240
Views
8
CrossRef citations to date
0
Altmetric
Review

Getting personal with myelodysplastic syndromes: is now the right time?

, ORCID Icon, ORCID Icon, ORCID Icon, &
Pages 215-224 | Received 30 Nov 2018, Accepted 06 Mar 2019, Published online: 12 Apr 2019

References

  • Shallis RM, Ahmad R, Zeidan AM. The genetic and molecular pathogenesis of myelodysplastic syndromes. Eur J Haematol. 2018;101(3):260–271.
  • Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89(6):2079–2088.
  • Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454–2465.
  • Malcovati L, Germing U, Kuendgen A, et al. Time-dependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes. J Clin Oncol. 2007;25(23):3503–3510.
  • Kantarjian H, O’Brien S, Ravandi F, et al. Proposal for a new risk model in myelodysplastic syndrome that accounts for events not considered in the original International Prognostic Scoring System. Cancer. 2008;113(6):1351–1361.
  • Swerdlow SH, Campo E, Harris NL, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. Revised 4th Edition. Vol. 2. WHO Classification of Tumours. Geneva, Switzerland: WHO Press; 2017.
  • Zeidan AM, Shallis RM, Wang R, et al. Epidemiology of myelodysplastic syndromes: why characterizing the beast is a prerequisite to taming it. Blood Rev. 2019 Mar;34:1–15. doi: 10.1016/j.blre.2018.09.001. Epub 2018 Sep 21.
  • Stahl M, Zeidan AM. Lenalidomide use in myelodysplastic syndromes: insights into the biologic mechanisms and clinical applications. Cancer. 2017;123(10):1703–1713.
  • Zeidan AM, Al Ali NH, Padron E, et al. Lenalidomide treatment for lower risk nondeletion 5q myelodysplastic syndromes patients yields higher response rates when used before azacitidine. Clin Lymphoma Myeloma Leuk. 2015;15(11):705–710.
  • Abou Zahr A, Saad Aldin E, Komrokji RS, et al. Clinical utility of lenalidomide in the treatment of myelodysplastic syndromes. J Blood Med. 2015;6:1–16.
  • Arber DA, Orazi A, Hasserjian R, et al. The revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–2405.
  • Della Porta MG, Travaglino E, Boveri E, et al. Minimal morphological criteria for defining bone marrow dysplasia: a basis for clinical implementation of WHO classification of myelodysplastic syndromes. Leukemia. 2015;29(1):66–75.
  • Mangan JK, Speck NA. RUNX1 mutations in clonal myeloid disorders: from conventional cytogenetics to next generation sequencing, a story 40 years in the making. Crit Rev Oncog. 2011;16(1–2):77–91.
  • Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med. 2011;364(26):2496–2506.
  • Traina F, Visconte V, Elson P, et al. Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia. 2014;28(1):78–87.
  • Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014;124(17):2705–2712.
  • Abou Zahr A, Saad Aldin E, Barbarotta L, et al. The clinical use of DNA methyltransferase inhibitors in myelodysplastic syndromes. Expert Rev Anticancer Ther. 2015;15(9):1019–1036.
  • DiNardo CD, Patel KP, Garcia-Manero G, et al. Lack of association of IDH1, IDH2 and DNMT3A mutations with outcome in older patients with acute myeloid leukemia treated with hypomethylating agents. Leuk Lymphoma. 2014;55(8):1925–1929.
  • Coombs CC, Sallman DA, Devlin SM, et al. Mutational correlates of response to hypomethylating agent therapy in acute myeloid leukemia. Haematologica. 2016;101(11):e457–e460.
  • Bejar R. Myelodysplastic syndromes diagnosis: what is the role of molecular testing? Curr Hematol Malig Rep. 2015;10(3):282–291.
  • Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28(2):241–247.
  • Steensma DP. How I use molecular genetic tests to evaluate patients who have or may have myelodysplastic syndromes. Blood. 2018;132(16):1657–1663.
  • Abou Zahr A, Bernabe Ramirez C, Wozney J, et al. New insights into the pathogenesis of MDS and the rational therapeutic opportunities. Expert Rev Hematol. 2016;9(4):377–388.
  • Moyo TK, Savona MR. Molecular testing in patients with suspected myelodysplastic syndromes. Curr Hematol Malig Rep. 2016;11(6):441–448.
  • Valent P, Orazi A, Steensma DP, et al. Proposed minimal diagnostic criteria for myelodysplastic syndromes (MDS) and potential pre-MDS conditions. Oncotarget. 2017;8(43):73483–73500.
  • Zeidan AM, Stahl M, Sekeres MA, et al. A call for action: increasing enrollment of untreated patients with higher-risk myelodysplastic syndromes in first-line clinical trials. Cancer. 2017;123(19):3662–3672.
  • Yohe S, Thyagarajan B. Review of clinical next-generation sequencing. Arch Pathol Lab Med. 2017;141(11):1544–1557.
  • Hardwick SA, Deveson IW, Mercer TR. Reference standards for next-generation sequencing. Nat Rev Genet. 2017;18(8):473–484.
  • Kohlmann A, Grossmann V, Nadarajah N, et al. Next-generation sequencing - feasibility and practicality in haematology. Br J Haematol. 2013;160(6):736–753.
  • Behjati S, Tarpey PS. What is next generation sequencing? Arch Dis Child Educ Pract Ed. 2013;98(6):236–238.
  • Steensma DP. The evolving role of genomic testing in assessing prognosis of patients with myelodysplastic syndromes. Best Pract Res Clin Haematol. 2017;30(4):295–300.
  • Papaemmanuil E, Gerstung M, Malcovati L, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122(22):3616–3627. quiz 99.
  • Bejar R, Stevenson KE, Caughey B, et al. Somatic mutations predict poor outcome in patients with myelodysplastic syndrome after hematopoietic stem-cell transplantation. J Clin Oncol. 2014;32(25):2691–2698.
  • Mossner M, Jann JC, Wittig J, et al. Mutational hierarchies in myelodysplastic syndromes dynamically adapt and evolve upon therapy response and failure. Blood. 2016;128(9):1246–1259.
  • Xu F, Wu LY, He Q, et al. Exploration of the role of gene mutations in myelodysplastic syndromes through a sequencing design involving a small number of target genes. Sci Rep. 2017;7:43113.
  • Wu SJ, Kuo YY, Hou HA, et al. The clinical implication of SRSF2 mutation in patients with myelodysplastic syndrome and its stability during disease evolution. Blood. 2012;120(15):3106–3111.
  • Young AL, Challen GA, Birmann BM, et al. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun. 2016;7:12484.
  • Bartels S, Schipper E, Hasemeier B, et al. Routine clinical mutation profiling using next generation sequencing and a customized gene panel improves diagnostic precision in myeloid neoplasms. Oncotarget. 2016;7(21):30084–30093.
  • Lew JL, Fenderson JL, Carmichael MG. Next-generation gene sequencing differentiates hypoplastic myelodysplastic syndrome from aplastic anemia. Hawaii J Med Public Health. 2017;76(11 Suppl 2):10–12.
  • Zhang X, List AF, Lancet JE, et al. Clinicopathological features and mutational analysis of patients with aplastic anemia and hypoplastic myelodysplastic syndrome. Blood. 2016;128(17):2175–2178.
  • Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic mutations and clonal hematopoiesis in aplastic anemia. N Engl J Med. 2015;373(1):35–47.
  • Shallis RM, Chokr N, Stahl M, et al. Immunosuppressive therapy in myelodysplastic syndromes: a borrowed therapy in search of the right place. Expert Rev Hematol. 2018;11(9):715–726.
  • Stahl M, DeVeaux M, de Witte T, et al. The use of immunosuppressive therapy in MDS: clinical outcomes and their predictors in a large international patient cohort. Blood Adv. 2018;2(14):1765–1772.
  • Kulasekararaj AG, Jiang J, Smith AE, et al. Somatic mutations identify a sub-group of aplastic anemia patients that progress to myelodysplastic syndrome. Blood. 2014;124(17):2698–2704.
  • Malcovati L, Cazzola M. The shadowlands of MDS: idiopathic cytopenias of undetermined significance (ICUS) and clonal hematopoiesis of indeterminate potential (CHIP). Hematology Am Soc Hematol Educ Program. 2015;2015:299–307.
  • Kwok B, Hall JM, Witte JS, et al. MDS-associated somatic mutations and clonal hematopoiesis are common in idiopathic cytopenias of undetermined significance. Blood. 2015;126(21):2355–2361.
  • Cargo CA, Rowbotham N, Evans PA, et al. Targeted sequencing identifies patients with preclinical MDS at high risk of disease progression. Blood. 2015;126(21):2362–2365.
  • Malcovati L, Galli A, Travaglino E, et al. Clinical significance of somatic mutation in unexplained blood cytopenia. Blood. 2017;129(25):3371–3378.
  • Heuser M, Thol F, Ganser A. Clonal hematopoiesis of indeterminate potential. Dtsch Arztebl Int. 2016;113(18):317–322.
  • Steensma DP, Bejar R, Jaiswal S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126(1):9–16.
  • Sellar RS, Jaiswal S, Ebert BL. Predicting progression to AML. Nat Med. 2018;24(7):904–906.
  • Hansen JW, Westman MK, Sjo LD, et al. Mutations in idiopathic cytopenia of undetermined significance assist diagnostics and correlate to dysplastic changes. Am J Hematol. 2016;91(12):1234–1238.
  • Snetsinger B, Heath E, Rauh MJ. Suspicious, non-MDS-diagnostic bone marrows have a high incidence of clonal hematopoiesis (CHIP), with MDS-like clone size but restricted mutation burden. Blood. 2015;126(23):1668.
  • Abelson S, Collord G, Ng SWK, et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature. 2018;559(7714):400–404.
  • Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–2498.
  • Bejar R, Papaemmanuil E, Haferlach T, et al. Somatic mutations in MDS patients are associated with clinical features and predict prognosis independent of the IPSS-R: analysis of combined datasets from the International Working Group for Prognosis in MDS-Molecular Committee. Blood. 2015;126(23):907.
  • Malcovati L, Karimi M, Papaemmanuil E, et al. SF3B1 mutation identifies a distinct subset of myelodysplastic syndrome with ring sideroblasts. Blood. 2015;126(2):233–241.
  • Bejar R. Implications of molecular genetic diversity in myelodysplastic syndromes. Curr Opin Hematol. 2017;24(2):73–78.
  • Nazha A, Narkhede M, Radivoyevitch T, et al. Incorporation of molecular data into the Revised International Prognostic Scoring System in treated patients with myelodysplastic syndromes. Leukemia. 2016;30(11):2214–2220.
  • Zeidan AM, Sekeres MA, Wang XF, et al. Comparing the prognostic value of risk stratifying models for patients with lower-risk myelodysplastic syndromes: is one model better? Am J Hematol. 2015;90(11):1036–1040.
  • Lee EJ, Podoltsev N, Gore SD, et al. The evolving field of prognostication and risk stratification in MDS: recent developments and future directions. Blood Rev. 2016;30(1):1–10.
  • Santos FP, Kantarjian H, Garcia-Manero G, et al. The search for better prognostic models in myelodysplastic syndromes. Curr Hematol Malig Rep. 2011;6(1):13–21.
  • Zeidan AM, Komrokji RS. There’s risk, and then there’s risk: the latest clinical prognostic risk stratification models in myelodysplastic syndromes. Curr Hematol Malig Rep. 2013;8(4):351–360.
  • Zeidan AM, Gore SD, Padron E, et al. Current state of prognostication and risk stratification in myelodysplastic syndromes. Curr Opin Hematol. 2015;22(2):146–154.
  • Zeidan AM, Sekeres MA, Garcia-Manero G, et al. Comparison of risk stratification tools in predicting outcomes of patients with higher-risk myelodysplastic syndromes treated with azanucleosides. Leukemia. 2016;30(3):649–657.
  • Zeidan AM, Smith BD, Komrokji RS, et al. Prognostication in myelodysplastic syndromes: beyond the International Prognostic Scoring System (IPSS). Am J Med. 2013;126(4):e25.
  • Nazha A, Sekeres MA, Gore SD, et al. Molecular testing in myelodysplastic syndromes for the practicing oncologist: will the progress fulfill the promise? Oncologist. 2015;20(9):1069–1076.
  • Lee EJ, Zeidan AM. Genome sequencing in myelodysplastic syndromes: can molecular mutations predict benefit from hypomethylating agent therapy? Expert Rev Hematol. 2015;8(2):155–158.
  • DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378(25):2386–2398.
  • Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130(6):722–731.
  • Itzykson R, Kosmider O, Cluzeau T, et al. Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias. Leukemia. 2011;25(7):1147–1152.
  • Tobiasson M, McLornan DP, Karimi M, et al. Mutations in histone modulators are associated with prolonged survival during azacitidine therapy. Oncotarget. 2016;7(16):22103–22115.
  • Takahashi K, Wang F, Sahil S, et al. Presence of 4 or more driver mutations predicts poor response to hypomethylating agent (HMA) therapy and poor overall survival in MDS. Blood. 2015;126(23):1663.
  • Takahashi K, Kantarjian HM, Patel K, et al. Mutated MDS patients respond equally to hypomethylating agents but have significantly shorter response duration compared to patients with wild type . Blood. 2015;126(23):1681.
  • Welch JS, Petti AA, Miller CA, et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med. 2016;375(21):2023–2036.
  • Jadersten M, Saft L, Smith A, et al. TP53 mutations in low-risk myelodysplastic syndromes with del(5q) predict disease progression. J Clin Oncol. 2011;29(15):1971–1979.
  • List A, Dewald G, Bennett J, et al. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med. 2006;355(14):1456–1465.
  • Fenaux P, Giagounidis A, Selleslag D, et al. A randomized phase 3 study of lenalidomide versus placebo in RBC transfusion-dependent patients with Low-/Intermediate-1-risk myelodysplastic syndromes with del5q. Blood. 2011;118(14):3765–3776.
  • Wei S, Chen X, McGraw K, et al. Lenalidomide promotes p53 degradation by inhibiting MDM2 auto-ubiquitination in myelodysplastic syndrome with chromosome 5q deletion. Oncogene. 2013;32(9):1110–1120.
  • Schneider RK, Schenone M, Ferreira MV, et al. Rps14 haploinsufficiency causes a block in erythroid differentiation mediated by S100A8 and S100A9. Nat Med. 2016;22(3):288–297.
  • Lode L, Menard A, Flet L, et al. Emergence and evolution of TP53 mutations are key features of disease progression in myelodysplastic patients with lower-risk del(5q) treated with lenalidomide. Haematologica. 2018;103(4):e143–e6.
  • Komrokji RS, Haider M, Al Ali NH, et al. Somatic gene mutations serve as molecular biomarkers predictive for response to immunosuppressive therapy (IST) in myelodysplastic syndromes (MDS). Blood. 2015;126(23):1664.
  • Mies A, Platzbecker U. Increasing the effectiveness of hematopoiesis in myelodysplastic syndromes: erythropoiesis-stimulating agents and transforming growth factor-beta superfamily inhibitors. Semin Hematol. 2017;54(3):141–146.
  • Platzbecker U, Germing U, Gotze KS, et al. Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): a multicentre, open-label phase 2 dose-finding study with long-term extension study. Lancet Oncol. 2017;18(10):1338–1347.
  • Fenaux P, Platzbecker U, Mufti GJ, et al. The medalist trial: results of a phase 3, randomized, double-blind, placebo-controlled study of luspatercept to treat anemia in patients with very low-, low-, or intermediate-risk myelodysplastic syndromes (MDS) with ring sideroblasts (RS) who require red blood cell (RBC) transfusions. Blood. 2018;132(Suppl 1):1.
  • Montalban-Bravo G, Takahashi K, Patel K, et al. Impact of the number of mutations in survival and response outcomes to hypomethylating agents in patients with myelodysplastic syndromes or myelodysplastic/myeloproliferative neoplasms. Oncotarget. 2018;9(11):9714–9727.
  • Greenberg PL, Stone RM, Al-Kali A, et al. Myelodysplastic syndromes, version 2.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2017;15(1):60–87.
  • Saber W, Horowitz MM. Transplantation for myelodysplastic syndromes: who, when, and which conditioning regimens. Hematology Am Soc Hematol Educ Program. 2016;2016(1):478–484.
  • de Witte T, Bowen D, Robin M, et al. Allogeneic hematopoietic stem cell transplantation for MDS and CMML: recommendations from an international expert panel. Blood. 2017;129(13):1753–1762.
  • Zeidan AM, Linhares Y, Gore SD. Current therapy of myelodysplastic syndromes. Blood Rev. 2013;27(5):243–259.
  • Zeidan AM, Gore SD. Should elderly patients with higher-risk myelodysplastic syndromes undergo allogeneic hematopoietic stem cell transplantation? Expert Rev Hematol. 2013;6(5):539–542.
  • Malcovati L, Papaemmanuil E, Ambaglio I, et al. Driver somatic mutations identify distinct disease entities within myeloid neoplasms with myelodysplasia. Blood. 2014;124(9):1513–1521.
  • Lindsley RC, Saber W, Mar BG, et al. Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation. N Engl J Med. 2017;376(6):536–547.
  • Della Porta MG, Galli A, Bacigalupo A, et al. Clinical effects of driver somatic mutations on the outcomes of patients with myelodysplastic syndromes treated with allogeneic hematopoietic stem-cell transplantation. J Clin Oncol. 2016;34(30):3627–3637.
  • Cimmino L, Dolgalev I, Wang Y, et al. Restoration of TET2 function blocks aberrant self-renewal and leukemia progression. Cell. 2017;170(6):1079–95 e20.
  • Zhang Q, Bykov VJN, Wiman KG, et al. APR-246 reactivates mutant p53 by targeting cysteines 124 and 277. Cell Death Dis. 2018;9(5):439.
  • Deneberg S, Cherif H, Lazarevic V, et al. An open-label phase I dose-finding study of APR-246 in hematological malignancies. Blood Cancer J. 2016;6(7):e447.
  • Lehmann S, Bykov VJ, Ali D, et al. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol. 2012;30(29):3633–3639.
  • Shapiro RM, Kim DDH. Next-generation sequencing-based minimal residual disease monitoring in patients receiving allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia or myelodysplastic syndrome. Curr Opin Hematol. 2018;25(6):425–432.
  • Schuurhuis GJ, Heuser M, Freeman S, et al. Minimal/measurable residual disease in AML: a consensus document from the European Leukemianet MRD Working Party. Blood. 2018;131(12):1275–1291.
  • Grimwade D, Freeman SD. Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for “prime time”? Blood. 2014;124(23):3345–3355.
  • Nassereddine S, Nishihori T, Padron E, et al. Integrating genomics in myelodysplastic syndrome to predict outcomes after allogeneic hematopoietic cell transplantation. Clin Lymphoma Myeloma Leuk. 2017;17(1):7–13.
  • Kharfan-Dabaja MA, Komrokji RS, Zhang Q, et al. TP53 and IDH2 somatic mutations are associated with inferior overall survival after allogeneic hematopoietic cell transplantation for myelodysplastic syndrome. Clin Lymphoma Myeloma Leuk. 2017;17(11):753–758.
  • Gibson CJ, Kennedy JA, Nikiforow S, et al. Donor-engrafted CHIP is common among stem cell transplant recipients with unexplained cytopenias. Blood. 2017;130(1):91–94.
  • Thol F, Gabdoulline R, Liebich A, et al. Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood. 2018;132(16):1703–1713.
  • Duncavage EJ, Jacoby MA, Chang GS, et al. Mutation clearance after transplantation for myelodysplastic syndrome. N Engl J Med. 2018;379(11):1028–1041.
  • Fu Y, Schroeder T, Zabelina T, et al. Postallogeneic monitoring with molecular markers detected by pretransplant next-generation or Sanger sequencing predicts clinical relapse in patients with myelodysplastic/myeloproliferative neoplasms. Eur J Haematol. 2014;92(3):189–194.
  • Pine AB, Chokr N, Stahl M, et al. Wide variation in use and interpretation of gene mutation profiling panels among health care providers of patients with Myelodysplastic Syndromes (MDS): results of a large web-based survey. Blood. 2018;132:1825.
  • Kanagal-Shamanna R, Loghavi S, DiNardo CD, et al. Bone marrow pathologic abnormalities in familial platelet disorder with propensity for myeloid malignancy and germline RUNX1 mutation. Haematologica. 2017;102(10):1661–1670.
  • Kratz CP, Achatz MI, Brugieres L, et al. Cancer screening recommendations for individuals with Li-Fraumeni syndrome. Clin Cancer Res. 2017;23(11):e38–e45.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.