Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1258 2023-05-03 00:10:57 |
2 format correct Meta information modification 1258 2023-05-05 02:38:39 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Awada, H.; Gurnari, C.; Xie, Z.; Bewersdorf, J.P.; Zeidan, A.M. Hypomethylating Agents Failure. Encyclopedia. Available online: https://encyclopedia.pub/entry/43690 (accessed on 27 July 2024).
Awada H, Gurnari C, Xie Z, Bewersdorf JP, Zeidan AM. Hypomethylating Agents Failure. Encyclopedia. Available at: https://encyclopedia.pub/entry/43690. Accessed July 27, 2024.
Awada, Hussein, Carmelo Gurnari, Zhuoer Xie, Jan Philipp Bewersdorf, Amer M. Zeidan. "Hypomethylating Agents Failure" Encyclopedia, https://encyclopedia.pub/entry/43690 (accessed July 27, 2024).
Awada, H., Gurnari, C., Xie, Z., Bewersdorf, J.P., & Zeidan, A.M. (2023, May 03). Hypomethylating Agents Failure. In Encyclopedia. https://encyclopedia.pub/entry/43690
Awada, Hussein, et al. "Hypomethylating Agents Failure." Encyclopedia. Web. 03 May, 2023.
Hypomethylating Agents Failure
Edit

Hypomethylating agents (HMA) such as azacitidine and decitabine are a mainstay in the current management of patients with myelodysplastic syndromes/neoplasms (MDS) and acute myeloid leukemia (AML) as either single agents or in multidrug combinations. Resistance to HMA is not uncommon, and it can result due to several tumor cellular adaptations. Several clinical and genomic factors have been identified as predictors of HMA resistance. However, the management of MDS/AML patients after the failure of HMA remains challenging in the absence of standardized guidelines.

hypomethylating agents myelodysplastic syndromes/neoplasms acute myeloid leukemia

1. Introduction

The hypomethylating agents (HMA) azacitidine (AZA) and decitabine (DEC) have been a mainstay of the treatment of myeloid neoplasms for almost two decades now. These two closely-related agents primarily act by epigenetic mechanisms, the disruption of which is central to the pathogenesis of these disorders, thereby reinducing the expression of silenced critical genes [1]. In particular, HMA inhibits DNA methyltransferase-1 (DNMT-1) by forming covalent bonds between this enzyme and the DNA containing the molecules, which are incorporated during DNA synthesis [1]. HMA also possesses direct cytotoxic effects, especially at higher doses [1]. Both HMAs have been approved in oral and injectable forms by the Food and Drug Administration (FDA) for specific indications that are not interchangeable in the treatment of myelodysplastic syndromes/neoplasms (MDS) and acute myeloid leukemia (AML) [1][2][3][4]. While allogeneic hematopoietic cell transplant (Allo-HCT) remains the only potentially curative therapeutic modality for MDS, most patients are older and considered ineligible for intensive therapies such as Allo-HCT, and instead opt for lower-intensity treatments with palliative intent [5][6]. Indeed, AZA demonstrated superior overall survival (OS) vs. conventional care regimens including supportive care, and low or intensive chemotherapy in HR-MDS patients in a randomized trial setting [7][8][9]. Although DEC has not shown a similar statistically significant OS benefit versus physician choice (median OS 10.5 vs. 8.1 months, p = 0.38), yet it demonstrated clinically meaningful benefits such as improvements in leukemia-free survival (median 6.6 vs. 3.0, p = 0.004) and quality of life (QoL) endpoints [7][10]. The survival benefit of HMA is somewhat limited to HR-MDS, as neither agent has conclusive evidence of prolonging OS in low-risk LR-MDS (defined as IPSS-R < 3.5) despite increasing the hematological response and reducing the risk of leukemic progression [11][12][13][14][15]. Paralleling the observations in the HR setting, no differences with regards to the injectable type of HMA used exist also in LR-MDS (when available to be used according to local regulations) [16][17].
As for patients with AML, the use of HMA is confined to those deemed ‘medically unfit’ for intensive antileukemic chemotherapy. Medical fitness is typically determined by a comprehensive assessment that includes subjective and objective tools such as the Eastern Cooperative Oncology Group (ECOG) performance scale and Charlson comorbidity index (CCI) scores, in addition to the validated and objective Ferrara Consensus criteria [18][19][20]. Age has also often been used as a proxy for fitness, although the correlation is far from perfect. For older and unfit patients, HMA-based approaches provide an alternative, less toxic treatment options. The preference for HMA in this setting is based on several randomized trials and prospective cohorts, in which HMA led to superior clinical outcomes with limited toxicity versus other low-intensity agents such as low-dose cytarabine, other targeted therapies, or supportive care [7][21][22]. In contrast to MDS, DEC has a similar survival advantage to that of AZA in AML [23][24][25][26][27][28]. Furthermore, the enhanced survival benefits gained by combining AZA with other agents such as Venetoclax (VEN) in AML have made HMA-based combinations an even more popular therapeutic option compared to single-agent HMA [29].
Despite the encouraging evidence, long-term outcomes of HR-MDS and AML patients treated with HMA remain suboptimal, especially for patients diagnosed at an older age with 5-year survival rates not exceeding 6% [30][31][32][33][34]. Outcomes are even worse in patients who do not respond to HMA at all (i.e., primary resistance/failure) or who experience disease progression after a transient period of response (i.e., secondary resistance/failure) [35][36][37]. As such, the management of patients with HMA failure is challenging due to the aggressive nature of the disease and the lack of FDA-approved therapies, as well as the patient characteristics (elderly patients not usually fit for intensive treatments) [35].

2. Hypomethylating Agents Failure: Definition, Mechanisms, and Prognosis

HMA failure is generally categorized into primary or secondary based on the patient’s initial response to treatment. In MDS, primary failure is generally defined as the lack of benefit defined by blast reduction or improvement in blood counts after at least four to six cycles of initial therapy, or MDS progression to higher-risk categories or transformation to AML. Secondary failure occurs in patients who progress despite initially responding to HMA. It is defined as worsening blood counts or progression of MDS to higher-risk categories or AML following the initial response to HMA. In AML, primary HMA failure is instead defined as failure to achieve a complete remission- or complete remission with incomplete count recovery (CR/CRi) while secondary failure occurs with the loss of CR/CRi [38]. To date, studies have suggested that the primary response to HMA in HR-MDS occurs in approximately 50%, while around 36% eventually become resistant and thus develop secondary failure [36][39][40]. Indeed, long-lasting remissions are achieved in only a minority of HR-MDS and AML patients receiving HMA [7][21].
Resistance to HMA remains poorly understood with several proposed mechanisms, most likely secondary to changes in the metabolism of these drugs [41]. Indeed, HMAs are prodrugs, whose activation requires phosphorylation by the uridine-cytidine kinase (UCK) and deoxycytidine kinase (DCK) [42]. Cell-line gene knock-out experiments suggest that silencing the expression of UCK and DCK correlate with decreased AZA and DEC activities, respectively [42][43]. Valencia et al. further corroborated this finding in a study of 57 MDS patients in whom resistant patients had lower UCK expression [44]. Similarly, Wu et al. noted a significant decrease in DCK at relapse (p = 0.012) among MDS patients with secondary failure, while no changes were noted in those with an ongoing response [45]. Alternatively, HMAs may lose their therapeutic effect if rapidly cleared by the cytidine deaminase (CDA) enzyme. CDA catalyzes the hydrolytic cleavage which results in the deamination-mediated inactivation of HMA [46]. Mahfouz et al. studied 90 MDS patients and observed that males had higher CDA levels compared to females [47]. Higher CDA levels were associated with HMA resistance and lower survival (median 563 vs. 1033 days, p = 0.01) [47]. Another mechanism generating a reduction in HMA potency may be a decrease in their influx or an increase in their efflux from leukemia cells, even though reports remain conflicting as of today. Indeed, two studies have unmasked significantly higher mRNA expression of the human equilibrative nucleoside transporter (hENT1), an HMA importer, in responders vs. non-responders [45][48]. However, a third study by Qin et al. did not observe such a correlation, concluding that hENT1 downregulation does not seem to play a significant role in developing resistance to HMA [44][49]. HMA resistance through increased cellular efflux is less studied, however, in vitro models suggest that the multidrug resistance-associated protein seven (MRP7) reduces HMA accumulation in the target cells [50]. Other proposed resistance mechanisms include altered responses to DNA damage, and changes in endosomal/exosomal and microvesicular cell communication [41].
As aforementioned, HMA failure is highly associated with reduced survival. Even for LR-MDS, the post-HMA median transformation-free survival reaches only 15 months while OS barely exceeds 17 months [35]. Among HR-MDS patients, the median OS is <6 months, with only 15% alive at 2 years [36]. Factors such as increasing age, male gender, high-risk cytogenetics and mutations, bone marrow blast count, and secondary rather than primary resistance influence post-HMA outcome [36]. In contrast, undergoing subsequent Allo-HCT or using investigational agents appear to marginally improve outcomes in HR-MDS post-HMA resistance [36]. Outcomes are even worse for AML patients, who after HMA failure have a median survival between 1.3 and 2 months [36][37].

References

  1. Short, N.J.; Kantarjian, H. Hypomethylating agents for the treatment of myelodysplastic syndromes and acute myeloid leukemia: Past discoveries and future directions. Am. J. Hematol. 2022, 97, 1616–1626.
  2. Sekeres, M.A.; Taylor, J. Diagnosis and Treatment of Myelodysplastic Syndromes: A Review. JAMA 2022, 328, 872–880.
  3. Wei, A.H.; Döhner, H.; Pocock, C.; Montesinos, P.; Afanasyev, B.; Dombret, H.; Ravandi, F.; Sayar, H.; Jang, J.H.; Porkka, K.; et al. Oral Azacitidine Maintenance Therapy for Acute Myeloid Leukemia in First Remission. N. Engl. J. Med. 2020, 383, 2526–2537.
  4. Garcia-Manero, G.; Griffiths, E.A.; Steensma, D.P.; Roboz, G.J.; Wells, R.; McCloskey, J.; Odenike, O.; DeZern, A.E.; Yee, K.; Busque, L.; et al. Oral cedazuridine/decitabine for MDS and CMML: A phase 2 pharmacokinetic/pharmacodynamic randomized crossover study. Blood 2020, 136, 674–683.
  5. Zeidan, A.M.; Shallis, R.M.; Wang, R.; Davidoff, A.; Ma, X. Epidemiology of myelodysplastic syndromes: Why characterizing the beast is a prerequisite to taming it. Blood Rev. 2019, 34, 1–15.
  6. Gurnari, C.; Xie, Z.; Zeidan, A.M. How I Manage Transplant Ineligible Patients with Myelodysplastic Neoplasms. Clin. Hematol. Int. 2022, 5, 8–20.
  7. Fenaux, P.; Mufti, G.J.; Hellstrom-Lindberg, E.; Santini, V.; Finelli, C.; Giagounidis, A.; Schoch, R.; Gattermann, N.; Sanz, G.; List, A.; 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, 223–232.
  8. Itzykson, R.; Thépot, S.; Quesnel, B.; Dreyfus, F.; Beyne-Rauzy, O.; Turlure, P.; Vey, N.; Recher, C.; Dartigeas, C.; Legros, L.; et al. Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine. Blood 2011, 117, 403–411.
  9. Itzykson, R.; Thépot, S.; Quesnel, B.; Dreyfus, F.; Recher, C.; Wattel, E.; Gardin, C.; Adès, L.; Fenaux, P. Long-term outcome of higher-risk MDS patients treated with azacitidine: An update of the GFM compassionate program cohort. Blood 2012, 119, 6172–6173.
  10. Lübbert, M.; Suciu, S.; Baila, L.; Rüter, B.H.; Platzbecker, U.; Giagounidis, A.; Selleslag, D.; Labar, B.; Germing, U.; Salih, H.R.; et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high-risk myelodysplastic syndrome (MDS) ineligible for intensive chemotherapy: Final results of the randomized phase III study of the European Organisation for Research and Treatment of Cancer Leukemia Group and the German MDS Study Group. J. Clin. Oncol. 2011, 29, 1987–1996.
  11. Pfeilstöcker, M.; Tuechler, H.; Sanz, G.; Schanz, J.; Garcia-Manero, G.; Solé, F.; Bennett, J.M.; Bowen, D.; Fenaux, P.; Dreyfus, F.; et al. Time-dependent changes in mortality and transformation risk in MDS. Blood 2016, 128, 902–910.
  12. Musto, P.; Maurillo, L.; Spagnoli, A.; Gozzini, A.; Rivellini, F.; Lunghi, M.; Villani, O.; Aloe-Spiriti, M.A.; Venditti, A.; Santini, V. Azacitidine for the treatment of lower risk myelodysplastic syndromes: A retrospective study of 74 patients enrolled in an Italian named patient program. Cancer 2010, 116, 1485–1494.
  13. Sanchez-Garcia, J.; Falantes, J.; Medina Perez, A.; Hernandez-Mohedo, F.; Hermosin, L.; Torres-Sabariego, A.; Bailen, A.; Hernandez-Sanchez, J.M.; Solé Rodriguez, M.; Casaño, F.J.; et al. Prospective randomized trial of 5 days azacitidine versus supportive care in patients with lower-risk myelodysplastic syndromes without 5q deletion and transfusion-dependent anemia. Leuk. Lymphoma 2018, 59, 1095–1104.
  14. Kantarjian, H.; Issa, J.P.; Rosenfeld, C.S.; Bennett, J.M.; Albitar, M.; DiPersio, J.; Klimek, V.; Slack, J.; de Castro, C.; Ravandi, F.; et al. Decitabine improves patient outcomes in myelodysplastic syndromes: Results of a phase III randomized study. Cancer 2006, 106, 1794–1803.
  15. Garcia-Manero, G.; Jabbour, E.; Borthakur, G.; Faderl, S.; Estrov, Z.; Yang, H.; Maddipoti, S.; Godley, L.A.; Gabrail, N.; Berdeja, J.G.; et al. Randomized open-label phase II study of decitabine in patients with low- or intermediate-risk myelodysplastic syndromes. J. Clin. Oncol. 2013, 31, 2548–2553.
  16. Jabbour, E.; Short, N.J.; Montalban-Bravo, G.; Huang, X.; Bueso-Ramos, C.; Qiao, W.; Yang, H.; Zhao, C.; Kadia, T.; Borthakur, 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, 1514–1522.
  17. Gurion, R.; Vidal, L.; Gafter-Gvili, A.; Belnik, Y.; Yeshurun, M.; Raanani, P.; Shpilberg, O. 5-azacitidine prolongs overall survival in patients with myelodysplastic syndrome—A systematic review and meta-analysis. Haematologica 2010, 95, 303–310.
  18. Budziszewska, B.K.; Pluta, A.; Sulek, K.; Wierzbowska, A.; Robak, T.; Giebel, S.; Holowiecka-Goral, A.; Sawicki, W.; Ejduk, A.; Patkowska, E.; et al. Treatment of elderly patients with acute myeloid leukemia adjusted for performance status and presence of comorbidities: A Polish Adult Leukemia Group study. Leuk. Lymphoma 2015, 56, 2331–2338.
  19. Ferrara, F.; Barosi, G.; Venditti, A.; Angelucci, E.; Gobbi, M.; Pane, F.; Tosi, P.; Zinzani, P.; Tura, S. Consensus-based definition of unfitness to intensive and non-intensive chemotherapy in acute myeloid leukemia: A project of SIE, SIES and GITMO group on a new tool for therapy decision making. Leukemia 2013, 27, 997–999.
  20. Palmieri, R.; Othus, M.; Halpern, A.B.; Percival, M.M.; Godwin, C.D.; Becker, P.S.; Walter, R.B. Accuracy of SIE/SIES/GITMO Consensus Criteria for Unfitness to Predict Early Mortality After Intensive Chemotherapy in Adults With AML or Other High-Grade Myeloid Neoplasm. J. Clin. Oncol. 2020, 38, 4163–4174.
  21. Dombret, H.; Seymour, J.F.; Butrym, A.; Wierzbowska, A.; Selleslag, D.; Jang, J.H.; Kumar, R.; Cavenagh, J.; Schuh, A.C.; Candoni, A.; et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood 2015, 126, 291–299.
  22. Kantarjian, H.M.; Thomas, X.G.; Dmoszynska, A.; Wierzbowska, A.; Mazur, G.; Mayer, J.; Gau, J.P.; Chou, W.C.; Buckstein, R.; Cermak, J.; et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J. Clin. Oncol. 2012, 30, 2670–2677.
  23. Maurillo, L.; Venditti, A.; Spagnoli, A.; Gaidano, G.; Ferrero, D.; Oliva, E.; Lunghi, M.; D’Arco, A.M.; Levis, A.; Pastore, D.; et al. Azacitidine for the treatment of patients with acute myeloid leukemia: Report of 82 patients enrolled in an Italian Compassionate Program. Cancer 2012, 118, 1014–1022.
  24. Welch, J.S.; Petti, A.A.; Miller, C.A.; Fronick, C.C.; O’Laughlin, M.; Fulton, R.S.; Wilson, R.K.; Baty, J.D.; Duncavage, E.J.; Tandon, B.; et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. N. Engl. J. Med. 2016, 375, 2023–2036.
  25. Bhatnagar, B.; Duong, V.H.; Gourdin, T.S.; Tidwell, M.L.; Chen, C.; Ning, Y.; Emadi, A.; Sausville, E.A.; Baer, M.R. Ten-day decitabine as initial therapy for newly diagnosed patients with acute myeloid leukemia unfit for intensive chemotherapy. Leuk. Lymphoma 2014, 55, 1533–1537.
  26. Ritchie, E.K.; Feldman, E.J.; Christos, P.J.; Rohan, S.D.; Lagassa, C.B.; Ippoliti, C.; Scandura, J.M.; Carlson, K.; Roboz, G.J. Decitabine in patients with newly diagnosed and relapsed acute myeloid leukemia. Leuk. Lymphoma 2013, 54, 2003–2007.
  27. Blum, W.; Garzon, R.; Klisovic, R.B.; Schwind, S.; Walker, A.; Geyer, S.; Liu, S.; Havelange, V.; Becker, H.; Schaaf, L.; 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. USA 2010, 107, 7473–7478.
  28. Zeidan, A.M.; Fenaux, P.; Gobbi, M.; Mayer, J.; Roboz, G.J.; Krauter, J.; Robak, T.; Kantarjian, H.M.; Novák, J.; Jedrzejczak, W.W.; et al. Prospective comparison of outcomes with azacitidine and decitabine in patients with AML ineligible for intensive chemotherapy. Blood 2022, 140, 285–289.
  29. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629.
  30. Zeidan, A.M.; Sekeres, M.A.; Garcia-Manero, G.; Steensma, D.P.; Zell, K.; Barnard, J.; Ali, N.A.; Zimmerman, C.; Roboz, G.; DeZern, A.; et al. Comparison of risk stratification tools in predicting outcomes of patients with higher-risk myelodysplastic syndromes treated with azanucleosides. Leukemia 2016, 30, 649–657.
  31. Zeidan, A.M.; Wang, R.; Wang, X.; Shallis, R.M.; Podoltsev, N.A.; Bewersdorf, J.P.; Huntington, S.F.; Neparidze, N.; Giri, S.; Gore, S.D.; et al. Clinical outcomes of older patients with AML receiving hypomethylating agents: A large population-based study in the United States. Blood Adv. 2020, 4, 2192–2201.
  32. Récher, C.; Röllig, C.; Bérard, E.; Bertoli, S.; Dumas, P.Y.; Tavitian, S.; Kramer, M.; Serve, H.; Bornhäuser, M.; Platzbecker, U.; et al. Long-term survival after intensive chemotherapy or hypomethylating agents in AML patients aged 70 years and older: A large patient data set study from European registries. Leukemia 2022, 36, 913–922.
  33. Zeidan, A.M.; Davidoff, A.J.; Long, J.B.; Hu, X.; Wang, R.; Ma, X.; Gross, C.P.; Abel, G.A.; Huntington, S.F.; Podoltsev, N.A.; et al. Comparative clinical effectiveness of azacitidine versus decitabine in older patients with myelodysplastic syndromes. Br. J. Haematol. 2016, 175, 829–840.
  34. Zeidan, A.M.; Stahl, M.; Hu, X.; Wang, R.; Huntington, S.F.; Podoltsev, N.A.; Gore, S.D.; Ma, X.; Davidoff, A.J. Long-term survival of older patients with MDS treated with HMA therapy without subsequent stem cell transplantation. Blood 2018, 131, 818–821.
  35. Jabbour, E.J.; Garcia-Manero, G.; Strati, P.; Mishra, A.; Al Ali, N.H.; Padron, E.; Lancet, J.; Kadia, T.; Daver, N.; O’Brien, S.; 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, 876–882.
  36. Prébet, T.; Gore, S.D.; Esterni, B.; Gardin, C.; Itzykson, R.; Thepot, S.; Dreyfus, F.; Rauzy, O.B.; Recher, C.; Adès, L.; et al. Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure. J. Clin. Oncol. 2011, 29, 3322–3327.
  37. Nanah, R.; McCullough, K.; Hogan, W.; Begna, K.; Patnaik, M.; Elliott, M.; Litzow, M.; Al-Kali, A. Outcome of elderly patients after failure to hypomethylating agents given as frontline therapy for acute myeloid leukemia: Single institution experience. Am. J. Hematol. 2017, 92, 866–871.
  38. Ilyas, R.; Johnson, I.M.; McCullough, K.; Al-Kali, A.; Alkhateeb, H.B.; Begna, K.; Mangaonkar, A.A.; Litzow, M.R.; Hogan, W.J.; Shah, M.V.; et al. Outcome of Patients with Acute Myeloid Leukemia Following Failure of Front-Line Venetoclax Plus Hypomethylating Agent Therapy. Blood 2022, 140, 1286–1287.
  39. Silverman, L.R.; McKenzie, D.R.; Peterson, B.L.; Holland, J.F.; Backstrom, J.T.; Beach, C.L.; Larson, R.A. Further analysis of trials with azacitidine in patients with myelodysplastic syndrome: Studies 8421, 8921, and 9221 by the Cancer and Leukemia Group B. J. Clin. Oncol. 2006, 24, 3895–3903.
  40. Garcia, J.S.; Jain, N.; Godley, L.A. An update on the safety and efficacy of decitabine in the treatment of myelodysplastic syndromes. OncoTargets Ther. 2010, 3, 1–13.
  41. Šimoničová, K.; Janotka, Ľ.; Kavcová, H.; Sulová, Z.; Breier, A.; Messingerova, L. Different mechanisms of drug resistance to hypomethylating agents in the treatment of myelodysplastic syndromes and acute myeloid leukemia. Drug Resist. Updates 2022, 61, 100805.
  42. Qin, T.; Jelinek, J.; Si, J.; Shu, J.; Issa, J.P. Mechanisms of resistance to 5-aza-2’-deoxycytidine in human cancer cell lines. Blood 2009, 113, 659–667.
  43. Gruber, E.; Franich, R.L.; Shortt, J.; Johnstone, R.W.; Kats, L.M. Distinct and overlapping mechanisms of resistance to azacytidine and guadecitabine in acute myeloid leukemia. Leukemia 2020, 34, 3388–3392.
  44. Valencia, A.; Masala, E.; Rossi, A.; Martino, A.; Sanna, A.; Buchi, F.; Canzian, F.; Cilloni, D.; Gaidano, V.; Voso, M.T.; et al. Expression of nucleoside-metabolizing enzymes in myelodysplastic syndromes and modulation of response to azacitidine. Leukemia 2014, 28, 621–628.
  45. Wu, P.; Geng, S.; Weng, J.; Deng, C.; Lu, Z.; Luo, C.; Du, X. The hENT1 and DCK genes underlie the decitabine response in patients with myelodysplastic syndrome. Leuk. Res. 2015, 39, 216–220.
  46. Griffiths, E.A.; Choy, G.; Redkar, S.; Taverna, P.; Azab, M.; Karpf, A.R. SGI-110: DNA Methyltransferase Inhibitor Oncolytic. Drugs Future 2013, 38, 535–543.
  47. Mahfouz, R.Z.; Jankowska, A.; Ebrahem, Q.; Gu, X.; Visconte, V.; Tabarroki, A.; Terse, P.; Covey, J.; Chan, K.; Ling, Y.; et al. Increased CDA expression/activity in males contributes to decreased cytidine analog half-life and likely contributes to worse outcomes with 5-azacytidine or decitabine therapy. Clin. Cancer Res. 2013, 19, 938–948.
  48. Wu, L.; Shi, W.; Li, X.; Chang, C.; Xu, F.; He, Q.; Wu, D.; Su, J.; Zhou, L.; Song, L.; et al. High expression of the human equilibrative nucleoside transporter 1 gene predicts a good response to decitabine in patients with myelodysplastic syndrome. J. Transl. Med. 2016, 14, 66.
  49. Qin, T.; Castoro, R.; El Ahdab, S.; Jelinek, J.; Wang, X.; Si, J.; Shu, J.; He, R.; Zhang, N.; Chung, W.; et al. Mechanisms of resistance to decitabine in the myelodysplastic syndrome. PLoS ONE 2011, 6, e23372.
  50. Hopper-Borge, E.; Xu, X.; Shen, T.; Shi, Z.; Chen, Z.S.; Kruh, G.D. Human multidrug resistance protein 7 (ABCC10) is a resistance factor for nucleoside analogues and epothilone B. Cancer Res. 2009, 69, 178–184.
More
Information
Subjects: Hematology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 280
Revisions: 2 times (View History)
Update Date: 05 May 2023
1000/1000
Video Production Service