Updated analysis of CALGB (Alliance) 100104 assessing lenalidomide versus placebo maintenance after single autologous stem-cell transplantation for multiple myeloma: a randomised, double-blind, phase 3 trial

Sarah A Holstein, Sin-Ho Jung, Paul G Richardson, Craig C Hofmeister, David D Hurd, Hani Hassoun, Sergio Giralt, Edward A Stadtmauer, Daniel J Weisdorf, Ravi Vij, Jan S Moreb, Natalie S Callander, Koen van Besien, Teresa G Gentile, Luis Isola, Richard T Maziarz, Asad Bashey, Heather Landau, Thomas Martin, Muzaffar H Qazilbash, Cesar Rodriguez, Brian McClune, Robert L Schlossman, Scott E Smith, Vera Hars, Kouros Owzar, Chen Jiang, Molly Boyd, Chelsea Schultz, Marcia Wilson, Parameswaran Hari, Marcelo C Pasquini, Mary M Horowitz, Thomas C Shea, Steven M Devine, Charles Linker, Kenneth C Anderson, Philip L McCarthy, Sarah A Holstein, Sin-Ho Jung, Paul G Richardson, Craig C Hofmeister, David D Hurd, Hani Hassoun, Sergio Giralt, Edward A Stadtmauer, Daniel J Weisdorf, Ravi Vij, Jan S Moreb, Natalie S Callander, Koen van Besien, Teresa G Gentile, Luis Isola, Richard T Maziarz, Asad Bashey, Heather Landau, Thomas Martin, Muzaffar H Qazilbash, Cesar Rodriguez, Brian McClune, Robert L Schlossman, Scott E Smith, Vera Hars, Kouros Owzar, Chen Jiang, Molly Boyd, Chelsea Schultz, Marcia Wilson, Parameswaran Hari, Marcelo C Pasquini, Mary M Horowitz, Thomas C Shea, Steven M Devine, Charles Linker, Kenneth C Anderson, Philip L McCarthy

Abstract

Background: In the CALGB (Alliance) 100104 study, lenalidomide versus placebo after autologous stem-cell transplantation (ASCT) was investigated for patients with newly diagnosed myeloma. That study showed improved time to progression and overall survival and an increase in second primary malignancies for lenalidomide at a median follow-up of 34 months. Here we report an updated intention-to-treat analysis of CALGB (Alliance) 100104 at a median follow-up of 91 months.

Methods: Patients were eligible for this randomised, double-blind, placebo-controlled, phase 3 trial if they had symptomatic disease requiring treatment; had received, at most, two induction regimens; and had achieved stable disease or better in the first 100 days after ASCT. We randomly assigned patients to either lenalidomide or placebo groups using permuted block randomisation, with a fixed block size of six. Randomisation was stratified by three factors: normal or elevated β2 microglobulin concentration at registration (≤2·5 mg/L vs >2·5 mg/L), previous use or non-use of thalidomide during induction therapy, and previous use or non-use of lenalidomide during induction therapy. The starting dose was two capsules (10 mg) per day, escalated to three capsules (15 mg) per day after 3 months. The primary endpoint was time to progression (time of progressive disease or death from any cause), with intention-to-treat analysis. This study is registered with ClinicalTrials.gov, identifier NCT00114101. New patients are no longer being recruited, but some patients remain on treatment and in follow-up.

Findings: Between April 14, 2005, and July 2, 2009, 460 patients were randomly assigned to receive either lenalidomide (n=231) or placebo (n=229). After three interim analyses, the study was unblinded at a median follow-up of 18 months, at which point 86 (67%) of 128 patients without progressive disease in the placebo group chose to cross over to the lenalidomide group. The median follow-up for the updated survival analysis, as of Oct 19, 2016, was 91 months (IQR 83·6-103·1). The median time to progression was 57·3 months (95% CI 44·2-73·3) for the lenalidomide group and 28·9 months (23·0-36·3) for the placebo group (hazard ratio 0·57, 95% CI 0·46-0·71; p<0·0001). The most common grade 3-4 adverse events were neutropenia (116 [50%] patients in the lenalidomide group and 41 [18%] patients in the placebo group) and thrombocytopenia (34 [15%] patients in the lenalidomide group and 12 [5%] patients in the placebo group). 18 (8%) haematological and 14 (6%) solid tumour second primary malignancies were diagnosed after randomisation and before disease progression in the lenalidomide group, compared with three (1%) haematological and nine (4%) solid tumour second primary malignancies in the placebo group. Three haematological and five solid tumour second primary malignancies in the placebo group were in the crossover subgroup.

Interpretation: Despite an increase in haematological adverse events and second primary malignancies, lenalidomide maintenance therapy after ASCT significantly improved time to progression and could be considered a standard of care.

Funding: The National Cancer Institute.

Copyright © 2017 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
CONSORT flow diagram of patient disposition at the current data cut-off.
Figure 2
Figure 2
Kaplan-Meier estimates of time to progression (A) and overall survival (B). ASCT denotes autologous stem cell transplant.
Figure 3
Figure 3
Kaplan-Meier estimates of survival time following progression.
Figure 4
Figure 4
Forest plot of time to progression (A) and overall survival (B). Hazard ratios from subgroup analyses are shown on a natural-log scale. The radii of the circles are proportional to the inverse of the square of the standard error.
Figure 5
Figure 5
Cumulative incidence risk (CIR) of progressive disease, death, and second primary malignancies (SPMs) by treatment arm. A) The CIR of progressive disease or death from any cause is higher with placebo compared to lenalidomide (p

References

    1. Morgan GJ, Gregory WM, Davies FE, et al. The role of maintenance thalidomide therapy in multiple myeloma: MRC Myeloma IX results and meta-analysis. Blood. 2012;119(1):7–15.
    1. McCarthy PL, Owzar K, Hofmeister CC, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012;366(19):1770–81.
    1. Attal M, Lauwers-Cances V, Marit G, et al. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012;366(19):1782–91.
    1. Palumbo A, Cavallo F, Gay F, et al. Autologous transplantation and maintenance therapy in multiple myeloma. N Engl J Med. 2014;271(10):895–905.
    1. Jackson GH, Davies FE, Pawlyn C, et al. Lenalidomide is a highly effective maintenance therapy in myeloma patients of all ages; results of the phase III Myeloma XI study. Blood (ASH Abstracts) 2016;128:1143.
    1. Attal M, Palumbo A, Holstein SA, et al. Lenalidomide (LEN) maintenance (MNTC) after high-dose melphalan and autologous stem cell transplant (ASCT) in multiple myeloma (MM): A meta-analysis (MA) of overall survival (OS) J Clin Oncol. 2016;34(15 suppl):8001.
    1. Blade J, Samson D, Reece D, et al. Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation. Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant. Br J Haematol. 1998;102(5):1115–23.
    1. Durie BG, Harousseau JL, Miguel JS, et al. International uniform response criteria for multiple myeloma. Leukemia. 2006;20(9):1467–73.
    1. Emerson SS, Fleming TR. Symmetric group sequential test designs. Biometrics. 1989;45(3):905–23.
    1. Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53:457–81.
    1. Cox DR, Oakes D. Monographs on Statistics and Applied Probability 21. Chapman & Hall; 1984. Analysis of Survival Data.
    1. Kalbfleisch JD, Prentice RL. The statistical analysis of failure time data. 2. Hoboken: John Wiley & Sons, Inc; 2002.
    1. Gray RJ. A class of K-sample tests for comparing the cumulative incidence of a competing risk. Ann Stat. 1988;16(3):1141–54.
    1. Fisher RA. The logic of inductive inference. J R Stat Soc. 1935;98:39–54.
    1. Palumbo A, Hajek R, Delforge M, et al. Continuous lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2012;366(19):1759–69.
    1. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014;371(10):906–17.
    1. Attal M, Lauwers-Cances V, Hulin C, et al. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med. 2017;376(14):1311–20.
    1. Stadtmauer EA, Pasquini MC, Blackwell B, et al. Comparison of autologous hematopoietic cell transplant (autoHCT), bortezomib, lenalidomide (Len) and dexamethasone (RVD) consolidation with Len maintenance (ACM), tandem autoHCT with Len maintenance (TAM) and autoHCT with Len maintenance (AM) for up-front treatment of patients with multiple myeloma (MM): primary results from the randomized phase III trial of the Blood and Marrow Transplant Clinical Trials Network (BMT CTN 0702-StaMINA trial) Blood (ASH Abstracts) 2016;128:LBA-1.
    1. Gay F, Oliva S, Petrucci MT, et al. Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicenter, phase 3 trial. Lancet Oncol. 2015;16(16):1617–29.
    1. Cavo M, Palumbo A, Zweegman S, et al. Upfront autologous stem cell transplantation (ASCT) versus novel agent-based therapy for multiple myeloma (MM): a randomized phase 3 study of the European Myeloma Network (EMN02/HO95 MM trial) J Clin Oncol. 2016;34(15 suppl):8000.
    1. Genovese G, Kahler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–87.
    1. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–98.
    1. Tzeng HE, Lin CL, Tsai CH, et al. Time trend of multiple myeloma and associated secondary primary malignancies in Asian patients: a Taiwan population-based study. PLoS One. 2013;8(7):e68041.
    1. Razavi P, Rand KA, Cozen W, et al. Patterns of second primary malignancy risk in multiple myeloma patients before and after the introduction of novel therapeutics. Blood Cancer J. 2013;3:e121.
    1. Mailankody S, Pfeiffer RM, Kristinsson SY, et al. Risk of acute myeloid leukemia and myelodysplastic syndromes after multiple myeloma and its precursor disease (MGUS) Blood. 2011;118(15):4086–92.
    1. Hasskarl J, Ihorst G, De Pasquale D, et al. Association of multiple myeloma with different neoplasms: systematic analysis in consecutive patients with myeloma. Leuk Lymphoma. 2011;52(2):247–59.
    1. Palumbo A, Bringhen S, Kumar SK, et al. Second primary malignancies with lenalidomide therapy for newly diagnosed myeloma: a meta-analysis of individual patient data. Lancet Oncol. 2014;15(3):333–42.
    1. Mullighan CG, Goorha S, Radtke I, et al. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature. 2007;446(7137):758–64.
    1. Mullighan CG, Miller CB, Radtke I, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453(7101):110–4.
    1. Iacobucci I, Storlazzi CT, Cilloni D, et al. Identification and molecular characterization of recurrent genomic deletions on 7p12 in the IKZF1 gene in a large cohort of BCR-ABL1-positive acute lymphoblastic leukemia patients: on behalf of Gruppo Italiano Malattie Ematologiche dell’Adulto Acute Leukemia Working Party (GIMEMA AL WP) Blood. 2009;114(10):2159–67.
    1. Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17(8):e328–46.
    1. Paiva B, Vidriales MB, Cervero J, et al. Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation. Blood. 2008;112(10):4017–23.
    1. Rawstron AC, Child JA, de Tute RM, et al. Minimal residual disease assessed by multiparatmeter flow cytometry in multiple myeloma: impact on outcome in the Medical Research Council Myeloma IX Study. J Clin Oncol. 2013;31(20):2540–7.

Source: PubMed

3
Se inscrever