Different mechanisms of drug resistance to hypomethylating agents in the treatment of myelodysplastic syndromes and acute myeloid leukemia

https://doi.org/10.1016/j.drup.2022.100805Get rights and content

Abstract

Resistance to the hypomethylating agents (HMAs) 5-azacytidine (AZA) and 5-aza-2′-deoxycytidine (DAC) represents a major obstacle in the treatment of elderly patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) which are not suitable for hematopoietic stem cells transplantation. Approximately 50 % of patients do not respond to HMA treatment because of intrinsic (primary) resistance, while others could acquire drug resistance during the repeated cycles of the treatment. To prevent, delay or surmount resistance development, the molecular mechanisms underlying drug resistance must be first identified. This is crucial as no further standard therapeutic opportunities are available for these patients who failed hypomethylating agents-based treatment. The current review provides an updated information about the different mechanisms that may contribute to the development of resistance to HMAs. Despite the similar structure and mechanism of action of HMA, several studies did not report the expected development of cross-resistance. It is clear that in addition to the common modalities of chemoresistance, there must be some specific mechanisms of drug resistance. Changes in transport and metabolism of HMAs are among the most studied mechanisms of resistance. Drug uptake provided by two solute carrier (SLC) families: SLC28 and SLC29 (also known as the concentrative and equilibrative nucleoside transporter families, respectively), could represent one of the mechanisms of cross-resistance. Changes in the metabolism of these drugs are the most likely mechanism responsible for the unique mode of resistance to AZA and DAC. Deoxycytidine kinase and uridine-cytidine kinase due to their necessity for drug activation, each could represent one of the response markers to treatment with DAC and AZA, respectively. Other mechanisms involved in the development of resistance common for both drugs involved: i. increased DNA repair (caused for example by constitutive activation of the ATM/BRCA1 pathway and inhibition of p53-dependent apoptosis); ii. changes in the regulation of apoptosis/disrupted apoptotic pathways (specifically increased levels of the anti-apoptotic protein BCL2) and iii. increased resilience of leukemic stem cells to multiple drugs including HMAs. Despite intense research on the resistance of MDS and AML patients to HMAs, the mechanisms that may reduce the response of these cells to HMAs are not known in detail. We herein highlight the most important directions that future research should take.

Introduction

Cancer is one of the main causes of morbidity and mortality in all regions of the world, and its incidence is constantly increasing (Bray et al., 2018). Hematologic malignancies represent a group of diseases derived from bone marrow cells and the lymphatic system with diverse etiologies, incidences, prognoses and survival (Loda et al., 2017). The myelodysplastic syndromes (MDS) comprise a heterogenous group of clonal hematopoietic malignancies characterized by ineffective hematopoiesis, progressive bone marrow failure, cytogenetic and molecular abnormalities and different risks of progression to acute myeloid leukemia (AML) (Cogle, 2015; Messingerova et al., 2016). AML is a heterogeneous neoplastic blood disease with characteristic clonal expansion and accumulation of abnormally differentiated leukemic myeloid blasts in the bone marrow, peripheral blood, and other tissues (Döhner et al., 2015; O’Donnell et al., 2017; Short et al., 2018). Therapy for patients with MDS is focused on reducing symptoms associated with disease and risk of disease progression to AML and improving quality of life. Because of the heterogeneity of MDS, treatment options include different approaches from supportive care to treatment with hypomethylating agents (HMAs) (Steensma, 2018). However, treatment efficacy is frequently limited by the chemoresistance phenotype of cancer cells (Assaraf et al., 2019; Gonen and Assaraf, 2012; Levin et al., 2019, 2021; Li et al., 2016). Chemoresistance is classified into two distinct categories: intrinsic or primary resistance, which is present in tumor cells from the beginning (based on the phenotype of the cells from which the tumor developed and the course of tumor transformation) before the chemotherapeutic agent is administered, and acquired or secondary resistance that develops in initially sensitive cells in response to the toxic stress of chemotherapeutics (Longley and Johnston, 2005). During the development of secondary resistance of tumor cells by repeated exposures to individual chemotherapeutic agents, cross-resistance to pharmacologically related substances and compounds with similar structures and biochemical properties (such as nucleoside analogs) may emerge, as these compounds are often dependent on the same transporters and specific metabolizing pathways or are aimed at the same intracellular targets (Moscow et al., 2010). Resistance to multiple drugs (multidrug resistance, MDR) can also develop, where cells are resistant to a wide range of substances, with no apparent relationship in chemical structure and a fundamentally different mechanism of action (Breier et al., 2013; Li et al., 2016;Wang et al., 2021). Primary and secondary resistance is also a significant problem in the treatment of hematological malignancies, including the treatment of patients with MDS and AML.

For AML and high-risk MDS patients who are not suitable for hematopoietic stem cell transplantation, a suitable treatment protocol is the application of HMAs such as 5-azacytidine (AZA) and 5-aza-2′-deoxycytidine (DAC), which have been found to be beneficial in the treatment of high-risk MDS (Steensma, 2018) and AML (Kubasch and Platzbecker, 2018). However, only 30–60 % of patients with MDS and 18–47 % of patients with AML respond to hypomethylation therapy. Another group of patients responds in the initial cycles of HMA therapy, but the response decreases or disappears during subsequent cycles of treatment. The occurrence of a reduced response to HMAs then significantly worsens the patient's further prognosis and survival time, as no follow-up therapy is approved for these patients (Duong et al., 2014). Resistance to treatment with HMAs is a complex phenomenon in which multiple molecular causes may be involved, working together to eliminate the cytotoxic effect of HMAs on myeloid blasts.

Section snippets

Resistance to chemotherapeutic treatment

As already mentioned, there is a group of patients with high-risk MDS and AML which are not suitable for induction therapy or hematopoietic stem cell transplantation who do not respond to HMAs. The causes of resistance may be specific for AZA and DAC or may be more general by activating drug detoxification, drug efflux, and accelerating cell renewal mechanisms, which may involve decreased sensitivity of myeloid blasts to other drugs with fundamentally different structures and mechanisms of

Different mechanisms of resistance to AZA and DAC

AZA and DAC are cytosine nucleoside analogs linked to either ribose or 2-deoxyribose, first synthesized in Czechoslovakia in 1964 (Pískala and Šorm, 1964; Pliml and Šorm, 1964). Both drugs have the carbon atom at position 5 of the pyrimidine ring replaced by a nitrogen atom. It is the carbon at this position that is usually methylated by DNA methyltransferases (DNMTs). The mechanism of action of AZA and DAC is not fully understood but could be the result of a combination of hypomethylating

Cross-resistance to hypomethylating agents

Despite the related structure, a different mechanism of primary resistance can be expected for AZA and DAC, as no correlation was observed between the IC50 values ​​for these drugs in human leukemia cell lines THP-1, OCI-AML2, OCI-AML3, MOLM-13, PL-21, HL-60, MV4−11, SIG-M5, ML2, NB4, KG1, MonoMac6, and HEL (Oellerich et al., 2019); or human leukemia and lymphoma cell lines HL60, ML-1, HEL, Raji, Jurkat, TF-1, U937, K562, and MOLT4; prostate cancer cell lines PC3 and DU145; colon cancer cell

Drug uptake

Nucleoside analogs used in cancer and antiviral therapy are transported into cells by two solute carrier (SLC) families: SLC28, also known as the concentrative nucleoside transporter (CNT) family, and SLC29, or the equilibrative nucleoside transporter (ENT) family. The SLC28 family comprises three Na+-dependent cotransporters with different substrate specificities. While CNT1 (encoded by the SLC28A1 gene) prefers pyrimidine as a transport substrate, CNT2 (encoded by the SLC28A2 gene) prefers

Drug efflux

Chemoresistant tumor cells can be not only cross-resistant to structurally and biochemically similar compounds but can also exhibit reduced sensitivity simultaneously to several structurally and functionally different substances. This phenomenon is called multidrug resistance (MDR). The first mechanism of MDR discovered was an increase in the expression of the ABCB1 (MDR1) gene encoding P-glycoprotein (Assaraf et al., 2019; Bellamy et al., 1990; Lepeltier et al., 2020; Li et al., 2016; Su et

Drug metabolism

Inside the cells, activation of the prodrugs AZA and DAC is necessary. The first rate-limiting step is their phosphorylation to the corresponding monophosphate, which is mediated by two different enzymes. While AZA is phosphorylated by uridine-cytidine kinase (UCK), DAC is phosphorylated by deoxycytidine kinase (DCK) (Qin et al., 2009). Changes in the expression of these enzymes could affect the sensitivity of cells to drugs.

In the NCI-60 cancer cell line panel, there was a correlation between

Alterations in DNA damage response

The DNA damage response (DDR) has a crucial role in the normal physiology of the cell, as the maintenance of genomic stability is required for its proper function. As a reaction to the occurrence of DNA damage, DDR will initiate DNA damage repair, but if the damage is too severe and cannot be repaired, apoptosis will be initiated (J. Y. J. Wang, 2019). Alterations in DDR represent a double-edged sword. Disruption of DNA repair can promote mutation propagation and lead to the development of

Alterations in regulation of apoptosis

One of the goals of chemotherapy treatment is the selective induction of apoptosis in neoplastic cells. Apoptosis is the most common mechanism of cell death induced by chemotherapeutics (Ouyang et al., 2012; Shahar and Larisch, 2020). Therefore, defects in apoptotic regulatory pathways may lead to the development of drug resistance (Carneiro and El-Deiry, 2020). For example, deregulation of proapoptotic APAF1 leads to chemoresistance and poor patient outcomes in melanomas (Fujimoto et al., 2004

Possible role of leukemic stem cells in chemoresistance

The presence of chemoresistant leukemic stem cells (LSCs) in niches in the bone marrow (BM) microenvironment may be another mechanism of hematologic malignancy relapse. Niches represent a BM area where hematopoietic stem cells (HSCs) reside under physiological conditions and are protected from environmental stress. The major mediator of HSC migration and homing in the BM is the CXCR4 receptor expressed on the surface of HSCs, which binds to its ligand CXCL12 (produced by BM stromal cells).

Conclusions

Despite the intense interest in the resistance of MDS and AML cells to HMAs, the mechanisms that may reduce the response of these cells to HMAs are not known in detail; therefore, this issue is not fully understood. However, given the many different results of individual studies, we must conclude that there are multiple cellular mechanisms of protection against HMAs. The possibility that only one mechanism of resistance to DAC and AZA alone or both together applies must be ruled out.

The most

Data availability

Data will be made available on request.

No data was used for the research described in the article.

Acknowledgment

This paper was supported by COST actionCA17104-STRATAGEM, and also by MVTS COSTCA17104. Our laboratory is supported by Slovak Research and Development Agency, grant No.: APVV-19-0093 and APVV-19-0094.

References (179)

  • J. Doskočil et al.

    The Effects of 5-Azacytidine and 5-Azauridine on Protein Synthesis in Escherichia coli

    Biochem. Biophys. Res. Commun.

    (1970)
  • Y. Dragan et al.

    Selective loss of nucleoside carrier expression in rat hepatocarcinomas

    Hepatology

    (2000)
  • Q. Ebrahem et al.

    Mechanisms of resistance to 5-Azacytidine/Decitabine in MDS-AML and pre-clinical in vivo proof of principle of rational solutions to extend response

    Blood

    (2015)
  • N. Gonen et al.

    Antifolates in cancer therapy: structure, activity and mechanisms of drug resistance

    Drug Resist. Updates

    (2012)
  • S. Harel et al.

    Outcome of patients with high risk Myelodysplastic Syndrome (MDS) and advanced Chronic Myelomonocytic Leukemia (CMML) treated with decitabine after azacitidine failure

    Leuk. Res.

    (2015)
  • N. Herold et al.

    With me or against me : tumor suppressor and drug resistance activities of SAMHD1

    Exp. Hematol.

    (2017)
  • G. Huls et al.

    Ibrutinib added to 10-day decitabine for older patients with AML and higher risk MDS

    Blood Adv.

    (2020)
  • S. Imanishi et al.

    Constitutive activation of the ATM/BRCA1 pathway prevents DNA damage-induced apoptosis in 5-azacytidine-resistant cell lines

    Biochem. Pharmacol.

    (2014)
  • E.D. Lagadinou et al.

    BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells

    Cell Stem Cell

    (2013)
  • W. Li et al.

    Overcoming ABC transporter-mediated multidrug resistance: molecular mechanisms and novel therapeutic drug strategies

    Drug Resist. Updates

    (2016)
  • Y. Adar et al.

    Imidazoacridinone-dependent lysosomal photodestruction: a pharmacological Trojan horse approach to eradicate multidrug-resistant cancers

    Cell Death Dis.

    (2012)
  • A.D. Adema et al.

    Overexpression of MRP4 (ABCC4) and MRP5 (ABCC5) confer resistance to the nucleoside analogs cytarabine and troxacitabine, but not gemcitabine

    Springer Plus

    (2014)
  • I. Aldoss et al.

    Efficacy of the combination of venetoclax and hypomethylating agents in relapsed/refractory acute myeloid leukemia

    Haematologica

    (2018)
  • R.C. Bargou et al.

    Constitutive nuclear factor-κb-RelA activation is required for proliferation and survival of Hodgkin’s disease tumor cells

    J. Clin. Invest.

    (1997)
  • M. Barkett et al.

    Control of apoptosis by Rel/NF-κB transcription factors

    Oncogene

    (1999)
  • W.T. Bellamy et al.

    The clinical relevance of multidrug resistance

    Cancer Invest.

    (1990)
  • M.P. Blaustein et al.

    Sodium/Calcium exchange : its physiological implications

    Physiol. Rev.

    (1999)
  • S. Boehrer et al.

    Suppression of the DNA damage response in acute myeloid leukemia versus myelodysplastic syndrome

    Oncogene

    (2009)
  • J.M. Bogenberger et al.

    BCL-2 family proteins as 5-Azacytidine-sensitizing targets and determinants of response in myeloid malignancies

    Leukemia

    (2014)
  • J.M. Bogenberger et al.

    Ex vivo activity of BCL-2 family inhibitors ABT-199 and ABT-737 combined with 5-azacytidine in myeloid malignancies

    Leuk. Lymphoma

    (2015)
  • P. Borst et al.

    Multidrug resistance-associated proteins 3, 4, and 5

    Pflugers Archiv Eur. J. Physiol.

    (2007)
  • G. Borthakur et al.

    Activity of decitabine in patients with myelodysplastic syndrome previously treated with azacitidine

    Leuk. Lymphoma

    (2008)
  • F. Bray et al.

    Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

    CA Cancer J. Clin.

    (2018)
  • A. Breier et al.

    New insight into P-Glycoprotein as a drug target

    Anticancer Agents Med. Chem.

    (2013)
  • C. Bull et al.

    Exposure to hypomethylating 5-aza-2′-deoxycytidine (decitabine) causes rapid, severe DNA damage, telomere elongation and mitotic dysfunction in human WIL2-NS cells

    Mutation Res. Genet. Toxicol. Environ. Mutagenesis

    (2021)
  • B.A. Carneiro et al.

    Targeting apoptosis in cancer therapy

    Nat. Rev. Clin. Oncol.

    (2020)
  • J.X. Cheng et al.

    RNA cytosine methylation and methyltransferases mediate chromatin organization and 5-azacytidine response and resistance in leukaemia

    Nat. Commun.

    (2018)
  • J. Ciccolini et al.

    Gender, cytidine deaminase, and 5-aza/decitabine - Letter

    Clin. Cancer Res.

    (2013)
  • T. Cluzeau et al.

    BCL2L10 is a predictive factor for resistance to Azacitidine in MDS and AML patients

    Oncotarget

    (2012)
  • T. Cluzeau et al.

    Phenotypic and genotypic characterization of azacitidine-sensitive and resistant SKM1 myeloid cell lines

    Oncotarget

    (2014)
  • C.R. Cogle

    Incidence and burden of the myelodysplastic syndromes

    Curr. Hematol. Malig. Rep.

    (2015)
  • C. Colomer et al.

    IKKα kinase regulates the DNA damage response and drives chemo-resistance in Cancer

    Mol. Cell

    (2019)
  • M.R. Corces-Zimmerman et al.

    Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission

    Proc. Natl. Acad. Sci.

    (2014)
  • C. Craddock et al.

    Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia

    Leukemia

    (2013)
  • V.L. Damaraju et al.

    Role of human nucleoside transporters in the uptake and cytotoxicity of azacitidine and decitabine

    Nucleosides Nucleotides Nucleic Acids

    (2012)
  • G. Damia et al.

    Platinum resistance in ovarian cancer: role of DNA repair

    Cancers

    (2019)
  • E.J.B. Derissen et al.

    Intracellular Pharmacokinetics of Pyrimidine Analogues used in Oncology and the Correlation with Drug Action

    Clin. Pharmacokinet.

    (2020)
  • J. Diesch et al.

    A clinical-molecular update on azanucleoside-based therapy for the treatment of hematologic cancers

    Clin. Epigenetics

    (2016)
  • C.D. DiNardo et al.

    Clinical experience with the BCL2-inhibitor venetoclax in combination therapy for relapsed and refractory acute myeloid leukemia and related myeloid malignancies

    Am. J. Hematol.

    (2018)
  • C.D. DiNardo et al.

    Azacitidine and venetoclax in previously untreated acute myeloid leukemia

    N. Engl. J. Med.

    (2020)
  • Cited by (16)

    • Targeting SAMHD1: To overcome multiple anti-cancer drugs resistance in hematological malignancies

      2023, Genes and Diseases
      Citation Excerpt :

      Decitabine and azacytidine are phosphorylated to the active form of decitabine-TP and azacytidine-TP by the action of cellular enzymes. Decitabine-TP and azacytidine-TP can incorporate into nucleic acid, while decitabine-TP mainly incorporates into DNA and azacytidine-TP incorporates into both DNA and RNA.72 Decitabine and azacytidine are mainly applied in AML and MDS.73–76

    • Understanding ER homeostasis and the UPR to enhance treatment efficacy of acute myeloid leukemia

      2022, Drug Resistance Updates
      Citation Excerpt :

      Dinaciclib (SCH727965) is an inhibitor of cyclin-dependent kinases (CDKs) 1,2,5 and 9. Deregulation of cell cycle control caused by abnormal CDKs activity is observed in most cancers, including AML (Cucchi et al., 2020; Šimoničová et al., 2022; Grant and Roberts, 2003). Exposure of human leukemic and myeloma cells to nanomolar concentrations of dinaciclib inhibited sXBP-1 nuclear localization and expression of BiP, which are prompted in response to treatment with the ER stress inducers tunicamycin and thapsigargin, increasing their cytotoxic effects (Nguyen and Grant, 2014).

    • Long non-coding RNA mediated drug resistance in breast cancer

      2022, Drug Resistance Updates
      Citation Excerpt :

      Various chemotherapeutic drugs used in the treatment of cancer act by inducing cell death by activating apoptosis pathways especially by caspases-mediated extrinsic or intrinsic mechanisms. In the pathophysiology of cancer progression, the apoptosis pathways are typically regulated through a wide variety of mechanisms which mostly include upregulation of antiapoptotic proteins and downregulation of proapoptotic proteins (Pfeffer and Singh, 2018; Shahar and Larisch, 2020; Cui et al., 2018; Kopecka et al., 2020; Šimoničová et al., 2022; Levin et al., 2021). In multiple investigations, lncRNAs associated to apoptotic pathways have been linked to MDR in BC.

    • Selenium and tellurium in the development of novel small molecules and nanoparticles as cancer multidrug resistance reversal agents

      2022, Drug Resistance Updates
      Citation Excerpt :

      Oncogenes are genes encoding proteins that act as promoters of cell cycle progression, for example growth factors and their receptors (e.g., HER2), signal transduction proteins (e.g., RAS), transcription factors (e.g., MYC) or proteins that inhibit apoptosis (e.g., BCL-2) (Gacche and Assaraf, 2018; Sciarrillo et al., 2020; Shahar and Larisch, 2020; Nussinov et al., 2021; Pecoraro et al., 2021; Chen et al., 2022). By inactivating tumor suppressor genes, the cells may lose their ability to regulate the cell cycle (e.g. RB, PTEN), induce apoptosis or maintain their genetic stability (e.g. BRCA1/BRCA2) (Šimoničová et al., 2022). One of the most commonly mutated tumor suppressor gene in cancers is p53 (Stiewe and Haran, 2018; Cao et al., 2020), which maintains the genetic stability by activating DNA repair proteins, arresting the cell cycle, and initiating apoptosis, if the DNA damage is unrepairable (Croce, 2008).

    View all citing articles on Scopus
    1

    Kristína Šimoničová and Ľuboš Janotka contributed equally to this work.

    View full text