1
|
Yong K, Wilson W, de Tute RM, Camilleri M, Ramasamy K, Streetly M, Sive J, Bygrave CA, Benjamin R, Chapman M, Chavda SJ, Phillips EH, Del Mar Cuadrado M, Pang G, Jenner R, Dadaga T, Kamora S, Cavenagh J, Clifton-Hadley L, Owen RG, Popat R. Upfront autologous haematopoietic stem-cell transplantation versus carfilzomib-cyclophosphamide-dexamethasone consolidation with carfilzomib maintenance in patients with newly diagnosed multiple myeloma in England and Wales (CARDAMON): a randomised, phase 2, non-inferiority trial. Lancet Haematol 2023; 10:e93-e106. [PMID: 36529145 DOI: 10.1016/s2352-3026(22)00350-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Standard-of-care treatment for patients with newly diagnosed multiple myeloma is bortezomib-based induction followed by high-dose melphalan and autologous haematopoietic stem-cell transplantation (HSCT) and lenalidomide maintenance. We aimed to evaluate whether an immunomodulatory-free carfilzomib-based induction, consolidation, and maintenance protocol without autologous HSCT was non-inferior to the same induction regimen followed by autologous HSCT and maintenance. METHODS CARDAMON is a randomised, open-label, phase 2 trial in 19 hospitals in England and Wales, UK. Newly diagnosed, transplantation-eligible patients with multiple myeloma aged 18 years or older with an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 received four 28-day cycles of carfilzomib (56 mg/m2 intravenously on days 1, 2, 8, 9, 15, and 16), cyclophosphamide (500 mg orally on days 1, 8, and 15), and dexamethasone (40 mg orally on days 1, 8, 15, and 22; KCd), followed by peripheral blood stem cell mobilisation. Patients with at least a partial response were randomly assigned (1:1) to either high-dose melphalan and autologous HSCT or four cycles of KCd. All randomised patients received 18 cycles of carfilzomib maintenance (56 mg/m2 intravenously on days 1, 8, and 15). The primary outcomes were the proportion of patients with at least a very good partial response after induction and difference in progression-free survival rate at 2 years from randomisation (non-inferiority margin 10%), both assessed by intention to treat. Safety was assessed in all patients who started treatment. The trial is registered with ClinicalTrials.gov (NCT02315716); recruitment is complete and all patients are in follow-up. FINDINGS Between June 16, 2015, and July 8, 2019, 281 patients were enrolled, with 218 proceeding to randomisation (109 assigned to the KCd consolidation group [99 of whom completed consolidation] and 109 to the HSCT group [104 of whom underwent transplantation]). A further seven patients withdrew before initiation of carfilzomib maintenance (two in the KCd consolidation group vs five in the HSCT group). Median age was 59 years (IQR 52 to 64); 166 (59%) of 281 patients were male and 115 (41%) were female. 152 (71%) of 214 patients with known ethnicity were White, 37 (17%) were Black, 18 (8%) were Asian, 5 (2%) identified as Mixed, and 2 (1%) identified as other. Median follow-up from randomisation was 40·2 months (IQR 32·7 to 51·8). After induction, 162 (57·7%; 95% CI 51·6 to 63·5) of 281 patients had at least a very good partial response. The 2-year progression-free survival was 75% (95% CI 65 to 82) in the HSCT group versus 68% (95% CI 58 to 76) in the KCd group (difference -7·2%, 70% CI -11·1 to -2·8), exceeding the non-inferiority margin. The most common grade 3-4 events during KCd induction and consolidation were lymphocytopenia (72 [26%] of 278 patients who started induction; 15 [14%] of 109 patients who started consolidation) and infection (50 [18%] of 278 for induction; 15 [14%] of 109 for consolidation), and during carfilzomib maintenance were hypertension (20 [21%] of 97 patients in the KCd consolidation group vs 23 [23%] of 99 patients in the HSCT group) and infection (16 [16%] of 97 patients vs 25 [25%] of 99). Treatment-related serious adverse events at any point during the trial were reported in 109 (39%) of 278 patients who started induction, with infections (80 [29%]) being the most common. Treatment-emergent deaths were reported in five (2%) of 278 patients during induction (three from infection, one from cardiac event, and one from renal failure) and one of 99 patients during maintenance after autologous HSCT (oesophageal carcinoma). INTERPRETATION KCd did not meet the criteria for non-inferiority compared with autologous HSCT, but the marginal difference in progression-free survival suggests that further studies are warranted to explore deferred autologous HSCT in some subgroups, such as individuals who are MRD negative after induction. FUNDING Cancer Research UK and Amgen.
Collapse
Affiliation(s)
- Kwee Yong
- Cancer Institute, University College London, London, UK; Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK.
| | - William Wilson
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Ruth M de Tute
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Marquita Camilleri
- Cancer Institute, University College London, London, UK; Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK
| | - Karthik Ramasamy
- Department of Clinical Haematology, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Matthew Streetly
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jonathan Sive
- Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK
| | - Ceri A Bygrave
- Department of Haematology, University Hospital of Wales, Cardiff, UK
| | | | - Michael Chapman
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | | | | | | | - Gavin Pang
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Richard Jenner
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Tushhar Dadaga
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Sumaiya Kamora
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - James Cavenagh
- Department of Haemato-Oncology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Laura Clifton-Hadley
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Roger G Owen
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Rakesh Popat
- Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
2
|
Goel U, Usmani S, Kumar S. Current approaches to management of newly diagnosed multiple myeloma. Am J Hematol 2022; 97 Suppl 1:S3-S25. [PMID: 35234302 DOI: 10.1002/ajh.26512] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/16/2022]
Abstract
Major developments in the treatment of multiple myeloma (MM) over the past decade have led to a continued improvement in survival. Significant progress has been made with deeper and longer remissions seen with newer treatment approaches-both for induction as well as maintenance therapy. The treatment approach to MM is guided by several factors including patient age, frailty, comorbidities, eligibility for autologous stem cell transplantation (ASCT), and risk stratification into standard-risk or high-risk MM. High-risk MM is defined by the presence of t(4;14), t(14;16), t(14;20), del (17p), TP53 mutation, or gain (1q). Transplant eligible patients should receive 4-6 cycles of induction followed by stem cell collection. Patients can then undergo ASCT, or continue induction therapy and shift to maintenance, delaying ASCT till first relapse. Transplant ineligible patients should receive induction therapy followed by maintenance. For induction therapy prior to ASCT, a proteasome inhibitor-IMiD combination remains standard with monoclonal antibody-based quadruplets preferred in high-risk patients. Among transplant ineligible patients, those with standard-risk MM should receive DRd continued until disease progression, while bortezomib containing regimens (VRd or VRd lite) can be considered for high-risk patients. Finally, standard-risk patients should receive lenalidomide maintenance after induction/ASCT, while proteasome inhibitor-IMiD combinations should be used for high-risk patients.
Collapse
Affiliation(s)
- Utkarsh Goel
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota USA
| | - Saad Usmani
- Multiple Myeloma Service, Department of medicine Memorial Sloan Kettering Cancer Center New York New York USA
| | - Shaji Kumar
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota USA
| |
Collapse
|
3
|
Auner HW, Brown SR, Walker K, Kendall J, Dawkins B, Meads D, Morgan GJ, Kaiser MF, Cook M, Roberts S, Parrish C, Cook G. Ixazomib with cyclophosphamide and dexamethasone in relapsed or refractory myeloma: MUKeight phase II randomised controlled trial results. Blood Cancer J 2022; 12:52. [PMID: 35365598 PMCID: PMC8972903 DOI: 10.1038/s41408-022-00626-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/05/2022] [Accepted: 01/24/2022] [Indexed: 11/15/2022] Open
Abstract
The all-oral combination of ixazomib, cyclophosphamide, and dexamethasone (ICD) is well tolerated and effective in newly diagnosed and relapsed multiple myeloma (MM). We carried out MUKeight, a randomised, controlled, open, parallel group, multi-centre phase II trial in patients with relapsed MM after prior treatment with thalidomide, lenalidomide, and a proteasome inhibitor (ISRCTN58227268), with the primary objective to test whether ICD has improved clinical activity compared to cyclophosphamide and dexamethasone (CD) in terms of progression-free survival (PFS). Between January 2016 and December 2018, 112 participants were randomised between ICD (n = 58) and CD (n = 54) in 33 UK centres. Patients had a median age of 70 years and had received a median of four prior lines of therapy. 74% were classed as frail. Median PFS in the ICD arm was 5.6 months, compared to 6.7 months with CD (hazard ratio (HR) = 1.21, 80% CI 0.9–1.6, p = 0.3634). Response rates and overall survival were not significantly different between ICD and CD. Dose modifications or omissions, and serious adverse events (SAEs), occurred more often in the ICD arm. In summary, the addition of ixazomib to cyclophosphamide and dexamethasone did not improve outcomes in the comparatively frail patients enroled in the MUKeight trial.
Collapse
Affiliation(s)
- Holger W Auner
- Department of Immunology and Inflammation and The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London, UK
| | - Sarah R Brown
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Katrina Walker
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Jessica Kendall
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Bryony Dawkins
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - David Meads
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Gareth J Morgan
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Martin F Kaiser
- The Institute of Cancer Research, London, UK.,The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Mark Cook
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - Sadie Roberts
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Christopher Parrish
- Department of Clinical Haematology, St. James's University Hospital, Leeds, UK
| | - Gordon Cook
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK. .,Department of Clinical Haematology, St. James's University Hospital, Leeds, UK.
| |
Collapse
|
4
|
Chen Y, Gopalakrishnan SK, Ooi M, Sultana R, Lim LH, Grigoropoulos N, Ong SY, Xu M, Chng WJ, Goh YT, Nagarajan C. A phase 2 study of carfilzomib, cyclophosphamide and dexamethasone as frontline treatment for transplant-eligible MM with high-risk features (SGH-MM1). Blood Cancer J 2021; 11:150. [PMID: 34480015 PMCID: PMC8417287 DOI: 10.1038/s41408-021-00544-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 01/02/2023] Open
Affiliation(s)
- Yunxin Chen
- Department of Haematology, Singapore General Hospital, Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Center, Singapore, Singapore
| | - Sathish Kumar Gopalakrishnan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore.,Department of Haematology, Health Sciences North, Sudbury, ON, Canada
| | - Melissa Ooi
- Department of Haematology, National University Hospital, Singapore, Singapore
| | - Rehena Sultana
- Centre for Quantitative Medicine, DUKE-NUS Medical School, Singapore, Singapore
| | - Li Hui Lim
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Nicholas Grigoropoulos
- Department of Haematology, Singapore General Hospital, Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Center, Singapore, Singapore
| | - Shin Yeu Ong
- Department of Haematology, Singapore General Hospital, Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Center, Singapore, Singapore
| | - Mingge Xu
- Department of Haematology, National University Hospital, Singapore, Singapore
| | | | - Wee Joo Chng
- Department of Haematology, National University Hospital, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yeow Tee Goh
- Department of Haematology, Singapore General Hospital, Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Center, Singapore, Singapore
| | - Chandramouli Nagarajan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore. .,SingHealth Duke-NUS Blood Cancer Center, Singapore, Singapore.
| |
Collapse
|
5
|
Wu J, Liu J. Research progress in proteasome inhibitor resistance to multiple myeloma. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:900-908. [PMID: 34565737 PMCID: PMC10929973 DOI: 10.11817/j.issn.1672-7347.2021.200430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Indexed: 11/03/2022]
Abstract
Multiple myeloma (MM) is a highly heterogeneous malignant plasma cell disease. Proteasome inhibitors (PIs) are the first line of medicine for MM. Bortezomib, ixazomib, and carfilzomib are also widely used for MM. Marizomib, oprozomib, and KZR-616 are in clinical trials. However, the drug resistance of PIs in MM is still a problem. The mechanisms for PIs resistance to MM include ubiquitin-proteasome pathway, autophagy lysosome pathway, endoplasmic reticulum stress pathway, cell survival signal pathway, exosome-mediated resistance, and bone marrow microenvironment-mediated resistance.
Collapse
Affiliation(s)
- Jiao Wu
- Department of Hematology, Loudi Gereral Hospital, Loudi Hunan 417000.
| | - Jing Liu
- Department of Hematology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
6
|
Visram A, Suska A, Jurczyszyn A, Gonsalves WI. Practical management and assessment of primary plasma cell leukemia in the novel agent era. Cancer Treat Res Commun 2021; 28:100414. [PMID: 34174530 DOI: 10.1016/j.ctarc.2021.100414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/08/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Affiliation(s)
- A Visram
- University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - A Suska
- Department of Hematology, Jagiellonian University Medical College, Kopernika 17, Krakow 31-501, Poland
| | - A Jurczyszyn
- Department of Hematology, Jagiellonian University Medical College, Kopernika 17, Krakow 31-501, Poland
| | - W I Gonsalves
- Division of Hematology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
7
|
Schjesvold F, Oriol A. Current and Novel Alkylators in Multiple Myeloma. Cancers (Basel) 2021; 13:2465. [PMID: 34070213 PMCID: PMC8158783 DOI: 10.3390/cancers13102465] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/22/2021] [Accepted: 05/11/2021] [Indexed: 12/22/2022] Open
Abstract
A large number of novel treatments for myeloma have been developed and approved; however, alkylating drugs continue to be part of standard regimens. Additionally, novel alkylators are currently being developed. We performed a non-systematized literary search for relevant papers and communications at large conferences, as well as exploiting the authors' knowledge of the field, to review the history, current use and novel concepts around the traditional alkylators cyclophosphamide, bendamustine and melphalan and current data on the newly developed pro-drug melflufen. Even in the era of targeted treatment and personalized medicine, alkylating drugs continue to be part of the standard-of-care in myeloma, and new alkylators are coming to the market.
Collapse
Affiliation(s)
- Fredrik Schjesvold
- Oslo Myeloma Center, Oslo University Hospital, 0450 Oslo, Norway
- K.G. Jebsen Centre for B-Cell Malignancies, University of Oslo, 4950 Oslo, Norway
| | - Albert Oriol
- Institut Josep Carreras and Institut Català d’Oncologia, Hospital Germans Trias I Pujol, 08916 Badalona, Spain;
| |
Collapse
|
8
|
Yong KL, Hinsley S, Auner HW, Bygrave C, Kaiser MF, Ramasamy K, De Tute RM, Sherratt D, Flanagan L, Garg M, Hawkins S, Williams C, Cavenagh J, Rabin NK, Croft J, Morgan G, Davies F, Owen RG, Brown SR. Carfilzomib or bortezomib in combination with cyclophosphamide and dexamethasone followed by carfilzomib maintenance for patients with multiple myeloma after one prior therapy: results from a multi-centre, phase II, randomized, controlled trial (MUK five). Haematologica 2021; 106:2694-2706. [PMID: 33910333 PMCID: PMC8485692 DOI: 10.3324/haematol.2021.278399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 11/28/2022] Open
Abstract
The proteasome inhibitors, carfilzomib and bortezomib, are widely used to treat myeloma but head-to-head comparisons have produced conflicting results. We compared the activity of these proteasome inhibitors in combination with cyclophosphamide and dexamethasone (KCd vs. VCd) in second-line treatment using fixed duration therapy and evaluated the efficacy of carfilzomib maintenance. MUKfive was a phase II controlled, parallel group trial that randomized patients (2:1) to KCd (n=201) or VCd (n=99); responding patients on carfilzomib were randomized to maintenance carfilzomib (n=69) or no further treatment (n=72). Primary endpoints were: (i) very good partial response (non-inferiority, odds ratio [OR] 0.8) at 24 weeks, and (ii) progression-free survival. More participants achieved a very good partial response or better with carfilzomib than with bortezomib (40.2% vs. 31.9%, OR=1.48, 90% confidence interval [CI]: 0.95, 2.31; non-inferior), with a trend for particular benefit in patients with adverse-risk disease. KCd was associated with higher overall response (partial response or better, 84.0% vs. 68.1%, OR=2.72, 90% CI: 1.62, 4.55, P=0.001). Neuropathy (grade ≥3 or ≥2 with pain) was more common with bortezomib (19.8% vs. 1.5%, P<0.0001), while grade ≥3 cardiac events and hypertension were only reported in the KCd arm (3.6% each). The median progression-free survival in the KCd arm was 11.7 months vs. 10.2 months in the VCd arm (hazard ratio [HR]=0.95, 80% CI: 0.77, 1.18). Carfilzomib maintenance was associated with longer progression-free survival, median 11.9 months vs. 5.6 months for no maintenance (HR 0.59, 80% CI: 0.46-0.77, P=0.0086). When used as fixed duration therapy in first relapase, KCd is at least as effective as VCd, and carfilzomib is an effective maintenance agent. This trial was registered with International Standard Randomised Controlled Trial Number (ISRCTN) identifier: ISRCTN17354232.
Collapse
Affiliation(s)
- Kwee L Yong
- Cancer Institute, University College London, London.
| | - Samantha Hinsley
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds
| | - Holger W Auner
- Department of Immunology and Inflammation and The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London
| | - Ceri Bygrave
- Cardiff and Vale University Health Board, Cardiff
| | - Martin F Kaiser
- The Institute of Cancer Research, London, UK and The Royal Marsden Hospital NHS Foundation Trust, London
| | - Karthik Ramasamy
- Department of Clinical Haematology, Oxford University Hospitals NHS Trust, Oxford
| | - Ruth M De Tute
- Department of Clinical Haematology, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Debbie Sherratt
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds
| | - Louise Flanagan
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds
| | - Mamta Garg
- Department of Haematology, University Hospitals of Leicester NHS Trust, Leicester
| | | | - Catherine Williams
- Centre for Clinical Haematology, Nottingham University Hospitals, Nottingham
| | - Jamie Cavenagh
- Department of Haematology, St Bartholomew's Hospital, London
| | - Neil K Rabin
- Department of Haematology, University College Hospital, London
| | - James Croft
- The Institute of Cancer Research, London, UK and The Royal Marsden Hospital NHS Foundation Trust, London
| | - Gareth Morgan
- Perlmutter Cancer Center, NYU Langone Health, New York
| | - Faith Davies
- Perlmutter Cancer Center, NYU Langone Health, New York
| | - Roger G Owen
- Haematological Malignancy Diagnostic Service (HMDS), St James's University Hospital, Leeds
| | - Sarah R Brown
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds
| |
Collapse
|
9
|
Engelhardt M, Bringhen S, Moreau P, Wäsch R, Waldschmidt J. In search of the optimal proteosome inhibitor. How, when and for whom? Haematologica 2021; 106:2539-2541. [PMID: 33910336 PMCID: PMC8485685 DOI: 10.3324/haematol.2021.278838] [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: 04/01/2021] [Indexed: 11/09/2022] Open
Affiliation(s)
- Monika Engelhardt
- University of Freiburg, Hematology and Oncology, Faculty of Freiburg, Hugstetterstr. 53, 79106 Freiburg
| | - Sara Bringhen
- Myeloma Unit, Division of Hematology, University of Turino, Azienda Ospedaliero-Universitaria Citta della Salute e della Scienza di Torino, Torino
| | | | - Ralph Wäsch
- University of Freiburg, Hematology and Oncology, Faculty of Freiburg, Hugstetterstr. 53, 79106 Freiburg
| | - Johannes Waldschmidt
- University of Freiburg, Hematology and Oncology, Faculty of Freiburg, Hugstetterstr. 53, 79106 Freiburg
| |
Collapse
|
10
|
Kaweme NM, Changwe GJ, Zhou F. Approaches and Challenges in the Management of Multiple Myeloma in the Very Old: Future Treatment Prospects. Front Med (Lausanne) 2021; 8:612696. [PMID: 33718400 PMCID: PMC7947319 DOI: 10.3389/fmed.2021.612696] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
The increasing incidence of geriatric patients with multiple myeloma has elevated concerns in clinical practice. While the introduction of novel therapeutic agents has substantially improved outcomes in younger patients with myeloma, poorer outcomes remain in older patients. Managing older patients requires a multidisciplinary team approach to consider factors that may influence both treatment selection and outcomes. Aging is associated with remodeling of vital organs, physiological downregulations of basal metabolism, susceptibility to multiple comorbidities with ultimate frailty, thereby contributing to the underrepresentation and exclusion of very old patients from clinical trials. Therefore, timely confirmation of a precise diagnosis is crucial for prompt initiation of treatment if the desired outcome is to be achieved. Adequate and judicious assessment using comprehensive geriatric assessment tools minimizes toxicities and treatment discontinuation. Initiating treatment with combinational therapy requires knowledge of indications and anticipated outcomes, as well as individualized therapy with appropriate dose-adjustment. Individualized therapy based on good clinical acumen and best practices obverts unwanted polypharmacy, preventing iatrogenic harm. This review will therefore address the approaches and challenges faced in managing myeloma in geriatric patients aged 80 years and older, highlighting recommended therapeutic strategies and future prospective regimens.
Collapse
Affiliation(s)
| | | | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Kumar SK, Jacobus SJ, Cohen AD, Weiss M, Callander N, Singh AK, Parker TL, Menter A, Yang X, Parsons B, Kumar P, Kapoor P, Rosenberg A, Zonder JA, Faber E, Lonial S, Anderson KC, Richardson PG, Orlowski RZ, Wagner LI, Rajkumar SV. Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol 2020; 21:1317-1330. [PMID: 32866432 DOI: 10.1016/s1470-2045(20)30452-6] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Bortezomib, lenalidomide, and dexamethasone (VRd) is a standard therapy for newly diagnosed multiple myeloma. Carfilzomib, a next-generation proteasome inhibitor, in combination with lenalidomide and dexamethasone (KRd), has shown promising efficacy in phase 2 trials and might improve outcomes compared with VRd. We aimed to assess whether the KRd regimen is superior to the VRd regimen in the treatment of newly diagnosed multiple myeloma in patients who were not being considered for immediate autologous stem-cell transplantation (ASCT). METHODS In this multicentre, open-label, phase 3, randomised controlled trial (the ENDURANCE trial; E1A11), we recruited patients aged 18 years or older with newly diagnosed multiple myeloma who were ineligible for, or did not intend to have, immediate ASCT. Participants were recruited from 272 community oncology practices or academic medical centres in the USA. Key inclusion criteria were the absence of high-risk multiple myeloma and an Eastern Cooperative Oncology Group performance status of 0-2. Enrolled patients were randomly assigned (1:1) centrally by use of permuted blocks to receive induction therapy with either the VRd regimen or the KRd regimen for 36 weeks. Patients who completed induction therapy were then randomly assigned (1:1) a second time to either indefinite maintenance or 2 years of maintenance with lenalidomide. Randomisation was stratified by intent for ASCT at disease progression for the first randomisation and by the induction therapy received for the second randomisation. Allocation was not masked to investigators or patients. For 12 cycles of 3 weeks, patients in the VRd group received 1·3 mg/m2 of bortezomib subcutaneously or intravenously on days 1, 4, 8, and 11 of cycles 1-8, and day 1 and day 8 of cycles nine to twelve, 25 mg of oral lenalidomide on days 1-14, and 20 mg of oral dexamethasone on days 1, 2, 4, 5, 8, 9, 11, and 12. For nine cycles of 4 weeks, patients in the KRd group received 36 mg/m2 of intravenous carfilzomib on days 1, 2, 8, 9, 15, and 16, 25 mg of oral lenalidomide on days 1-21, and 40 mg of oral dexamethasone on days 1, 8, 15, and 22. The coprimary endpoints were progression-free survival in the induction phase, and overall survival in the maintenance phase. The primary analysis was done in the intention-to-treat population and safety was assessed in patients who received at least one dose of their assigned treatment. The trial is registered with ClinicalTrials.gov, NCT01863550. Study recruitment is complete, and follow-up of the maintenance phase is ongoing. FINDINGS Between Dec 6, 2013, and Feb 6, 2019, 1087 patients were enrolled and randomly assigned to either the VRd regimen (n=542) or the KRd regimen (n=545). At a median follow-up of 9 months (IQR 5-23), at a second planned interim analysis, the median progression-free survival was 34·6 months (95% CI 28·8-37·8) in the KRd group and 34·4 months (30·1-not estimable) in the VRd group (hazard ratio [HR] 1·04, 95% CI 0·83-1·31; p=0·74). Median overall survival has not been reached in either group. The most common grade 3-4 treatment-related non-haematological adverse events included fatigue (34 [6%] of 527 patients in the VRd group vs 29 [6%] of 526 in the KRd group), hyperglycaemia (23 [4%] vs 34 [6%]), diarrhoea (23 [5%] vs 16 [3%]), peripheral neuropathy (44 [8%] vs four [<1%]), dyspnoea (nine [2%] vs 38 [7%]), and thromboembolic events (11 [2%] vs 26 [5%]). Treatment-related deaths occurred in two patients (<1%) in the VRd group (one cardiotoxicity and one secondary cancer) and 11 (2%) in the KRd group (four cardiotoxicity, two acute kidney failure, one liver toxicity, two respiratory failure, one thromboembolic event, and one sudden death). INTERPRETATION The KRd regimen did not improve progression-free survival compared with the VRd regimen in patients with newly diagnosed multiple myeloma, and had more toxicity. The VRd triplet regimen remains the standard of care for induction therapy for patients with standard-risk and intermediate-risk newly diagnosed multiple myeloma, and is a suitable treatment backbone for the development of combinations of four drugs. FUNDING US National Institutes of Health, National Cancer Institute, and Amgen.
Collapse
Affiliation(s)
| | - Susanna J Jacobus
- ECOG-ACRIN Biostatistics Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Natalie Callander
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | | | - Terri L Parker
- Department of Hematology, Yale University, Hamden, CT, USA
| | | | | | | | | | | | - Aaron Rosenberg
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jeffrey A Zonder
- Department of Malignant Hematology, Barbara Ann Karmanos Cancer Institute and Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Sagar Lonial
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | | | | | - Lynne I Wagner
- Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | | |
Collapse
|