Addressing MDS Treatment Gap Post Hypomethylating Failure

Publication
Article
Targeted Therapies in OncologyMarch I, 2024
Volume 13
Issue 3
Pages: 84

The frontline standard-of-care regimen for patients with higher-risk MDS and some with lower-risk MDS remain hypomethylating agents, such as azacitidine, decitabine, and decitabine/cedazuridine.

a close-up of red blood cells flowing through a vein, displaying the characteristic sickle shape Generative AI: © catalin - stock.adobe.com

A close-up of red blood cells flowing through a vein, displaying the characteristic sickle shape Generative AI: © catalin - stock.adobe.com

Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic stem cell disorders characterized by bone marrow dysplasia, cytopenia, and a propensity to progress to acute monocytic leukemia (AML).1 Treatment decisions in MDS have traditionally been based on various prognostic factors, such as peripheral blood counts, bone marrow blast percentage, and cytogenetics, that predict survival and progression to AML, with the International Prognostic Scoring System (IPSS) and revised IPSS (IPSS-R) being the most widely used prognostic systems.2,3 Although multiple studies have demonstrated the impact of various somatic mutations on prognoses over a decade, it was not until the recently published Molecular IPSS (IPSS-M) that a risk stratification system incorporated molecular patient factors.4-8

The frontline standard-of-care regimen for patients with higher-risk MDS and some with lower-risk MDS remain hypomethylating agents (HMAs), such as azacitidine (Vidaza), decitabine (Dacogen), and decitabine/cedazuridine (Inqovi).9-17 The use of these therapeutic agents results in improved cytopenias and survival, but these benefits are transient and modest in magnitude.18 Patients who are refractory to or progress after responding to HMA therapy are faced with a phenomenon known as HMA failure.19 The median overall survival for individuals with higher-risk MDS post-HMA failure is approximately 4 to 6 months and approximately 17 months for those with lower-risk HMA failure MDS.20-22 Furthermore, HMA failure MDS is associated with extremely high health care resource utilization burdens.23,24

There have been several studies looking at predictive biomarkers for response to HMA, including bone marrow blast percentage, previous treatments, cytogenetics, molecular studies, and gene expression profiles.25-31 However, these are not readily used, as alternative treatments to HMAs are unavailable. Therefore, the improvement of frontline therapy and the development of novel therapeutics after HMA failure remain urgent and unmet needs in the field of MDS.

Therapeutic Options for HMA Failure MDS

Until recently, there were no FDA-approved therapeutic options for patients with MDS who do not respond to or have lost their response to HMA therapy. Ivosidenib (Tibsovo) recently obtained approval for patients with IDH1-mutated relapsed or refractory MDS after HMAs after demonstrating a complete response rate of 39%.32 However, no other treatments are commercially available for those without IDH1 mutations. Given that IDH1 mutations are extremely rare in MDS (approximately 5% of MDS patients have IDH1/2 mutations5,33,34), most patients still do not have any FDA-approved therapies.

There are many clinical trials researching novel therapeutics in HMA failure MDS.35 In addition to IDH1/2 inhibitors, these investigational agents can be divided into immune checkpoint, inflammation pathway, antiapoptotic, multikinase, and NEDDylation inhibitors. Therefore, referral to a tertiary care center for clinical trial options is warranted for patients with HMA failure MDS.

Managing HMA Failure MDS

In the case of suspected HMA failure MDS, complete laboratory and bone marrow examination is recommended. Although the IPSS-R was shown to lose predictive power in those who have already been treated with HMA and the IPSS-M does not provide much additional prognostic value at this time,36,37 having an understanding of the genomic components at this time is paramount, either for clinical trial enrollment or obtaining ivosidenib with IDH1-mutated disease. It is also possible that patients may have transformed to secondary AML38; though outcomes are dismal, more therapies are available in that situation, including induction chemotherapy regimens and BCL2 inhibition with venetoclax (Venclexta).

Fit patients with higher-risk MDS by IPSS-R or lower-risk MDS by IPSS-R with poor-risk genetic features at diagnosis are candidates for allogeneic stem cell transplantation (alloSCT),39 which remains the only curative treatment option. In the case of HMA failure and relapsed/refractory MDS, all fit patients are recommended to undergo alloSCT, as it results in prolonged survival compared with other treatment modalities.21 Although there was a higher risk of post-alloSCT relapse in patients with HMA failure MDS compared with those who underwent alloSCT during response,40 results are still more encouraging than the dismal survival outcomes detailed above.

Summary Points

  • HMA failure MDS is associated with poor survival outcomes.
  • The only commercially available treatment option is ivosidenib for patients with IDH1-mutated MDS after HMA failure.
  • Referral to a tertiary cancer center for clinical trial options is paramount.
  • Consider alloSCT in all fit patients with MDS after HMA failure.

REFERENCES:
1. Nimer SD. Myelodysplastic syndromes. Blood. 2008;111(10):4841-4851. doi:10.1182/blood-2007-08-078139
2. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89(6):2079-2088.
3. Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454-2465. doi:10.1182/blood-2012-03-420489
4. 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. doi:10.1056/NEJMoa1013343
5. Papaemmanuil E, Gerstung M, Malcovati L, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122(22):3616-3699. doi:10.1182/blood-2013-08-518886
6. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28(2):241-247. doi:10.1038/leu.2013.336
7. Bersanelli M, Travaglino E, Meggendorfer M, et al. Classification and personalized prognostic assessment on the basis of clinical and genomic features in myelodysplastic syndromes. J Clin Oncol. 2021;39(11):1223-1233. doi:10.1200/JCO.20.01659
8. Bernard E, Nannya Y, Hasserjian RP, et al. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat Med. 2020;26(10):1549-1556. doi:10.1038/s41591-020-1008-z
9. Silverman LR, Demakos EP, Peterson BL, et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 2002;20(10):2429-2440. doi:10.1200/JCO.2002.04.117
10. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10(3):223-232. doi:10.1016/S1470-2045(09)70003-8
11. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer. 2006;106(8):1794-1803. doi:10.1002/cncr.21792
12. Kantarjian H, Oki Y, Garcia-Manero G, et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood. 2007;109(1):52-57. doi:10.1182/blood-2006-05-021162
13. Jabbour E, Short NJ, Montalban-Bravo G, et al. Randomized phase 2 study of low-dose decitabine vs low-dose azacitidine in lower-risk MDS and MDS/MPN. Blood. 2017;130(13):1514-1522. doi:10.1182/blood-2017-06-788497
14. Sasaki K, Jabbour E, Montalban-Bravo G, et al. Low-dose decitabine versus low-dose azacitidine in lower-risk MDS. NEJM Evid. 2022;1(10). doi:10.1056/EVIDoa2200034
15. Savona MR, Odenike O, Amrein PC, et al. An oral fixed-dose combination of decitabine and cedazuridine in myelodysplastic syndromes: a multicentre, open-label, dose-escalation, phase 1 study. Lancet Haematol. 2019;6(4):e194-e203. doi:10.1016/S2352-3026(19)30030-4
16. Garcia-Manero G, Griffiths EA, Steensma DP, et al. Oral cedazuridine/decitabine for MDS and CMML: a phase 2 pharmacokinetic/pharmacodynamic randomized crossover study. Blood. 2020;136(6):674-683. doi:10.1182/blood.2019004143
17. Garcia-Manero G, McCloskey JK, Griffiths EA, et al. Oral decitabine/cedazuridine in patients with lower risk myelodysplastic syndrome: a longer-term follow-up of from the Ascertain study. Blood. 2021;138:66. doi:10.1182/blood-2021-144648
18. Zeidan AM, Stahl M, Hu X, et al. Long-term survival of older patients with MDS treated with HMA therapy without subsequent stem cell transplantation. Blood. 2018;131(7):818-821. doi:10.1182/blood-2017-10-811729
19. Santini V. How I treat MDS after hypomethylating agent failure. Blood. 2019;133(6):521-529. doi:10.1182/blood-2018-03-785915
20. Jabbour E, Garcia-Manero G, Batty N, et al. Outcome of patients with myelodysplastic syndrome after failure of decitabine therapy. Cancer. 2010;116(16):3830-3834. doi:10.1002/cncr.25247
21. Prébet T, Gore SD, Esterni B, et al. Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure. J Clin Oncol. 2011;29(24):3322-3327. doi:10.1200/JCO.2011.35.8135
22. Jabbour EJ, Garcia-Manero G, Strati P, et al. Outcome of patients with low-risk and intermediate-1-risk myelodysplastic syndrome after hypomethylating agent failure: a report on behalf of the MDS Clinical Research Consortium. Cancer. 2015;121(6):876-882. doi:10.1002/cncr.29145
23. Stein EM, Bonifacio G, Latrémouille-Viau D, et al. Healthcare resource utilization and costs in patients with myelodysplastic syndromes treated with hypomethylating agents: a SEER-Medicare analysis. J Med Econ. 2021;24(1):234-243. doi:10.1080/13696998.2021.1876714
24. Cheng WY, Satija A, Cheung HC, et al. Persistence to hypomethylating agents and clinical and economic outcomes among patients with myelodysplastic syndromes. Hematology. 2021;26(1):261-270. doi:10.1080/16078454.2021.1889161
25. Itzykson R, Thépot S, Quesnel B, et al. Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine. Blood. 2011;117(2):403-411. doi:10.1182/blood-2010-06-289280
26. 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. doi:10.1182/blood-2014-06-582809
27. Garzon R, Heaphy CE, Havelange V, et al. MicroRNA 29b functions in acute myeloid leukemia. Blood. 2009;114(26):5331-5341. doi:10.1182/blood-2009-03-211938
28. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc Natl Acad Sci U S A. 2010;107(16):7473-7478. doi:10.1073/pnas.1002650107
29. Valencia A, Masala E, Rossi A, et al. Expression of nucleoside-metabolizing enzymes in myelodysplastic syndromes and modulation of response to azacitidine. Leukemia. 2014;28(3):621-628. doi:10.1038/leu.2013.330
30. Sripayap P, Nagai T, Uesawa M, et al. Mechanisms of resistance to azacitidine in human leukemia cell lines. Exp Hematol. 2014;42(4):294-306.e2. doi:10.1016/j.exphem.2013.12.004
31. Yang H, Bueso-Ramos C, DiNardo C, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014;28(6):1280-1288. doi:10.1038/leu.2013.355
32. DiNardo C, Roboz G, M. Watts J, et al. Updated substudy results for ivosidenib in idh1-mutant relapsed/refractory myelodysplastic syndrome. Hemasphere. 2023;7(suppl 3):e75740ab. doi:10.1097/01.HS9.0000969800.75740.ab
33. DiNardo CD, Jabbour E, Ravandi F, et al. IDH1 and IDH2 mutations in myelodysplastic syndromes and role in disease progression. Leukemia. 2016;30(4):980-984. doi:10.1038/leu.2015.211
34. Kosmider O, Gelsi-Boyer V, Slama L, et al. Mutations of IDH1 and IDH2 genes in early and accelerated phases of myelodysplastic syndromes and MDS/myeloproliferative neoplasms. Leukemia. 2010;24(5):1094-1096. doi:10.1038/leu.2010.52
35. Rodriguez-Sevilla JJ, Adema V, Garcia-Manero G, Colla S. Emerging treatments for myelodysplastic syndromes: biological rationales and clinical translation. Cell Rep Med. 2023;4(2):100940. doi:10.1016/j.xcrm.2023.100940
36. Blum S, Gattermann N, Martins F, et al. Is the IPSS-R useful for patients with MDS receiving disease-modifying treatment? Blood.2016;128(22):5526. doi:10.1182/blood.V128.22.5526.5526
37. Urrutia S, Chien KS, Li Z, et al. Performance of IPSS-M in patients with myelodysplastic syndrome after hypomethylating agent failure. Am J Hematol. 2023;98(10):E281-E284. doi:10.1002/ajh.27043
38. Short NJ, Venugopal S, Qiao W, et al. Impact of frontline treatment approach on outcomes in patients with secondary AML with prior hypomethylating agent exposure. J Hematol Oncol.2022;15(1):12. doi:10.1186/s13045-022-01229-z
39. 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. doi:10.1182/blood-2016-06-724500
40. Festuccia M, Baker K, Gooley TA, Sandmaier BM, Deeg HJ, Scott BL. Hematopoietic cell transplantation in myelodysplastic syndromes after treatment with hypomethylating agents. Biol Blood Marrow Transplant. 2017;23(9):1509-1514. doi:10.1016/j.bbmt.2017.05.034
Related Videos
Andrew Kuykendall, MD, an expert on polycythemia vera
Andrew Kuykendall, MD, an expert on polycythemia vera
Andrew Kuykendall, MD, an expert on polycythemia vera
Andrew Kuykendall, MD, an expert on polycythemia vera
Andrew Kuykendall, MD, an expert on polycythemia vera
Andrew Kuykendall, MD, an expert on polycythemia vera
Andrew Kuykendall, MD, an expert on polycythemia vera
Andrew Kuykendall, MD, an expert on polycythemia vera
Prithviraj Bose, MD, an expert on myelofibrosis
Prithviraj Bose, MD, an expert on myelofibrosis
Related Content