
- Emily C Liang
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Lori S Muffly
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Parveen Shiraz
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Judith A Shizuru
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Laura Johnston
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Sally Arai
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Matthew J Frank
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Wen-Kai Weng
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Robert Lowsky
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Andrew Rezvani
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Everett H Meyer
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Robert Negrin
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - David B Miklos
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Surbhi Sidana
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California..
| |
Collapse
|
112
|
Johnsrud A, Ladha A, Muffly L, Shiraz P, Goldstein G, Osgood V, Shizuru JA, Johnston L, Arai S, Weng WK, Lowsky R, Rezvani AR, Meyer EH, Frank MJ, Negrin RS, Miklos DB, Sidana S. Stem Cell Mobilization in Multiple Myeloma: Comparing Safety and Efficacy of Cyclophosphamide +/- Plerixafor versus Granulocyte Colony-Stimulating Factor +/- Plerixafor in the Lenalidomide Era. Transplant Cell Ther 2021; 27:590.e1-590.e8. [PMID: 33915323 DOI: 10.1016/j.jtct.2021.04.016] [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: 02/18/2021] [Revised: 04/05/2021] [Accepted: 04/18/2021] [Indexed: 11/17/2022]
Abstract
Growth factor and chemotherapy-based stem cell mobilization strategies are commonly used to treat patients with multiple myeloma. We retrospectively compared 398 patients mobilized between 2017 and 2020 using either cyclophosphamide (4 g/m2) plus granulocyte colony-stimulating factor (G-CSF) or G-CSF alone, with on demand plerixafor (PXF) in both groups. Although total CD34+ yield was higher after chemomobilization compared with G-CSF +/- PXF (median, 13.6 × 106/kg versus 4.4 × 106/kg; P < .01), achievement of ≥2 × 106 CD34+ cells (95% versus 93.7%; P = .61) and rates of mobilization failure (5% versus 6.3%; P = .61) were similar. Fewer patients required PXF with chemomobilization (12.3% versus 49.5%; P < .01), and apheresis sessions were fewer (median, 1 [range, 1 to 4] versus 2 [range, 1 to 5]). The rate of complications, including neutropenic fever, emergency department visits, and hospitalizations, was higher after chemomobilization (30% versus 7.4%; P < .01). Previous use of ≤6 cycles of lenalidomide did not impair cell yield in either group. The median cost of mobilization was 17.4% lower in the G-CSF +/- PXF group (P = .01). Between group differences in time to engraftment were not clinically significant. Given similar rates of successful mobilization, similar engraftment time, and less toxicity and lower costs compared with chemomobilization, G-CSF with on-demand PXF may be preferable in myeloma patients with adequate disease control and limited lenalidomide exposure.
Collapse
Affiliation(s)
- Andrew Johnsrud
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Abdullah Ladha
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California; Division of Hematology, University of Southern California, Los Angeles, California
| | - Lori Muffly
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Parveen Shiraz
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Gary Goldstein
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Victoria Osgood
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Judith A Shizuru
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Laura Johnston
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Sally Arai
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Wen-Kai Weng
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Robert Lowsky
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Andrew R Rezvani
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Everett H Meyer
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Matthew J Frank
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Robert S Negrin
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - David B Miklos
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California
| | - Surbhi Sidana
- Stanford Cancer Institute, Stanford, California; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, Stanford, California.
| |
Collapse
|
113
|
Poczta A, Rogalska A, Marczak A. Treatment of Multiple Myeloma and the Role of Melphalan in the Era of Modern Therapies-Current Research and Clinical Approaches. J Clin Med 2021; 10:1841. [PMID: 33922721 PMCID: PMC8123041 DOI: 10.3390/jcm10091841] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/01/2021] [Accepted: 04/16/2021] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) accounts for 10% of all hematological malignancies, and it is the second most common hematological neoplasm for which chemotherapy is an important pharmacological treatment. High dose melphalan followed by autologous stem cell transplantation remains the standard of treatment for transplant-eligible patients with MM. In this review, we describe aspects of the pharmacokinetics and pharmacodynamics of melphalan therapy and related compounds. In addition, we describe the use of melphalan in innovative therapies for the treatment of MM, including the development of drug carriers to reduce systemic toxicity, combination therapy to improve the effectiveness of cancer therapy, and the chemical modification of the melphalan molecule to improve antitumor activity.
Collapse
Affiliation(s)
- Anastazja Poczta
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, Institute of Biophysics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.R.); (A.M.)
| | | | | |
Collapse
|
114
|
Uckun FM. Overcoming the Immunosuppressive Tumor Microenvironment in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13092018. [PMID: 33922005 PMCID: PMC8122391 DOI: 10.3390/cancers13092018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary This article provides a comprehensive review of new and emerging treatment strategies against multiple myeloma that employ precision medicines and/or drugs capable of improving the ability of the immune system to prevent or slow down the progression of multiple myeloma. These rationally designed new treatment methods have the potential to change the therapeutic landscape in multiple myeloma and improve the long-term survival outcome. Abstract SeverFigurel cellular elements of the bone marrow (BM) microenvironment in multiple myeloma (MM) patients contribute to the immune evasion, proliferation, and drug resistance of MM cells, including myeloid-derived suppressor cells (MDSCs), tumor-associated M2-like, “alternatively activated” macrophages, CD38+ regulatory B-cells (Bregs), and regulatory T-cells (Tregs). These immunosuppressive elements in bidirectional and multi-directional crosstalk with each other inhibit both memory and cytotoxic effector T-cell populations as well as natural killer (NK) cells. Immunomodulatory imide drugs (IMiDs), protease inhibitors (PI), monoclonal antibodies (MoAb), adoptive T-cell/NK cell therapy, and inhibitors of anti-apoptotic signaling pathways have emerged as promising therapeutic platforms that can be employed in various combinations as part of a rationally designed immunomodulatory strategy against an immunosuppressive tumor microenvironment (TME) in MM. These platforms provide the foundation for a new therapeutic paradigm for achieving improved survival of high-risk newly diagnosed as well as relapsed/refractory MM patients. Here we review the scientific rationale and clinical proof of concept for each of these platforms.
Collapse
Affiliation(s)
- Fatih M. Uckun
- Norris Comprehensive Cancer Center and Childrens Center for Cancer and Blood Diseases, University of Southern California Keck School of Medicine (USC KSOM), Los Angeles, CA 90027, USA;
- Department of Developmental Therapeutics, Immunology, and Integrative Medicine, Drug Discovery Institute, Ares Pharmaceuticals, St. Paul, MN 55110, USA
- Reven Pharmaceuticals, Translational Oncology Program, Golden, CO 80401, USA
| |
Collapse
|
115
|
Prolonged Lenalidomide Therapy Does Not Impact Autologous Peripheral Blood Stem Cell Mobilization and Collection in Multiple Myeloma Patients: A Single-Center Retrospective Analysis. Transplant Cell Ther 2021; 27:661.e1-661.e6. [PMID: 33895403 DOI: 10.1016/j.jtct.2021.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/22/2021] [Accepted: 04/13/2021] [Indexed: 12/22/2022]
Abstract
Since the introduction of lenalidomide into induction therapy for multiple myeloma (MM), there have been conflicting reports about its impact on autologous peripheral blood stem cell (PBSC) mobilization. We evaluated the impact of previous lenalidomide exposure in a large cohort of patients with MM undergoing mobilization and collection at a tertiary stem cell transplantation center. We hypothesized that collection of PBSCs is feasible even with a prolonged duration of previous lenalidomide therapy. We examined patients with MM who attempted stem cell mobilization and collection, seen at our center between January 2012 and July 2015. The patients were categorized into 3 groups for analysis: (1) patients with previous receipt of >6 cycles lenalidomide, (2) patients with previous receipt of ≤6 cycles of lenalidomide, and (3) patients without previous lenalidomide exposure. We compared collection yields and days of apheresis among the 3 groups using linear regression analysis. We identified 297 patients with MM who underwent mobilization of PBSCs. Of these, 35 had received >6 cycles of lenalidomide (median, 8 cycles; range, 7 to 25 cycles), 156 had received ≤6 cycles of lenalidomide (median, 4 cycles; range, 1 to 6 cycles), and 106 had received no lenalidomide. Prior lenalidomide exposure did not have a statistically significant impact on the absolute number of CD34+ cells collected or on the duration of collection based on a multivariate linear regression analysis for association between receipt of >6 cycles of lenalidomide. In this retrospective analysis of MM patients undergoing autologous PBSC transplantation, we show that the duration of previous lenalidomide exposure does not impact the total number of PBSCs collected or the number of days of apheresis. These data suggest that longer courses of induction therapy with lenalidomide-containing regimens to achieve a maximum response can be safe without impairing the ability to collect PBSCs, and that limiting lenalidomide use before mobilization does not appear warranted in all cases.
Collapse
|
116
|
Lim JY, Kim TW, Ryu DB, Park SS, Lee SE, Kim BS, Min CK. Myeloma-Secreted Galectin-1 Potently Interacts with CD304 on Monocytic Myeloid-Derived Suppressor Cells. Cancer Immunol Res 2021; 9:503-513. [PMID: 33771821 DOI: 10.1158/2326-6066.cir-20-0663] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/11/2021] [Accepted: 03/24/2021] [Indexed: 11/16/2022]
Abstract
Progression of multiple myeloma is regulated by factors intrinsic to the clonal plasma cells (PC) and by the immune effector cells in the tumor microenvironment. In this study, we investigated the interaction between CD304 expression on myeloid-derived suppressor cells (MDSC) and galectin-1 from malignant PCs in the context of autologous stem cell transplantation (ASCT) for multiple myeloma. Using high-throughput screening, CD304 expression on circulating monocytic MDSCs (M-MDSC; CD14+HLA-DRlow/-) was compared before and after ASCT. There was a significantly higher M-MDSC expression of CD304 before ASCT and a clear correlation between circulating pre-ASCT M-MDSC frequency and serum galectin-1 concentration. Treatment of pre-ASCT M-MDSCs, but not post-ASCT M-MDSCs, with galectin-1 in vitro expanded the M-MDSC population and increased expression of CD304. High galectin-1 expression by malignant PCs was associated with poor clinical outcomes. M-MDSC development and expression of CD304 were differentially induced when healthy donor peripheral blood mononuclear cells were cultured with the human multiple myeloma cell lines RPMI-8226 and JJN3, which express high and low galectin-1, respectively. Inhibition of galectin-1 reduced M-MDSC proliferation induced by RPMI-8226 cells but not by JJN3 cells, and blockade of CD304 reduced M-MDSC migration induced by RPMI-8226 cells but not by JJN3 cells. In addition, blockade of CD304 reversed suppression of the in vitro cytotoxic effect of melphalan by pre-ASCT M-MDSCs. Our data demonstrate that multiple myeloma-derived galectin-1 could mediate the tumor-promoting effect of M-MDSCs through its interaction with CD304 on M-MDSCs and contribute to multiple myeloma progression after ASCT.See related Spotlight on p. 488.
Collapse
Affiliation(s)
- Ji-Young Lim
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Tae-Woo Kim
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Da-Bin Ryu
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sung-Soo Park
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Byung-Soo Kim
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
117
|
Oral ixazomib, lenalidomide, and dexamethasone for transplant-ineligible patients with newly diagnosed multiple myeloma. Blood 2021; 137:3616-3628. [PMID: 33763699 PMCID: PMC8462404 DOI: 10.1182/blood.2020008787] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 02/05/2021] [Indexed: 01/14/2023] Open
Abstract
Addition of ixazomib to Rd in nontransplant NDMM patients resulted in a nonstatistically significant increase in PFS (HR, 0.830; P = .073). Ixazomib-Rd is a feasible and tolerable all-oral triplet regimen in this setting, with a well-characterized and manageable safety profile.
Continuous lenalidomide-dexamethasone (Rd)-based regimens are among the standards of care in transplant-ineligible newly diagnosed multiple myeloma (NDMM) patients. The oral proteasome inhibitor ixazomib is suitable for continuous dosing, with predictable, manageable toxicities. In the double-blind, placebo-controlled TOURMALINE-MM2 trial, transplant-ineligible NDMM patients were randomized to ixazomib 4 mg (n = 351) or placebo (n = 354) plus Rd. After 18 cycles, dexamethasone was discontinued and treatment was continued using reduced-dose ixazomib (3 mg) and lenalidomide (10 mg) until progression/toxicity. The primary endpoint was progression-free survival (PFS). Median PFS was 35.3 vs 21.8 months with ixazomib-Rd vs placebo-Rd, respectively (hazard ratio [HR], 0.830; 95% confidence interval, 0.676-1.018; P = .073; median follow-up, 53.3 and 55.8 months). Complete (26% vs 14%; odds ratio [OR], 2.10; P < .001) and ≥ very good partial response (63% vs 48%; OR, 1.87; P < .001) rates were higher with ixazomib-Rd vs placebo-Rd. In a prespecified high-risk cytogenetics subgroup, median PFS was 23.8 vs 18.0 months (HR, 0.690; P = .019). Overall, treatment-emergent adverse events (TEAEs) were mostly grade 1/2. With ixazomib-Rd vs placebo-Rd, 88% vs 81% of patients experienced grade ≥3 TEAEs, 66% vs 62% serious TEAEs, and 35% vs 27% TEAEs resulting in regimen discontinuation; 8% vs 6% died on study. Addition of ixazomib to Rd was tolerable with no new safety signals and led to a clinically meaningful PFS benefit of 13.5 months. Ixazomib-Rd is a feasible option for certain patients who can benefit from an all-oral triplet combination. This trial was registered at www.clinicaltrials.gov as #NCT01850524.
Collapse
|
118
|
Up-front ixazomib in older patients with myeloma. Blood 2021; 137:3584-3586. [PMID: 34196682 DOI: 10.1182/blood.2021011262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
119
|
Lopes R, Ferreira BV, Caetano J, Barahona F, Carneiro EA, João C. Boosting Immunity against Multiple Myeloma. Cancers (Basel) 2021; 13:1221. [PMID: 33799565 PMCID: PMC8001641 DOI: 10.3390/cancers13061221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/12/2021] [Accepted: 03/01/2021] [Indexed: 01/10/2023] Open
Abstract
Despite the improvement of patient's outcome obtained by the current use of immunomodulatory drugs, proteasome inhibitors or anti-CD38 monoclonal antibodies, multiple myeloma (MM) remains an incurable disease. More recently, the testing in clinical trials of novel drugs such as anti-BCMA CAR-T cells, antibody-drug conjugates or bispecific antibodies broadened the possibility of improving patients' survival. However, thus far, these treatment strategies have not been able to steadily eliminate all malignant cells, and the aim has been to induce a long-term complete response with minimal residual disease (MRD)-negative status. In this sense, approaches that target not only myeloma cells but also the surrounding microenvironment are promising strategies to achieve a sustained MRD negativity with prolonged survival. This review provides an overview of current and future strategies used for immunomodulation of MM focusing on the impact on bone marrow (BM) immunome.
Collapse
Affiliation(s)
- Raquel Lopes
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Bruna Velosa Ferreira
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Joana Caetano
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Filipa Barahona
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
| | - Cristina João
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
120
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021. [DOI: 10.37349/etat.2020.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3University of Montpellier, UFR Medicine, 34093 Montpellier, France 4 Institut Universitaire de France (IUF), 75000 Paris France
| |
Collapse
|
121
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:65-106. [PMID: 36046090 PMCID: PMC9400753 DOI: 10.37349/etat.2021.00034] [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: 10/17/2020] [Accepted: 01/06/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3UFR Medicine, University of Montpellier, 34093 Montpellier, France 4Institut Universitaire de France (IUF), 75000 Paris, France
| |
Collapse
|
122
|
Low-Dose Cyclophosphamide versus Intermediate-High-Dose Cyclophosphamide versus Granulocyte Colony-Stimulating Factor Alone for Stem Cell Mobilization in Multiple Myeloma in the Era of Novel Agents: A Multicenter Retrospective Study. Transplant Cell Ther 2021; 27:244.e1-244.e8. [PMID: 33781522 DOI: 10.1016/j.jtct.2020.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/13/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022]
Abstract
The optimal stem cell (SC) mobilization strategy for patients with multiple myeloma (MM) remains a matter of debate. Possible approaches include low or high doses of cyclophosphamide (Cy), other chemotherapeutic agents, or granulocyte colony-stimulating factor (G-CSF) alone. The scope of the study was to compare low-dose Cy plus G-CSF versus intermediate-high-dose Cy plus G-CSF versus G-CSF alone for SC mobilization in MM, in terms of efficacy and safety. We retrospectively analyzed 422 MM patients undergoing SC mobilization in 6 Italian centers, including 188 patients who received low-dose Cy (LD-Cy group, defined as 2 g/m2), 163 patients who received intermediate-high-dose Cy (HD-Cy group, defined as ≥ 3 g/m2), and 71 patients who received G-CSF alone (G-CSF group). The median peak of circulating CD34+ cells was 77/µL in the LD-Cy group, 92/µL in the HD-Cy group, and 55/µL in the G-CSF group (P = .0001). The median amount of SCs collected was 9.1 × 106/kg, 9.7 × 106/kg, and 5.6 × 106/kg in the 3 groups, respectively (P = .0001). The rate of mobilization failure (defined as failure to collect ≥2 × 106/kg) was 3.7% in the LD-Cy group, 3.4% in the HD-Cy group, and 4.3% in the G-CSF group (P = .9). The target SC dose of at least 4 × 106/kg was reached in 90.4%, 91.1%, and 78.6% of the patients in these 3 groups, respectively (P = .014). The "on demand" use of plerixafor was higher in the G-CSF group (76%) compared with the LD-Cy group (19%) and the HD-Cy group (6%). In multivariate analysis, G-CSF mobilization and previous use of melphalan or radiotherapy were independently associated with failure to collect the target SC dose of ≥4 × 106/kg. No impacts of age, blood counts, or previous treatment with lenalidomide, bortezomib, or carfilzomib were observed. Our results suggest that LD-Cy may be considered for successful SC mobilization in patients with MM.
Collapse
|
123
|
Belantamab Mafodotin to Treat Multiple Myeloma: A Comprehensive Review of Disease, Drug Efficacy and Side Effects. ACTA ACUST UNITED AC 2021; 28:640-660. [PMID: 33494319 PMCID: PMC7924384 DOI: 10.3390/curroncol28010063] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/09/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by excessive clonal proliferation of plasma cells. The treatment of multiple myeloma presents a variety of unique challenges due to the complex molecular pathophysiology and incurable status of the disease at this time. Given that MM is the second most common blood cancer with a characteristic and unavoidable relapse/refractory state during the course of the disease, the development of new therapeutic modalities is crucial. Belantamab mafodotin (belamaf, GSK2857916) is a first-in-class therapeutic, indicated for patients who have previously attempted four other treatments, including an anti-CD38 monoclonal antibody, a proteosome inhibitor, and an immunomodulatory agent. In November 2017, the FDA designated belamaf as a breakthrough therapy for heavily pretreated patients with relapsed/refractory multiple myeloma. In August 2020, the FDA granted accelerated approval as a monotherapy for relapsed or treatment-refractory multiple myeloma. The drug was also approved in the EU for this indication in late August 2020. Of note, belamaf is associated with the following adverse events: decreased platelets, corneal disease, decreased or blurred vision, anemia, infusion-related reactions, pyrexia, and fetal risk, among others. Further studies are necessary to evaluate efficacy in comparison to other standard treatment modalities and as future drugs in this class are developed.
Collapse
|
124
|
Gut Microbiota Influence in Hematological Malignancies: From Genesis to Cure. Int J Mol Sci 2021; 22:ijms22031026. [PMID: 33498529 PMCID: PMC7864170 DOI: 10.3390/ijms22031026] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Hematological malignancies, including multiple myeloma, lymphoma, and leukemia, are a heterogeneous group of neoplasms that affect the blood, bone marrow, and lymph nodes. They originate from uncontrolled growth of hematopoietic and lymphoid cells from different stages in their maturation/differentiation and account for 6.5% of all cancers around the world. During the last decade, it has been proven that the gut microbiota, more specifically the gastrointestinal commensal bacteria, is implicated in the genesis and progression of many diseases. The immune-modulating effects of the human microbiota extend well beyond the gut, mostly through the small molecules they produce. This review aims to summarize the current knowledge of the role of the microbiota in modulating the immune system, its role in hematological malignancies, and its influence on different therapies for these diseases, including autologous and allogeneic stem cell transplantation, chemotherapy, and chimeric antigen receptor T cells.
Collapse
|
125
|
Treatment pathways and disease journeys differ before and after introduction of novel agents in newly diagnosed multiple myeloma in Taiwan. Sci Rep 2021; 11:1112. [PMID: 33441901 PMCID: PMC7806818 DOI: 10.1038/s41598-020-80607-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/17/2020] [Indexed: 01/10/2023] Open
Abstract
Limited real-world data are available regarding treatment practices and outcomes of multiple myeloma (MM) in Asia. We conducted a retrospective cohort study using the Taiwan National Healthcare Insurance Research database and Taiwan Death Registry and used a Markov model to describe disease progression and outcomes in 4092 patients newly diagnosed with MM from 01-Jan-2007 to 31-Dec-2015. We observed marked differences in the characteristics, length and outcome of the clinical journey between patients who did/did not receive autologous stem cell transplant, and between patients initiated on novel agents versus other treatment regimens. In transplant recipients, initiation with combined thalidomide + bortezomib increased over time (12.2–77.5%). Progression-free survival after first-line treatment improved and a lower percentage of patients died. Lenalidomide in second and third-line regimens increased (15.5–31.5%). In non-transplanted patients, initiation with novel agents increased (17.5–54.6%), but death rates remained high (60.3%). The treatment landscape of MM in Taiwan has evolved, with increased use of combined bortezomib + thalidomide for first-line and lenalidomide for second/third-line but many patients die before receiving second-line treatment. Novel agents with different modes of action should be used as early as possible to maximize their benefits. Improved MM treatments remains a critical medical need.
Collapse
|
126
|
ALADAĞ E, DEMİROĞLU H, BÜYÜKAŞIK Y, TURGUT M, AKSU S, SAYINALP N, HAZNEDAROĞLU İC, ÖZCEBE Oİ, GÖKER H. CD34+ hematopoietic progenitor cell dose as a predictor of engraftment and survival in multiple myeloma patients undergoing autologous stem cell transplantation. Turk J Med Sci 2020; 50:1851-1856. [PMID: 32512672 PMCID: PMC7775700 DOI: 10.3906/sag-2001-173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/05/2020] [Indexed: 01/11/2023] Open
Abstract
Background/aim High-dose melphalan and autologous hematopoietic stem cell transplantation (AHSCT) is the standard treatment strategy for multiple myeloma (MM) patients who are eligible for it. The recommended dose of CD34+ hematopoietic progenitor cells (HPCs) for adequate engraftment is above 2 × 106/kg. The aim of this study was to evaluate the relationship between the dose of CD34+HPCs and survival in MM patients who underwent AHSCT at a tertiary care center. Materials and methods Enrolled in this study were 271 MM patients who underwent AHSCT between 2003 and 2019. Clinical characteristics of the patients, disease status pre-AHSCT, reinfused CD34+ cell doses, and neutrophil and platelet engraftment days were recorded, retrospectively. The patients were divided into 2 groups according to whether the dose of reinfused CD 34+ HPCs was <5 × 106/kg or ≥5 × 106/kg. The groups were compared in terms of engraftment and overall survival (OS) times. Results The median age of the patients was 54.8 (33–76) years. The median dose of infused CD34+ HPCs was 5.94 × 106/kg (1.47–59.5 × 106/kg). The median follow-up period was 54 months (4–211). The median OS of the patients was 103 months (11–144). The median neutrophil and platelet engraftment time was 10 (8–24) and 11 (7–40) days. Doses of <5 × 106/kg and ≥5 × 106/kg CD34+ HPC were reinfused in 38.1% and 61.9% of the patients, respectively. There was a negative significant correlation between the reinfused CD34+cell level and neutrophil/platelet engraftment times (r = –0.32, P < 0.001; r = –0.27, P < 0.001, respectively). The median OS times were observed as 103 months (11–144) and 145 months (123–166) for patients who had been administered <5 × 106/kg and ≥5 × 106/kg of CD34+ HPCs, respectively (P = 0.009). Conclusion The increased amount of CD34+ autologous hematopoietic stem cell dose after high dose melphalan chemotherapy in MM patients shortened the platelet and neutrophil engraftment time and increased OS. Early platelet engraftment and administration of a CD34+ HPC count that is ≥5 × 106/kg can be considered as predictors of better survival in patients.
Collapse
Affiliation(s)
- Elifcan ALADAĞ
- Department of Hematology, Faculty of Medicine, Hacettepe University, AnkaraTurkey
| | - Haluk DEMİROĞLU
- Department of Hematology, Faculty of Medicine, Hacettepe University, AnkaraTurkey
| | - Yahya BÜYÜKAŞIK
- Department of Hematology, Faculty of Medicine, Hacettepe University, AnkaraTurkey
| | - Mehmet TURGUT
- Department of Hematology, Faculty of Medicine, Ondokuz Mayıs University, SamsunTurkey
| | - Salih AKSU
- Department of Hematology, Faculty of Medicine, Hacettepe University, AnkaraTurkey
| | - Nilgün SAYINALP
- Department of Hematology, Faculty of Medicine, Hacettepe University, AnkaraTurkey
| | | | - Osman İlhami ÖZCEBE
- Department of Hematology, Faculty of Medicine, Hacettepe University, AnkaraTurkey
| | - Hakan GÖKER
- Department of Hematology, Faculty of Medicine, Hacettepe University, AnkaraTurkey
| |
Collapse
|
127
|
Piriyakhuntorn P, Tantiworawit A, Rattanathammethee T, Hantrakool S, Chai-Adisaksopha C, Rattarittamrong E, Norasetthada L. Outcomes of Non-Cryopreserved Versus Cryopreserved Peripheral Blood Stem Cells for Autologous Stem Cell Transplantation in Multiple Myeloma. Ann Transplant 2020; 25:e927084. [PMID: 33303730 PMCID: PMC7737409 DOI: 10.12659/aot.927084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Autologous stem cell transplantation (ASCT) has become a standard procedure in multiple myeloma (MM) patients. Cryopreservation (CRYO) of stem cells may be associated with adverse reactions of dimethyl sulfoxide. Previous studies showed that stem cell storage at 4°C (non-cryopreserved [NC] method) may have some advantages. This analysis focused on comparing the transplant-related outcomes of the 2 preservation methods. MATERIAL AND METHODS This was a cohort study of consecutive MM patients who underwent ASCT at Chiang Mai University from 2014 to 2019. Primary outcomes were time to neutrophil and platelet engraftment. Key secondary outcomes were the incidence of infusion reactions, duration of hospitalization, cost, and survival. RESULTS A total of 42 MM patients underwent ASCT. Of these, 26 patients and 16 patients underwent NC and CRYO stem cell collections, respectively. There was no difference in time to neutrophil engraftment (median 12 vs. 10.5 days, P=0.203) or platelet engraftment (median 14 vs. 12 days, P=0.809) between groups. The incidence of infusion reactions and duration of hospitalization were similar in both groups. The average cost of ASCT was 10% lower in the NC group. There was no difference in progression-free survival (median 16 vs. 22 months, P=0.701) or overall survival between NC and CRYO groups. CONCLUSIONS ASCT in MM using the NC preservation method is effective and safe compared to the CRYO method in both short-term and survival outcomes.
Collapse
Affiliation(s)
- Pokpong Piriyakhuntorn
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Adisak Tantiworawit
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanawat Rattanathammethee
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sasinee Hantrakool
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chatree Chai-Adisaksopha
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Ekarat Rattarittamrong
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Lalita Norasetthada
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
128
|
Liam CCK, Boo YL, Lee YL, Law KB, Ho KW, Shahnaz SAKS, Tan JTC, Lau NS, Ong TC, Tan SM, Sathar J. Autologous stem cell transplantation (ASCT) outcome for multiple myeloma in a tertiary referral centre in Malaysia. BLOOD CELL THERAPY 2020; 4:1-8. [PMID: 36712843 PMCID: PMC9851782 DOI: 10.31547/bct-2020-009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/15/2020] [Indexed: 02/01/2023]
Abstract
Background Multiple Myeloma (MM) is characterized by the presence of clonal plasma cells. These often result in complications including bone destruction, hypercalcemia, renal insufficiency, and anaemia. Induction with a triplet or quadruplet regimen followed by autologous stem cell transplantation (ASCT) has been the standard of care for transplant eligible patients to achieve durable remission. Purpose This is a retrospective analytical study to determine the outcome of Multiple Myeloma patients who underwent ASCT in Ampang Hospital. Materials and Methods We included a 5-year cohort of patients transplanted from 1st July 2014 to 30th June 2019. Data were obtained through electronic medical records. Prognostic factors for progression-free survival (PFS) and overall survival (OS) were analyzed using simple and multiple Cox proportional hazard regression analysis. All analyses were done using software R version 3.6.2 with validated statistical packages. Results 139 patients were analyzed. The median age at transplant was 56 years old and 56.1% are males (n=78). The most common subtype is IgG Kappa (n=67, 48.2%). Only 93 patients in which the International Staging System (ISS) could be determined, and among them, 33.3% of patients (n=31) have advanced stage Ⅲ disease. The most common induction received before ASCT was a bortezomib based regimen and/or an immunomodulatory (IMiD) based regimen. 63.3% of patients achieved at least a very good partial response (VGPR) before ASCT. Most patients received myeloablative conditioning (MAC) (n=119, 85.6%). The mean cell dose is 3.68×106/kg. The median time to engraftment was 11 days for both platelet and absolute neutrophil count (ANC). With the median follow-up of 17.3 (range, 6.2-33.4) months, the median OS and PFS were not reached. The 1-year and 2-year PFS were 75% (95% CI 66-82%) and 52% (95% CI 42-62%), respectively. The 1-year and 2-year OS were 82% (95% CI 74-88%) and 70% (95% CI 60-78%), respectively. 6 patients (4.3%) had transplant-related mortality (TRM). IgA subtype was found to adversely affect PFS. Maintenance therapy and the absence of renal impairment was associated with better PFS and OS. Discussion and Conclusions Our study found that ASCT following induction treatment is safe and beneficial to achieve a deeper remission status. In our study, the addition of maintenance therapy is associated with an improved outcome in PFS and OS.
Collapse
Affiliation(s)
| | - Yang Liang Boo
- Department of Haematology, Ampang Hospital, Selangor, Malaysia
| | - Yi Lin Lee
- Centre for Clinical Trial, Ampang Hospital, Selangor, Malaysia
| | - Kian Boon Law
- Centre for Clinical Trial, Ampang Hospital, Selangor, Malaysia
| | - Kim Wah Ho
- Department of Haematology, Ampang Hospital, Selangor, Malaysia
| | | | | | - Ngee Siang Lau
- Department of Haematology, Ampang Hospital, Selangor, Malaysia
| | - Tee Chuan Ong
- Department of Haematology, Ampang Hospital, Selangor, Malaysia
| | - Sen Mui Tan
- Department of Haematology, Ampang Hospital, Selangor, Malaysia
| | - Jameela Sathar
- Department of Haematology, Ampang Hospital, Selangor, Malaysia
| |
Collapse
|
129
|
Hanafi HHW, Husin A, Yaacob NM, Abdullah AD. Relapsed/Progressive Disease and Its Prognostic Factors among Multiple Myeloma Patients Receiving Novel Agent Treatment in North East Peninsular Malaysia: A Single Centre Experience. Malays J Med Sci 2020; 27:62-77. [PMID: 33154703 PMCID: PMC7605841 DOI: 10.21315/mjms2020.27.5.7] [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: 03/11/2020] [Accepted: 08/10/2020] [Indexed: 11/25/2022] Open
Abstract
Background Some multiple myeloma (MM) patients still relapse/progress despite novel agent therapy and relapse/progression in MM is therefore a vital area of ongoing research in the novel treatment era. This retrospective cohort study aimed to evaluate the time to relapse/progression (TTP) among MM patients who received novel agents and to determine the associated prognostic factors. Methods This study included 89 MM patients treated at Hospital Universiti Sains Malaysia. We analysed the TTP and the type of relapse/progression (biochemical versus clinical), and a Cox proportional hazard model was used to identify the significant prognostic factors. Results Sixty-four percent of patients had biochemical relapse/progression. The overall median TTP among MM patients who received the novel agent(s) was 29.33 months (95% CI: 21.36–37.29). The type of paraprotein at diagnosis (P = 0.026, P = 0.228), International Staging System (ISS) score (P = 0.036, P = 0.067) and autologous stem cell transplant (ASCT) (P = 0.002) were prognostic factors for relapse/progression by simple Cox regression, but ASCT was the only significant predictor detected by multiple Cox regression (P = 0.003). Conclusion Our study reflects the importance of paraprotein monitoring to detect early features of relapse/progression. ASCT is the most prognostic factor that may lengthen the TTP.
Collapse
Affiliation(s)
- Hany Haqimi Wan Hanafi
- Haematopoietic Stem Cell Transplant Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Azlan Husin
- Haematopoietic Stem Cell Transplant Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Najib Majdi Yaacob
- Unit of Biostatistics and Research Methodology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Abu Dzarr Abdullah
- Haematopoietic Stem Cell Transplant Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
130
|
Gatenby RA, Brown JS. Integrating evolutionary dynamics into cancer therapy. Nat Rev Clin Oncol 2020; 17:675-686. [PMID: 32699310 DOI: 10.1038/s41571-020-0411-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 12/28/2022]
Abstract
Many effective drugs for metastatic and/or advanced-stage cancers have been developed over the past decade, although the evolution of resistance remains the major barrier to disease control or cure. In large, diverse populations such as the cells that compose metastatic cancers, the emergence of cells that are resistant or that can quickly develop resistance is virtually inevitable and most likely cannot be prevented. However, clinically significant resistance occurs only when the pre-existing resistant phenotypes are able to proliferate extensively, a process governed by eco-evolutionary dynamics. Attempts to disrupt the molecular mechanisms of resistance have generally been unsuccessful in clinical practice. In this Review, we focus on the Darwinian processes driving the eco-evolutionary dynamics of treatment-resistant cancer populations. We describe a variety of evolutionarily informed strategies designed to increase the probability of disease control or cure by anticipating and steering the evolutionary dynamics of acquired resistance.
Collapse
Affiliation(s)
- Robert A Gatenby
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA.
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, Tampa, FL, USA.
- Diagnostic Imaging Department, Moffitt Cancer Center, Tampa, FL, USA.
| | - Joel S Brown
- Cancer Biology and Evolution Program, Moffitt Cancer Center, Tampa, FL, USA
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, Tampa, FL, USA
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
131
|
Sweiss K, Vemu B, Hofmeister CC, Wenzler E, Calip GS, Galvin JP, Mahmud N, Rondelli D, Johnson JJ, Patel P. Development of a method for clinical pharmacokinetic testing to allow for targeted Melphalan dosing in multiple myeloma patients undergoing autologous transplant. Br J Clin Pharmacol 2020; 86:2165-2173. [PMID: 32285957 PMCID: PMC7576633 DOI: 10.1111/bcp.14308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 11/28/2022] Open
Abstract
AIMS High dose melphalan (HDM) and autologous stem cell transplant (ASCT) is standard of care for multiple myeloma (MM), but there is significant variability in melphalan exposure (area under the plasma drug concentration-time curve, AUC) when using body surface area-based dosing. Our aim was to establish a method of pharmacokinetic (PK) testing for real-time melphalan dose adjustments. METHODS We performed a prospective PK study of melphalan 140 or 200 mg/m2 in MM patients undergoing ASCT. Twenty MM patients were administered HDM on days -2 and - 1, with PK sampling at 8-10 time points. PK testing was performed on day -2 in all patients, and on day -1 in 5 patients. RESULTS Less than 20% interpatient variation in the day -2 and - 1 AUC was observed. The day -2 range in AUC (4.95-11.28 mg h/L) confirmed significant interpatient variability. The hypothetical total dose ranged from 133-302 mg/m2 to achieve the total median AUC. A 4-time point AUC (0, 30, 150 and 240 min) highly correlated with the AUC from the 8-time point schedule. A higher AUC correlated with increased risk of febrile neutropenia (P = .05). CONCLUSION Here we outline the methods to establish novel melphalan dosing using PK testing in MM patients undergoing ASCT to target a desired melphalan AUC.
Collapse
Affiliation(s)
- Karen Sweiss
- Department of Pharmacy PracticeUniversity of Illinois at ChicagoChicagoILUSA
- Cancer CenterUniversity of IllinoisChicagoILUSA
| | - Bhaskar Vemu
- Department of Pharmacy PracticeUniversity of Illinois at ChicagoChicagoILUSA
| | - Craig C. Hofmeister
- Department of Hematology and Medical OncologyWinship Cancer Institute of Emory UniversityAtlantaGAUSA
| | - Eric Wenzler
- Department of Pharmacy PracticeUniversity of Illinois at ChicagoChicagoILUSA
| | - Gregory Sampang Calip
- Cancer CenterUniversity of IllinoisChicagoILUSA
- Center for Pharmacoepidemiology and Pharmacoeconomic ResearchUniversity of Illinois at ChicagoChicagoILUSA
| | - John P. Galvin
- Cancer CenterUniversity of IllinoisChicagoILUSA
- Division of Hematology/OncologyUniversity of Illinois at ChicagoChicagoILUSA
| | - Nadim Mahmud
- Cancer CenterUniversity of IllinoisChicagoILUSA
- Division of Hematology/OncologyUniversity of Illinois at ChicagoChicagoILUSA
| | - Damiano Rondelli
- Cancer CenterUniversity of IllinoisChicagoILUSA
- Division of Hematology/OncologyUniversity of Illinois at ChicagoChicagoILUSA
| | - Jeremy James Johnson
- Department of Pharmacy PracticeUniversity of Illinois at ChicagoChicagoILUSA
- Cancer CenterUniversity of IllinoisChicagoILUSA
| | - Pritesh Patel
- Cancer CenterUniversity of IllinoisChicagoILUSA
- Division of Hematology/OncologyUniversity of Illinois at ChicagoChicagoILUSA
| |
Collapse
|
132
|
Betticher C, Bacher U, Legros M, Zimmerli S, Banz Y, Mansouri Taleghani B, Pabst T. Prophylactic corticosteroid use prevents engraftment syndrome in patients after autologous stem cell transplantation. Hematol Oncol 2020; 39:97-104. [PMID: 32979278 DOI: 10.1002/hon.2813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 11/08/2022]
Abstract
Engraftment syndrome (ES) following autologous stem cell transplantation (ASCT) at the time of neutrophil recovery may comprise fever, rash, pulmonary edema, or diarrhea. Usually, ES is easily manageable using corticosteroids but may prolong hospitalization. In two consecutive cohorts of subsequent patients with myeloma, lymphomas, and testicular/germ cell cancer, we assessed the benefit of corticosteroid use to prevent incidence and severity of ES following ASCT. Whereas Cohort A (82 patients) received no prophylactic corticosteroids, corticosteroids (4 mg dexamethasone oral daily) were started in Cohort B (60 patients) at day +9 until day +13 following ASCT. Steroid prophylaxis significantly reduced the incidence of ES (6/60; 10% vs. 33/82; 40%; p < 0.001). Hospitalization duration was longer in patients with ES than in patients without ES within both cohorts (in Cohort A: p = 0.007; and B: p = 0.011), but did not differ significantly between cohorts A and B. Finally, in Cohort A, there was a trend to an inferior 2-year overall survival rate in patients without ES compared to patients with ES (p = 0.067), but definite conclusions are not yet allowed. Our results suggest that corticosteroid prophylaxis from days +9 to +13 following ASCT significantly reduces the risk of ES and shortens hospitalization duration.
Collapse
Affiliation(s)
- Christophe Betticher
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ulrike Bacher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Myriam Legros
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stefan Zimmerli
- Institute of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Behrouz Mansouri Taleghani
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
133
|
Mateos MV, Bladé J, Bringhen S, Ocio EM, Efebera Y, Pour L, Gay F, Sonneveld P, Gullbo J, Richardson PG. Melflufen: A Peptide-Drug Conjugate for the Treatment of Multiple Myeloma. J Clin Med 2020; 9:E3120. [PMID: 32992506 PMCID: PMC7601491 DOI: 10.3390/jcm9103120] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/15/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the availability of new therapies that have led to improved outcomes for patients with multiple myeloma, most patients will eventually relapse. With triplet and even quadruplet combination therapies becoming standard in the first and second line, many patients will have few treatment options after second-line treatment. Melflufen (melphalan flufenamide) is a first-in-class peptide-drug conjugate (PDC) that targets aminopeptidases and rapidly releases alkylating agents into tumor cells. Once inside the tumor cells, melflufen is hydrolyzed by peptidases to release alkylator molecules, which become entrapped. Melflufen showed anti-myeloma activity in myeloma cells that were resistant to bortezomib and the alkylator melphalan. In early phase studies (O-12-M1 and HORIZON [OP-106]), melflufen plus dexamethasone has demonstrated encouraging clinical activity and a manageable safety profile in heavily pretreated patients with relapsed/refractory multiple myeloma, including those with triple-class refractory disease and extramedullary disease. The Phase III OCEAN study (OP-104) is further evaluating melflufen plus dexamethasone in patients with relapsed/refractory multiple myeloma. The safety profile of melflufen is characterized primarily by clinically manageable hematologic adverse events. Melflufen, with its novel mechanism of action, has the potential to provide clinically meaningful benefits to patients with relapsed/refractory multiple myeloma, including those with high unmet needs.
Collapse
Affiliation(s)
| | - Joan Bladé
- Hematology Department, Institut d’Investigacions Biomèdiques August Pi I Sunyer, Hospital Clinic, 08036 Barcelona, Spain;
| | - Sara Bringhen
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (S.B.); (F.G.)
| | - Enrique M Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain;
| | - Yvonne Efebera
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
| | - Luděk Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 62500 Brno, Czech Republic;
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (S.B.); (F.G.)
| | - Pieter Sonneveld
- Erasmus MC Cancer Institute, 3075 EA Rotterdam, The Netherlands;
| | - Joachim Gullbo
- Department of Medical Sciences, Division of Clinical Pharmacology, Uppsala University, 751 85 Uppsala, Sweden;
| | - Paul G. Richardson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
134
|
Daratumumab: A Review in Combination Therapy for Transplant-Eligible Newly Diagnosed Multiple Myeloma. Drugs 2020; 80:1455-1464. [DOI: 10.1007/s40265-020-01385-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
135
|
Atrash S, Bhutani M, Paul B, Voorhees PM, Usmani SZ. Management of newly diagnosed transplant ineligible multiple myeloma. Leuk Lymphoma 2020; 61:2549-2560. [PMID: 32623918 DOI: 10.1080/10428194.2020.1786558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Multiple myeloma (MM) is a chronically managed blood cancer with a median age of 69 years at the time of diagnosis. Although high dose melphalan and autologous stem cell transplantation (ASCT) remains a standard of care for eligible patients, more than half of the newly diagnosed MM patients are deemed ineligible due to comorbidities or complications of the disease by itself. In this setting, where ASCT is deemed inappropriate, patients could still achieve durable and deep responses if given the appropriate treatment plan. The key concepts of optimizing induction and maintenance strategies while minimizing side-effects are discussed in this review, especially in the context of employing novel agent combinations. It is important to understand the balance between safety and efficacy for each regimen, utilizing maintenance strategy and the best supportive care measures (bone health, infection prevention, and treatment, pain management, etc.). Here, we examine the evidence behind each of those principles and review results from clinical trials for transplant-ineligible (TI) MM.
Collapse
Affiliation(s)
- Shebli Atrash
- Department of Hematologic Oncology & Blood Disorders, Division of Plasma Cell Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Manisha Bhutani
- Department of Hematologic Oncology & Blood Disorders, Division of Plasma Cell Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Barry Paul
- Department of Hematologic Oncology & Blood Disorders, Division of Plasma Cell Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Peter M Voorhees
- Department of Hematologic Oncology & Blood Disorders, Division of Plasma Cell Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Saad Z Usmani
- Department of Hematologic Oncology & Blood Disorders, Division of Plasma Cell Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| |
Collapse
|
136
|
Lad DP, Malhotra P, Patil AN, Nampoothiri RV, Kasudhan KS, Khadwal A, Prakash G, Jain A, Malhotra S, Varma N, Attri SV, Varma S. Long-term outcomes of innovator versus generic melphalan formulation in autologous hematopoietic cell transplantation for multiple myeloma. Hematol Oncol Stem Cell Ther 2020; 14:114-118. [PMID: 32622756 DOI: 10.1016/j.hemonc.2020.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Most data on autologous hematopoietic cell transplantation (auto-HCT) in myeloma are based on the use of innovator formulation of melphalan. Comparative bioequivalence and efficacy studies of generic melphalan are lacking. METHODS In this retrospective study, we report long-term outcomes of auto-HCT in myeloma using innovator (Alkeran, Aspen Pharma; n = 41) and generic melphalan (Alkacel, Celon Labs, India; n = 55) formulations. All consecutive patients at a single center from the period 2011-2018 were included. RESULTS The median follow-up in the innovator and generic groups was 61.7 and 32.5 months, respectively. Both groups were matched for age, sex, stage, and myeloma response. There were significantly more patients in the innovator melphalan group who were administered melphalan at a reduced dose at physician discretion (26.8% vs. 3.6%, p = .001). There were significantly more patients with grade 3 or higher mucositis (68.3% vs. 38.1%, p < .0001) and grade 3 or higher diarrhea (85.4% vs. 50.1%, p < .0001) in the innovator group. The median duration of hospital stay was significantly longer in the innovator group (19 days vs. 15.5 days, p < .0001). There were significantly more patients in the generic group who received standard maintenance (94.5% vs. 34.1%, p < .0001). Despite the differences in the melphalan dose and post-transplant strategies, the 4-year progression-free survival and overall survival were not significantly different in the two groups (58% vs. 63%, p = .7, 71% vs. 72%, p = .4, respectively). CONCLUSION Long-term efficacy comparison is helpful in the absence of postmarketing bioequivalence studies of generic melphalan.
Collapse
Affiliation(s)
- Deepesh P Lad
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Pankaj Malhotra
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amol N Patil
- Department of Clinical Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ram V Nampoothiri
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kripa Shanker Kasudhan
- Department of Clinical Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arihant Jain
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Samir Malhotra
- Department of Clinical Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Varma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Savita Verma Attri
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Subhash Varma
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
137
|
Kostopoulos IV, Ntanasis-Stathopoulos I, Gavriatopoulou M, Tsitsilonis OE, Terpos E. Minimal Residual Disease in Multiple Myeloma: Current Landscape and Future Applications With Immunotherapeutic Approaches. Front Oncol 2020; 10:860. [PMID: 32537439 PMCID: PMC7267070 DOI: 10.3389/fonc.2020.00860] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/30/2020] [Indexed: 01/10/2023] Open
Abstract
The basic principle that deeper therapeutic responses lead to better clinical outcomes in cancer has emerged technologies capable of detecting rare residual tumor cells. The need for ultra-sensitive approaches for minimal residual disease (MRD) detection is particularly evident in Multiple Myeloma (MM), where patients will ultimately relapse despite the achievement of complete remission, which is commonplace due to remarkable therapeutic advances. Consequently, current response criteria on MM have been amended based on MRD status and MRD negativity is now considered the most dominant prognostic factor and the most valuable indicator for a subsequent relapse. However, there are particular limitations and several aspects for MRD assessment that remain open. This review summarizes current data on MRD in the clinical management of MM, highlights open issues and discusses the challenges and the endless opportunities arising for both patients and clinicians. Furthermore, it focuses on the current status of MRD in clinical trials, its dynamics in addressing debatable aspects in the clinical handling and its potential role as the prevailing factor for future MRD-driven tailored therapies.
Collapse
Affiliation(s)
- Ioannis V Kostopoulos
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania E Tsitsilonis
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
138
|
Culos KA, Gatwood KS, Byrne M. Maintenance Strategies After Hematopoietic Cell Transplantation. Pharmacotherapy 2020; 40:727-740. [PMID: 32343426 DOI: 10.1002/phar.2407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hematopoietic cell transplantation (HCT) is an essential component of potentially curative therapy for patients with hematologic malignancies. High-dose chemotherapy with autologous (auto) stem cell rescue is used to overcome chemoresistance in multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Alternatively, poor-risk acute leukemias rely on the graft versus leukemia effect of allogeneic (allo) products. Long-term remissions are feasible with both auto- and allo-HCT; however, disease relapse is the leading cause of death after HCT for many patients. In recognition of this, novel therapies are being investigated in the upfront, relapsed/refractory, and post-HCT maintenance settings to deepen response and maintain disease control. To date, the most robust data to support this approach are in multiple myeloma, where post-transplant maintenance therapy has improved clinical outcomes. In Hodgkin lymphoma, patients with high-risk features may benefit from post-auto-HCT vedotin (BV) regardless of pre-HCT BV exposure. Apart from mantle cell lymphoma, where rituximab maintenance is generally accepted, post-auto-HCT maintenance in other forms of NHL is less established. In patients who undergo allo-HCT, the utilization of maintenance therapy is an important component of improving post-HCT outcomes, however, an individualized approach that considers patient factors such as residual toxicity from HCT, an immature graft with poor graft function, infection, and graft-versus-host disease create a complex environment for aggressive interventions. Initiation of directed agents in patients with identified mutations prior to allo-HCT, including FLT3 in acute myeloid leukemia and Philadelphia chromosome in acute lymphoid leukemia have generally improved post-HCT outcomes. Ongoing studies are exploring the safety and efficacy of additional maintenance strategies post-allo-HCT in an effort to further improve post-HCT outcomes.
Collapse
Affiliation(s)
- Kathryn A Culos
- Department of Pharmacy, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Katie S Gatwood
- Department of Pharmacy, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael Byrne
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
139
|
Byun JM, Yoon SS, Koh Y, Min CK, Lee JH, Jo J, Park H, Lee J, Kang KW, Lee Y. Incorporating hematopoietic stem-cell transplantation after second-line carfilzomib-lenalidomide-dexamethasone (KRd). Ther Adv Hematol 2020; 11:2040620720921046. [PMID: 32523660 PMCID: PMC7236395 DOI: 10.1177/2040620720921046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Traditionally believed to be an integral part of multiple myeloma (MM) treatment, the role of hematopoietic stem-cell transplantation (HSCT) is being challenged. As such, we sought to evaluate the impact of HSCT in the era of novel agents. Methods: A multicenter, retrospective, longitudinal cohort study was carried out between January 2016 and December 2018. A total of 55 patients who received VTD (bortezomib-thalidomide-dexamethasone) as first-line treatment and KRd (carfilzomib-lenalidomide-dexamethasone) as second-line treatment were analyzed for outcomes. Results: The enrolled patients were divided into Group 1, defined as those who continued KRd treatment until progression (n = 41), versus Group 2, defined as those who underwent HSCT after a certain number of cycles of KRd (n = 14). Both groups showed a generally favorable response to KRd, with overall response rate (ORR) of 87.9% and clinical benefit rate of 92.8% after a median of seven cycles in Group 1, and ORR 92.8% and clinical benefit rate 100% after median of five cycles in Group 2. However, significantly poorer progression-free survival (PFS) (p = 0.004) was observed in Group 1 (median 12 months) compared with Group 2 (median not reached). Multivariate analyses identified HSCT after KRd as potential risk factors associated with PFS. Also, in Group 1, bortezomib refractoriness was associated with significantly shorter PFS compared with those who were responsive (median 12 months versus 14 months, respectively, p = 0.039). Conclusions: In conclusion, even with the advent of novel agents, HSCT still remains a valuable treatment modality with additive efficacy.
Collapse
Affiliation(s)
- Ja Min Byun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, Korea
| | - Jae Hoon Lee
- Department of Internal Medicine, Gachon University Gil Hospital, Incheon, Korea
| | - Jaemin Jo
- Department of Internal Medicine, Jeju National University Hospital, Jeju, Korea
| | - Hyunkyung Park
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Jiyun Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ka-Won Kang
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Yoojin Lee
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | | |
Collapse
|
140
|
Ma LL, Liu Y, Jia SX, Lv HC, Fang MY, Xia YL. A case report: high dose melphalan as a conditioning regimen for multiple myeloma induces sinus arrest. CARDIO-ONCOLOGY 2020; 6:4. [PMID: 32190351 PMCID: PMC7069049 DOI: 10.1186/s40959-020-0059-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/20/2020] [Indexed: 11/10/2022]
Abstract
High dose melphalan is commonly used as a conditioning regimen for autologous stem cell transplantation in multiple myeloma. There are reports of adverse cardiac events with melphalan manifested by supraventricular tachycardia and atrial fibrillation. Here, we report a rare case of a 58 year old female with multiple myeloma, who developed sinus arrest after autologous stem cell transplantation using high dose melphalan as a conditioning regimen. It was severe and rare, therefore, monitoring for cardiac toxicity in patients receiving high-dose melphalan is mandatory.
Collapse
Affiliation(s)
- Liang-Liang Ma
- 1Department of Hematology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Liu
- 2Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116021 Liaoning China
| | - Si-Xun Jia
- 3Department of Hematology, Affiliated Zhongshan Hospital of Dalian University, 6 Jiefang Road, Dalian, 116001 Liaoning China
| | - Hai-Chen Lv
- 2Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116021 Liaoning China
| | - Mei-Yun Fang
- 3Department of Hematology, Affiliated Zhongshan Hospital of Dalian University, 6 Jiefang Road, Dalian, 116001 Liaoning China
| | - Yun-Long Xia
- 2Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116021 Liaoning China
| |
Collapse
|
141
|
Hradska K, Kascak M, Hajek R, Jelinek T. Identifying and treating candidates for checkpoint inhibitor therapies in multiple myeloma and lymphoma. Expert Rev Hematol 2020; 13:375-392. [PMID: 32116068 DOI: 10.1080/17474086.2020.1733405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: One of the hallmarks of cancerogenesis is the ability of tumor cells to evade the immune system. They can achieve it by abusing inhibitory immune checkpoint pathways, which, under normal circumstances, maintain peripheral tolerance during infection. Immune checkpoint inhibitors, especially anti-PD-1/PD-L1 monoclonal antibodies, currently represent a widely discussed treatment option not only in solid oncology, but in hematology-oncology as well.Areas covered: The manuscript is focused on clinical research concerning PD-1/PD-L1 blockade in lymphoma and multiple myeloma in order to identify the patients who would profit the most from this treatment modality. The authors reviewed articles on the topic on PubMed and relevant clinical trials on clinicaltrials.gov before October 2019.Expert opinion: So far, nivolumab and pembrolizumab have been approved for treating patients with relapsed/refractory classical Hodgkin lymphoma and primary mediastinal B cell lymphoma. Nevertheless, monotherapy alone is not curative and a combinational approach is needed. Modern treatment strategies and combinations are comprehensively summarized in this manuscript. There is no approved immune checkpoint inhibitor for the multiple myeloma indication. Although the combination of PD-1/PD-L1 inhibitors with immunomodulatory agents initially seemed promising, unexpected immune related toxicities have stopped any further development. Novel strategies and more potent combinations in myeloma and lymphoma are further discussed in the manuscript.
Collapse
Affiliation(s)
- Katarina Hradska
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Michal Kascak
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hajek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Tomas Jelinek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.,Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
142
|
Abbasi S, Britt A, McClune B, Shune L, Kambhampati S, Mohyuddin GR. Inpatient hospitalization's associated cost and mortality in myeloma patients undergoing autologous stem cell transplant: a 13-year analysis of the National Inpatient Sample. Leuk Lymphoma 2020; 61:1254-1256. [PMID: 32031039 DOI: 10.1080/10428194.2020.1719094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Saqib Abbasi
- Division of Hematologic Malignancies and Biomolecular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alec Britt
- Division of Hematologic Malignancies and Biomolecular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brian McClune
- Division of Hematologic Malignancies and Biomolecular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Leyla Shune
- Division of Hematologic Malignancies and Biomolecular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Ghulam Rehman Mohyuddin
- Division of Hematologic Malignancies and Biomolecular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
143
|
Ntanasis-Stathopoulos I, Gavriatopoulou M, Kastritis E, Terpos E, Dimopoulos MA. Multiple myeloma: Role of autologous transplantation. Cancer Treat Rev 2019; 82:101929. [PMID: 31770695 DOI: 10.1016/j.ctrv.2019.101929] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/31/2019] [Accepted: 11/05/2019] [Indexed: 01/07/2023]
Abstract
Autologous stem cell transplantation (ASCT) has been the mainstay of multiple myeloma (MM) treatment for approximately 30 years. Although the continuous introduction of novel agents in the armamentarium against MM has questioned its value, ASCT remains a backbone treatment for fit MM patients. However, there is no unanimous approach for several aspects including the positioning of ASCT in the therapeutic algorithm either upfront or following the first relapse, the need for single or tandem ASCT, as well as the role of ASCT as salvage therapy. Furthermore, the anti-CD38 monoclonal antibodies along with the next generation proteasome inhibitors and immunomodulatory drugs provide a platform for optimizing the induction and consolidation/maintenance regimens. In this review, we present current data pertaining to all aspects of ASCT in MM, whereas we highlight the open issues that should be addressed in the design of future clinical trials in the field.
Collapse
Affiliation(s)
- Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| |
Collapse
|
144
|
Kazandjian D, Landgren O. Delaying the use of high-dose melphalan with stem cell rescue in multiple myeloma is ready for prime time. CLINICAL ADVANCES IN HEMATOLOGY & ONCOLOGY : H&O 2019; 17:559-568. [PMID: 31730582 PMCID: PMC7451402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The significant advances made in the treatment of multiple myeloma (MM) have allowed for a paradigm shift away from the early use of high-dose melphalan with autologous stem cell transplant (HDM-ASCT). In 2015 alone, the US Food and Drug Administration (FDA) approved 4 novel drugs for MM. Novel drugs and regimens have shown unprecedented efficacy, which has increased the tempo of new drug development. Furthermore, the FDA recently approved a diagnostic test to detect minimal residual disease (MRD) that will allow community clinicians to conduct real-time testing of MRD. Most importantly, frontline regimens based on immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs) have shown a large clinical benefit. The next era has begun, as several 4-drug combinations that include monoclonal antibodies are being evaluated in clinical trials in the attempt to replace HDM-ASCT in the treatment of MM. We and others have therefore questioned the need for early HDM-ASCT, especially in light of the possible complications. HDM-ASCT is associated not only with acute toxicities-cytopenia, infection, and even death-but also an increased risk of developing secondary cancers. This article discusses the historic context of HDM-ASCT, the modern role of HDM-ASCT given the availability of highly sensitive MRD testing, and the likely future of quadruplet treatment. In summary, patients who attain deep responses using IMiD- and PI-based regimens may not require early HDM-ASCT. A delayed approach to this treatment is acceptable, and might be preferred by patients.
Collapse
Affiliation(s)
- Dickran Kazandjian
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ola Landgren
- Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
145
|
Yoon SS. Optimizing carfilzomib use in multiple myeloma treatment. Blood Res 2019; 54:159-161. [PMID: 31730678 PMCID: PMC6779934 DOI: 10.5045/br.2019.54.3.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 11/17/2022] Open
Affiliation(s)
- Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
146
|
Affiliation(s)
- Reinhold Munker
- Department of Internal Medicine, Division of Hematology & BMT, University of Kentucky, Lexington, KY 40536, USA
| | - Gregory Monohan
- Department of Internal Medicine, Division of Hematology & BMT, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
147
|
Goswami C, Poonia S, Kumar L, Sengupta D. Staging System to Predict the Risk of Relapse in Multiple Myeloma Patients Undergoing Autologous Stem Cell Transplantation. Front Oncol 2019; 9:633. [PMID: 31355145 PMCID: PMC6640159 DOI: 10.3389/fonc.2019.00633] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Over the last decade autologous stem cell transplantation (ASCT) has emerged as the standard of care in the management of Multiple Myeloma (MM). However, the cases of early relapse (within 36 months) after the stem cell rescue remains a significant challenge. For a lot of practical purposes, it is crucial to identify whether a patient undergoing ASCT falls into the high-risk group (likely to relapse within 36 months) or a low risk one. Our analysis showed that existing MM staging systems (International Staging System or ISS and Durie Salmon Staging or DSS) are not sufficient to discriminate between the risk groups significantly. To address this, we gathered a total of 39 clinical and laboratory parameters of 347 patients from the Department of Medical Oncology of All India Institute of Medical Sciences (AIIMS). We employed a stacked machine learning model consisting spectral clustering and Fast and Frugal Tree (FFT) technique to come up with a 3-factor multivariate 2-stage staging scheme, which turns out to be extremely decisive about the outcome of the stem cell rescue. Our model comes up with a three-factor (1. if patients has relapsed following remission, 2. response to induction, 3. pre-transplant Glomerular Filtration Rate or GFR) staging scheme. The resulting model stratifies patients into high-risk and low-risk groups with markedly distinct progression-free (median survival—24 months vs. 91 months) and overall survival (median survival—51 months vs. 135 months) patterns.
Collapse
Affiliation(s)
- Chitrita Goswami
- Department of Computer Science and Engineering, Indraprastha Institute of Information Technology, New Delhi, India
| | - Sarita Poonia
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, India
| | - Lalit Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Debarka Sengupta
- Department of Computer Science and Engineering, Indraprastha Institute of Information Technology, New Delhi, India.,Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, India.,Infosys Center for Artificial Intelligence, Indraprastha Institute of Information Technology, New Delhi, India
| |
Collapse
|
148
|
Shinde P, Melinkeri S, Santra MK, Kale V, Limaye L. Autologous Hematopoietic Stem Cells Are a Preferred Source to Generate Dendritic Cells for Immunotherapy in Multiple Myeloma Patients. Front Immunol 2019; 10:1079. [PMID: 31164886 PMCID: PMC6536579 DOI: 10.3389/fimmu.2019.01079] [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: 12/13/2018] [Accepted: 04/29/2019] [Indexed: 11/13/2022] Open
Abstract
In multiple myeloma (MM), dendritic cells (DCs), and their precursors are prone to malignant cell-mediated regulation of function leading to low efficacy of DC vaccine. DCs taken directly from MM patient's body or derived from monocytes are fewer in numbers and are also dysfunctional. Here, we investigated the functionality of Hematopoietic stem cell-derived DCs (SC-DCs) from MM patients. Mature-MM-SC-DCs showed all essential functions like antigen uptake, allogenic T cells simulation and migration comparable to those derived from healthy donor (HD) samples. A comparison of Mo-DCs and SC-DCs obtained from the same MM patients' samples revealed that the expression of IL-6 was higher in the precursors of Mo-DCs leading to their impaired migration. In addition, expression of CCR7 which is responsible for DCs migration was found to be lower in MM-Mo-DCs. The chromatin permissiveness as observed by H3K4me3 histone modification at the Ccr7 promoter in MM-Mo-DCs was significantly lower than those in MM-SC-DCs. Levels of Zbtb46- a hall mark DC transcription factor mRNA was also found to be reduced in MM-Mo-DCs. Cytotoxic T cells generated from MM-SC-DCs from autologous naïve T cells exhibited reduced antitumor activity because the T cells were exhausted. Blocking of CTLA-4 on autologous T cells could partially restore T cell proliferation and activation. Thus, a combination of MM-SC-DC vaccine and anti-CTLA-4 antibody may serve as a better candidate for immunotherapy of MM. This study has implications in increasing the efficacy of cancer immunotherapy in MM.
Collapse
Affiliation(s)
- Prajakta Shinde
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Sameer Melinkeri
- Blood and Marrow Transplant Unit, Deenanath Mangeshkar Hospital, Pune, India
| | - Manas Kumar Santra
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Vaijayanti Kale
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Lalita Limaye
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| |
Collapse
|