Open access peer-reviewed chapter

Roads of Drug Resistance in Acute Myeloid Leukemia – Is It a Dead End?

Written By

Yanitsa Davidkova, Milan Jagurinoski, Gueorgui Balatzenko and Margarita Guenova

Submitted: 27 July 2022 Reviewed: 02 December 2022 Published: 22 December 2022

DOI: 10.5772/intechopen.109304

From the Edited Volume

Leukemia - From Biology to Clinic

Edited by Margarita Guenova and Gueorgui Balatzenko

Chapter metrics overview

86 Chapter Downloads

View Full Metrics

Abstract

Acute myeloid leukemia (AML) is a biologically and clinically heterogeneous neoplasm, which is characterized by abnormal proliferation, impaired apoptosis, and differentiation of leukemic immature cells. Nowadays, the first line treatment of AML is the chemotherapy regimen, which combines both cytosine arabinoside and anthracycline. Despite that complete remission (CR) can be achieved in 40–80% of patients depending on age, a considerable number will eventually relapse (acquired resistance) or have refractory disease (primary resistance). Finally, the estimated 5-year overall survival (OS) is less than 30%. Recent investigations reveal various mechanisms, responsible for drug resistance leading to AML persistence and recurrence. In order to improve clinical outcomes and develop successful therapeutic strategies, it is necessary to better explore the major adverse factors for escape from treatment, as well as to explore ways to predict and prevent or target drug resistance.

Keywords

  • acute myeloid leukemia
  • chemotherapy
  • drug resistance

1. Introduction

Acute myeloid leukemia (AML) comprises approximately 80% of all acute leukemias in adults [1]. The median age of the disease is about 69 years with a progressing incidence with advanced age [2]. AML represents a clinically and genetically heterogeneous disease, which is characterized by uncontrolled clonal proliferation, impaired apoptosis, and differentiation of leukemic immature cells. It has poor survival and fatal outcome in those who are untreated. Despite the progress in understanding the pathophysiology of AML and the discovery of novel therapeutic agents, the treatment approach has not changed essentially since the 1970s. According to the European LeukemiaNet (ELN) recommendations for the diagnosis and management of AML in adults (2017), the first-line therapy for the patients, eligible for intensive chemotherapy remains the conventional “3 + 7 regimen” (3 days of anthracycline + 7 days of cytarabine) [3]. Unfortunately, recent studies report that the disease is curable in only 5–15% of patients above 60 years and 35–40% in younger adults with intensive chemotherapy, which is unsatisfactory [4, 5]. Additionally, research data reveals that 10–40% of newly diagnosed AMLs fail to achieve CR after frontline therapy and are classified as primary refractory AML [6, 7, 8]. Lately, ELN determines primary refractory AML as a lack of obtaining CR or complete remission with incomplete hematologic recovery (CRi) after at least 2 courses of intensive induction chemotherapy [3]. One of the major reasons for treatment failure is considered to be drug resistance. Besides, chemoresistance is basically divided into two groups intrinsic (primary) and acquired (secondary). The primary drug-resistant leukemic cells are present already at diagnosis, while the secondary resistance emerges during or after therapy, probably as a result of additionally occurring genetic disorders. In 30–40% of relapsed AML patients, newly developed gene mutations occur, yet in about 25% no molecular alterations emerge [9]. So, these data raise the question that gene alterations are drivers of relapse or refractoriness of AML and whether mutational status is the only cause of disease progression. This review will discuss the potential molecular pathways underlying drug resistance in blast cells and the interactions with the leukemic microenvironment.

Advertisement

2. Leukemogenesis in AML

Two decades ago, Gilliland and Griffin introduced the “two-hit model”. In this model, the collaboration of two lesions of two different classes of mutations contributes synergistically to inflict AML [10]. Class I mutations (FLT3, c-KIT, NRAS) comprise mutations that activate signal-transduction pathways and thereby give a proliferation advantage to the hematopoietic progenitor cells. Class II mutations (recurring chromosomal aberrations, which produce fusion transcripts) affect transcription factors and cause impaired differentiation and following apoptosis. However, recent studies report that it is difficult to divide functions between the two classes of mutations [11]. Further, in the last years, genomic sequencing research has encountered new epigenetic genes associated with AML (chromatin-modifying genes: MLL fusions, ASXL1, and EZH2 mutations; methylation-related genes: DNMT3A, TET2, IDH1/2 mutations), which expands the complexity and heterogeneity of AML [12, 13]. Thus, the “two-hit model” of Gilliland and Griffin turns out to be insufficient to explain AML leukemogenesis.

A novel hypothesis for AML development has been formulated recently, based on three types of AML-associated mutations, investigated in mouse models [14]. According to this functional classification the first “type A mutations” (fusion genes) are necessary to initiate or maintain the leukemic phenotype, “type B mutations” (ABL, PDGFR, KIT, FLT3, etc.) support the proliferation and survival of leukemic cells and “type C mutations” (epigenetic modifiers), also called “seed mutations”, provide a growth advantage, but still not sufficient to induce leukemia. The study research demonstrates that the combination of mutations of any two types, that is, A + B or A + C or B + C, may result in AML [14]. The model of clonal evolution of AML is considered to be a process of losing specific mutations or gaining a feature, which leads toward resistance. Emerging evidence has revealed that a relapse may present with re-occurring of the initial leukemic clone, assuming that the chemotherapy was unsuccessful or due to further clonal evolution following AML treatment [15, 16].

Advertisement

3. Mechanisms underlying drug resistance

Relapsed and refractory AML (R/R AML) is associated with unfavorable prognosis, due to poor response to conventional antileukemic therapy [17, 18]. Thus, a better understanding of the mechanisms, underlying drug resistance, would improve the therapeutic approach using novel strategies. Data are available showing that chemoresistance in AML may be due to numerous factors, which include proteins and enzymes with altered function, dysregulation of signaling pathways, mutations in cell cycle control genes, epigenetic modifiers, microRNA as well as impaired interactions with the bone marrow environment, and changes in the immune tolerance (Table 1).

FactorsMoleculesFunctionReference
Proteins and enzymesP-gp, GST, MCL-1, MRP1/LRP, Topo II, PKCAffect the drug transport; altered enzyme function[19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43]
Signal pathwaysNF/kB, PI3K/Akt/mTORCoordinate complex cellular changes[44, 45]
Genes and epigeneticsASLX1, DNMT3, EZH2, FLT3, IDH1/2, TET2, WT1Cell proliferation and differentiation; regulation of DNA and histones[46, 47, 48, 49, 50, 51, 52, 53]
microRNAmiRNA-155, miRNA-125, miRNA-100, miRNA-223Control of cell division, self-renewal, DNA damage[54, 55, 56]
MicroenvironmentCD44, FGF2/FGFR1, SDF1/CXCR4, VCAM/VLA4Cell-to-cell, cell-to-matrix interactions[57, 58, 59, 60, 61, 62, 63, 64, 65, 66]

Table 1.

Different mechanisms of drug resistance.

ASXL1, additional sex combs-like 1; CXCR4, C-X-C motif chemokine receptor 4; DNMT3A, DNA methyltransferase 3A; EZH2, enhancer of zeste homolog 2; IDH1/2, isocitrate dehydrogenase 1/2; FGF2, fibroblast growth factor 2; FGFR1, fibroblast growth factor receptor 1; FLT3, FMS-like tyrosine kinase 3; GST, glutathione S-transferase; LRP, lung resistance protein; MCL-1, myeloid cell leukemia 1; microRNA, microribonucleic acid; MRP1, multidrug resistance-related protein; mTOR, mammalian target of rapamycin; NF/kB, nuclear factor kappa B; P-gp, P-glycoprotein; PI3K, phosphatidylinositol-3-kinase; PKC, protein kinase C; SDF-1, stromal cell-derived factor 1; TET2, ten eleven translocation methylcytosine dioxygenase 2; Topo II, topoisomerase II; VLA-4, very late antigen-4.

3.1 Proteins and enzymes with altered function

Overexpression of the transporter proteins P-glycoprotein (P-gp) and multidrug resistance-related protein (MRP) 1 plays an important role in cross-resistance to drugs. In 1976 Juliano and Ling firstly described the ATP-dependent membrane P-gp, which acts as an efflux pump conferring resistance [19]. P-gp is a 170-kDa protein, encoded by the MDR1 gene (multidrug resistance gene 1) and belongs to the ABC transporter family. It pumps out chemotherapy drugs, maintaining lower drug concentrations intracellularly continuously, so that drug resistance is developed [20]. P-gp is considered to be an independent adverse prognostic factor for response and survival in newly diagnosed or R/R AML [21, 22]. However, according to other research studies, no correlation was identified between the MDR parameters and overall survival of the AML patients [23] and also P-gp activity is not consistently upregulated in relapsed AML [24]. Broxterman et al. found no correlation between the expression of P-gp and the complete response rate, event-free survival, or overall survival after idarubicine-containing induction [25]. The steady-state cellular accumulation of lipophilic idarubicine may circumvent the P-gp-mediated drug resistance in AML patients. The increased expression of P-gp and MRP1 are associated with advanced age, leukocytosis, poor chromosomal abnormalities, shorter overall survival, and also are detected with higher incidence in R/R and secondary AML comparing with de novo cases [26, 27]. Interestingly, an association has been observed between the expression of P-gp and MRP1 and the flow cytometric antigens (CD34 and CD7) and the FAB (French-American-British) classification of AML morphology (M2, M5a, and M7 types) [26]. Moreover, a recommendation to observe the higher MRP1 expression by flow cytometry as an adverse prognostic marker in AML was proposed by Legrand et al. [28]. In contrast, another study revealed that AML patients relapse despite the lower expression of MRP1, suggesting the involvement of other intracellular mechanisms, possibly leading to cytarabine resistance [29].

LRP (lung resistance protein), a drug efflux transporter, is also assumed to play a role in drug resistance. However, the published data show conflicting evidence of the involvement of LRP in the process of chemoresistance. The overexpression of LRP is found to predict an inferior response in AML, but another study described that higher bone marrow expression of LRP predicts significant favorable therapeutic outcome with increased CR rate and 1-year DFS (disease-free survival) and OS [22, 30, 31].

Glutathione S-transferase (GST) is a drug-metabolizing enzyme, consisting of α, μ, θ, and π-types. It is responsible for controlling cellular oxidative balance, catalyzing the reduced glutathione which leads to diminished cytotoxic drug effects. The decreased enzymatic activity due to GST polymorphisms is associated with cancerogenesis and AML [32]. Indeed, changed detoxification contributes toward drug resistance in AML. Furthermore, patients with higher expression of GSTμ tend to have also MRP1 overexpression which results in increasing the survival of tumor cells and protects them from apoptosis [33, 34]. Recent investigation by Pei et al. demonstrated that primitive leukemic cells acquire aberrant glutathione metabolism and may be selectively eliminated by target therapy against the glutathione pathway [35].

Topoisomerase II (Topo II) is an essential ribozyme that alters the topological properties of DNA. The inhibitors of Topo II may trigger chromosomal translocations that are associated with therapy-related secondary leukemia, often bearing 11q23 translocations involving the MLL gene [36]. Decreased or increased expression or mutation in the topoisomerase II genes may lead to chemoresistance to topoisomerase II inhibitors [37, 38].

Somatic mutations in protein kinase genes play a significant role in proliferation, resistance, and apoptosis. The overexpression of PKC (protein kinase C) in AML results in a decline in CR induction and DFS by diminishing intracellular concentration of daunorubicine [39]. The connection between the activation of PKC and the upregulation of P-gp further contributes to chemoresistance in AML [40].

As a BCL-2 family protein member, MCL-1 (myeloid cell leukemia 1) prevents apoptosis. It is upregulated in several hematologic malignancies such as multiple myeloma, AML, and non-Hodgkin lymphoma and is associated with treatment resistance and inferior prognosis [41]. The increased expression or amplification of MCL-1 gene protects tumor cells from programmed cell death and decreases their sensitivity to conventional chemotherapy which appears to be a potential drug resistance mechanism [42]. Besides, the overexpression of MCL-1 correlates with resistance to venetoclax [41]. MCL-1 is described to be regulated by cyclin-dependent kinase (CDK). Recent studies reveal that the treatment with both MCL-1 inhibitors and BCL-2 inhibitors may overcome the acquired resistance to BCL-2 inhibition [43].

3.2 Signaling pathways

Knowledge of aberrantly regulated signal pathways in AML allowed the identification of novel therapy targets. The combination of conventional chemotherapy with targeted agents may potentially overcome resistance. An example is the PI3K/Akt/mTOR signal pathway which is responsible for cell metabolism, proliferation, differentiation, and survival. The upregulation of the PI3K/Akt/mTOR pathway in AML is caused by mutations in the receptor tyrosine kinases. The FLT3 mutation leads to deregulation of PI3K/Akt/mTOR signaling which results in cytokine-independent survival and proliferation of hematopoietic cells and myeloproliferative neoplasms [44]. In addition, according to lately published data the PI3K/Akt/mTOR pathway plays a significant role in the regulation of therapy-resistant leukemic cells through the pro-inflammatory transcription factor NF-κB (nuclear factor-kappa-light-chain-enhancer of activated B cells). Thus, the mediated chemoresistance is caused by upregulation of anti-apoptotic genes, which leads to tumor cell growth and resistance of apoptosis [45].

3.3 Genes and epigenetic modifiers

FLT3 is normally expressed by hematopoietic stem and progenitor cells, but in AML acts as a protooncogene that stimulates cell proliferation, differentiation, and survival. In approximately 30% de novo AML patients mutations in FLT3 gene emerge. There are two types of mutations, that is, internal tandem duplication (ITD), which is present in about 25% of cases with adverse prognostic impact, and the tyrosine kinase domain (TKD) in about 5%, which prognostic value remains disputable [46]. According to the 2016 revision of the WHO classification of myeloid neoplasms and acute leukemia, the potent significance of the FLT3-ITD mutation depends on the allelic ratio and the presence of NPM1 (nucleophosmin) gene mutations [47]. Published data demonstrated that FLT3-ITD mutation can constitutively activate the receptor and force uncontrollable cell proliferaton, which turns leukemic cells resistant to conventional chemotherapeutic agents [48, 49]. In addition, authors suggested that the mechanisms of drug resistance consist of clonal evolution of resistant leukemic cells, adaptive cellular mechanisms and a protective leukemic microenvironment [50]. FLT3-inhibitors the relapse may occur due to leukemic cells harboring FLT3-TKD mutations or non-FLT3 clones, carrying epigenetic mutations such as IDH1/2, ASXL1, or TET2 [50]. By the advanced methods of whole genome or exome sequencing, several epigenetic modifiers have been determined in AML, regulating DNA methylation (DNMT3A, IDH1/2, TET2) and histone modification (EZH2 and ASXL1) [51]. Research data suggest that epigenetics-modifying gene mutations promote genetic instability and induce FLT3-ITD, leading to drug resistance and relapse [52]. However, the prognostic impact and the precise contribution of these genes to leukemogenesis have not been fully elucidated yet [53].

3.4 microRNA

MicroRNAs are small, 19–24 nucleotide-long, non-coding single-stranded RNAs which play a key role in the control of the expression of several genes involved in the differentiation of hematopoietic stem cells and the development of cancers [54, 55]. The impaired regulation of microRNAs may contribute to the chemoresistance of tumor cells by affecting cell survival and apoptosis-related signaling pathways [56]. Research efforts in the last decade have demonstrated the unquestionable role of microRNAs in reversing drug resistance. However, their implementation into clinical practice is hampered by the inability to ensure sufficient safe and specific entry into tumor cells and further studies are needed [56].

3.5 Tumor microenvironment

The bone marrow microenvironment supports normal hematopoiesis through signaling cascades and affects the evolution, progression, and chemotherapy resistance of AML [57]. The bone marrow consists of two distinct niches, that is, the osteoblastic (endosteal) and the vascular (endothelial), which act synergistically in order to regulate cell self-renewal, proliferation, and differentiation [58]. Hematopoietic stem cells are maintained by stem cell factor (SCF), CXCL12 (C-X-C Motif Chemokine Receptor 4), Notch ligands, and transforming growth factor-β [59]. Mesenchymal stromal cells secrete SCF and CXCL12, which regulate leukocyte migration [60]. The binding of CXCL12 to its receptor CXCR4 initiates the phosphorylation of CXCR4 and activates prosurvival signaling pathways such as MEK/ERK, JAK/STAT, and PI3K/AKT cascades [58]. Of note, CXCR4 signaling is associated with increased retention to the bone marrow, enhanced chemoresistance of leukemic cells, and poor prognosis in AML [61]. The interaction between the very late antigen-4 (VLA-4) and fibronectin take part in chemokine-mediated homing and mobilization [62]. The adhesion receptor VLA-4 binds to the fibronectin and vascular cell adhesion molecule-1 (VCAM-1), resulting in the retention of the leukemic cells within the bone marrow niche [63]. Wang et al. observed that a higher level of expression of VLA-4 is characterized by poorer survival [58]. The surface marker CD44 is a glycoprotein, that mediates cell adhesion, migration, and homing of leukemic cells [64, 65]. The antigen CD44 is expressed on both hematopoietic and leukemic cells, binding to E-selectin and L-selectin. The inhibition of E-selectin strengthens the influence of the chemotherapeutics daunorubicin and cytarabine [66], and lowers the leukemia burden [57]. As components of the microenvironment have been shown to contribute to drug resistance in AML, novel targeted therapies have been advanced in order to overcome it.

Advertisement

4. Conclusion

The development of drug resistance has emerged as an insurmountable challenge in the treatment of patients with R/R AML. The incompletely understood molecular mechanisms which cause therapeutic failure remain аs a major obstacle to the long-term success of leukemic therapy, inferior prognosis, and reduced survival. Further investigations are needed to delineate more precise, genomic-guided, individualized clinical approaches.

Advertisement

Acknowledgments

This work was carried out in collaboration among all authors. All of them read and approved the final manuscript.

Advertisement

Conflict of interest

The authors declare no conflict of interest. The funders had no role in the design of the study and in the writing of the manuscript.

Advertisement

Acronyms and abbreviations

AMLacute myeloid leukemia
ASXL1additional sex combs-like 1
ATPadenosine triphosphate
CDKcyclin-dependent kinase
CRcomplete remission
CRiincomplete hematologic recovery
CXCR4C-X-C motif chemokine receptor 4
DFSdisease-free survival
DNMT3ADNA methyltransferase 3A
ELNEuropean LeukemiaNet
EZH2enhancer of zeste homolog 2
FABFrench-American-British
FGF2fibroblast growth factor 2
FGFR1fibroblast growth factor receptor 1
FLT3FMS-like tyrosine kinase 3
GSTglutathione S-transferase
IDH1/2isocitrate dehydrogenase 1/2
ITDinternal tandem duplication
LRPlung resistance protein
MCL-1myeloid cell leukemia 1
MDRmultidrug resistance
MRP1multidrug resistance-related protein 1
mTORmammalian target of rapamycin
NF/kBnuclear factor-kappa-light-chain-enhancer of activated B cells
NPM1nucleophosmin
P-gpP-glycoprotein
PI3Kphosphatidylinositol-3-kinase
PKCprotein kinase C
R/R AMLrelapsed and refractory acute myeloid leukemia
RNAribonucleic acid
SCFstem cell factor
SDF-1stromal cell-derived factor 1
TET2ten eleven translocation methylcytosine dioxygenase 2
TKDtyrosine kinase domain
Topo IItopoisomerase II
VCAM-1vascular cell adhesion molecule-1
VLA-4very late antigen-4
WHOWorld Health Organization

References

  1. 1. Thein MS, Ershler WB, Jemal A, Yates JW, Baer MR. Outcome of older patients with acute myeloid leukemia: An analysis of SEER data over 3 decades. Cancer. 2013;119(15):2720-2727
  2. 2. Juliusson G, Antunovic P, Derolf A, Lehmann S, Möllgård L, Stockelberg D, et al. Age and acute myeloid leukemia: Real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood. 2009;113(18):4179-4187
  3. 3. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424-447
  4. 4. Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. The New England Journal of Medicine. 17 Sep 2015;373(12):1136-1152. DOI: 10.1056/NEJMra1406184. PMID: 26376137
  5. 5. Eleni LD, Nicholas ZC, Alexandros S. Challenges in treating older patients with acute myeloid leukemia. Journal of Oncology. 2010;2010:943823
  6. 6. Koenig K, Mims A. Relapsed or primary refractory AML: Moving past MEC and FLAG-ida. Current Opinion in Hematology. 2020;27(2):108-114
  7. 7. Thol F, Schlenk RF, Heuser M, Ganser A. How I treat refractory and early relapsed acute myeloid leukemia. Blood. 2015;126(3):319-327
  8. 8. Ferguson P, Hills RK, Grech A, Betteridge S, Kjeldsen L, Dennis M, et al. An operational definition of primary refractory acute myeloid leukemia allowing early identification of patients who may benefit from allogeneic stem cell transplantation. Haematologica. 2016;101(11):1351-1358
  9. 9. Vosberg S, Greif PA. Clonal evolution of acute myeloid leukemia from diagnosis to relapse. Genes, Chromosomes & Cancer. 2019;58(12):839-849
  10. 10. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 1 Sep 2002;100(5):1532-1542. DOI: 10.1182/blood-2002-02-0492 PMID: 12176867
  11. 11. Naoe T, Kiyoi H. Gene mutations of acute myeloid leukemia in the genome era. International Journal of Hematology. Feb 2013;97(2):165-174. DOI: 10.1007/s12185-013-1257-4 Epub 2013 Jan 29. PMID: 23359299
  12. 12. Steensma DP. Clinical consequences of clonal hematopoiesis of indeterminate potential. Blood Advances. 27 Nov 2018;2(22):3404-3410. DOI: 10.1182/bloodadvances.2018020222 PMID: 30482770; PMCID: PMC6258914
  13. 13. Gebhard C, Glatz D, Schwarzfischer L, Wimmer J, Stasik S, Nuetzel M, et al. Profiling of aberrant DNA methylation in acute myeloid leukemia reveals subclasses of CG-rich regions with epigenetic or genetic association. Leukemia. Jan 2019;33(1):26-36. DOI: 10.1038/s41375-018-0165-2 Epub 2018 Jun 20. PMID: 29925905
  14. 14. Fisher JN, Kalleda N, Stavropoulou V, Schwaller J. The impact of the cellular origin in acute myeloid leukemia: Learning from mouse models. HemaSphere. 2019;3(1):e152
  15. 15. Gruszka AM, Valli D, Alcalay M. Understanding the molecular basis of acute myeloid leukemias: Where are we now? International Journal of Hematologic Oncology. 2017;6(2):43-53. DOI: 10.2217/ijh-2017-0002
  16. 16. Grove CS, Vassiliou GS. Acute myeloid leukaemia: A paradigm for the clonal evolution of cancer? Disease Models & Mechanisms. 2014;7(8):941-951. DOI: 10.1242/dmm.015974
  17. 17. Thol F, Heuser M. Treatment for relapsed/refractory acute myeloid leukemia. Hema. 1 Jun 2021;5(6):e572. DOI: 10.1097/HS9.0000000000000572 PMID: 34095756; PMCID: PMC8171365
  18. 18. Brandwein JM, Saini L, Geddes MN, Yusuf D, Liu F, Schwann K, et al. Outcomes of patients with relapsed or refractory acute myeloid leukemia: A population-based real-world study. American Journal of Blood Research. 25 Aug 2020;10(4):124-133 PMID: 32923092; PMCID: PMC7486485
  19. 19. Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochimica et Biophysica Acta. 11 Nov 1976;455(1):152-162. DOI: 10.1016/0005-2736(76)90160-7 PMID: 990323
  20. 20. Zeng DF, Zhang J, Zhu LD, Kong PY, Li JP, Zhang X, et al. Analysis of drug resistance-associated proteins expressions of patients with the recurrent of acute leukemia via protein microarray technology. European Review for Medical and Pharmacological Sciences. 2014;18(4):537-543 PMID: 24615181
  21. 21. van den Heuvel-Eibrink MM, Wiemer EA, de Boevere MJ, van der Holt B, Vossebeld PJ, Pieters R, et al. MDR1 gene-related clonal selection and P-glycoprotein function and expression in relapsed or refractory acute myeloid leukemia. Blood. 1 Jun 2001;97(11):3605-3611. DOI: 10.1182/blood.v97.11.3605. PMID: 11369657.
  22. 22. Zhang J, Gu Y, Chen B. Mechanisms of drug resistance in acute myeloid leukemia. Oncotargets and Therapy. 11 Mar 2019;12:1937-1945. DOI: 10.2147/OTT.S191621 PMID: 30881045; PMCID: PMC6417008
  23. 23. van der Kolk DM, de Vries EG, van Putten WJ, Verdonck LF, Ossenkoppele GJ, Verhoef GE, et al. P-glycoprotein and multidrug resistance protein activities in relation to treatment outcome in acute myeloid leukemia. Clinical Cancer Research. Aug 2000;6(8):3205-3214 PMID: 10955805
  24. 24. van der Kolk DM, de Vries EG, Noordhoek L, van den Berg E, van der Pol MA, Müller M, et al. Activity and expression of the multidrug resistance proteins P-glycoprotein, MRP1, MRP2, MRP3 and MRP5 in de novo and relapsed acute myeloid leukemia. Leukemia. Oct 2001;15(10):1544-1553. DOI: 10.1038/sj.leu.2402236 PMID: 11587212
  25. 25. Broxterman HJ, Sonneveld P, van Putten WJ, Lankelma J, Eekman CA, Ossenkoppele GJ, et al. P-glycoprotein in primary acute myeloid leukemia and treatment outcome of idarubicin/cytosine arabinoside-based induction therapy. Leukemia. Jun 2000;14(6):1018-1024. DOI: 10.1038/sj.leu.2401796 PMID: 10865967
  26. 26. da Silveira Júnior LS, Soares VL, Jardim da Silva AS, Gil EA, Pereira de Araújo MDG, Merces Gonçalves CA, et al. P-glycoprotein and multidrug resistance-associated protein-1 expression in acute myeloid leukemia: Biological and prognosis implications. International Journal of Laboratory Hematology. Oct 2020;42(5):594-603. DOI: 10.1111/ijlh.13241 Epub 2020 May 26. PMID: 32452631
  27. 27. Do JH, Oh SH, Song EJ, Chung JS, Kang CD, Lee EY. Treatment outcome of multidrug resistance related mRNA expression and c-jun-N-terminal kinase activity in patients with acute myeloid leukemia. The Korean Journal of Laboratory Medicine. Aug 2007;27(4): 229-236. Korean. DOI: 10.3343/kjlm.2007.27.4.229. PMID: 18094581
  28. 28. Legrand O, Simonin G, Beauchamp-Nicoud A, Zittoun R, Marie JP. Simultaneous activity of MRP1 and Pgp is correlated with in vitro resistance to daunorubicin and with in vivo resistance in adult acute myeloid leukemia. Blood. 1 Aug 1999;94(3):1046-1056 PMID: 10419897
  29. 29. Funato T, Harigae H, Abe S, Sasaki T. Assessment of drug resistance in acute myeloid leukemia. Expert Review of Molecular Diagnostics. Sep 2004;4(5):705-713. DOI: 10.1586/14737159.4.5.705 PMID: 15347263
  30. 30. Kulsoom B, Shamsi TS, Afsar NA. Lung resistance-related protein (LRP) predicts favorable therapeutic outcome in acute myeloid leukemia. Scientific Reports. 23 Jan 2019;9(1):378. DOI: 10.1038/s41598-018-36780-8 PMID: 30674943; PMCID: PMC6344578
  31. 31. Scheffer GL, Wijngaard PL, Flens MJ, Izquierdo MA, Slovak ML, Pinedo HM, et al. The drug resistance-related protein LRP is the human major vault protein. Nature Medicine. Jun 1995;1(6):578-582. DOI: 10.1038/nm0695-578 PMID: 7585126
  32. 32. Voso MT, D’Alo’ F, Putzulu R, Mele L, Scardocci A, Chiusolo P, et al. Negative prognostic value of glutathione S-transferase (GSTM1 and GSTT1) deletions in adult acute myeloid leukemia. Blood. 15 Oct 2002;100(8):2703-2707. DOI: 10.1182/blood.V100.8.2703 PMID: 12351375
  33. 33. Sargent JM, Williamson C, Hall AG, Elgie AW, Taylor CG. Evidence for the involvement of the glutathione pathway in drug resistance in AML. Advances in Experimental Medicine and Biology. 1999;457:205-209. DOI: 10.1007/978-1-4615-4811-9_22. PMID: 10500795
  34. 34. Karwicka E. Role of glutathione in the multidrug resistance in cancer. Advances in Cell Biology. 2010;2(3):105-124. DOI: 10.2478/v10052-010-0006-6
  35. 35. Pei S, Minhajuddin M, Callahan KP, Balys M, Ashton JM, Neering SJ, et al. Targeting aberrant glutathione metabolism to eradicate human acute myelogenous leukemia cells. The Journal of Biological Chemistry. 22 Nov 2013;288(47):33542-33558. DOI: 10.1074/jbc.M113.511170 Epub 2013 Oct 2. PMID: 24089526; PMCID: PMC3837103
  36. 36. Cowell IG, Austin CA. Mechanism of generation of therapy related leukemia in response to anti-topoisomerase II agents. International Journal of Environmental Research and Public Health. Jun 2012;9(6):2075-2091. DOI: 10.3390/ijerph9062075 Epub 2012 May 31. PMID: 22829791; PMCID: PMC3397365
  37. 37. Beck WT, Morgan SE, Mo YY, Bhat UG. Tumor cell resistance to DNA topoisomerase II inhibitors: New developments. Drug Resistance Updates. Dec 1999;2(6): 382-389. DOI: 10.1054/drup.1999.0110. PMID: 11498354
  38. 38. Okada Y, Tosaka A, Nimura Y, Kikuchi A, Yoshida S, Suzuki M. Atypical multidrug resistance may be associated with catalytically active mutants of human DNA topoisomerase II alpha. Gene. 11 Jul 2001;272(1-2):141-148. DOI: 10.1016/s0378-1119(01)00554-6 PMID: 11470519
  39. 39. Nicholson R, Menezes AC, Azevedo A, Leckenby A, Davies S, Seedhouse C, et al. Protein kinase C epsilon overexpression is associated with poor patient outcomes in AML and promotes daunorubicin resistance through p-glycoprotein-mediated drug efflux. Frontiers in Oncology. 30 May 2022;12:840046. DOI: 10.3389/fonc.2022.840046 PMID: 35707351; PMCID: PMC9191576
  40. 40. Shtil AA, Ktitorova OV, Kakpakova ES, Holian O. Differential effects of the MDR1 (multidrug resistance) gene-activating agents on protein kinase C: Evidence for redundancy of mechanisms of acquired MDR in leukemia cells. Leukemia & Lymphoma. Dec 2000;40(1-2):191-195. DOI: 10.3109/10428190009054896 PMID: 11426620
  41. 41. Carter BZ, Mak PY, Tao W, Warmoes M, Lorenzi PL, Mak D, et al. Targeting MCL-1 dysregulates cell metabolism and leukemia-stroma interactions and resensitizes acute myeloid leukemia to BCL-2 inhibition. Haematologica. 1 Jan 2022;107(1):58-76. DOI: 10.3324/haematol.2020.260331 PMID: 33353284; PMCID: PMC8719086
  42. 42. Wang H, Guo M, Wei H, Chen Y. Targeting MCL-1 in cancer: Current status and perspectives. Journal of Hematology & Oncology. 21 Apr 2021;14(1):67. DOI: 10.1186/s13045-021-01079-1 PMID: 33883020; PMCID: PMC8061042
  43. 43. Tibes R, Bogenberger JM. Transcriptional silencing of MCL-1 through cyclin-dependent kinase inhibition in acute myeloid leukemia. Frontiers in Oncology. 12 Dec 2019;9:1205. DOI: 10.3389/fonc.2019.01205 PMID: 31921615; PMCID: PMC6920180
  44. 44. Nogami A, Oshikawa G, Okada K, Fukutake S, Umezawa Y, Nagao T, et al. FLT3-ITD confers resistance to the PI3K/Akt pathway inhibitors by protecting the mTOR/4EBP1/Mcl-1 pathway through STAT5 activation in acute myeloid leukemia. Oncotarget. 20 Apr 2015;6(11):9189-9205. DOI: 10.18632/oncotarget.3279 PMID: 25826077; PMCID: PMC4496211
  45. 45. Darici S, Alkhaldi H, Horne G, Jørgensen HG, Marmiroli S, Huang X. Targeting PI3K/Akt/mTOR in AML: Rationale and clinical evidence. Journal of Clinical Medicine. 11 Sep 2020;9(9):2934. DOI: 10.3390/jcm9092934 PMID: 32932888; PMCID: PMC7563273
  46. 46. Daver N, Venugopal S, Ravandi F. FLT3 mutated acute myeloid leukemia: 2021 treatment algorithm. Blood Cancer Journal. 27 May 2021;11(5):104. DOI: 10.1038/s41408-021-00495-3 PMID: 34045454; PMCID: PMC8159924
  47. 47. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 27 May 2016;127(20):2391-2405. DOI: 10.1182/blood-2016-03-643544 Epub 2016 Apr 11. PMID: 27069254
  48. 48. El Fakih R, Rasheed W, Hawsawi Y, Alsermani M, Hassanein M. Targeting FLT3 mutations in acute myeloid leukemia. Cell. 8 Jan 2018;7(1):4. DOI: 10.3390/cells7010004 PMID: 29316714; PMCID: PMC5789277
  49. 49. Scholl S, Fleischmann M, Schnetzke U, Heidel FH. Molecular mechanisms of resistance to FLT3 inhibitors in acute myeloid leukemia: Ongoing challenges and future treatments. Cell. 17 Nov 2020;9(11):2493. DOI: 10.3390/cells9112493 PMID: 33212779; PMCID: PMC7697863
  50. 50. Lam SSY, Leung AYH. Overcoming resistance to FLT3 inhibitors in the treatment of FLT3-mutated AML. International Journal of Molecular Sciences. 24 Feb 2020;21(4):1537. DOI: 10.3390/ijms21041537 PMID: 32102366; PMCID: PMC7073218
  51. 51. Venney D, Mohd-Sarip A, Mills KI. The impact of epigenetic modifications in myeloid malignancies. International Journal of Molecular Sciences. 9 May 2021;22(9):5013. DOI: 10.3390/ijms22095013 PMID: 34065087; PMCID: PMC8125972
  52. 52. Wakita S, Yamaguchi H, Omori I, Terada K, Ueda T, Manabe E, et al. Mutations of the epigenetics-modifying gene (DNMT3a, TET2, IDH1/2) at diagnosis may induce FLT3-ITD at relapse in de novo acute myeloid leukemia. Leukemia. Apr 2013;27(5):1044-1052. DOI: 10.1038/leu.2012.317 Epub 2012 Nov 8. PMID: 23135354
  53. 53. Conway O’Brien E, Prideaux S, Chevassut T. The epigenetic landscape of acute myeloid leukemia. Advances in Hematology. 2014;2014:103175. DOI: 10.1155/2014/103175 Epub 2014 Mar 23. PMID: 24778653; PMCID: PMC3980839
  54. 54. Gabra MM, Salmena L. microRNAs and acute myeloid leukemia chemoresistance: A mechanistic overview. Frontiers in Oncology. 30 Oct 2017;7:255. DOI: 10.3389/fonc.2017.00255 PMID: 29164055; PMCID: PMC5674931
  55. 55. Tutar Y. miRNA and cancer; computational and experimental approaches. Current Pharmaceutical Biotechnology. 2014;15(5):429. DOI: 10.2174/138920101505140828161335 PMID: 25189575
  56. 56. Si W, Shen J, Zheng H, Fan W. The role and mechanisms of action of microRNAs in cancer drug resistance. Clinical Epigenetics. 11 Feb 2019;11(1):25. DOI: 10.1186/s13148-018-0587-8 PMID: 30744689; PMCID: PMC6371621
  57. 57. Rashidi A, Uy GL. Targeting the microenvironment in acute myeloid leukemia. Current Hematologic Malignancy Reports. Jun 2015;10(2):126-131. DOI: 10.1007/s11899-015-0255-4 PMID: 25921388; PMCID: PMC4447552
  58. 58. Wang A, Zhong H. Roles of the bone marrow niche in hematopoiesis, leukemogenesis, and chemotherapy resistance in acute myeloid leukemia. Hematology. Dec 2018;23(10):729-739. DOI: 10.1080/10245332.2018.1486064 Epub 2018 Jun 14. PMID: 29902132
  59. 59. Shin DY. Human acute myeloid leukemia stem cells: Evolution of concept. Blood Research. 30 Apr 2022;57(S1):67-74. DOI: 10.5045/br.2022.2021221 PMID: 35483929; PMCID: PMC9057671
  60. 60. Menter T, Tzankov A. Tumor microenvironment in acute myeloid leukemia: Adjusting niches. Frontiers in Immunology. 22 Feb 2022;13:811144. DOI: 10.3389/fimmu.2022.811144 PMID: 35273598; PMCID: PMC8901718
  61. 61. Ahn JY, Seo K, Weinberg OK, Arber DA. The prognostic value of CXCR4 in acute myeloid leukemia. Applied Immunohistochemistry & Molecular Morphology. Jan 2013;21(1):79-84. DOI: 10.1097/PAI.0b013e3182606f4d PMID: 22914607
  62. 62. Matsunaga T, Takemoto N, Sato T, Takimoto R, Tanaka I, Fujimi A, Akiyama T, et al. Interaction between leukemic-cell VLA-4 and stromal fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia. Nature Medicine. Sep 2003;9(9):1158-1165. doi: 10.1038/nm909. Epub 2003 Aug 3. Erratum in: Nat Med. 2005 May;11(5):578. PMID: 12897778
  63. 63. Becker PS, Kopecky KJ, Wilks AN, Chien S, Harlan JM, Willman CL, et al. Very late antigen-4 function of myeloblasts correlates with improved overall survival for patients with acute myeloid leukemia. Blood. 22 Jan 2009;113(4):866-874. DOI: 10.1182/blood-2007-12-124818 Epub 2008 Oct 16. PMID: 18927435; PMCID: PMC2630271
  64. 64. Quéré R, Andradottir S, Brun AC, Zubarev RA, Karlsson G, Olsson K, et al. High levels of the adhesion molecule CD44 on leukemic cells generate acute myeloid leukemia relapse after withdrawal of the initial transforming event. Leukemia. Mar 2011;25(3):515-526. DOI: 10.1038/leu.2010.281 Epub 2010 Nov 30. PMID: 21116281; PMCID: PMC3072510
  65. 65. Yu X, Munoz-Sagredo L, Streule K, Muschong P, Bayer E, Walter RJ, et al. CD44 loss of function sensitizes AML cells to the BCL-2 inhibitor venetoclax by decreasing CXCL12-driven survival cues. Blood. 23 Sep 2021;138(12):1067-1080. DOI: 10.1182/blood.2020006343 PMID: 34115113
  66. 66. Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, et al. A role for the bone marrow microenvironment in drug resistance of acute myeloid leukemia. Cell. 21 Oct 2021;10(11):2833. DOI: 10.3390/cells10112833 PMID: 34831055; PMCID: PMC8616250

Written By

Yanitsa Davidkova, Milan Jagurinoski, Gueorgui Balatzenko and Margarita Guenova

Submitted: 27 July 2022 Reviewed: 02 December 2022 Published: 22 December 2022