Page 171 - 2019_07 resto del Mondo-web
P. 171

Maintenance, response status, and subclonal structure at MM relapse
CRBN mutation. It is likely that the mutation occurred by chance, but that, in true Darwinian fashion, in the pres- ence of lenalidomide, it conferred a survival advantage to the malignant cells harboring it. Recent analyses indicating that prolonged exposure to lenalidomide is not linked to an increased incidence of second primary hematologic malignancies when used in combinations excluding oral melphalan may be supported by a lack of evidence of mutagenesis leading to treatment resistance in this work, although it is acknowledged that larger analyses using patients with prolonged remissions are required.24,57 It is important to note that the patients studied here relapsed early and that they were selected because of this charac- teristic, as it had previously been suggested that exposure to lenalidomide could enhance progression of such cases post maintenance. However, we did not find any evidence to support such a hypothesis.
It does remain important to evaluate the impact of maintenance on low-risk cases who are long-term sur- vivors, a question not addressed in this study. In addition, we acknowledge that clonal structure may be further assessed using single cell analysis. However, for the moment, the challenges of obtaining individual malignant plasma cells from large series of patients at multiple dis- ease time points has proved to be a significant barrier. In this series of patients, however, we do see evidence of par- allel evolution, as previously described using single cell technology, further validating the clonal changes described here.23 An example is seen in a patient with bi-
allelic inactivation of TP53 at presentation characterized by del(17p) and TP53 mutation, but only the presence of a TP53 mutation at relapse. In this case the clone with del(17p) was no longer detectable at relapse, suggesting it was treatment sensitive and/or out-completed by a more aggressive treatment insensitive clone harboring a TP53 mutation that had expanded following treatment, con- firmed by a higher CCF at relapse (0.55 vs. 0.83).
In this group of high-risk early-relapsing patients, we show that relapse is a result of the dynamic interplay of evolutionary processes based on the capacity of clonal cells to adapt to their bone marrow microenvironment as a result of new mutations, copy number change, and the selective pressure of the treatment used. Specifically, the depth of response is a critical feature that impacts the evo- lutionary patterns seen at relapse, rather than the use of lenalidomide maintenance.
Acknowledgments
We are thankful to all the patients who have taken part in the Myeloma XI trial, from which samples used in this analysis were obtained. We thank the Data Monitoring and Ethics Committee (DMEC) and Trial Steering Group Committee (TSG) for their support and guidance throughout the trial.
Funding
Myeloma XI was funded by Cancer Research UK and received unrestricted educational grants from Celgene, Takeda and Merck.
References
1. Kandoth C, McLellan MD, Vandin F, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013; 502(7471):333-339.
2. Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499(7457):214-218.
3. Lonial S, Anderson KC. Association of response endpoints with survival outcomes in multiple myeloma. Leukemia. 2014; 28(2):258-268.
4. Pawlyn C, Morgan GJ. Evolutionary biology of high-risk multiple myeloma. Nat Rev Cancer. 2017;17(9):543-556.
5. Kumar SK, Dispenzieri A, Lacy MQ, et al. Continued improvement in survival in mul- tiple myeloma: changes in early mortality and outcomes in older patients. Leukemia. 2014;28(5):1122-1128.
6. Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood. 2008;111(5):2516-2520.
7. Pulte D, Gondos A, Brenner H. Improvement in survival of older adults with multiple myeloma: results of an updat- ed period analysis of SEER data. Oncologist. 2011;16(11):1600-1603.
8. Rawstron AC, Gregory WM, de Tute RM, et al. Minimal residual disease in myeloma by flow cytometry: independent prediction of survival benefit per log reduction. Blood. 2015;125(12):1932-1935.
9. Oliva S, Gambella M, Gilestro M, et al. Minimal residual disease after transplanta-
tion or lenalidomide-based consolidation in myeloma patients: a prospective analysis. Oncotarget. 2017;8(4):5924-5935.
10. Paiva B, van Dongen JJ, Orfao A. New crite- ria for response assessment: role of minimal residual disease in multiple myeloma. Blood. 2015;125(20):3059-3068.
11. 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):1782-1791.
12. Palumbo A, Hajek R, Delforge M, et al. Continuous lenalidomide treatment for newly diagnosed multiple myeloma. N Engl J Med. 2012;366(19):1759-1769.
13. McCarthy PL, Owzar K, Hofmeister CC, et al. Lenalidomide after stem-cell transplanta- tion for multiple myeloma. N Engl J Med. 2012;366(19):1770-1781.
14. Bjorklund CC, Lu L, Kang J, et al. Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Blood Cancer J. 2015;5:e354.
15. Fischer ES, Bohm K, Lydeard JR, et al. Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Nature. 2014;512(7512):49-53.
16. Lopez-Girona A, Mendy D, Ito T, et al. Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalido- mide. Leukemia. 2012;26(11):2326-2335.
17. Quach H, Ritchie D, Stewart AK, et al. Mechanism of action of immunomodulato- ry drugs (IMiDS) in multiple myeloma. Leukemia. 2010;24(1):22-32.
18. Weinhold N, Ashby C, Rasche L, et al. Clonal selection and double-hit events involving tumor suppressor genes underlie relapse in myeloma. Blood. 2016; 128(13): 1735-1744.
19. Keats JJ, Chesi M, Egan JB, et al. Clonal com- petition with alternating dominance in mul- tiple myeloma. Blood. 2012; 120(5):1067- 1076.
20. Magrangeas F, Avet-Loiseau H, Gouraud W, et al. Minor clone provides a reservoir for relapse in multiple myeloma. Leukemia. 2013;27(2):473-481.
21. Bolli N, Avet-Loiseau H, Wedge DC, et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun. 2014;5:2997.
22. Kortum KM, Langer C, Monge J, et al. Longitudinal analysis of 25 sequential sam- ple-pairs using a custom multiple myeloma mutation sequencing panel (M(3)P). Ann Hematol. 2015;94(7):1205-1211.
23. Melchor L, Brioli A, Wardell CP, et al. Single- cell genetic analysis reveals the composition of initiating clones and phylogenetic pat- terns of branching and parallel evolution in myeloma. Leukemia. 2014; 28(8):1705-1715.
24. Jones JR, Cairns DA, Gregory WM, et al. Second malignancies in the context of lenalidomide treatment: an analysis of 2732 myeloma patients enrolled to the Myeloma XI trial. Blood Cancer J. 2016;6(12):e506.
25. 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- 346.
haematologica | 2019; 104(7)
1449


































































































   169   170   171   172   173