Autophagy in Acute Myeloid Leukemia: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Autophagy is a highly conserved cellular degradation process that regulates cellular metabolism and homeostasis under normal and pathophysiological conditions. Autophagy and metabolism are linked in the hematopoietic system, playing a critical role in the self-renewal, survival and differentiation of hematopoietic stem and progenitor cells, and in cell death, particularly influencing the cell fate of the hematopoietic stem cell pool. In leukemia, autophagy supports leukemia cell growth, contributes to leukemia stem cell survival and resistance to chemotherapy. Acute myeloid leukemia (AML), a common type of acute leukemia with poor survival and prognosis, 

  • autophagy
  • hematopoiesis
  • acute myeloid leukemia
  • metabolism
  • therapy resistance

1. Autophagy and Acute Myeloid Leukemia Stem Cells

During leukemic development, LSCs can adapt their metabolism and autophagy mechanisms to supply high energy and nutrients required for LSCs proliferation and survival under conditions of nutrient deficiency, starvation, hypoxia, or during chemotherapy treatments [1][2][3][4][5].
Mitophagy, mitochondrial function, and integrity may affect the viability, proliferation, and differentiation potential and longevity of normal hematopoietic stem cells (HSCs) [6], and are important in the survival strategy of acute myeloid leukemia (AML) stem cells (LSCs).
In particular, the adenosine5′-monophosphate (AMP)-activated protein kinase (AMPK), a protein complex fundamental in mitochondrial metabolism and mitophagy, is constitutively activated in LSCs, increasing mitochondrial clearance to support LSCs growth and survival through its downstream target FIS1, the mitochondrial fission 1 protein component of a mitochondrial complex that promotes mitochondrial fission [7]. AMPK/FIS1-mediated mitophagy is required for the self-renewal and survival of LSCs [7]. An overexpression of FIS1 was also found in AML cells while FIS1 depletion impairs mitophagy, weakening the self-renewal capacity of LSCs and determining the induction of myeloid differentiation by GSK3 inactivation (glycogen synthase kinase 3), thus indicating mitophagy as being a regulatory mechanism for the progression of AML [7]. More recently, the loss of sequestosome 1 (SQSTM1), also known as p62, a selective autophagy receptor crucial for the development and progression of AML in vivo, induces the accumulation of damaged mitochondria and mitochondrial superoxide, thus compromising the survival of leukemia cells. Then, the loss of SQSTM1 impairs leukemia progression in AML mouse models, underlying the role of mitophagy in the survival of LSCs [8]. Altogether, these studies demonstrate that enhanced autophagic activity of LSCs is required for malignant progression into AML.
However, in contrast to the autophagy activation observed in AML, a loss of autophagy in healthy HSCs triggers the expansion of a population of progenitor cells in the bone marrow, giving rise to severe and invasive myeloproliferation, such as in human AML [9]. This apparent paradox can be explained by the distinct roles that autophagy can play during AML progression, which may be different at various stages of leukemogenesis [10][11]. Autophagy in normal HSCs may prevent the onset of cancer, as a tumor suppressive mechanism. Indeed, autophagy removes damaged organelles, such as mitochondria, and protects hematopoietic cells from genomic instability and inflammation, thus preventing the onset of leukemia. Particularly, increased DNA damage, high ROS levels, aneuploidy, and an aberrant accumulation of p62/SQSTM1 have been correlated with an impaired autophagic process, indicating a key role of autophagy in preventing tumor initiation [12]. Conversely, in established cancer, autophagy may function as a favorable pathway that promotes survival and tumor growth, by helping tumor cells to escape metabolic stress and death stimuli.
Some studies have also shown that the activation of autophagic flow plays a role only in the initiation of AML, with a transformation from HSCs to LSCs, and, therefore, after this phase, autophagy is not required for disease maintenance [13]. Studies in MLL-AF9 AML, the most common alteration in childhood AML, indicate ATG5 or ATG7 as necessary for the onset of AML, but once the leukemic condition is established, autophagy is not required for LSC function in vivo [13][14]. However, in a different MLL-ENL AML mouse model, Atg5 or Atg7 knockout reduced the number of functional LSCs, increased mitochondrial activation and ROS levels in these cells, and prolonged the survival of leukemic mice [15]. In this context, during the leukemogenesis process, histone methylation can regulate core autophagy effectors and upstream autophagic regulators such as ATG 5 and ATG7 to influence autophagy level indirectly [16]. Together, these studies suggest a highly complex and context-dependent role for autophagy in leukemic transformation with respect to the maintenance properties of LSCs in AML.
The dual role of autophagy in AML, as a promoter or suppressor of cancer in AML, is still a matter of debate. Studies have shown that autophagy can act as a pro or anti-proliferative mechanism depending on the lineage and the molecular genotypic context of the disease, reflecting the degree of heterogeneity of AML [17].

2. Regulation of Autophagy Genes in Acute Myeloid Leukemia Cells

Numerous studies have shown that increased autophagy in AML cells confers protection from chemotherapeutic treatment and promotes AML cell survival.
Increased ATG7-mediated autophagy has been associated with poor clinical outcomes and a short duration of remission in AML patients [18]. More recently, some proteins involved in leukemic cell survival, and overexpressed in AML, have been related to ATG overexpression, underlying the interplay between autophagy and protein overexpression promoting leukemic cell survival [19]. Hu et al. have shown that a high expression of SIRT1 (Sirtuin 1), a key player in mitochondrial biogenesis and autophagy-related protein, is associated with high CXCR4 expression, a negative prognostic marker in AML, and with other autophagy-related proteins such as ATG5 and LC3 in primary human AML samples, indicating a potential role of the SDF-1α-CXCR4 signaling pathway in autophagy induction in AML cells, which further promotes their survival under stress [20].
The transient receptor potential melastatin 2 (TRPM2) ion channel, involved in maintaining cell survival following oxidant injury, is highly expressed in AML [21]. By performing TRPM2-depletion, Chen SJ et al. [21] have shown that ULK1, Atg7, and Atg5 protein levels are decreased in TRPM2-depleted cells, leading to autophagy inhibition. Importantly, the depletion of TRPM2 in AML inhibits leukemia proliferation and increases the doxorubicin sensitivity of AML cells [21].
Functional studies in normal CD34+ CB cells indicated that the inhibition of VMP1 expression reduced autophagic flux, with decreased hematopoietic stem and progenitor cell (HSPC) expansion, delayed differentiation, increased apoptosis, and impaired cell function and in vivo engraftment. Similar results were observed in leukemic cell lines and primary AML CD34+ cells. Furthermore, ultrastructural analysis indicated that leukemic cells overexpressing VMP1 have a reduced number of mitochondrial structures, and the number of lysosomal-degrading structures has increased. VMP1 (vacuole membrane protein-1) overexpression increased autophagic flux and improved mitochondrial quality, which coincided with an increased threshold for venetoclax-induced loss of mitochondrial outer membrane permeabilization (MOMP) and apoptosis in leukemia cells [22].
Heterozygous deletions, missense mutations, or changes in the number of copies of key autophagy genes have been found with a high frequency in AML patients, especially AML patients with complex karyotypes [15][23]. In particular, a heterozygous chromosomal loss of 5q, 16q, or 17p correlates with regions encoding autophagy genes ATG10 and ATG12, GABARAPL2 and MAP1LC3B, or GABARAP, respectively [15], and several others autophagy genes have a low level of expression in human AML blasts, a decreased autophagic flow, and high levels of ROS [15]. In addition, a study suggested that key autophagy genes such as ULK1, ATG3, ATG4D, and ATG5 were significantly downregulated in primary AML cells compared to normal granulocytes [24].

3. Autophagic Biomarkers

Significant progress has recently been made to identify specific autophagy-related genes for the prediction of clinical outcomes in AML. Along with the ATG genes previously described, several microRNAs implicated in leukemogenesis and chemoresistance have been also involved in the activation of autophagy, and may be used as biomarkers [25]. In particular, miR-17-5p overexpression in leukemia promotes AML proliferation by inhibiting autophagy through BECN1 targeting [26][27][28]. Ganesan et al. demonstrated that stromal cells downregulate miR-23a-5p levels in leukemic cells, leading to the upregulation of protective autophagy in these cells, thereby increasing their resistance to chemotherapy toxicity [29]. MiR-143 overexpression was shown to enhance the sensitivity of AML cells to the cytotoxicity of cytarabine (Ara-C) treatment by inhibiting autophagy through ATG7 and ATG2B targeting [30]. An overexpression of miR-15a-5p is involved in the chemoresistance of AML patients, through autophagy-related genes ATG9A, ATG14, GABARAPL1, and SMPD1 targeting AML cells [31].
Recent advances in bioinformatics have yielded an autophagy-related signature that can help to predict overall survival (OS) and/or the clinical outcomes of AML patients. Several studies have shown that the progression of AML depends on the autophagy-associated gene signature [32]. A recent bioinformatics study has built a model containing 10 autophagy-related genes to predict the survival of AML patients, showing that groups at high risk of AML have an increased expression of immune checkpoint genes and a higher percentage of CD4 T and NK cells [33]. In addition, this research was able to predict OS in AML through the signature of 10 genes, indicating this model as an effective prognostic predictor for AML patients, useful to guide patient stratification for immunotherapies and drugs [33]. The bioinformatics study LASSO Cox regression that identified a critical risk signature for AML, consisting of the autophagy genes BAG3, CALCOCO2, CAMKK2, CANX, DAPK1, P4HB, TSC2, and ULK1, had excellent predictive power for AML prognosis [34]. Notably, the immunosuppressive cytokines were found to be significantly increased in the tumor microenvironment of patients with a high-risk of AML, predicted on the basis of the autophagy-related signature of these patients [34]. However, the prognostic value of the ATG signature in the clinical setting is still debated. Therefore, the roles of the ATG signature and autophagy in the pathogenesis of AML should be further investigated.
Furthermore, an interesting study indicated that an autophagy-related lncRNA signature containing six lncRNAs (HYMAI, MIR155HG, MGC12916, DIRC3, C1orf220, and HCP5) may have an important prognostic value [35]. A recent study indicated four autophagy-associated lncRNAs (MIR133A1HG, AL359715.1, MIRLET7BHG, and AL356752.1) as a signature to potentially use as a biomarker to predict the survival of AML patients [36].
Collectively, these data indicate that the role of autophagy in tumor development clearly depends on the type of AML and stage of tumor development. Furthermore, autophagy may provide cancer cells with a survival strategy, suggesting a therapeutic use for autophagy inhibition. On the other hand, autophagy can induce cell death, pointing to autophagy activation as a novel strategy in cancer therapy. Therefore, it is necessary to determine the role played by autophagy in the molecular subtypes of AML, or the degree of tumor development, to verify whether its modulation could lead to benefits for the treated patient.

4. Autophagy and Genetic Alterations in Acute Myeloid Leukemia

The AML phenotype results from multiple molecular, genetic, and epigenetic alterations affecting the differentiation, proliferation, and apoptosis of myeloid progenitors. The World Health Organization has classified AMLs according to the presence of particular genetic alterations, frequently originating from chromosomal translocations or other genome rearrangements such as t(8;21), t(15;17), inv (16), inv(3), t(6;9), t(9;11) or t(11;19), or mutations in receptor kinases, in key signaling mediators, proto-oncogenes, or epigenetic enzymes, e.g., mutations in FLT3 (FMS-like tyrosine kinase 3), TP53, c-KIT or IDH1/2, NPM1 (nucleophosmin 1), and CCAAT enhancer-binding protein (CEBPA) [37][38][39]. These mutations in AMLs have an impact on the choice of the most suitable therapy.
The association between autophagy and recurrent genetic alterations has been described in several studies in AML, but needs further investigation [40][41]. Here, researchers summarize and update the recent advances that have highlighted the link between autophagy and fusion genes and recurrent oncogenic mutations in AML and the involvement of autophagy in chemotherapy treatment (Figure 1).
Figure 1. Gene mutations in AML and autophagy. Several recurrent genetic abnormalities in AML are involved in the deregulation of autophagy, leading to leukemia progression.

4.1. Fusion Genes in Acute Myeloid Leukemia and Autophagy

Most cases of APL are caused by a de novo t (15;17) (q22; q21) translocation, which results in the fusion of the RARA gene with the PML gene [42][43]. APL cells that have a lower expression of autophagy-related genes than normal cells have a reduced autophagic activity. By using differentiating agents, such as all-trans retinoid acid (ATRA) and arsenic trioxide (ATO) currently used in clinical settings, the expression level of autophagy-related genes increases, thus restoring autophagy in APL cells [44]. Both agents can activate ETosis, a type of cell death mediated by the release of neutrophil extracellular traps (ETs). In addition, mTOR-dependent autophagic action is required for ATO-induced NETosis in APL cells [45]. Of note, rapamycin, the inhibitor of mTOR pathway, synergizes with ATO in the eradication of leukemia-initiating cells (LIC) through the activation of NETosis in both APL cells and an in vivo APL model [45].
Mixed lineage leukemia (MLL) gene translocations 11q23 were observed in approximately 80% of pediatric AML. In these, the MLL gene can, by genomic translocation, be fused with >60 different fusion partners [46]. Treatment with the RAS oncogene inhibitor, tipifamib, leads to the inhibition of AML with the t(6;11) translocation by inducing both apoptosis and autophagy [47]. Another study demonstrated that ATG5 participates in the development of MLL-AF9-driven leukemia, but not in AML-sensitive chemotherapy mice expressing MLL-AF9 [48].
Acute myeloid leukemia with core binding factor (CBF-AML) is characterized by the presence of t(8;21) (q22; q22), or inv (16) (p13q22)/t(16;16), which leads to the formation of RUNX1/RUNX1T1 (AML1/ETO) and CBFbeta-MYH11, respectively [49]. The activation of ULK1-mediated autophagy may control and delay AML1-ETO9a -guided leukemogenesis in an AML CASPASE-3 knockout mouse model [50], suggesting that CASPASE-3 is an important regulator of autophagy in AML. The results of these studies highlight the different roles of autophagy in the initiation, progression, and chemotherapeutic responses in AML cells, depending on the different type of aberrant oncoprotein.

4.2. Gene Mutations in Acute Myeloid Leukemia and Autophagy

FLT3
Among the most common genetic alterations in AML, the tyrosine kinase 3 (FLT3) gene mutation occurs in approximately 30% of AML cases.
The most frequent aberrations affecting FLT3 gene, associated with a poor prognosis in AML, are the internal tandem duplication (FLT3-ITD) in the juxtamembrane domain, and point mutations, involving the tyrosine kinase domain of FLT3 (FLT3-TKD) [51]. FLT3-ITD expression increases basal autophagy in AML cells through a mechanism involving transcription factor ATF4 (activating transcription factor 4) [52]. In addition, the inhibition of autophagy in FLT3-TKD cells, which are resistant to the FLT3 inhibitor quizartinib (AC220), also inhibits proliferation both in vitro and in vivo [52]. More recently, the acquired D835Y mutation induced resistance to the FLT3 inhibitor sorafenib, and activated autophagy in FLT3-ITD-positive cell lines. By inhibiting autophagy, the authors were able to overcome resistance to sorafenib in FLT3-ITD-positive AML, improving its efficacy [53]. Recently, a study showed that the inhibition of autophagy reduces the repopulation potential of FLT3-ITD AML LSCs associated with mitochondrial accumulation [54]. In addition, the authors showed that autophagy inhibition improves p53 activity and increases the TKI-mediated inhibition of AML progenitors [54].
Autophagy not only contributes to downstream proliferation of the FLT3-ITD receptor, but may also be involved in mutated receptor degradation. In fact, in one study, the frequent activation of the receptor tyrosine kinase RET was observed in several AML subtypes [55]. RET mediates autophagy suppression in an mTORC1-dependent manner, leading to the stabilization of the mutant FLT3 receptor. The genetic or pharmacological inhibition of RET decreased the growth of FLT3-dependent AML cells, with the upregulation of autophagy and FLT3 depletion [55]. These results suggest that restoring autophagy in FLT3-dependent AML may result in the degradation of mutated FLT3, and therefore may represent an interesting therapeutic approach. It has also been shown that the inhibition of the FLT3-ITD protein leads to an increase in ceramide synthesis and mediates ceramide-dependent mitophagy, leading to AML cell death [56][57].
KIT
KIT mutations are associated with an increased proliferation of leukemic cells and an increased risk of AML recurrence [58][59]. A recent study reported that the KIT D816V mutation in AML cells increases basal autophagy, stimulating AML cell proliferation and survival via STAT3 signaling [60]. A different point mutation in c-KIT (N822K T > A) constitutively activates this receptor, making AML cells highly sensitive to sunitinib (a tyrosine kinase inhibitor), resulting in AML cell death through the activation of both apoptosis and autophagy processes [61].
NPM1
Mutations in NPM1 (nucleophosmin 1) are the most frequent genetic alterations in adult AML, responsible for the aberrant localization of the NPM1 protein in the cytoplasm [62]. Increased autophagic activity found in NPM1 -mutated AML cells is involved in leukemic cell survival [63]. Mutant NPM1 can also interact with the tumor suppressor protein PML (leukemia pro-myelocytic protein), leading to PML delocalization and stabilization that, in turn, can activate autophagy via AKT signaling [63]. In another study, it was shown that in AML patients carrying mutant NPM1, the glycolytic enzyme PKM2 (pyruvate kinase M2) induced autophagy via phosphorylation of the autophagic protein Beclin 1, contributing to cell survival [64]. Finally, the NPM1 mutant protein can also interact with the autophagic protein ULK1, stimulating the TRAF6-dependent ubiquitination of ULK1 via miR-146, thereby maintaining ULK1 stability and functionality and promoting autophagic cell survival [65]. Furthermore, it was observed that the expression of RASGRP3, a protein associated with tumor progression, is upregulated in patients with AML with NPM1 mutation compared to patients with AML without mutant NPM1. The authors demonstrated that NPM1 -mut blocks the degradation of the RASGRP3 protein through binding to the ubiquitin ligase E3 MID1 protein, leading to RASGRP3 overexpression, as well as promoting the downstream activation of EGFR-STAT3, which in turn promoted proliferation and autophagy in AML cells [66].
P53
Alterations in the tumor suppressor gene TP53 are found in about 5–15% of AML cases, and, frequently, in older patients [67][68]. It has been proposed that the role of autophagy in the development of AML can be determined by the status of TP53. For wild-type TP53 AML, researchers have shown that pharmacological autophagy blockade achieves therapeutic benefits, while AMLs harboring TP53 mutations do not respond to the inhibition of autophagy by hydroxychloroquine (HCQ) [69][70]. The use of autophagic inhibitors may be a potential therapeutic strategy to use, particularly for the treatment of TP53 wild-type AML. For AML with TP53 mutations, autophagic pathways may be a therapeutic option to use for the elimination of mutant T TP53.
Another study demonstrated that macroautophagy stimulation by 17-AAG, a HSP90 inhibitor, causes the degradation of TP53 R248Q in AML cells and also enhances the transcription of autophagy-associated genes [71]. In addition, accumulated evidence indicates that TP53 activated by a variety of cellular stresses can trigger autophagy through the transactivation of pro-autophagy genes, including DRAM1 (autophagic modulator regulated by DNA damage 1), SESN1 (sestrin 1), and (sestrin SESN2) [71][72][73][74].
A recent study highlighted the role of autophagy in AML cells, in the context of p53-mediated apoptosis, which is associated with increased cytotoxicity to treatment with MDM2 inhibitors and Ara-C when miR-10a is inhibited [75]. The antileukemic strategy based on the use of MDM2/X inhibitors in wild-type p53 tumors to restore the normal and active conformation of p53, MDM2, and MDMX has not been extensively tested [76]. Thus, the use of a combination of treatments, including MDM2 inhibitors with autophagic modulators, may be a new strategy to improve the treatment of wild-type p53 AML.
Pharmacological treatments that modulate autophagy in AML patients carrying p53 mutations participate in the degradation of aberrant p53 proteins. The point mutation of TP53 at the amino acid residue R428 (R248Q), with gain-of-function activity, gives rise to malignant activity in lung cancer cells [77] and a loss of tumor suppressor function in AML [78].
Interestingly, the treatment with the Hsp90 inhibitor (17-AAG) results in the activation of chaperone-mediated autophagy, which induces the degradation of the aberrant protein p53R248Q in AML cells. In particular, under conditions of metabolic stress, 17-AAG induces the interaction between p53R248Q and the chaperone protein Hsc70, triggering chaperone-mediated autophagy to degrade p53R248Q [71]. These data open new opportunities for future studies that may elucidate the functional involvement of different types of autophagy and their connection with molecular mechanisms to improve anticancer therapies against AML harboring the different TP53 variants.
IDH1/2 (isocitrate dehydrogenase)
Recent advances in bioinformatics have enabled the identification of several epigenetic mutations affecting AML, including IDH1/2, Tet methylcytosine dioxygenase 2 (TET2), DNA methyltransferase 3A (DNMT3A), and ASXL1, all of which are associated with the pathogenesis of AML [79][80][81]. IDH proteins are isocitrate dehydrogenases, implicated in various biological processes, such as energy metabolism, histone demethylation, DNA modification, and adaptation to hypoxia. Further studies are needed to investigate innovative therapies based on targeted autophagy in combination with DNA hypomethylation to treat AMLs harboring certain types of epigenetic alterations.
DNMT3A
Mutations in the DNMT3A gene, an enzyme involved in the methylation of CpG dinucleotides, are present in 20–23% of adult patients with de novo AML [81]. Several studies have shown that the treatment of AML patients with DNA methyltransferase inhibiting agents, such as azacitidine (5-aza-2′-deoxycytidine), induces autophagic activity in AML leukemia cells [82]. A study performed on a DNMT3A R878H conditional knock-in mouse model, used to predict the specific long non-coding RNAs (lncRNAs) regulated by DNMT3A mutations in AML, first identified 23 differentially expressed lncRNAs, then the downstream target genes regulated by these lncRNAs, including ATP6V1A, a critical autophagy-related gene, the overexpression of which is associated with poor prognosis in AML [83]. However, there is still little evidence of a direct involvement of DNMT3A gene mutations with autophagic activity in AML.
Further studies are needed to understand the functional significance of autophagy associated with different genetic mutations in AML cells.

This entry is adapted from the peer-reviewed paper 10.3390/cells12111553

References

  1. Stergiou, I.E.; Kapsogeorgou, E.K. Autophagy and Metabolism in Normal and Malignant Hematopoiesis. Int. J. Mol. Sci. 2021, 22, 8540.
  2. Bednarczyk, M.; Kociszewska, K.; Grosicka, O.; Grosicki, S. The role of autophagy in acute myeloid leukemia development. Expert Rev. Anticancer Ther. 2023, 23, 5–18.
  3. Maynard, R.S.; Hellmich, C.; Bowles, K.M.; Rushworth, S.A. Acute Myeloid Leukaemia Drives Metabolic Changes in the Bone Marrow Niche. Front. Oncol. 2022, 12, 924567.
  4. Schiliro, C.; Firestein, B.L. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021, 10, 1056.
  5. Castro, I.; Sampaio-Marques, B.; Ludovico, P. Targeting Metabolic Reprogramming in Acute Myeloid Leukemia. Cells 2019, 8, 967.
  6. Lin, Q.; Chen, J.; Gu, L.; Dan, X.; Zhang, C.; Yang, Y. New insights into mitophagy and stem cells. Stem Cell Res. Ther. 2021, 12, 452.
  7. Pei, S.; Minhajuddin, M.; Adane, B.; Khan, N.; Stevens, B.M.; Mack, S.C.; Sisi, L.; Rich, J.N.; Inguva, A.; Shannon, K.M.; et al. AMPK/FIS1-Mediated Mitophagy Is Required for Self-Renewal of Human AML Stem Cells. Cell Stem Cell 2018, 23, 86–100.
  8. Nguyen, T.D.; Shaid, S.; Vakhrusheva, O.; Koschade, S.E.; Klann, K.; Thölken, M.; Baker, F.; Zhang, J.; Oellerich, T.; Sürün, D.; et al. Loss of the selective autophagy receptor p62 impairs murine myeloid leukemia progression and mitophagy. Blood 2019, 133, 168–179, Erratum in: Blood 2019, 134, 94..
  9. Mortensen, M.; Soilleux, E.J.; Djordjevic, G.; Tripp, R.; Lutteropp, M.; Sadighi-Akha, E.; Stranks, A.J.; Glanville, J.; Knight, S.; Jacobsen, S.W.; et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 2011, 208, 455–467.
  10. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410.
  11. Gewirtz, D.A. The four faces of autophagy: Implications for cancer therapy. Cancer Res. 2014, 74, 647–651.
  12. Chen, H.Y.; White, E. Role of autophagy in cancer prevention. Cancer Prev. Res. 2011, 4, 973–983.
  13. Chen, X.; Clark, J.; Wunderlich, M.; Fan, C.; Davis, A.; Chen, S.; Guan, J.L.; Mulloy, J.C.; Kumar, A.; Zheng, Y. Autophagy is dispensable for Kmt2a/Mll-Mllt3/Af9 AML maintenance and anti-leukemic effect of chloroquine. Autophagy 2017, 13, 955–966.
  14. Porter, H.A.; Leveque-El Mouttie, L.; Vu, T.; Bruedigam, C.; Sutton, J.; Jacquelin, S.; Hill, G.R.; MacDonald, K.P.A.; Lane, S.W. Acute myeloid leukemia stem cell function is preserved in the absence of autophagy. Haematologica 2017, 102, 344–347.
  15. Watson, A.S.; Riffelmacher, T.; Stranks, A.; Williams, O.; De Boer, J.; Cain, K.; MacFarlane, M.; McGouran, J.; Kessler, B.; Khandwala, S.; et al. Autophagy limits proliferation and glycolytic metabolism in acute myeloid leukemia. Cell Death Discov. 2015, 1, 15008.
  16. Shu, F.; Xiao, H.; Li, Q.N.; Ren, X.S.; Liu, Z.G.; Hu, B.W.; Wang, H.S.; Wang, H.; Jiang, G.M. Epigenetic and post-translational modifications in autophagy: Biological functions and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 32.
  17. Djavaheri-Mergny, M.; Giuriato, S.; Tschan, M.P.; Humbert, M. Therapeutic Modulation of Autophagy in Leukaemia and Lymphoma. Cells 2019, 8, 103.
  18. Piya, S.; Kornblau, S.M.; Ruvolo, V.R.; Mu, H.; Ruvolo, P.P.; McQueen, T.; Davis, R.E.; Hail, N., Jr.; Kantarjian, H.; Andreeff, M.; et al. Atg7 suppression enhances chemotherapeutic agent sensitivity and overcomes stroma-mediated chemoresistance in acute myeloid leukemia. Blood 2016, 128, 1260–1269.
  19. Du, W.; Xu, A.; Huang, Y.; Cao, J.; Zhu, H.; Yang, B.; Shao, X.; He, Q.; Ying, M. The role of autophagy in targeted therapy for acute myeloid leukemia. Autophagy 2021, 17, 2665–2679.
  20. Hu, X.; Mei, S.; Meng, W.; Xue, S.; Jiang, L.; Yang, Y.; Hui, L.; Chen, Y.; Guan, M.X. CXCR4-mediated signaling regulates autophagy and influences acute myeloid leukemia cell survival and drug resistance. Cancer Lett. 2018, 425, 1–12.
  21. Chen, S.J.; Bao, L.; Keefer, K.; Shanmughapriya, S.; Chen, L.; Lee, J.; Wang, J.F.; Zhang, X.Q.; Hirschler-Laszkiewicz, I.; Merali, S.; et al. Transient receptor potential ion channel TRPM2 promotes AML proliferation and survival through modulation of mitochondrial function, ROS, and autophagy. Cell Death Dis. 2020, 11, 247.
  22. Folkerts, H.; Wierenga, A.T.; van den Heuvel, F.A.; Woldhuis, R.R.; Kluit, D.S.; Jaques, J.; Schuringa, J.J.; Vellenga, E. Elevated VMP1 expression in acute myeloid leukemia amplifies autophagy and is protective against venetoclax-induced apoptosis. Cell Death Dis. 2019, 10, 421.
  23. Visconte, V.; Przychodzen, B.; Han, Y.; Nawrocki, S.T.; Thota, S.; Kelly, K.R.; Patel, B.J.; Hirsch, C.; Advani, A.S.; Carraway, H.E.; et al. Complete mutational spectrum of the autophagy interactome: A novel class of tumor suppressor genes in myeloid neoplasms. Leukemia 2017, 31, 505–510.
  24. Jin, J.; Britschgi, A.; Schläfli, A.M.; Humbert, M.; Shan-Krauer, D.; Batliner, J.; Federzoni, E.A.; Ernst, M.; Torbett, B.E.; Yousefi, S.; et al. Low Autophagy (ATG) Gene Expression Is Associated with an Immature AML Blast Cell Phenotype and Can Be Restored during AML Differentiation Therapy. Oxid. Med. Cell Longev. 2018, 2018, 1482795.
  25. Gabra, M.M.; Salmena, L. microRNAs and Acute Myeloid Leukemia Chemoresistance: A Mechanistic Overview. Front. Oncol. 2017, 7, 255.
  26. Fischer, J.; Rossetti, S.; Datta, A.; Eng, K.; Beghini, A.; Sacchi, N. miR-17 deregulates a core RUNX1-miRNA mechanism of CBF acute myeloid leukemia. Mol. Cancer. 2015, 14, 7.
  27. Mi, S.; Li, Z.; Chen, P.; He, C.; Cao, D.; Elkahloun, A.; Lu, J.; Pelloso, L.A.; Wunderlich, M.; Huang, H.; et al. Aberrant overexpression and function of the miR-17-92 cluster in MLL-rearranged acute leukemia. Proc. Natl. Acad. Sci. USA 2010, 107, 3710–3715.
  28. Wang, W.; Liu, J.; Chen, K.; Wang, J.; Dong, Q.; Xie, J.; Yuan, Y. Vitamin D promotes autophagy in AML cells by inhibiting miR-17-5p-induced Beclin-1 overexpression. Mol. Cell Biochem. 2021, 476, 3951–3962.
  29. Ganesan, S.; Palani, H.K.; Lakshmanan, V.; Balasundaram, N.; Alex, A.A.; David, S.; Venkatraman, A.; Korula, A.; George, B.; Balasubramanian, P.; et al. Stromal cells downregulate miR-23a-5p to activate protective autophagy in acute myeloid leukemia. Cell Death Dis. 2019, 10, 736.
  30. Zhang, H.; Kang, J.; Liu, L.; Chen, L.; Ren, S.; Tao, Y. MicroRNA-143 sensitizes acute myeloid leukemia cells to cytarabine via targeting ATG7- and ATG2B-dependent autophagy. Aging 2020, 12, 20111–20126.
  31. Bollaert, E.; Claus, M.; Vandewalle, V.; Lenglez, S.; Essaghir, A.; Demoulin, J.B.; Havelange, V. MiR-15a-5p Confers Chemoresistance in Acute Myeloid Leukemia by Inhibiting Autophagy Induced by Daunorubicin. Int. J. Mol. Sci. 2021, 22, 5153.
  32. Chen, X.X.; Li, Z.P.; Zhu, J.H.; Xia, H.T.; Zhou, H. Systematic Analysis of Autophagy-Related Signature Uncovers Prognostic Predictor for Acute Myeloid Leukemia. DNA Cell Biol. 2020, 39, 1595–1605.
  33. Huang, L.; Lin, L.; Fu, X.; Meng, C. Development and validation of a novel survival model for acute myeloid leukemia based on autophagy-related genes. PeerJ 2021, 9, 11968.
  34. Fu, D.; Zhang, B.; Wu, S.; Zhang, Y.; Xie, J.; Ning, W.; Jiang, H. Prognosis and Characterization of Immune Microenvironment in Acute Myeloid Leukemia Through Identification of an Autophagy-Related Signature. Front. Immunol. 2021, 12, 695865.
  35. Gu, S.; Zi, J.; Han, Q.; Song, C.; Ge, Z. The Autophagy-Related Long Non-Coding RNA Signature for Acute Myeloid Leukemia. 16 April 2020. Available online: https://ssrn.com/abstract=3578746 (accessed on 16 April 2020).
  36. Zhao, C.; Wang, Y.; Tu, F.; Zhao, S.; Ye, X.; Liu, J.; Zhang, J.; Wang, Z. A Prognostic Autophagy-Related Long Non-coding RNA (ARlncRNA) Signature in Acute Myeloid Leukemia (AML). Front. Genet. 2021, 12, 681867.
  37. Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377.
  38. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719.
  39. Nazha, A.; Zarzour, A.; Al-Issa, K.; Radivoyevitch, T.; Carraway, H.E.; Hirsch, C.M.; Przychodzen, B.; Pate, B.J.; Clemente, M.; Sanikommu, S.R.; et al. The complexity of interpreting genomic data in patients with acute myeloid leukemia. Blood Cancer J. 2016, 6, 510.
  40. Torgersen, M.L.; Engedal, N.; Bøe, S.O.; Hokland, P.; Simonsen, A. Targeting autophagy potentiates the apoptotic effect of histone deacetylase inhibitors in t(8;21) AML cells. Blood 2013, 122, 2467–2476.
  41. Nakatani, K.; Matsuo, H.; Harata, Y.; Higashitani, M.; Koyama, A.; Noura, M.; Nishinaka-Arai, Y.; Kamikubo, Y.; Adachi, S. Inhibition of CDK4/6 and autophagy synergistically induces apoptosis in t(8;21) acute myeloid leukemia cells. Int. J. Hematol. 2021, 113, 243–253.
  42. Mannan, A.; Muhsen, N.I.; Barragán, E.; Sanz, M.A.; Mohty, M.; Hashmi, S.K.; Aljurf, M. Genotypic and Phenotypic Characteristics of Acute Promyelocytic Translocation Variants. Hematol. Oncol. Stem Cell Ther. 2020, 13, 189–201.
  43. De Thé, H.; Pandolfi, P.P.; Chen, Z. Acute Promyelocytic Leukemia: A Paradigm for Oncoprotein-Targeted Cure. Cancer Cell 2017, 32, 552–560.
  44. Moosavi, M.A.; Djavaheri-Mergny, M. Autophagy: New Insights into Mechanisms of Action and Resistance of Treatment in Acute Promyelocytic leukemia. Int. J. Mol. Sci. 2019, 20, 3559.
  45. Li, T.; Ma, R.; Zhang, Y.; Mo, H.; Yang, X.; Hu, S.; Wang, L.; Novakovic, V.A.; Chen, H.; Kou, J.; et al. Arsenic trioxide promoting ETosis in acute promyelocytic leukemia through mTOR-regulated autophagy. Cell Death Dis. 2018, 9, 75.
  46. Meyer, C.; Kowarz, E.; Hofmann, J.; Renneville, A.; Zuna, J.; Trka, J.; Ben Abdelali, R.; Macintyre, E.; De Braekeleer, E.; De Braekeleer, M.; et al. New insights to the MLL recombinome of acute leukemias. Leukemia 2009, 23, 1490–1499.
  47. Manara, E.; Baron, E.; Tregnago, C.; Aveic, S.; Bisio, V.; Bresolin, S.; Masetti, R.; Locatelli, F.; Basso, G.; Pigazzi, M. MLL-AF6 fusion oncogene sequesters AF6 into the nucleus to trigger RAS activation in myeloid leukemia. Blood 2014, 124, 263–272.
  48. Liu, Q.; Chen, L.; Atkinson, J.M.; Claxton, D.F.; Wang, H.G. Atg5-dependent autophagy contributes to the development of acute myeloid leukemia in an MLL-AF9-driven mouse model. Cell Death Dis. 2016, 7, 2361.
  49. Müller, A.M.; Duque, J.; Shizuru, J.A.; Lübbert, M. Complementing mutations in core binding factor leukemias: From mouse models to clinical applications. Oncogene 2008, 27, 5759–5773.
  50. Man, N.; Tan, Y.; Sun, X.J.; Liu, F.; Cheng, G.; Greenblatt, S.M.; Martinez, C.; Karl, D.L.; Ando, K.; Sun, M.; et al. Caspase-3 controls AML1-ETO-driven leukemogenesis via autophagy modulation in a ULK1-dependent manner. Blood 2017, 129, 2782–2792.
  51. Sakaguchi, M.; Yamaguchi, H.; Najima, Y.; Usuki, K.; Ueki, T.; Oh, I.; Mori, S.; Kawata, E.; Uoshima, N.; Kobayashi, Y.; et al. Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia. Blood Adv. 2018, 2, 2744–2754.
  52. Heydt, Q.; Larrue, C.; Saland, E.; Bertoli, S.; Sarry, J.E.; Besson, A.; Manenti, S.; Joffre, C.; Mansat-De Mas, V. Oncogenic FLT3-ITD supports autophagy via ATF4 in acute myeloid leukemia. Oncogene 2018, 37, 787–797.
  53. Xu, D.; Chen, Y.; Yang, Y.; Yin, Z.; Huang, C.; Wang, Q.; Jiang, L.; Jiang, X.; Yin, C.; Liu, Q.; et al. Autophagy activation mediates resistance to FLT3 inhibitors in acute myeloid leukemia with FLT3-ITD mutation. J. Transl. Med. 2022, 20, 300.
  54. Qiu, S.; Kumar, H.; Yan, C.; Li, H.; Paterson, A.J.; Anderson, N.R.; He, J.; Yang, J.; Xie, M.; Crossman, D.K.; et al. Autophagy inhibition impairs leukemia stem cell function in FLT3-ITD AML but has antagonistic interactions with tyrosine kinase inhibition. Leukemia 2022, 36, 2621–2633.
  55. Rudat, S.; Pfaus, A.; Cheng, Y.Y.; Holtmann, J.; Ellegast, J.M.; Bühler, C.; Marcantonio, D.D.; Martinez, E.; Göllner, S.; Wickenhauser, C.; et al. RET-mediated autophagy suppression as targetable co-dependence in acute myeloid leukemia. Leukemia 2018, 32, 2189–2202.
  56. Dany, M.; Gencer, S.; Nganga, R.; Thomas, R.J.; Oleinik, N.; Baron, K.D.; Szulc, Z.M.; Ruvolo, P.; Kornblau, S.; Andreeff, M.; et al. Targeting FLT3-ITD signaling mediates ceramide-dependent mitophagy and attenuates drug resistance in AML. Blood 2016, 128, 1944–1958.
  57. Zalpoor, H.; Bakhtiyari, M.; Akbari, A.; Aziziyan, F.; Shapourian, H.; Liaghat, M.; Zare-Badie, Z.; Yahyazadeh, S.; Tarhriz, V.; Ganjalikhani-Hakemi, M. Potential role of autophagy induced by FLT3-ITD and acid ceramidase in acute myeloid leukemia chemo-resistance: New insights. Cell Commun. Signal. 2022, 20, 172.
  58. Ishikawa, Y.; Kawashima, N.; Atsuta, Y.; Sugiura, I.; Sawa, M.; Dobashi, N.; Yokoyama, H.; Doki, N.; Tomita, A.; Kiguchi, T.; et al. Prospective evaluation of prognostic impact of KIT mutations on acute myeloid leukemia with RUNX1-RUNX1T1 and CBFB-MYH11. Blood Adv. 2020, 4, 66–75.
  59. Ayatollahi, H.; Shajiei, A.; Sadeghian, M.H.; Sheikhi, M.; Yazdandoust, E.; Ghazanfarpour, M.; Shams, S.F.; Shakeri, S. Prognostic Importance of C-KIT Mutations in Core Binding Factor Acute Myeloid Leukemia: A Systematic Review. Hematol. Oncol. Stem Cell Ther. 2017, 10, 1–7.
  60. Larrue, C.; Heydt, Q.; Saland, E.; Boutzen, H.; Kaoma, T.; Sarry, J.E.; Joffre, C.; Récher, C. Oncogenic KIT mutations induce STAT3-dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis 2019, 8, 39.
  61. Xu, J.; Zheng, J.; Fu, X.; Wu, W.; Tao, L.; Li, D.; Lin, D. Inhibition of N822K T>A mutation-induced constitutive c-KIT activation in AML cells triggers apoptotic and autophagic pathways leading to death. Int. J. Med. Sci. 2019, 16, 757–765.
  62. Chen, Y.; Hu, J. Nucleophosmin1 (NPM1) abnormality in hematologic malignancies, and therapeutic targeting of mutant NPM1 in acute myeloid leukemia. Ther. Adv. Hematol. 2020, 11, 2040620719899818.
  63. Zou, Q.; Tan, S.; Yang, Z.; Zhan, Q.; Jin, H.; Xian, J.; Zhang, S.; Yang, L.; Wang, L.; Zhang, L. NPM1 Mutant Mediated PML Delocalization and Stabilization Enhances Autophagy and Cell Survival in Leukemic Cells. Theranostics 2017, 7, 2289–2304.
  64. Wang, L.; Yang, L.; Yang, Z.; Tang, Y.; Tao, Y.; Zhan, Q.; Lei, L.; Jing, Y.; Jiang, X.; Jin, H.; et al. Glycolytic Enzyme PKM2 Mediates Autophagic Activation to Promote Cell Survival in NPM1-Mutated Leukemia. Int. J. Biol. Sci. 2019, 15, 882–894.
  65. Tang, Y.; Tao, Y.; Wang, L.; Yang, L.; Jing, Y.; Jiang, X.; Lei, L.; Yang, Z.; Wang, X.; Peng, M.; et al. NPM1 mutant maintains ULK1 protein stability via TRAF6-dependent ubiquitination to promote autophagic cell survival in leukemia. FASEB J. 2021, 35, 21192.
  66. Wang, Y.; Pan, C.; Zhang, X.; Zhao, A.; Dong, Y. Mutant NPM1 maintains RASGRP3 protein stability via interaction with MID1 to promote acute myeloid leukemia cell proliferation and autophagy. J. Leukoc. Biol. 2023, 113, 504–517.
  67. Ok, C.Y.; Patel, K.P.; Garcia-Manero, G.; Routbort, M.J.; Peng, J.; Tang, G.; Goswami, M.; Young, K.H.; Singh, R.; Medeiros, L.J.; et al. TP53 mutation characteristics in therapy-related myelodysplastic syndromes and acute myeloid leukemia is similar to de novo diseases. J. Hematol. Oncol. 2015, 8, 45.
  68. Prokocimer, M.; Molchadsky, A.; Rotter, V. Dysfunctional diversity of p53 proteins in adult acute myeloid leukemia: Projections on diagnostic workup and therapy. Blood 2017, 130, 699–712.
  69. Folkerts, H.; Hilgendorf, S.; Wierenga, A.T.J.; Jaques, J.; Mulder, A.B.; Coffer, P.J.; Schuringa, J.J.; Vellenga, E. Inhibition of autophagy as a treatment strategy for p53 wild-type acute myeloid leukemia. Cell Death Dis. 2017, 8, 2927.
  70. Gomez-Puerto, M.C.; Folkerts, H.; Wierenga, A.T.; Schepers, K.; Schuringa, J.J.; Coffer, P.J.; Vellenga, E. Autophagy proteins ATG5 and ATG7 are essential for the maintenance of human CD34 (+) hematopoietic stem-progenitor cells. Stem Cells 2016, 34, 1651–1663.
  71. Allende-Vega, N.; Villalba, M. Metabolic stress controls mutant p53 R248Q stability in acute myeloid leukemia cells. Sci. Rep. 2019, 9, 5637.
  72. Maiuri, M.C.; Galluzzi, L.; Morselli, E.; Kepp, O.; Malik, S.A.; Kroemer, G. Autophagy regulation by p53. Curr. Opin. Cell Biol. 2010, 22, 181–185.
  73. Crighton, D.; Wilkinson, S.; O’Prey, J.; Syed, N.; Smith, P.; Harrison, P.R.; Gasco, M.; Garrone, O.; Crook, T.; Ryan, K.M. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006, 126, 121–134.
  74. Feng, Z.; Zhang, H.; Levine, A.J.; Jin, S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc. Natl. Acad. Sci. USA 2005, 102, 8204–8209.
  75. Vu, T.T.; Stölzel, F.; Wang, K.W.; Röllig, C.; Tursky, M.L.; Molloy, T.J.; Ma, D.D. miR-10a as a therapeutic target and predictive biomarker for MDM2 inhibition in acute myeloid leukemia. Leukemia 2021, 35, 1933–1948.
  76. Burgess, A.; Chia, K.M.; Haupt, S.; Thomas, D.; Haupt, Y.; Lim, E. Clinical Overview of MDM2/X-Targeted Therapies. Front. Oncol. 2016, 6, 7.
  77. Nakazawa, S.; Sakata, K.I.; Liang, S.; Yoshikawa, K.; Iizasa, H.; Tada, M.; Hamada, J.I.; Kashiwazaki, H.; Kitagawa, Y.; Yamazaki, Y. Dominant-negative p53 mutant R248Q increases the motile and invasive activities of oral squamous cell carcinoma cells. BioMed. Res. 2019, 40, 37–49.
  78. Yao, Y.; Chai, X.; Gong, C.; Zou, L. WT1 inhibits AML cell proliferation in a p53-dependent manner. Cell Cycle 2021, 20, 1552–1560.
  79. Wilson, E.R.; Helton, N.M.; Heath, S.E.; Fulton, R.S.; Payton, J.E.; Welch, J.S.; Walter, M.J.; Westervelt, P.; DiPersio, J.F.; Link, D.C.; et al. Focal disruption of DNA methylation dynamics at enhancers in IDH-mutant AML cells. Leukemia 2022, 36, 935–945.
  80. Tulstrup, M.; Soerensen, M.; Hansen, J.W.; Gillberg, L.; Needhamsen, M.; Kaastrup, K.; Helin, K.; Christensen, K.; Weischenfeldt, J.; Grønbæk, K. TET2 mutations are associated with hypermethylation at key regulatory enhancers in normal and malignant hematopoiesis. Nat. Commun. 2021, 12, 6061.
  81. Chaudry, S.F.; Chevassut, T.J.T. Epigenetic Guardian: A Review of the DNA Methyltransferase DNMT3A in Acute Myeloid Leukaemia and Clonal Haematopoiesis. BioMed. Res. Int. 2017, 2017, 5473197.
  82. Nawrocki, S.T.; Han, Y.; Visconte, V.; Przychodzen, B.; Espitia, C.M.; Phillips, J.; Anwer, F.; Advani, A.; Carraway, H.E.; Kelly, K.R.; et al. The novel autophagy inhibitor ROC-325 augments the antileukemic activity of azacitidine. Leukemia 2019, 33, 2971–2974.
  83. Dai, Y.-J.; Hu, F.; He, S.-Y.; Wang, Y.-Y. Epigenetic landscape analysis of lncRNAs in acute myeloid leukemia with DNMT3A mutations. Ann. Transl. Med. 2020, 8, 318.
More
This entry is offline, you can click here to edit this entry!
Video Production Service