Next Article in Journal
Docking and Selectivity Studies of Covalently Bound Janus Kinase 3 Inhibitors
Next Article in Special Issue
Attenuation of Polycyclic Aromatic Hydrocarbon (PAH)-Induced Carcinogenesis and Tumorigenesis by Omega-3 Fatty Acids in Mice In Vivo
Previous Article in Journal
Fabrication and Characterization of Functional Biobased Membranes from Postconsumer Cotton Fabrics and Palm Waste for the Removal of Dyes
Previous Article in Special Issue
Effects of CYP3A43 Expression on Cell Proliferation and Migration of Lung Adenocarcinoma and Its Clinical Significance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Updated Overview of the Role of CYP450 during Xenobiotic Metabolization in Regulating the Acute Myeloid Leukemia Microenvironment

1
Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile
2
Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile
3
Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
4
School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK
5
Núcleo Científico y Tecnológico en Biorecursos (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(7), 6031; https://doi.org/10.3390/ijms24076031
Submission received: 15 February 2023 / Revised: 8 March 2023 / Accepted: 16 March 2023 / Published: 23 March 2023
(This article belongs to the Special Issue Cytochrome P450 (CYP) 2.0)

Abstract

:
Oxidative stress is associated with several acute and chronic disorders, including hematological malignancies such as acute myeloid leukemia, the most prevalent acute leukemia in adults. Xenobiotics are usually harmless compounds that may be detrimental, such as pharmaceuticals, environmental pollutants, cosmetics, and even food additives. The storage of xenobiotics can serve as a defense mechanism or a means of bioaccumulation, leading to adverse effects. During the absorption, metabolism, and cellular excretion of xenobiotics, three steps may be distinguished: (i) inflow by transporter enzymes, (ii) phases I and II, and (iii) phase III. Phase I enzymes, such as those in the cytochrome P450 superfamily, catalyze the conversion of xenobiotics into more polar compounds, contributing to an elevated acute myeloid leukemia risk. Furthermore, genetic polymorphism influences the variability and susceptibility of related myeloid neoplasms, infant leukemias associated with mixed-lineage leukemia (MLL) gene rearrangements, and a subset of de novo acute myeloid leukemia. Recent research has shown a sustained interest in determining the regulators of cytochrome P450, family 2, subfamily E, member 1 (CYP2E1) expression and activity as an emerging field that requires further investigation in acute myeloid leukemia evolution. Therefore, this review suggests that CYP2E1 and its mutations can be a therapeutic or diagnostic target in acute myeloid leukemia.

1. Background

Acute myeloid leukemia is a cancerous condition that affects hemopoietic stem cells or progenitors and is defined by the stopping of myeloid lineage development and abnormal proliferation [1]. The most prevalent acute leukemia in adults is acute myeloid leukemia, which has a wide range of genetic variations. In 2021, more than 20,000 new cases of acute myeloid leukemia were expected in the US [2]. Traditionally, acute myeloid leukemia has been categorized based on immunophenotype and morphology. However, genetic abnormalities, such as chromosomal translocations and transcription factor involvement, must be considered in acute myeloid leukemia diagnostic algorithms [3,4]. These factors led to the classification of acute myeloid leukemia into six groups [3]: myeloid proliferations linked to Down syndrome, myeloid sarcoma, recurring genetic abnormalities, therapy-related myeloid neoplasms, and acute myeloid leukemia with myelodysplasia-related alterations.
Oxidative stress is implicated in several acute and chronic diseases, including hematological malignancies such as acute myeloid leukemia, which is the most common acute leukemia in adults, with an increasing incidence with age and high relapse rates [1]. Despite current advancements in the treatment of acute myeloid leukemia, refractory disease remains prevalent, with disease relapse being the major cause of treatment failure [5]. The current acute myeloid leukemia management guidelines largely rely on high-dose chemotherapy with cytarabine- and anthracycline-based regimes and allogeneic hematopoietic stem cell transplantation [6].
Cytarabine with anthracycline induction therapy is the standard of care for acute myeloid leukemia. The most commonly used regimen includes anthracycline daunorubicin (45–90 mg/m2) on days 1–3 and cytarabine (100–200 mg/m2) in continuous infusion on days 1–7 [7]. In this regard, systematic reviews have assessed the efficiency of cytarabine and daunorubicin regimens and determined that 62.1% of patients achieve full remission. In addition, it has been noted that, when cytarabine or daunorubicin doses are raised throughout treatment, the rate of full remission increases [8]. However, the dose intensification of cytarabine, such as doses of 1–2 g/m2/12 h [8,9] or the extension of the treatment duration to 10 days [10], did not result in improved results and was associated with increased toxicity [8,9].
Even in individuals who receive doses of myeloablative chemotherapy or radiation given for hematopoietic stem cell transplantation, the malignant stem cell might undergo further mutations [11]. The graft-versus-leukemia effect, which results from the elimination of these stem cells by T- and natural killer (NK) lymphocytes of the donor, is one of the advantages of allogeneic hematopoietic stem cell transplantation [12,13]. However, the presence of comorbidities greatly compromises the results of hematopoietic stem cell transplantation, as a result of which this therapeutic option is not recommended for these patients [14].
Acute myeloid leukemia treatment is becoming more customized based on the molecular characteristics of the disease because of better knowledge of its pathophysiology [5,15]. This allows for greater risk assessment and more personalized drugs. Seven of the nine novel drugs approved for the treatment of relapsed or refractory acute myeloid leukemia (R/R AML)—azacitidine, enasidenib, glasdegib, gemtuzumab ozogamicin, gilteritinib, low-dose cytarabine, midostaurin, venetoclax and, venetoclax plus low-dose cytarabine—act via a molecularly defined target, as opposed to standard cytotoxic chemotherapy [16,17,18,19,20,21,22]. There are still certain unmet needs, despite the development and popularity of these new medicines for the treatment of acute myeloid leukemia. These include the fact that many individuals with R/R AML who do not currently have targetable mutations still have few therapy choices. Aside from the limited percentage of patients who continue with an allogeneic hematopoietic cell transplant, none of the recently approved medicines are curative [23]. Therefore, this article reviews current acute myeloid leukemia pathogenesis and novel therapies. Figure 1 illustrates the flow chart for the study selection process.

2. Pathophysiology of Acute Myeloid Leukemia

2.1. Cytogenetic Abnormalities

Acute myeloid leukemia is characterized by mutations in hematopoiesis-related genes [24]. Ineffective erythropoiesis and bone marrow failure are caused by these mutations, which cause a clonal increase in undifferentiated myeloid progenitors (blasts) in the peripheral blood and bone marrow. Recent research has suggested that it could result from several recurring genetic changes in hematopoietic stem cells that accumulate over time [15,25,26,27,28,29,30]. Acute myeloid leukemia often develops from scratch in a previously healthy person. Although the precise source of genetic abnormalities is unknown, a few known risk factors include smoking, chemotherapy, and radiation exposure [31]. Aplastic anemia, paroxysmal nocturnal hemoglobinuria, myelodysplastic syndrome, and myeloproliferative diseases can all develop into acute myeloid leukemia [32,33].
Genetic mutations that have familial causes should also be considered (Table 1). The most prevalent mutational subset in acute myeloid leukemia is type 1 mutations, which are present in about two-thirds of patients and result in abnormal activation and proliferation of cellular signaling pathways (e.g., FMS-like tyrosine kinase 3 (FLT3); Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS); NRAS Proto-Oncogene, GTPase (NRAS); Tyrosine-protein phosphatase non-receptor type 11 (PTPN11); neurofibromin 1 (NF1); and KIT proto-oncogene, receptor tyrosine kinase (KIT)). It is interesting to note that mutations in this class are usually found in subclonal cellular fractions, indicating that they are frequently late clonal events in the development of illness [15].
Somatic mutations within key epigenetic regulators are identified in >50% of acute myeloid leukemia, and are now recognized as a key, and often an inciting, component of leukemogenesis [15]. It is of interest that age-related clonal hematopoiesis, identified in >10% of individuals over age 65, is predominantly defined by the clonal outgrowth of preleukemic clones harboring mutations in one of the genes within this epigenetic class [34,35].
Along with FLT3 and DNA methyltransferase 3 alpha (DNMT3A; https://www.ncbi.nlm.nih.gov/gene/1788; accessed on 14 January 2023), nucleophosmin 1 (NPM1) is one of the three most frequent driver mutations in acute myeloid leukemia. The regulation of pathways for cell proliferation, differentiation, adhesion, and death by receptor tyrosine kinase (RTKs) signaling pathways in acute myeloid leukemia is crucial for the onset and spread of malignancy. Around 20 separate subfamilies make up the RTKs, which include class III and TYRO3, AXL, and MERTK (TAM) family RTKs [53]. Class III RTKs, which include c-Kit, CSF1R, FLT3, and platelet-derived growth factor receptors (PDGFR), have been discovered to have a major effect on leukemogenesis and transformation into acute myeloid leukemia. Class III RTKs have been linked to aberrant activation that promotes proliferation in leukemia. Particularly, FLT3 expression and c-Kit mutations are critical for acute myeloid leukemia. Both RTKs have been crucial targets in the development of antileukemic therapies, since they are both associated with worse prognoses. TYRO3, AXL, and MERTK are members of the TAM family of RTKs. These are essential for platelet activation and stabilization, for the normal hematopoiesis of certain innate immune cells, and have been linked to erythropoiesis [37]. TAM RTKs are critical for normal hematopoietic development, but they can activate pathways for proliferation and survival in cancer cells, particularly in acute leukemia [54]. TAM RTKs, particularly AXL and MERTK, have been linked to hematologic malignancies and have grown in interest as potential targets for creating novel treatments [54,55,56,57,58].
In addition, compared to the genomes of other cancers, acute myeloid leukemia genomes are typically less mutated [59], with similar distributions of mutations before and following relapse [60,61]. Of these mutations, many frequently occur in genes involved in DNA methylation and epigenetic regulation, such as DNMT3A, TET1/2, and IDH1/2 [62]. Hassan et al. [63] point to a greater need for understanding acute myeloid leukemia through a non-genetic lens, focusing on DNA methylation and other epigenetic modalities. They also suggest relative independence between the progression of acute myeloid leukemia and the disease’s strictly genetic landscape.
IDH1 gene mutations are present in around 6–10% of individuals with acute myeloid leukemia [64]. Given its capacity to create cytoplasmic NADPH and glucose sensing, IDH1 is implicated in controlling cellular metabolism, particularly lipid metabolism [65,66]. Isocitrate is oxidized to α-ketoglutarate by wild-type isocitrate dehydrogenases [67]. It has been hypothesized that the IDH1 Arg132 mutation alters the way the enzyme functions, causing α-ketoglutarate to be converted to R(—)-2-hydroxyglutarate [68]. This excess of R(—)-2-hydroxyglutarate causes cellular proliferation to rise and cellular differentiation to be compromised [69].
Nucleophosmin 1 mutations, occurring almost exclusively within exon 12 of the gene, occur in approximately one-third of adults with acute myeloid leukemia, and in more than 50% of NK-AML. The NPM1 gene encodes for the nuclear chaperone protein NPM, which shuttles between the nucleus and cytoplasm and plays a role in diverse cellular functions, including protein formation, ribosome biogenesis, DNA replication, and the cell cycle. NPM1 mutations are typically stable throughout the disease course, are identified in nearly all leukemic cells, and impart a distinct expression profile [70]. NPM1 mutations in the setting of mutant DNMT3A, particularly in the setting of FLT3 internal tandem duplication (FLT3-ITD), confer a markedly poor prognosis [15,71].
Approximately 20% to 25% of adults with acute myeloid leukemia have mutations involving the myeloid transcription factors Runt-related transcription factor 1 (RUNX1), CEBPA, and/or GATA binding protein 2 (GATA2) [24]. In this sense, RUNX1 is known to act as a direct transcriptional activator of several proteins important for platelet function and as a transcriptional repressor of others, including MYH10 [72,73,74,75] and ANKRD26 [76]. In addition, CEBPA encodes a master hematopoietic transcription factor that acts as a critical regulator of granulocyte and monocyte differentiation [77], while GATA2 encodes a zinc finger transcription factor critical for normal hematopoiesis [78,79] and lymphatic vascular development [80,81].
Tumor protein p53 (TP53) is a key tumor suppressor with highly variable functions related to the maintenance of genomic stability, including regulation of cellular senescence, apoptosis, metabolism, and DNA repair. Although uncommon in de novo acute myeloid leukemia, TP53 mutations occur in ~15% of therapy-related acute myeloid leukemia or acute myeloid leukemia with myelodysplastic syndrome-related changes, and are predominantly associated with complex cytogenetics, advanced age, chemotherapy resistance, and poor survival [47,48]. Irrespective of age or treatment modality, TP53 mutations in acute myeloid leukemia portend lower response rates and inferior outcomes compared with TP53 wild-type acute myeloid leukemia patients [49].
Frequently mutated in myelodysplastic syndrome and myeloproliferative neoplasms, mutations in splicing factors (SF3B1, SRSF2, U2AF1, and ZRSR2) are identified in ~10% of patients with acute myeloid leukemia and are associated with older age, less proliferative disease, poor rates of response to standard treatment, and decreased survival. Spliceosome mutations are postulated to promote malignancy through the missplicing of various genes involved in epigenetic regulation, transcription, and genome integrity [50].
The structural maintenance of chromosomes (SMC3 and SMC1A), RAD cohesin complex component (RAD21), and cohesin subunit SA (STAG1/STAG2) make up the four core elements of cohesin with a ring shape. Cohesin helps several other subunits, such as NIPBL, MAU2, WAPL, PDS5A, PDS5B, and sororin, to form cohesion during the cell cycle [82,83,84,85]. Consequently, the ring-shaped cohesin controls the sister chromatids’ separation, DNA replication, and repair of the broken double-strand DNA during the advancement of the cell cycle [86,87,88,89,90]. To control chromatin structure and gene expression, the cohesin complex can also interact with the transcriptional repressor CTCF, promoters, mediators, enhancers, initiation and elongation forms of RNA polymerase II (RNAPII), or transcription factors (TFs) [87,91,92,93,94,95].
Acute myeloid leukemia prognosis is exceedingly variable and unpredictable. It can be caused by molecular changes, chromosomal translocations, or genetic mutations. Genetic mutations have been shown to occur in around 97% of cases. Table 2 shows an updated classification of acute myeloid leukemia based on the National Comprehensive Cancer Network’s cytogenetic and molecular criteria [96,97]. The following are examples of cytogenetic subsets: (1) chromosomal translocations [t(15;17)(q22,q21)] and core binding factor acute myeloid leukemia (CBF-AML), both of which are cytogenetic/molecular subgroups of inversion 16 [inv16(p13;q22)] or t(16;16)(p13;q22); (2) individuals with cytogenetically normal acute myeloid leukemia (CN-AML) who have monosomy 5 or 7 or t(9 or 11) have a low risk (40–50% of patients) [85,86,87,88,89,90]; (3) individuals with t(6;9), inv (3), or 11q changes (11q23 translocations) [85,86,87,88,89,90]; (4) and those with other karyotypes have been demonstrated to have a higher risk of treatment failure and mortality [98,99,100,101,102,103]. Furthermore, people with translocations involving the MECOM (myelodysplastic syndrome-1 and ecotropic viral integration site 1 (EVI1) complex locus) gene on chromosome 3q26.2 have a very poor prognosis [104,105].

2.2. Mutations

Modern molecular technologies have permitted the identification of a wide spectrum of genetic disorders. Six genes have already been incorporated into the European Leukemia Net risk categories [5], including FLT3, NPM1, CCAAT/enhancer binding protein α (CEBPA), RUNX1, additional sexcombs-like 1 (ASXL1), and TP53 [5]. Other recurrent gene mutations in acute myeloid leukemia patients have been discovered [106,107,108,109,110,111]. Furthermore, research has been undertaken on the essential roles played by recurrent gene mutations in the pathogenesis of acute myeloid leukemia, as well as the development of medicines that precisely target gene alterations [112,113,114,115,116,117,118]. Preleukemic cell detection, particularly in acute myeloid leukemia patients with mutant DNMT3A and TET2 genes, has been linked to leukemia genesis [119,120]. Mutations in the DNMT3A and TET2 genes are common in patients with clonal hematopoiesis of undetermined potential [34,35,121,122]; these mutations may serve as markers for the identification of preleukemia cells [119,120]. A summary of the pathophysiology of acute myeloid leukemia is shown in Figure 2.

3. Acute Myeloid Leukemia Microenvironment

Currently, niches are thought of as microenvironments that mix non-hematopoietic cells with the structure of the bone marrow to encourage hematopoietic stem cell self-renewal and differentiation by offering beneficial and crucial components [123,124]. The non-hematopoietic progenitors known as mesenchymal stromal cells are an essential component of the bone marrow niche.
Due to their ability to directly govern the development and differentiation of hematopoietic stem cells and their ability to release a range of soluble growth factors and cytokines, mesenchymal stromal cells really play an important role in immunomodulation [125,126,127]. Mesenchymal stromal cells express significant hematopoietic factors, such as stem cell factor and stromal cell-derived factor 1. Additionally, they give off trophic factors that control the immune system and turn on the body’s own stem cells to repair damaged tissues [128,129,130,131].
The ability of allogeneic stem cells to differentiate into various stromal marrow components, such as pericytes, bone marrow stromal cells, myofibroblasts, osteoblasts, osteocytes, and endothelial cells, is also essential for allogeneic stem cell transplantation to be successful [132,133]. Bone marrow mesenchymal stromal cells are commonly recognized as significant contributors to tumor genesis, recurrence, and treatment resistance in the context of acute myeloid leukemia due to their ability to provide leukemic blasts with survival and anti-apoptotic signals [134,135]. According to several studies, co-culturing acute myeloid leukemia blasts with stromal or mesenchymal stem cells has been linked to increased in vitro tumor cell viability [136,137], aberrant phenotypic expression [138,139,140], and decreased chemoresistance [134,141,142].
Additionally, animal models of myeloproliferative neoplasms [143,144,145,146,147], myelodysplastic syndrome [148,149], and acute myeloid leukemia [150,151] have been used to illustrate niche-induced disease onset in vivo. Ex vivo expanded mesenchymal stromal niche cells from myelodysplastic syndrome, and acute myeloid leukemia patients, have been shown to have a variety of functional and molecular alterations, including chromosomal aberrations [152,153], transcriptional changes [154], and epigenetic changes [155], as well as functional changes in their ability to differentiate and hematopoietic stem cell-supportive behaviors [156,157].
By using array comparative hybridization and transcriptome profiling, it was also discovered that CD271+ mesenchymal stromal cells directly obtained from myelodysplastic syndrome patients had genetic and transcriptomic changes [158,159,160]. The down-regulation of dicer 1, ribonuclease III (DICER), and SBDS ribosome maturation factor (SBDS) [149,157,160] and the activation of beta-catenin in osteoblastic cells are two processes that have been proven to start malignant transformation in mice and have also been documented in patient-derived mesenchymal cells [150]. More recently, it was discovered that mice with nestin+ cells developed juvenile myelomonocytic leukemia when a PTPN11 mutant was expressed [147].
Despite the above-mentioned, few studies have used environmental sampling [161,162] and biomarkers such as malondialdehyde, total antioxidant capacity, thiobarbituric acid reactive substances, protein carbonyl, and lipid hydroperoxide evaluation [163,164,165,166] to better characterize chemical exposures, which could provide powerful insights to better understand the continuum between routes of exposure, chemical body burden, and risk of acute myeloid leukemia. Several studies have found that gene polymorphisms in xenobiotic pathways, such as cytochrome P450 family 2 subfamily E member 1 (CYP2E1), glutathione S-transferase Mu 1 (GSTM1), NAD(P)H: quinone oxidoreductase (NQO1), N-acetyltransferase 2 (NAT2), and multidrug resistance protein 1 (MDR1), influence leukemia risk alone or in combination with chemical exposure [166].

4. Mechanisms of Liver and Bone Marrow CYP2E1 Induction, Activity, and Degradation

CYPs are a family of heme-containing proteins that play an essential function in the metabolism of a wide variety of xenobiotics [167]. CYP450 proteins have an essential function in tumorigenesis by activating or deactivating carcinogens, which influences tumour start and progression [168]. Recent research has demonstrated that CYP2E1 is not only markedly elevated in the liver, but also expressed in bone marrow [169]. Drugs and plants (isoniazid, Salvia miltiorrhiza, Schisandra chinensis), pollutants (phenylamine), food additives (coffee and cocoa polyphenols), and industrial material and environmental contaminants (benzene) can stimulate CYP2E1 activity [170,171].
Benzene has been recognized as an environmental contaminant that can generate hematotoxicity and leukemogenicity [172,173,174]. Many studies have hypothesized that the conversion of benzene to reactive metabolites by hepatic enzymes, specifically CYP2E1, is a precondition for the cyto- and genotoxic effects of benzene exposure [175,176,177]. Hydroquinone, phenol, trans–trans muconic acid, and catechol are the principal benzene metabolites [178]. These phenolic metabolites work synergistically to increase benzene toxicity [179,180,181]. In terms of the mechanism of its toxicity and carcinogenicity, this process of multimetabolite genotoxicity is another distinguishing feature of benzene compared to other compounds. Thereafter, benzene metabolites undergo further activation by myeloperoxidase, which is abundant in bone marrow tissue. Inducing not only hemopoietic cellular damage [182,183,184] but also bone marrow stromal cell dysfunction [185].
Among the biochemical mechanisms abnormally elevated in malignancies, including acute myeloid leukemia, is the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin route (PI3K-Akt-mTOR pathway) [186]. PI3K enzymes have crucial functions in cell metabolism, proliferation, and survival [187,188]. PI3K activation triggers pathways of signaling that stimulate cell differentiation, metabolism, migration, proliferation, and survival [189]. Ethanol-induced suppression of Akt phosphorylation and pharmacological modulation of Akt can result in CYP2E1-induced hepatic oxidative stress, which could be a viable treatment for ethanol-induced fatty liver [190,191]. Therefore, the effect of alcohol on CYP2E1 induction and the involvement of PI3K/Akt in guarding against the cytotoxicity of CYP2E1 suggest that CYP2E1 overexpression may reduce the expression of critical proteins in the PI3K signaling pathway [190,191].

5. Mechanisms of CYP-Mediated Carcinogenesis and the Roles of Their Isoforms

CYP450 enzymes serve a critical role in preventing oxidative damage; they metabolize several exogenous and endogenous genotoxic chemicals, such as hydrogen peroxide, by inserting an oxygen atom into the substrate [192].
Specific single-nucleotide polymorphisms at the CYP450 loci (CYP2D6, CYP1A1, CYP3A5, and CYP2E1) may very well be categorized as risk factors for numerous kinds of cancers, due to the inactivation of enzymatic activity [193,194,195,196,197,198,199,200].
At the CYP2B6 gene locus, the G516T polymorphism has been recognized as a nonsense polymorphism that decreases the activity of the protein complex. Hence, people who have the T allele (TT) have a reduced enzymatic activity than people who have the wild-type G allele (GG), but individuals holding the genotype GT exhibit intermediate activity [201].
Ethnicity causes CYP3A enzyme activity to vary up to 50-fold. Most genetic variants in the CYP3A4 gene result in lower enzyme activity. Single-nucleotide polymorphisms in the CYP3A4 promoter region (A290G) (i.e., CYP3A4*1B) have been implicated as a possible cause of this variability; hence, it may be considered a risk factor for cancer. However, the implications of these single-nucleotide polymorphisms are not well-understood [202,203,204,205]. Hence, diverse allelic expressions of the CYP450 gene have distinct pathogenic effects and prognostic characteristics in various hematological cancers. Figure 3 shows the main processes involved in ROS production during acute myeloid leukemia.

6. Xenobiotics and CYP450 Activation

The broad category of xenobiotics includes substances that are generally safe but may be harmful, such as medications, environmental toxins, cosmetics, and even elements included in our diet, such as food additives [206,207,208,209]. Storage of xenobiotics can serve as a defense mechanism or a way for bioaccumulation to result in harmful consequences. The physiologic connection between the storage depot and the target tissues for a particular toxin determines this possible hazardous pathway [206,208].
Xenobiotic metabolism increases their water solubility, thus enhancing their elimination from the body [210]. When xenobiotics are consumed orally, they go through the upper gastrointestinal tract and, if they are absorbed, are then transferred to the liver via the hepatic portal vein. The liver chemically converts both endogenous and exogenous substances, utilizing the CYP450 family of enzymes [211,212]. In fact, in the absorption, metabolism, and cellular excretion of xenobiotics, three steps may be distinguished: (i) inflow by transporter enzymes, (ii) phases I and II, mediated by drug-metabolizing enzymes, and (iii) phase III, the excretion mediated mainly by transporter enzymes [195,206,213]. Non-metabolized and unexcreted xenobiotics build up in the body and can cause chronic illnesses and inflammation [214].
The inflow of xenobiotics is mediated by sodium taurocholate cotransporting polypeptide, organic anion transporting polypeptides, and organic anion transporters [195]. Phase I enzymes, such as those in Cytochrome P450 (CYP450), flavin-containing monooxygenases (FMOs), monoamine oxidases (MAOs), and xanthine oxidase/aldehyde oxidase (XO/AO) superfamily, catalyze the conversion of predominantly lipophilic xenobiotics into more polar compounds by oxidation, reduction, or hydrolysis [115,116,117,167]. Phase I processes that introduce polar groups create the sites needed for conjugation reactions, which are carried out by Phase II enzymes [118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220]. Phase I metabolites are frequently conjugated with glucuronic acid, glutathione, sulfate, amino acids, or methyl or acetyl groups, even though Phase II enzymes can directly operate on the parent substance [221]. The addition of these large anionic groups, which may detoxify reactive electrophiles (either parent chemicals or Phase I metabolites), results in Phase II metabolites with enhanced hydrophilicity and molecular weight, which cannot penetrate the phospholipid membrane barrier [115,172,221,222,223]. The anionic groups of phase III xenobiotic transporters operate as affinity tags for a variety of membrane carriers belonging to two major clusters: ATP binding cassette transporters, including the multidrug resistance protein family, and solute carrier transporters [224,225,226].
There are significant inter- and intra-individual differences in the ability to metabolize, detoxify, and expel xenobiotics. These are genetic, epigenetic, environmental, and physiological pathophysiological in nature, and they change during life [215,225,227,228,229,230]. Most xenobiotics are detoxified and eliminated via a complicated network of numerous enzymes and pathways. The interaction of xenobiotic local or cellular concentration, specific enzyme affinity, tissue-specific enzyme expression, stability, and cofactor availability frequently determines which metabolic processes prevail at any given time [167,206,231].

7. CYP2E1 Expression and Regulation in Acute Myeloid Leukemia

CYP2E1 is a hepatic monooxygenase involved in the metabolism of xenobiotics. The CYP2E1 gene is linked to the metabolism of many carcinogens. CYP2E1 is essential for the metabolism of endogenous compounds (such as acetone and fatty acids) as well as external substrates such as medications, contaminants, and ethanol [232]. The mechanism of both CYP2E1-mediated metabolism (e.g., styrene metabolism) and enzyme-associated toxicity, such as methemoglobinemia and acetaminophen-induced liver necrosis, has recently piqued researchers’ attention. Electrons are transferred to the substrates through CYP2E1-mediated oxidation of reduced nicotinamide, adenine dinucleotide phosphate, and molecular oxygen. This reaction adds additional polar groups to the substrates and generates hazardous intermediates such as epoxides or aldehydes [233].
An in silico approach for simulating the CYP2E1 active site as a sheet of hexagonal blocks has been devised, which might directly relate to the two-dimensional structure of chemicals. For a core part of the CYP2E1 active site, a region with the shape of benzopyrene was suggested as a model [234]. This was carried out to predict how the drug would be broken down at both sites and in what order.
CYP2E1 polymorphisms have been linked to potential mechanisms of tumor initiation. The CYP2E1*5 allele is linked to an increased risk of developing acute myeloid leukemia and acute lymphoblastic leukemia [235]. When lymphocytes with the CYP2E1 single-nucleotide polymorphisms rs2070673TT and rs2030920CC are exposed to phenol, they exhibit increased transcription and enzyme activity as well as increased DNA damage [236]. The CYP2E1*5B(C-1019T) polymorphism has not been linked to therapy-related acute myeloid leukemia or myelodysplastic syndrome [237]. In individuals with chronic lymphocytic leukemia/small lymphocytic lymphoma, the CYP2E1*07 (rs2070673) allele has been linked to a higher survival rate [238]. As a result, distinct allelic expressions of the CYP2E1 gene have diverse pathogenic and prognostic consequences in various hematological malignancies. Furthermore, the existence of polymorphisms does not always correspond with the phenotypic functional activity of CYP2E1, and total functional evaluation is more accurate than testing for polymorphisms. Increased CYP2E1 expression has been linked to liver illnesses such as alcoholic hepatitis and non-alcoholic steatohepatitis and is considered to play a role in their etiology [239,240].
Therapy-related myeloid neoplasms [241], infant leukemia associated with mixed-lineage leukemia (MLL) gene rearrangements [242], and a subtype of de novo acute myeloid leukemia [243], have low NQO1 activity. These findings support the idea that common ambient pollutants detoxified by NQO1 are risk factors for acute leukemia [243]. In fact, the NQO1 polymorphism had the strongest connection with acute myeloid leukemia and inv(16)/CBF-MYH11 in a study concentrating on de novo acute myeloid leukemia [243]. While the translocation has been postulated to disrupt the NQO1 gene, which is located on chromosome 16q22.1 [243], it is also possible that myeloid cells with this chromosomal abnormality are more sensitive to environmental pollutants. Furthermore, CYP2E1 was one of the four most differentially expressed genes in acute myeloid leukemia with inv(16)/CBF-MYH11, being raised 3.3-fold in acute myeloid leukemia with inv(16) [244].

8. Common Treatments of Acute Myeloid Leukemia

Between the treatments used for acute myeloid leukemia, we can find azacitidine, which is a pyrimidine nucleoside analog of cytidine that can be directly integrated into RNA, disrupting RNA, protein production, and metabolism [245]. It is only minimally integrated into DNA, covalently linking to DNA methyltransferases and directing their destruction. Without methyltransferases, daughter cells are hypomethylated, and repressed gene expression is reactivated during DNA synthesis [245,246]. Azacitidine, a DNA methyltransferase inhibitor, has been described as restoring tumor suppressor gene function and cell differentiation in patients with myelodysplastic syndrome and acute myeloid leukemia [247].
Azacitidine is available for intravenous and subcutaneous injection, with equivalent absorption for both routes [248]. The cytochrome P450 enzyme, uridine diphosphate, and glutathione transferase do not metabolise azacitidine, according to in vitro investigations. Instead, they are deaminated by cytidine deaminase and excreted primarily through the kidneys [249].
Other treatments for acute myeloid leukemia are based in enasidenib, a novel, mutant IDH2 protein-targeting inhibitor used to treat relapsed or resistant acute myeloid leukemia [250,251,252,253]. Enasidenib reduces the oncometabolite 2-hydroxyglutarate by 90.6% [250,251,252,253]. In vitro research shows that enasidenib inhibits several CYP enzymes and transporters and induces CYP3A4 [254]. Since enasidenib may induce or inhibit drug-metabolizing enzymes and transporters, the co-administration of enasidenib may increase or reduce the concentrations of combination drugs [254]. However, among patients with relapsed or refractory acute myeloid leukemia, the overall response rate is approximately 40.3% [17].
Finally, drugs against acute myeloid leukemia, such as glasdegib, gilteritinib, midostaurin, cytarabine, and venetoclax, have high oral bioavailability and are widely used [255,256,257,258,259,260]. Most, such as glasdegib, gilteritinib, midostaurin, cytarabine, and venetoclax, are eliminated by oxidative metabolism, mainly CYP3A4, with a minor contribution from glucuronidation by uridine diphosphate glucuronosyltransferase 1A [258,259,260,261]. However, because glasdegib, gilteritinib, midostaurin, cytarabine, and venetoclax are a substrate of CYP3A4 enzyme-mediated metabolism, plasma levels of glasdegib tend to decline; CYP3A inhibitors such as ketoconazole should be administered to increase glasdegib levels [256,257,258,259,260,262]. Table 3 shows one-year survival for each drug approved for the treatment of acute myeloid leukemia.

9. Carotenoids, CYP2E1 Expression, and Regulation in Acute Myeloid Leukemia

Dietary phytochemicals are one specific class of dietary components having anti-cancer action, among other dietary variables that are well known for their chemo-preventive effects. In addition to being non-essential nutrients, phytochemicals can significantly contribute to the prevention of disease. Numerous phytochemicals have strong anti-oxidant and anti-carcinogenic properties, including polyphenols, flavonoids, allyl sulphides, and carotenoids [271].
Carotenoids are pigments found in fruits, vegetables, and whole grains that are yellow, orange, or red. Patients with asthma, cataracts, and heart disease have been shown to benefit from beta-carotene, the main source of vitamin A and its derivative retinoic acid [272,273,274]. It has also been connected to a lower risk of prostate cancer. While some research has suggested that antioxidants, such as β-carotene, may increase CYP2E1 activity after moderate alcohol consumption and β-carotene supplementation [275], other studies have found that it is possible to prevent the degree of hepatic steatosis produced by various alcohol doses in order to prevent the progression to more serious injuries [276,277]. Therefore, it is unknown whether β-carotene, when combined with other vitamins, medications, or dietary components, has the capacity to reprogramme epigenetic activity.

10. Limitations

The purpose of this review was to describe the pathophysiology of acute myeloid leukemia as well as the role of CYP2E1 in the xenobiotic metabolism that governs the myeloid leukemia microenvironment. Still, this review found some problems that could make it hard to combine scientific evidence. These problems include: (1) a lack of information because there were so few articles; (2) a lack of NQO1 levels during CYP2E1 activity in acute myeloid leukemia; (3) a lack of information about how gene polymorphisms affect the encoded protein; and (4) a lack of thought about how genes and the environment interact.

11. Conclusions

Through the conversion of a range of xenobiotics into hazardous intermediates such as reactive oxygen species and free radicals, CYP2E1 contributes to an elevated acute myeloid leukemia risk. CYP2E1-related disorders relate to protein levels, and there are inter-individual variances in CYP2E1 expression levels, according to research. Furthermore, genetic polymorphism, drugs, plants, pollutants, food additives, and industrial material and environmental contaminants influence the variability and susceptibility to related myeloid neoplasms, infant leukemias associated with MLL gene rearrangements, and a subset of de novo acute myeloid leukemia. Recent research has shown a sustained interest in determining the regulators of CYP2E1 expression and activity as an emerging field that requires further investigation in acute myeloid leukemia evolution. This research has the potential to give insight into novel strategies for the treatment of acute myeloid leukemia via CYP2E1.

Author Contributions

C.S. conceived of the presented idea. C.S., Y.C., K.G. and J.F. conducted the database research and wrote the paper. C.S. and Y.C. drew the figures and flow gram. C.S. and K.G. edited the paper. C.S. supervised the project. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by [Programa de Formación de Investigadores Postdoctorales, Universidad de La Frontera] grant number [2022]; [DIUFRO, Universidad de La Frontera] grant number [DI22-0007], and [Programa LANCE, Universidad de La Frontera] grant number [Consorcio Ci2030].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. De Kouchkovsky, I.; Abdul-Hay, M. Acute myeloid leukemia: A comprehensive review and 2016 update. Blood Cancer J. 2016, 6, e441. [Google Scholar] [CrossRef] [Green Version]
  2. Shallis, R.M.; Wang, R.; Davidoff, A.; Ma, X.; Zeidan, A.M. Epidemiology of acute myeloid leukemia: Recent progress and enduring challenges. Blood Rev. 2019, 36, 70–87. [Google Scholar] [CrossRef] [PubMed]
  3. Swerdlow, S.H.; Campo, E.; Harris, N.L.; Jaffe, E.S.; Pileri, S.A.; Stein, H.; Thiele, J.; Vardiman, J.W. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, 4th ed.; IARC Press: Lyon, France, 2017; pp. 110–144. [Google Scholar]
  4. Jaffe, E.S.; Harris, N.L.; Stein, H.; Vardiman, J.W. Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues, 3rd ed.; IARC Press: Lyon, France, 2001; pp. 75–105. [Google Scholar]
  5. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017b. ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [Green Version]
  6. Dombret, H.; Gardin, C. An update of current treatments for adult acute myeloid leukemia. Blood 2016, 127, 53–61. [Google Scholar] [CrossRef] [PubMed]
  7. Megías-Vericat, J.E.; Martínez-Cuadrón, D.; Sanz, M.A.; Poveda, J.L.; Montesinos, P. Daunorubicin and cytarabine for certain types of poor-prognosis acute myeloid leukemia: A systematic literature review. Exp. Rev. Clin. Pharmacol. 2019, 12, 197–218. [Google Scholar] [CrossRef]
  8. Weick, J.K.; Kopecky, K.J.; Appelbaum, F.R.; Head, D.R.; Kingsbury, L.L.; Balcerzak, S.P.; Bickers, J.N.; Hynes, H.E.; Welborn, J.L.; Simon, S.R.; et al. A randomized investigation of high-dose versus standard-dose cytosine arabinoside with daunorubicin in patients with previously untreated acute myeloid leukemia: A Southwest Oncology Group study. Blood 1996, 88, 2841–2851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Volger, W.R.; Weiner, R.S.; Moore, J.O.; Omura, G.A.; Bartolucci, A.A.; Stagg, M. Long-term follow-up of a randomized postinduction therapy trial in acute myelogenous leukemia (a Southeastern Cancer Study Group trial). Leukemia 1995, 9, 1456–1460. [Google Scholar] [PubMed]
  10. Preisler, H.; Davis, R.B.; Kirshner, J.; Dupre, E.; Richards, F.; Hoagland, H.C.; Kopel, S.; Levy, R.N.; Carey, R.; Schulman, P. Comparison of three remission induction regimens and two postinduction strategies for the treatment of acute nonlymphocytic leukemia: A cancer and leukemia group B study. Blood 1987, 69, 1441–1449. [Google Scholar]
  11. Ding, L.; Ley, T.J.; Larson, D.E.; Miller, C.A.; Kodoldt, D.C.; Welch, J.S.; Ritchey, J.K.; Young, M.A.; Lamprecht, T.; McLellan, M.D.; et al. Clonal evaluation in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 2012, 481, 506–510. [Google Scholar] [CrossRef] [Green Version]
  12. Horowitz, M.M.; Gale, R.P.; Sondel, P.M.; Goldman, J.M.; Kersey, J.; Kolb, H.J.; Rimm, A.A.; Ringdén, O.; Rozman, C.; Speck, B. Graft-versus-leukemia reactions after bone marrow transplantation. Blood 1990, 75, 555–562. [Google Scholar] [CrossRef] [Green Version]
  13. Velardi, A.; Ruggeri, L.; Mancusi, A. Killer-cell immunoglobulin-like receptors reactivity and outcome of stem cell transplant. Curr. Opin. Hematol. 2012, 19, 319–323. [Google Scholar] [CrossRef]
  14. Sorror, M.L.; Giralt, S.; Sandmaier, B.M.; De Lima, M.; Shahjahan, M.; Maloney, D.G.; Deeg, H.J.; Appelbaum, F.R.; Storer, B.; Storb, R. Hematopoietic cell transplantation specific comorbidity index as an outcome predictor for patients with acute myeloid leukemia in first remission: Combined FHCRC and MDACC experiences. Blood 2007, 110, 4606–4613. [Google Scholar] [CrossRef] [Green Version]
  15. Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N.; et al. Genomic classification and prognosis in acute myeloid leukemia. N. Engl. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef] [PubMed]
  16. Castaigne, S.; Pautas, C.; Terré, C.; Raffoux, E.; Bordessoule, D.; Bastie, J.N.; Legrand, O.; Thomas, X.; Turlure, P.; Reman, O.; et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): A randomised, open-label, phase 3 study. Lancet 2012, 379, 1508–1516. [Google Scholar] [CrossRef]
  17. Stein, E.M.; DiNardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, K.; Marcucci, G.; et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
  19. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef]
  20. Cortes, J.E.; Heidel, F.H.; Hellmann, A.; Fiedler, W.; Smith, B.D.; Robak, T.; Montesinos, P.; Pollyea, D.A.; DesJardins, P.; Ottmann, O.; et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia 2019, 33, 379–389. [Google Scholar] [CrossRef] [Green Version]
  21. Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C.; et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740, Erratum in N. Engl. J. Med. 2022, 386, 1868. [Google Scholar] [CrossRef] [PubMed]
  22. Wei, A.H.; Montesinos, P.; Ivanov, V.; DiNardo, C.D.; Novak, J.; Laribi, K.; Kim, I.; Stevens, D.; Fiedler, W.; Pagoni, M.; et al. Venetoclax plus LDAC for patients with untreated AML ineligible for intensive chemotherapy: Phase 3 randomized placebo-controlled trial. Blood 2020, 135, 2137–2145. [Google Scholar] [CrossRef]
  23. Bewersdorf, J.P.; Derkach, A.; Gowda, L.; Menghrajani, K.; DeWolf, S.; Ruiz, J.D.; Ponce, D.M.; Shaffer, B.C.; Tamari, R.; Young, J.W.; et al. Venetoclax-based combinations in AML and high-risk MDS prior to and following allogeneic hematopoietic cell transplant. Leuk. Lymphoma 2021, 62, 3394–3401. [Google Scholar] [CrossRef]
  24. DiNardo, C.D.; Cortes, J.E. Mutations in AML: Prognostic and therapeutic implications. Hematol. Am. Soc. Hematol. Educ. Program 2016, 2016, 348–355. [Google Scholar] [CrossRef] [Green Version]
  25. Badar, T.; Patel, K.P.; Thompson, P.A.; DiNardo, C.; Takahashi, K.; Cabrero, M.; Borthakur, G.; Cortes, J.; Konopleva, M.; Kadia, T.; et al. Detectable FLT3-ITD or RAS mutation at the time of transformation from MDS to AML predicts for very poor outcomes. Leuk. Res. 2015, 39, 1367–1374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Burgess, M.R.; Hwang, E.; Firestone, A.J.; Huang, T.; Xu, J.; Zuber, J.; Bohin, N.; Wen, T.; Kogan, S.C.; Haigis, K.M.; et al. Preclinical efficacy of MEK inhibition in Nras-mutant AML. Blood 2014, 124, 3947–3955. [Google Scholar] [CrossRef] [PubMed]
  27. Borthakur, G.; Popplewell, L.; Boyiadzis, M.; Foran, J.; Platzbecker, U.; Vey, N.; Walter, R.B.; Olin, R.; Raza, A.; Giagounidis, A.; et al. Activity of the oral mitogen-activated protein kinase kinase inhibitor trametinib in RASmutant relapsed or refractory myeloid malignancies. Cancer 2016, 122, 1871–1879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Pollard, J.A.; Alonzo, T.A.; Gerbing, R.B.; Ho, P.A.; Zeng, R.; Ravindranath, Y.; Dahl, G.; Lacayo, N.J.; Becton, D.; Chang, M.; et al. Prevalence and prognostic significance of KIT mutations in pediatric patients with core binding factor AML enrolled on serial pediatric cooperative trials for de novo AML. Blood 2010, 115, 2372–2379. [Google Scholar] [CrossRef] [Green Version]
  29. Klein, K.; Kaspers, G.; Harrison, C.J.; Beverloo, H.B.; Reedijk, A.; Bongers, M.; Cloos, J.; Pession, A.; Reinhardt, D.; Zimmerman, M.; et al. Clinical impact of additional cytogenetic aberrations, cKIT and RAS mutations, and treatment elements in pediatric t(8;21)-AML: Results from an international retrospective study by the International Berlin-Frankfurt-Munster Study Group. J. Clin. Oncol. 2015, 33, 4247–4258. [Google Scholar] [CrossRef] [Green Version]
  30. Marcucci, G.; Geyer, S.; Zhao, W.; Caroll, A.J.; Bucci, D.; Uy, G.L.; Blum, W.; Pardee, T.; Wetzler, M.; Stock, W.; et al. Adding KIT inhibitor dasatinib (DAS) to chemotherapy overcomes the negative impact of KIT mutation/over-expression in core binding factor (CBF) acute myeloid leukemia (AML): Results from CALGB 10801 (Alliance). Blood 2014, 124, 8. [Google Scholar] [CrossRef]
  31. Vakiti, A.; Mewawalla, P. Acute Myeloid Leukemia. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  32. Sun, L.; Babushok, D.V. Secondary myelodysplastic syndrome and leukemia in acquired aplastic anemia and paroxysmal nocturnal hemoglobinuria. Blood 2020, 136, 36–49. [Google Scholar] [CrossRef]
  33. Hasle, H. Myelodysplastic and myeloproliferative disorders of childhood. Hematol. Am. Soc. Hematol. Educ. Program 2016, 2016, 598–604. [Google Scholar] [CrossRef] [Green Version]
  34. Jaiswal, S.; Fontanillas, P.; Flannick, J.; Manning, A.; Grauman, P.V.; Mar, B.G.; Lindsley, R.C.; Mermel, C.H.; Burtt, N.; Chavez, A.; et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 2014, 371, 2488–2498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Genovese, G.; Kähler, A.K.; Handsaker, R.E.; Lindberg, J.; Rose, S.A.; Bakhoum, S.F.; Chambert, K.; Mick, E.; Neale, B.M.; Fromer, M.; et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 2014, 371, 2477–2487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Döhner, K.; Schlenk, R.F.; Habdank, M.; Scholl, C.; Rücker, F.G.; Corbacioglu, A.; Bullinger, L.; Fröhling, S.; Döhner, H. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: Interaction with other gene mutations. Blood 2005, 106, 3740–3746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Falini, B.; Mecucci, C.; Tiacci, E.; Alcalay, M.; Rosati, R.; Pasqualucci, L.; La Starza, R.; Diverio, D.; Colombo, E.; Santucci, A.; et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N. Engl. J. Med. 2005, 352, 254–266. [Google Scholar] [CrossRef]
  38. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405. [Google Scholar] [CrossRef]
  39. Wouters, B.J.; Löwenberg, B.; Erpelinck-Verschueren, C.A.; van Putten, W.L.; Valk, P.J.; Delwel, R. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood 2009, 113, 3088–3091. [Google Scholar] [CrossRef] [Green Version]
  40. Pabst, T.; Eyholzer, M.; Fos, J.; Mueller, B.U. Heterogeneity within AML with CEBPA mutations; only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis. Br. J. Cancer 2009, 100, 1343–1346. [Google Scholar] [CrossRef] [Green Version]
  41. Gaidzik, V.I.; Teleanu, V.; Papaemmanuil, E.; Weber, D.; Paschka, P.; Hahn, J.; Wallrabenstein, T.; Kolbinger, B.; Köhne, C.H.; Horst, H.A.; et al. RUNX1 mutations in acute myeloid leukemia are associated with distinct clinico-pathologic and genetic features. Leukemia 2016, 30, 2160–2168. [Google Scholar] [CrossRef]
  42. Jacob, B.; Osato, M.; Yamashita, N.; Wang, C.Q.; Taniuchi, I.; Littman, D.R.; Asou, N.; Ito, Y. Stem cell exhaustion dueto Runx1 deficiency is prevented by Evi5 activation in leukemogenesis. Blood 2010, 115, 1610–1620. [Google Scholar] [CrossRef] [Green Version]
  43. Mendler, J.H.; Maharry, K.; Radmacher, M.D.; Mrózek, K.; Becker, H.; Metzeler, K.H.; Schwind, S.; Whitman, S.P.; Khalife, J.; Kohlschmidt, J.; et al. RUNX1 mutations are associated with poor outcome in younger and older patients with cytogenetically normal acute myeloid leukemia and with distinct gene and microRNA expression signatures. J. Clin. Oncol. 2012, 30, 3109–3118. [Google Scholar] [CrossRef] [Green Version]
  44. Wlodarski, M.W.; Hirabayashi, S.; Pastor, V.; Starý, J.; Hasle, H.; Masetti, R.; Dworzak, M.; Schmugge, M.; van den Heuvel-Eibrink, M.; Ussowicz, M.; et al. Prevalence, clinical characteristics, and prognosis of GATA2-related myelodysplastic syndromes in children and adolescents. Blood 2016, 127, 1387–1397. [Google Scholar] [CrossRef]
  45. Green, C.L.; Tawana, K.; Hills, R.K.; Bödör, C.; Fitzgibbon, J.; Inglott, S.; Ancliff, P.; Burnett, A.K.; Linch, D.C.; Gale, R.E. GATA2 mutations in sporadic and familial acute myeloid leukaemia patients with CEBPA mutations. Br. J. Haematol. 2013, 161, 701–705. [Google Scholar] [CrossRef]
  46. Lindsley, R.C.; Mar, B.G.; Mazzola, E.; Grauman, P.V.; Shareef, S.; Allen, S.L.; Pigneux, A.; Wetzler, M.; Stuart, R.K.; Erba, H.P.; et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015, 125, 1367–1376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Devillier, R.; Mansat-De Mas, V.; Gelsi-Boyer, V.; Demur, C.; Murati, A.; Corre, J.; Prebet, T.; Bertoli, S.; Brecqueville, M.; Arnoulet, C.; et al. Role of ASXL1and TP53 mutations in the molecular classification and prognosis of acute myeloid leukemias with myelodysplasia-related changes. Oncotarget 2015, 6, 8388–8396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Rücker, F.G.; Schlenk, R.F.; Bullinger, L.; Kayser, S.; Teleanu, V.; Kett, H.; Habdank, M.; Kugler, C.M.; Holzmann, K.; Gaidzik, V.I.; et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood 2012, 119, 2114–2121. [Google Scholar] [CrossRef] [Green Version]
  49. Dvinge, H.; Kim, E.; Abdel-Wahab, O.; Bradley, R.K. RNA splicing factors as oncoproteins and tumour suppressors. Nat. Rev. Cancer 2016, 16, 413–430. [Google Scholar] [CrossRef] [PubMed]
  50. Lee, S.C.; Dvinge, H.; Kim, E.; Cho, H.; Micol, J.B.; Chung, Y.R.; Durham, B.H.; Yoshimi, A.; Kim, Y.J.; Thomas, M.; et al. Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins. Nat. Med. 2016, 22, 672–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Han, C.; Gao, X.; Li, Y.; Zhang, J.; Yang, E.; Zhang, L.; Yu, L. Characteristics of Cohesin Mutation in Acute Myeloid Leukemia and Its Clinical Significance. Front. Oncol. 2021, 11, 579881. [Google Scholar] [CrossRef]
  52. Zhang, N.; Jiang, Y.; Mao, Q.; Demeler, B.; Tao, Y.J.; Pati, D. Characterization of the interaction between the cohesin subunits Rad21 and SA1/2. PLoS ONE 2013, 8, e69458. [Google Scholar] [CrossRef] [Green Version]
  53. Berenstein, R. Class III receptor tyrosine kinases in acute leukemia—Biological functions and modern laboratory analysis. Biomark. Insights 2015, 10, BMI-S22433. [Google Scholar] [CrossRef] [Green Version]
  54. Brandão, L.; Migdall-Wilson, J.; Eisenman, K.; Graham, D.K. TAM receptors in leukemia: Expression, signaling, and therapeutic implications. Crit. Rev. Oncog. 2011, 16, 47–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Lee-Sherick, A.B.; Eisenman, K.M.; Sather, S.; McGranahan, A.; Armistead, P.M.; McGary, C.S.; Hunsucker, S.A.; Schlegel, J.; Martinson, H.; Cannon, C.; et al. Aberrant Mer receptor tyrosine kinase expression contributes to leukemogenesis in acute myeloid leukemia. Oncogene 2013, 32, 5359–5368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Rochlitz, C.; Lohri, A.; Bacchi, M.; Schmidt, M.; Nagel, S.; Fopp, M.; Fey, M.F.; Herrmann, R.; Neubauer, A. Axl expression is associated with adverse prognosis and with expression of Bcl-2 and CD34 in de novo acute myeloid leukemia (AML): Results from a multicenter trial of the Swiss Group for Clinical Cancer Research (SAKK). Leukemia 1999, 13, 1352–1358. [Google Scholar] [CrossRef] [Green Version]
  57. Ben-Batalla, I.; Schultze, A.; Wroblewski, M.; Erdmann, R.; Heuser, M.; Waizenegger, J.S.; Riecken, K.; Binder, M.; Schewe, D.; Sawall, S.; et al. Axl, a prognostic and therapeutic target in acute myeloid leukemia mediates paracrine crosstalk of leukemia cells with bone marrow stroma. Blood 2013, 122, 2443–2452. [Google Scholar] [CrossRef] [PubMed]
  58. Crosier, P.S.; Hall, L.R.; Vitas, M.R.; Lewis, P.M.; Crosier, K.E. Identification of a novel receptor tyrosine kinase expressed in acute myeloid leukemic blasts. Leuk. Lymphoma 1995, 18, 443–449. [Google Scholar] [CrossRef]
  59. Cancer Genome Atlas Research Network; Ley, T.J.; Miller, C.; Ding, L.; Raphael, B.J.; Mungall, A.J.; Robertson, A.; Hoadley, K.; Triche, T.J., Jr.; Laird, P.W.; et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 2013, 368, 2059–2074. [Google Scholar]
  60. Li, S.; Garrett-Bakelman, F.E.; Chung, S.S.; Sanders, M.A.; Hricik, T.; Rapaport, F.; Patel, J.; Dillon, R.; Vijay, P.; Brown, A.L.; et al. Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia. Nat. Med. 2016, 22, 792–799. [Google Scholar] [CrossRef] [Green Version]
  61. Li, S.; Mason, C.; Melnick, A. Genetic and epigenetic heterogeneity in acute myeloid leukemia. Curr. Opin. Genet. Dev. 2016, 36, 100–106. [Google Scholar] [CrossRef] [Green Version]
  62. Guillamot, M.; Cimmino, L.; Aifantis, I. The impact of DNA methylation in hematopoietic malignancies. Trends Cancer 2016, 2, 70–83. [Google Scholar] [CrossRef] [Green Version]
  63. Hassan, C.; Afshinnekoo, E.; Li, S.; Wu, S.; Mason, C.E. Genetic and epigenetic heterogeneity and the impact on cancer relapse. Exp. Hematol. 2017, 54, 26–30. [Google Scholar] [CrossRef] [Green Version]
  64. DiNardo, C.D.; Stein, E.M.; de Botton, S.; Roboz, G.J.; Altman, J.K.; Mims, A.S.; Swords, R.; Collins, R.H.; Mannis, G.N.; Pollyea, D.A.; et al. Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. N. Engl. J. Med. 2018, 378, 2386–2398. [Google Scholar] [CrossRef]
  65. Koh, H.J.; Lee, S.M.; Son, B.G.; Lee, S.H.; Ryoo, Z.Y.; Chang, K.T.; Park, J.W.; Park, D.C.; Song, B.J.; Veech, R.L.; et al. Cytosolic NADP+-dependent isocitrate dehydrogenase plays a key role in lipid metabolism. J. Biol. Chem. 2004, 279, 39968–39974. [Google Scholar] [CrossRef] [Green Version]
  66. Joseph, J.W.; Jensen, M.V.; Ilkayeva, O.; Palmieri, F.; Alárcon, C.; Rhodes, C.J.; Newgard, C.B. The mitochondrial citrate/isocitrate carrier plays a regulatory role in glucose-stimulated insulin secretion. J. Biol. Chem. 2006, 281, 35624–35632. [Google Scholar] [CrossRef] [Green Version]
  67. Cairns, R.A.; Mak, T.W. Oncogenic isocitrate dehydrogenase mutations: Mechanisms, models, and clinical opportunities. Cancer Discov. 2013, 3, 730–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Dang, L.; White, D.W.; Gross, S.; Bennett, B.D.; Bittinger, M.A.; Driggers, E.M.; Fantin, V.R.; Jang, H.G.; Jin, S.; Keenan, M.C.; et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009, 462, 739–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Losman, J.A.; Looper, R.E.; Koivunen, P.; Lee, S.; Schneider, R.K.; McMahon, C.; Cowley, G.S.; Root, D.E.; Ebert, B.L.; Kaelin, W.G., Jr. (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science 2013, 339, 1621–1625. [Google Scholar] [CrossRef]
  70. Becker, H.; Marcucci, G.; Maharry, K.; Radmacher, M.D.; Mrózek, K.; Margeson, D.; Whitman, S.P.; Wu, Y.Z.; Schwind, S.; Paschka, P.; et al. Favorable prognostic impact of NPM1 mutations in older patients with cytogenetically normal de novo acute myeloid leukemia and associated gene- and microRNA-expression signatures: A Cancer and Leukemia Group B study. J. Clin. Oncol. 2010, 28, 596–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Alpermann, T.; Schnittger, S.; Eder, C.; Dicker, F.; Meggendorfer, M.; Kern, W.; Schmid, C.; Aul, C.; Staib, P.; Wendtner, C.M.; et al. Molecular subtypes of NPM1 mutations have different clinical profiles, specific patterns of accompanying molecular mutations and varying outcomes in intermediate risk acute myeloid leukemia. Haematologica 2016, 101, e55–e58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Jalagadugula, G.; Mao, G.; Kaur, G.; Goldfinger, L.E.; Dhanasekaran, D.N.; Rao, A.K. Regulation of platelet myosin light chain (MYL9) by RUNX1: Implications for thrombocytopenia and platelet dysfunction in RUNX1 haplodeficiency. Blood 2010, 116, 6037–6045. [Google Scholar] [CrossRef] [Green Version]
  73. Jalagadugula, G.; Mao, G.; Kaur, G.; Dhanasekaran, D.N.; Rao, A.K. Platelet protein kinase C-theta deficiency with human RUNX1 mutation: PRKCQ is a transcriptional target of RUNX1. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 921–927. [Google Scholar] [CrossRef] [Green Version]
  74. Kaur, G.; Jalagadugula, G.; Mao, G.; Rao, A.K. RUNX1/core binding factor A2 regulates platelet 12-lipoxygenase gene (ALOX12): Studies in human RUNX1 haplodeficiency. Blood 2010, 115, 3128–3135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Antony-Debre, I.; Bluteau, D.; Itzykson, R.; Baccini, V.; Renneville, A.; Boehlen, F.; Morabito, M.; Droin, N.; Deswarte, C.; Chang, Y.; et al. MYH10 protein expression in platelets as a biomarker of RUNX1 and FLI1 alterations. Blood 2012, 120, 2719–2722. [Google Scholar] [CrossRef] [PubMed]
  76. Bluteau, D.; Balduini, A.; Balayn, N.; Currao, M.; Nurden, P.; Deswarte, C.; Leverger, G.; Noris, P.; Perrotta, S.; Solary, E.; et al. Thrombocytopenia-associated mutations in the ANKRD26 regulatory region induce MAPK hyperactivation. J. Clin. Investig. 2014, 124, 580–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Paz-Priel, I.; Friedman, A. C/EBPalpha dysregulation in AML and ALL. Crit. Rev. Oncog. 2011, 16, 93–102. [Google Scholar] [CrossRef] [PubMed]
  78. Tsai, F.Y.; Keller, G.; Kuo, F.C.; Weiss, M.; Chen, J.; Rosenblatt, M.; Alt, F.W.; Orkin, S.H. An early haematopoietic defect in mice lacking the transcription factor GATA-2. Nature 1994, 371, 221–226. [Google Scholar] [CrossRef]
  79. Rodrigues, N.P.; Janzen, V.; Forkert, R.; Dombkowski, D.M.; Boyd, A.S.; Orkin, S.H.; Enver, T.; Vyas, P.; Scadden, D.T. Haploinsufficiency of GATA-2 perturbs adult hematopoietic stem-cell homeostasis. Blood 2005, 106, 477–484. [Google Scholar] [CrossRef] [Green Version]
  80. Kazenwadel, J.; Secker, G.A.; Liu, Y.J.; Rosenfeld, J.A.; Wildin, R.S.; Cuellar-Rodriguez, J.; Hsu, A.P.; Dyack, S.; Fernandez, C.V.; Chong, C.E.; et al. Loss-of-function germline GATA2 mutations in patients with MDS/AML or MonoMAC syndrome and primary lymphedema reveal a key role for GATA2 in the lymphatic vasculature. Blood 2012, 119, 1283–1291. [Google Scholar] [CrossRef] [Green Version]
  81. Godley, L.A. Inherited predisposition to acute myeloid leukemia. Semin. Hematol. 2014, 51, 306–321. [Google Scholar] [CrossRef] [Green Version]
  82. Solomon, D.A.; Kim, J.S.; Waldman, T. Mutational inactivation of STAG2 causes aneuploidy in human cancer. Science 2011, 333, 1039–1043. [Google Scholar] [CrossRef] [Green Version]
  83. Losada, A. Cohesin in cancer: Chromosome segregation and beyond. Nat. Rev. Cancer 2014, 14, 389–393. [Google Scholar] [CrossRef]
  84. Litwin, I.; Wysocki, R. New insights into cohesin loading. Curr. Genet. 2018, 64, 53–61. [Google Scholar] [CrossRef] [PubMed]
  85. Dauban, L.; Montagne, R.; Thierry, A.; Lazar-Stefanita, L.; Bastié, N.; Gadal, O.; Cournac, A.; Koszul, R.; Beckouët, F. Regulation of Cohesin-Mediated Chromosome Folding by Eco1 and Other Partners. Mol. Cell 2020, 77, 1279–1293.e4. [Google Scholar] [CrossRef] [PubMed]
  86. Cuartero, S.; Innes, A.J.; Merkenschlager, M. Towards a Better Understanding of Cohesin Mutations in AML. Front. Oncol. 2019, 9, 867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Kagey, M.H.; Newman, J.J.; Bilodeau, S.; Zhan, Y.; Orlando, D.; Am van Berkumm, N.L.; Ebmeier, C.C.; Goossens, J.; Rahl, P.B.; Levine, S.S.; et al. Mediator and cohesin connect gene expression and chromatin architecture. Nature 2010, 467, 430–435. [Google Scholar] [CrossRef] [Green Version]
  88. Guillou, E.; Ibarra, A.; Coulon, V.; Casado-Vela, J.; Rico, D.; Casal, I.; Schwob, E.; Losada, A.; Méndez, J. Cohesin organizes chromatin loops at DNA replication factories. Genes Dev. 2010, 24, 2812–2822. [Google Scholar] [CrossRef] [Green Version]
  89. McAleenan, A.; Clemente-Blanco, A.; Cordon-Preciado, V.; Sen, N.; Esteras, M.; Jarmuz, A.; Aragón, L. Post-replicative repair involves separase-dependent removal of the kleisin subunit of cohesin. Nature 2012, 493, 250–254. [Google Scholar] [CrossRef] [PubMed]
  90. Panigrahi, A.K.; Pati, D. Higher-order orchestration of hematopoiesis: Is cohesin a new player? Exp. Hematol. 2012, 40, 967–973. [Google Scholar] [CrossRef] [Green Version]
  91. Guo, Y.; Monahan, K.; Wu, H.; Gertz, J.; Varley, K.E.; Li, W.; Myers, R.M.; Maniatis, T.; Wu, Q. CTCF/cohesin-mediated DNA looping is required for protocadherin alpha promoter choice. Proc. Natl. Acad. Sci. USA 2012, 109, 21081–21086. [Google Scholar] [CrossRef] [Green Version]
  92. Haarhuis, J.H.I.; van der Weide, R.H.; Blomen, V.A.; Yáñez-Cuna, J.O.; Amendola, M.; van Ruiten, M.S.; Krijger, P.H.L.; Teunissen, H.; Medema, R.H.; van Steensel, B.; et al. The cohesin release factor wapl restricts chromatin loop extension. Cell 2017, 169, 693–707. [Google Scholar] [CrossRef] [Green Version]
  93. Schwarzerm, W.; Abdennur, N.; Goloborodko, A.; Pekowska, A.; Fudenberg, G.; Loe-Mie, Y.; Fonseca, N.A.; Huber, W.; Haering, C.H.; Mirny, L.; et al. Two independent modes of chromatin organization revealed by cohesin removal. Nature 2017, 551, 51–56. [Google Scholar] [CrossRef] [Green Version]
  94. Rao, S.S.; Huang, S.C.; Glenn St Hilaire, B.; Engreitz, J.M.; Perez, E.M.; Kieffer-Kwon, K.R.; Sanborn, A.L.; Johnstone, S.E.; Bascom, G.D.; Bochkov, I.D.; et al. Cohesin loss eliminates all loop domains. Cell 2017, 171, 305–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bintu, B.; Mateo, L.J.; Su, J.H.; Sinnott-Armstrong, N.A.; Parker, M.; Kinrot, S.; Yamaya, K.; Boettiger, A.N.; Zhuang, X. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 2018, 362, eaau1783. [Google Scholar] [CrossRef] [Green Version]
  96. National Comprehensive Cancer Network. NCCN Guidelines & Clinical Resources. Adapted from the National Cancer Centers Network (NCCN). 2020. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1410 (accessed on 14 December 2022).
  97. 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. [Google Scholar] [CrossRef]
  98. Grimwade, D.; Hills, R.K.; Moorman, A.V.; Walker, H.; Chatters, S.; Goldstone, A.H.; Wheatley, K.; Harrison, C.J.; Burnett, A.K.; National Cancer Research Institute Adult Leukaemia Working Group. Refinement of cytogenetic classification in acute myeloid leukemia: Determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010, 116, 354–365. [Google Scholar] [CrossRef] [Green Version]
  99. Schoch, C.; Schnittger, S.; Klaus, M.; Kern, W.; Hiddemann, W.; Haferlach, T. AML with 11q23/MLL abnormalities as defined by the WHO classification: Incidence, partner chromosomes, FAB subtype, age distribution, and prognostic impact in an unselected series of 1897 cytogenetically analyzed AML cases. Blood 2003, 102, 2395–2402. [Google Scholar] [CrossRef] [Green Version]
  100. Ayatollahi, H.; Bazi, A.; Sadeghian, M.H.; Fani, A.; Siyadat, P.; Sheikhi, M.; Sargazi-aval, O. The Survival of Patients with t(15;17)(q22;q12) Positive Acute Promyelocytic Leukemia: A Study in North-East of Iran. Iran. J. Pathol. 2020, 15, 175–181. [Google Scholar] [CrossRef] [PubMed]
  101. Eghtedar, A.; Borthakur, G.; Ravandi, F.; Jabbour, E.; Cortes, J.; Pierce, S.; Kantarjian, H.; Garcia-Manero, G. Characteristics of translocation (16;16)(p13;q22) acute myeloid leukemia. Am. J. Hematol. 2012, 87, 317. [Google Scholar] [CrossRef] [Green Version]
  102. Rücker, F.G.; Agrawal, M.; Corbacioglu, A.; Weber, D.; Kapp-Schwoerer, S.; Gaidzik, V.I.; Jahn, N.; Schroeder, T.; Wattad, M.; Lübbert, M.; et al. Measurable residual disease monitoring in acute myeloid leukemia with t(8;21)(q22;q22.1): Results from the AML Study Group. Blood 2019, 134, 1608–1618. [Google Scholar] [CrossRef] [PubMed]
  103. Stölzel, F.; Mohr, B.; Kramer, M.; Oelschlägel, U.; Bochtler, T.; Berdel, W.E.; Kaufmann, M.; Baldus, C.D.; Schäfer-Eckart, K.; Stuhlmann, R.; et al. Karyotype complexity and prognosis in acute myeloid leukemia. Blood Cancer J. 2016, 6, e386. [Google Scholar] [CrossRef] [Green Version]
  104. Groschel, S.; Schlenk, R.F.; Engelmann, J.; Rockova, V.; Teleanu, V.; Kühn, M.W.; Eiwen, K.; Erpelinck, C.; Havermans, M.; Lübbert, M.; et al. Deregulated expression of EVI1 defines a poor prognostic subset of MLL-rearranged acute myeloid leukemias: A study of the German-Austrian Acute Myeloid Leukemia Study Group and the Dutch-Belgian-Swiss HOVON/SAKK Cooperative Group. J. Clin. Oncol. 2013, 31, 95–103. [Google Scholar] [CrossRef]
  105. Liu, K.; Tirado, C.A. MECOM: A Very Interesting Gene Involved also in Lymphoid Malignancies. J. Assoc. Genet. Technol. 2019, 45, 109–114. [Google Scholar] [PubMed]
  106. Yu, J.; Li, Y.; Li, T.; Li, Y.; Xing, H.; Sun, H.; Sun, L.; Wan, D.; Liu, Y.; Xie, X.; et al. Gene mutational analysis by NGS and its clinical significance in patients with myelodysplastic syndrome and acute myeloid leukemia. Exp. Hematol. Oncol. 2020, 6, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Han, X.; Li, W.; He, N.; Feng, P.; Pang, Y.; Ji, C.; Ma, D. Gene mutation patterns of Chinese acute myeloid leukemia patients by targeted next-generation sequencing and bioinformatic analysis. Clin. Chim. Acta 2018, 479, 25–37. [Google Scholar] [CrossRef] [PubMed]
  108. Chung, W.; Kelly, A.D.; Kropf, P.; Fung, H.; Jelinek, J.; Su, X.Y.; Roboz, G.J.; Kantarjian, H.M.; Azab, M.; Issa, J.P.J. Genomic and epigenomic predictors of response to guadecitabine in relapsed/refractory acute myelogenous leukemia. Clin. Epigenet. 2019, 11, 106. [Google Scholar] [CrossRef]
  109. Yang, L.; Shen, K.; Zhang, M.; Zhang, W.; Cai, H.; Lin, L.; Long, X.; Xing, S.; Tang, Y.; Xiong, J.; et al. Clinical features and microRNA expression patterns between AML patients with DNMT3A R882 and frameshift mutations. Front Oncol. 2019, 24, 1133. [Google Scholar] [CrossRef] [Green Version]
  110. Folta, A.; Culen, M.; Jeziskova, I.; Herudkova, Z.; Tom, N.; Hlubinkova, T.; Janeckova, V.; Durinikova, A.; Vydra, J.; Semerad, L.; et al. Prognostic significance of mutation profile at diagnosis and mutation persistence during disease remission in adult acute myeloid leukaemia patients. Br. J. Haematol. 2019, 186, 300–310. [Google Scholar] [CrossRef]
  111. Vetro, C.; Haferlach, T.; Meggendorfer, M.; Stengel, A.; Jeromin, S.; Kern, W.; Haferlach, C. Cytogenetic and molecular genetic characterization of KMT2A-PTD positive acute myeloid leukemia in comparison to KMT2A-rearranged acute myeloid leukemia. Cancer Genet. 2020, 240, 15–22. [Google Scholar] [CrossRef]
  112. Antar, A.I.; Otrock, Z.K.; Jabbour, E.; Mohty, M.; Bazarbachi, A. FLT3 inhibitors in acute myeloid leukemia: Ten frequently asked questions. Leukemia 2020, 34, 682–696. [Google Scholar] [CrossRef]
  113. Darracq, A.; Pak, H.; Bourgoin, V.; Zmiri, F.; Dellaire, G.; Affar, E.B.; Milot, E. NPM and NPM-MLF1 interact with chromatin remodeling complexes and influence their recruitment to specific genes. PLoS Genet. 2019, 15, e1008463. [Google Scholar] [CrossRef]
  114. Patel, S.S.; Kuo, F.C.; Gibson, C.J.; Steensma, D.P.; Soiffer, R.J.; Alyea, E.P., 3rd; Chen, Y.A.; Fathi, A.T.; Graubert, T.A.; Brunner, A.M.; et al. High NPM1-mutant allele burden at diagnosis predicts unfavorable outcomes in de novo AML. Blood 2018, 131, 2816–2825. [Google Scholar] [CrossRef] [Green Version]
  115. Cucchi, D.G.J.; Denys, B.; Kaspers, G.J.L.; Janssen, J.J.W.M.; Ossenkoppele, G.J.; de Haas, V.; Zwaan, C.M.; van den Heuvel-Eibrink, M.M.; Philippé, J.; Csikós, T.; et al. RNA-based FLT3-ITD allelic ratio is associated with outcome and ex vivo response to FLT3 inhibitors in pediatric AML. Blood 2018, 131, 2485–2489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Boileau, M.; Shirinian, M.; Gayden, T.; Harutyunyan, A.S.; Chen, C.C.L.; Mikael, L.G.; Duncan, H.M.; Neumann, A.L.; Arreba-Tutusaus, P.; De Jay, N.; et al. Mutant H3 histones drive human pre-leukemic hematopoietic stem cell expansion and promote leukemic aggressiveness. Nat. Commun. 2019, 10, 2891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Tallman, M. Prognostic significance of molecular markers and targeted regimens in the management of acute myeloid leukemia. J. Natl. Compr. Cancer Netw. 2018, 16, 656–659. [Google Scholar] [CrossRef] [PubMed]
  118. Tallman, M.S.; Wang, E.S.; Altman, J.K.; Appelbaum, F.R.; Bhatt, V.R.; Bixby, D.; Coutre, S.E.; De Lima, M.; Fathi, A.T.; Fiorella, M.; et al. Acute myeloid leukemia, version 3.2019, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Cancer Netw. 2019, 17, 721–749. [Google Scholar] [CrossRef] [Green Version]
  119. Corces-Zimmerman, M.R.; Hong, W.J.; Weissman, I.L.; Medeiros, B.C.; Majeti, R. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc. Natl. Acad. Sci. USA 2014, 111, 2548–2553. [Google Scholar] [CrossRef] [Green Version]
  120. Shlush, L.I.; Zandi, S.; Mitchell, A.; Chen, W.C.; Brandwein, J.M.; Gupta, V.; Kennedy, J.A.; Schimmer, A.D.; Schuh, A.C.; Yee, K.W.; et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 2014, 506, 328–333. [Google Scholar] [CrossRef] [Green Version]
  121. Xie, M.; Lu, C.; Wang, J.; McLellan, M.D.; Johnson, K.J.; Wendl, M.C.; McMichael, J.F.; Schmidt, H.K.; Yellapantula, V.; Miller, C.A.; et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 2014, 20, 1472–1478. [Google Scholar] [CrossRef]
  122. Buscarlet, M.; Provost, S.; Zada, Y.F.; Bourgoin, V.; Mollica, L.; Dubé, M.P.; Busque, L. Lineage restriction analyses in CHIP indicate myeloid bias for TET2 and multipotent stem cell origin for DNMT3A. Blood 2018, 132, 277–280. [Google Scholar] [CrossRef] [Green Version]
  123. Morrison, S.J.; Spradling, A.C. Stem Cells and Niches: Mechanisms that Promote Stem Cell Maintenance throughout Life. Cell 2008, 132, 598–611. [Google Scholar] [CrossRef] [Green Version]
  124. Szade, K.; Gulati, G.S.; Chan, C.K.F.; Kao, K.S.; Miyanishi, M.; Marjon, K.D.; Sinha, R.; George, B.M.; Chen, J.Y.; Weissman, I.L. Where Hematopoietic Stem Cells Live: The Bone Marrow Niche. Antioxid. Redox Signal. 2018, 29, 191–204. [Google Scholar] [CrossRef]
  125. Dorshkind, K. Regulation of Hemopoiesis by Bone Marrow Stromal Cells and Their Products. Annu. Rev. Immunol. 1990, 8, 111–137. [Google Scholar] [CrossRef] [PubMed]
  126. Dexter, T.M. Regulation of Hemopoietic Cell Growth and Development: Experimental and Clinical Studies. Leukemia 1989, 3, 469–474. [Google Scholar]
  127. Saleh, M.; Shamsasanjan, K.; Movassaghpourakbari, A.; Akbarzadehlaleh, P.; Molaeipour, Z. The Impact of Mesenchymal Stem Cells on Differentiation of Hematopoietic Stem Cells. Adv. Pharm. Bull. 2015, 5, 299–304. [Google Scholar] [CrossRef] [Green Version]
  128. Shafat, M.S.; Gnaneswaran, B.; Bowles, K.M.; Rushworth, S.A. The bone marrow microenvironment—Home of the leukemic blasts. Blood Rev. 2017, 31, 277–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Md Yusof, M.Y.; Psarras, A.; El-Sherbiny, Y.M.; Hensor, E.M.A.; Dutton, K.; Ul-Hassan, S.; Zayat, A.S.; Shalbaf, M.; Alase, A.; Wittmann, M.; et al. Prediction of autoimmune connective tissue disease in an at-risk cohort: Prognostic value of a novel two-score system for interferon status. Ann. Rheum. Dis. 2018, 77, 1432–1439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Waclawiczek, A.; Hamilton, A.; Rouault-Pierre, K.; Abarrategi, A.; Albornoz, M.G.; Miraki-Moud, F.; Bah, N.; Gribben, J.; Fitzgibbon, J.; Taussig, D.; et al. Mesenchymal niche remodeling impairs hematopoiesis via stanniocalcin 1 in acute myeloid leukemia. J. Clin. Investig. 2020, 130, 3038–3050. [Google Scholar] [CrossRef] [PubMed]
  131. Rodríguez-Fuentes, D.E.; Fernández-Garza, L.E.; Samia-Meza, J.A.; Barrera-Barrera, S.A.; Caplan, A.I.; Barrera-Saldaña, H.A. Mesenchymal Stem Cells Current Clinical Applications: A Systematic Review. Arch. Med. Res. 2021, 52, 93–101. [Google Scholar] [CrossRef]
  132. Aggarwal, S.; Pittenger, M.F. Human Mesenchymal Stem Cells Modulate Allogeneic Immune Cell Responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
  133. Muguruma, Y.; Yahata, T.; Miyatake, H.; Sato, T.; Uno, T.; Itoh, J.; Kato, S.; Ito, M.; Hotta, T.; Ando, K. Reconstitution of the Functional Human Hematopoietic Microenvironment Derived from Human Mesenchymal Stem Cells in the Murine Bone Marrow Compartment. Blood 2006, 107, 1878–1887. [Google Scholar] [CrossRef]
  134. Garrido, S.M.; Appelbaum, F.R.; Willman, C.L.; Banker, D.E. Acute Myeloid Leukemia Cells Are Protected from Spontaneous and Drug-Induced Apoptosis by Direct Contact with a Human Bone Marrow Stromal Cell Line (HS-5). Exp. Hematol. 2001, 29, 448–457. [Google Scholar] [CrossRef]
  135. Chen, S.; Zambetti, N.A.; Bindels, E.M.; Kenswill, K.; Mylona, A.M.; Adisty, N.M.; Hoogenboezem, R.M.; Sanders, M.A.; Cremers, E.M.; Westers, T.M.; et al. Massive parallel RNA sequencing of highly purified mesenchymal elements in low-risk MDS reveals tissue-context-dependent activation of inflammatory programs. Leukemia 2016, 30, 1938–1942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Moshaver, B.; van der Pol, M.A.; Westra, A.H.; Ossenkoppele, G.J.; Zweegman, S.; Schuurhuis, G.J. Chemotherapeutic Treatment of Bone Marrow Stromal Cells Strongly Affects Their Protective Effect on Acute Myeloid Leukemia Cell Survival. Leuk. Lymphoma 2008, 49, 134–148. [Google Scholar] [CrossRef]
  137. Brenner, A.K.; Nepstad, I.; Bruserud, Ø. Mesenchymal Stem Cells Support Survival and Proliferation of Primary Human Acute Myeloid Leukemia Cells through Heterogeneous Molecular Mechanisms. Front. Immunol. 2017, 8, 1331. [Google Scholar] [CrossRef] [Green Version]
  138. Ciciarello, M.; Corradi, G.; Loscocco, F.; Visani, G.; Monaco, F.; Cavo, M.; Curti, A.; Isidori, A. The Yin and Yang of the Bone Marrow Microenvironment: Pros and Cons of Mesenchymal Stromal Cells in Acute Myeloid Leukemia. Front. Oncol. 2019, 9, 1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Civini, S.; Jin, P.; Ren, J.; Sabatino, M.; Castiello, L.; Jin, J.; Wang, H.; Zhao, Y.; Marincola, F.; Stroncek, D. Leukemia Cells Induce Changes in Human Bone Marrow Stromal Cells. J. Transl. Med. 2013, 11, 298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Long, X.; Yu, Y.; Perlaky, L.; Man, T.-K.; Redell, M.S. Stromal CYR61 Confers Resistance to Mitoxantrone via Spleen Tyrosine Kinase Activation in Human Acute Myeloid Leukaemia. Br. J. Haematol. 2015, 170, 704–718. [Google Scholar] [CrossRef] [Green Version]
  141. Konopleva, M.; Konoplev, S.; Hu, W.; Zaritskey, A.; Afanasiev, B.; Andreeff, M. Stromal Cells Prevent Apoptosis of AML Cells by Up-Regulation of Anti-Apoptotic Proteins. Leukemia 2002, 16, 1713–1724. [Google Scholar] [CrossRef] [Green Version]
  142. Gynn, L.E.; Anderson, E.; Robinson, G.; Wexler, S.A.; Upstill-Goddard, G.; Cox, C.; May, J.E. Primary Mesenchymal Stromal Cells in Co-Culture with Leukaemic HL-60 Cells Are Sensitised to Cytarabine-Induced Genotoxicity, While Leukaemic Cells Are Protected. Mutagenesis 2021, 36, 419–428. [Google Scholar] [CrossRef]
  143. Walkley, C.R.; Shea, J.M.; Sims, N.A.; Purton, L.E.; Orkin, S.H. Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment. Cell 2007, 129, 1081–1095. [Google Scholar] [CrossRef] [Green Version]
  144. Walkley, C.R.; Olsen, G.H.; Dworkin, S.; Fabb, S.A.; Swann, J.; McArthur, G.A.; Westmoreland, S.V.; Chambon, P.; Scadden, D.T.; Purton, L.E. A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency. Cell 2007, 129, 1097–1110. [Google Scholar] [CrossRef] [Green Version]
  145. Kim, Y.W.; Koo, B.K.; Jeong, H.W.; Yoon, M.J.; Song, R.; Shin, J.; Jeong, D.C.; Kim, S.H.; Kong, Y.Y. Defective Notch activation in microenvironment leads to myeloproliferative disease. Blood 2008, 112, 4628–4638. [Google Scholar] [CrossRef]
  146. Wang, L.; Zhang, H.; Rodriguez, S.; Cao, L.; Parish, J.; Mumaw, C.; Zollman, A.; Kamoka, M.M.; Mu, J.; Chen, D.Z.; et al. Notch-dependent repression of miR-155 in the bone marrow niche regulates hematopoiesis in an NF-κB-dependent manner. Cell Stem Cell 2014, 15, 51–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Dong, L.; Yu, W.M.; Zheng, H.; Loh, M.L.; Bunting, S.T.; Pauly, M.; Huang, G.; Zhou, M.; Broxmeyer, H.E.; Scadden, D.T.; et al. Leukaemogenic effects of Ptpn11 activating mutations in the stem cell microenvironment. Nature 2016, 539, 304–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Rupec, R.A.; Jundt, F.; Rebholz, B.; Eckelt, B.; Weindl, G.; Herzinger, T.; Flaig, M.J.; Moosmann, S.; Plewig, G.; Dörken, B.; et al. Stroma-mediated dysregulation of myelopoiesis in mice lacking I kappa B alpha. Immunity 2005, 22, 479–491. [Google Scholar] [CrossRef] [Green Version]
  149. Raaijmakers, M.H.; Mukherjee, S.; Guo, S.; Zhang, S.; Kobayashi, T.; Schoonmaker, J.A.; Ebert, B.L.; Al-Shahrour, F.; Hasserjian, R.P.; Scadden, E.O.; et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 2010, 464, 852–857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Kode, A.; Manavalan, J.S.; Mosialou, I.; Bhagat, G.; Rathinam, C.V.; Luo, N.; Khiabanian, H.; Lee, A.; Murty, V.V.; Friedman, R.; et al. Leukaemogenesis induced by an activating β-catenin mutation in osteoblasts. Nature 2014, 506, 240–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Kode, A.; Mosialou, I.; Manavalan, S.J.; Rathinam, C.V.; Friedman, R.A.; Teruya-Feldstein, J.; Bhagat, G.; Berman, E.; Kousteni, S. FoxO1-dependent induction of acute myeloid leukemia by osteoblasts in mice. Leukemia 2016, 30, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Huang, J.C.; Basu, S.K.; Zhao, X.; Chien, S.; Fang, M.; Oehler, V.G.; Appelbaum, F.R.; Becker, P.S. Mesenchymal stromal cells derived from acute myeloid leukemia bone marrow exhibit aberrant cytogenetics and cytokine elaboration. Blood Cancer J. 2015, 5, e302. [Google Scholar] [CrossRef]
  153. von der Heide, E.K.; Neumann, M.; Vosberg, S.; James, A.R.; Schroeder, M.P.; Ortiz-Tanchez, J.; Isaakidis, K.; Schlee, C.; Luther, M.; Jöhrens, K.; et al. Molecular alterations in bone marrow mesenchymal stromal cells derived from acute myeloid leukemia patients. Leukemia 2017, 31, 1069–1078. [Google Scholar] [CrossRef]
  154. Medyouf, H.; Mossner, M.; Jann, J.C.; Nolte, F.; Raffel, S.; Herrmann, C.; Lier, A.; Eisen, C.; Nowak, V.; Zens, B.; et al. Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit. Cell Stem Cell 2014, 14, 824–837. [Google Scholar] [CrossRef] [Green Version]
  155. Kim, Y.; Jekarl, D.W.; Kim, J.; Kwon, A.; Choi, H.; Lee, S.; Kim, Y.J.; Kim, H.J.; Kim, Y.; Oh, I.H.; et al. Genetic and epigenetic alterations of bone marrow stromal cells in myelodysplastic syndrome and acute myeloid leukemia patients. Stem Cell Res. 2015, 14, 177–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Geyh, S.; Oz, S.; Cadeddu, R.P.; Fröbel, J.; Brückner, B.; Kündgen, A.; Fenk, R.; Bruns, I.; Zilkens, C.; Hermsen, D.; et al. Insufficient stromal support in MDS results from molecular and functional deficits of mesenchymal stromal cells. Leukemia 2013, 27, 1841–1851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Santamaría, C.; Muntión, S.; Rosón, B.; Blanco, B.; López-Villar, O.; Carrancio, S.; Sánchez-Guijo, F.M.; Díez-Campelo, M.; Alvarez-Fernández, S.; Sarasquete, M.E.; et al. Impaired expression of DICER, DROSHA, SBDS and some microRNAs in mesenchymal stromal cells from myelodysplastic syndrome patients. Haematologica 2012, 97, 1218–1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Chen, P.; Jin, Q.; Fu, Q.; You, P.; Jiang, X.; Yuan, Q.; Huang, H. Induction of Multidrug Resistance of Acute Myeloid Leukemia Cells by Cocultured Stromal Cells via Upregulation of the PI3K/Akt Signaling Pathway. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2016, 24, 215–223. [Google Scholar] [CrossRef]
  159. Lopez-Villar, O.; Garcia, J.L.; Sanchez-Guijo, F.M.; Robledo, C.; Villaron, E.M.; Hernández-Campo, P.; Lopez-Holgado, N.; Diez-Campelo, M.; Barbado, M.V.; Perez-Simon, J.A.; et al. Both expanded and uncultured mesenchymal stem cells from MDS patients are genomically abnormal, showing a specific genetic profile for the 5q- syndrome. Leukemia 2009, 23, 664–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Zambetti, N.A.; Ping, Z.; Chen, S.; Kenswil, K.J.G.; Mylona, M.A.; Sanders, M.A.; Hoogenboezem, R.M.; Bindels, E.M.J.; Adisty, M.N.; Van Strien, P.M.H.; et al. Mesenchymal inflammation drives genotoxic stress in hematopoietic stem cells and predicts disease evolution in human pre-leukemia. Cell Stem Cell 2016, 19, 613–627. [Google Scholar] [CrossRef] [Green Version]
  161. Whitehead, T.P.; Metayer, C.; Ward, M.H.; Colt, J.S.; Gunier, R.B.; Deziel, N.C.; Rappaport, S.M.; Buffler, P.A. Persistent organic pollutants in dust from older homes: Learning from lead. Am. J. Public Health 2014, 104, 1320–1326. [Google Scholar] [CrossRef]
  162. Lagorio, S.; Ferrante, D.; Ranucci, A.; Negri, S.; Sacco, P.; Rondelli, R.; Cannizzaro, S.; Torregrossa, M.V.; Cocco, P.; Forastiere, F.; et al. Exposure to benzene and childhood leukaemia: A pilot case-control study. BMJ Open 2013, 3, e002275. [Google Scholar] [CrossRef]
  163. Whitehead, T.P.; Crispo Smith, S.; Park, J.S.; Petreas, M.X.; Rappaport, S.M.; Metayer, C. Concentrations of persistent organic pollutants in California children’s whole blood and residential dust. Environ. Sci. Technol. 2015, 49, 9331–9340. [Google Scholar] [CrossRef]
  164. Whitehead, T.P.; Crispo Smith, S.; Park, J.S.; Petreas, M.X.; Rappaport, S.M.; Metayer, C. Concentrations of persistent organic pollutants in California women’s serum and residential dust. Environ. Res. 2015, 136, 57–66. [Google Scholar] [CrossRef] [Green Version]
  165. Zhang, Y.; Gao, Y.; Shi, R.; Chen, D.; Wang, X.; Kamijima, M.; Sakai, K.; Nakajima, T.; Khalequzzaman, M.; Zhou, Y.; et al. Household pesticide exposure and the risk of childhood acute leukemia in Shanghai, China. Environ. Sci. Pollut. Res. Int. 2015, 22, 11755–11763. [Google Scholar] [CrossRef] [PubMed]
  166. Chokkalingam, A.P.; Chun, D.S.; Noonan, E.J.; Pfeiffer, C.M.; Zhang, M.; Month, S.R.; Taggart, D.R.; Wiemels, J.L.; Metayer, C.; Buffler, P.A. Blood levels of folate at birth and risk of childhood leukemia. Cancer Epidemiol. Biomark. Prev. 2013, 22, 1088–1094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Rendic, S.; Guengerich, F.P. Survey of Human Oxidoreductases and Cytochrome P450 Enzymes Involved in the Metabolism of Xenobiotic and Natural Chemicals. Chem. Res. Toxicol. 2015, 28, 38–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Lewis, D.F.; Ioannides, C.; Parke, D.V. Cytochromes P450 and Species Differences in Xenobiotic Metabolism and Activation of Carcinogen. Environ. Health Perspect. 1998, 106, 633–641. [Google Scholar] [CrossRef]
  169. Bernauer, U.; Vieth, B.; Ellrich, R.; Heinrich-Hirsch, B.; Jänig, G.R.; Gundert-Remy, U. CYP2E1 expression in bone marrow and its intra- and interspecies variability: Approaches for a more reliable extrapolation from one species to another in the risk assessment of chemicals. Arch. Toxicol. 2000, 73, 618–624. [Google Scholar] [CrossRef]
  170. Tillement, J.P.; Tremblay, D. Clinical Pharmacokinetic Criteria for Drug Research. In Comprehensive Medicinal Chemistry II; Taylor, J.B., Triggle, D.J., Eds.; Elsevier: Amsterdam, The Netherlands, 2007; pp. 11–30. [Google Scholar]
  171. Chen, L.; Guo, P.; Zhang, H.; Li, W.; Gao, C.; Huang, Z.; Fan, J.; Zhang, Y.; Li, X.; Liu, X.; et al. Benzene-induced mouse hematotoxicity is regulated by a protein phosphatase 2A complex that stimulates transcription of cytochrome P4502E1. J. Biol. Chem. 2019, 294, 2486–2499. [Google Scholar] [CrossRef] [Green Version]
  172. Henderson, R.F. Species differences in the metabolism of benzene. Environ. Health Perspect. 1996, 104 (Suppl. 6), 1173–1175. [Google Scholar]
  173. Yoon, B.I.; Li, G.X.; Kitada, K.; Kawasaki, Y.; Igarashi, K.; Kodama, Y.; Inoue, T.; Kobayashi, K.; Kanno, J.; Kim, D.Y.; et al. Mechanisms of benzene-induced hematotoxicity and leukemogenicity: cDNA microarray analyses using mouse bone marrow tissue. Environ. Health Perspect. 2003, 111, 1411–1420. [Google Scholar] [CrossRef] [Green Version]
  174. Snyder, R. Leukemia and Benzene. Int. J. Environ. Res. Public Health 2012, 9, 2875–2893. [Google Scholar] [CrossRef] [Green Version]
  175. Gut, I.; Nedelcheva, V.; Soucek, P.; Stopka, P.; Tichavská, B. Cytochromes P450 in benzene metabolism and involvement of their metabolites and reactive oxygen species in toxicity. Environ. Health Perspect. 1996, 104 (Suppl. S6), 1211–1218. [Google Scholar]
  176. Snyder, R.; Hedli, C.C. An overview of benzene metabolism. Environ. Health Perspect. 1996, 104 (Suppl. S6), 1165–1171. [Google Scholar] [PubMed]
  177. Valentine, J.L.; Lee, S.S.; Seaton, M.J.; Asgharian, B.; Farris, G.; Corton, J.C.; Gonzalez, F.J.; Medinsky, M.A. Reduction of benzene metabolism and toxicity in mice that lack CYP2E1 expression. Toxicol. Appl. Pharmacol. 1996, 141, 205–213. [Google Scholar] [CrossRef] [PubMed]
  178. Ross, D. Metabolic basis of benzene toxicity. Eur. J. Haematol. Suppl. 1996, 60 (Suppl. S60), 111–118. [Google Scholar] [CrossRef] [PubMed]
  179. Chen, H.; Eastmond, D.A. Topoisomerase inhibition by phenolic metabolites: A potential mechanism for benzene’s clastogenic effects. Carcinogenesis 1995, 16, 2301–2307. [Google Scholar] [CrossRef]
  180. Eastmond, D.A.; Smith, M.T.; Irons, R.D. An interaction of benzene metabolites reproduces the myelotoxicity observed with benzene exposure. Toxicol. Appl. Pharmacol. 1987, 91, 85–95. [Google Scholar] [CrossRef]
  181. Subrahmanyam, V.V.; Ross, D.; Eastmond, D.A.; Smith, M.T. Potential role of free radicals in benzene-induced myelotoxicity and leukemia. Free Radic. Biol. Med. 1991, 11, 495–515. [Google Scholar] [CrossRef]
  182. Farris, G.M.; Robinson, S.N.; Gaido, K.W.; Wong, B.A.; Wong, V.A.; Hahn, W.P. Benzene-induced hematotoxicity and bone marrow compensation in B6C3F1 mice. Fundam. Appl. Toxicol. 1997, 36, 119–129. [Google Scholar] [CrossRef]
  183. Kolachana, P.; Subrahmanyam, V.V.; Meyer, K.B.; Zhang, L.; Smith, M.T. Benzene and its phenolic metabolites produce oxidative DNA damage in HL60 cells in vitro and in the bone marrow in vivo. Cancer Res. 1993, 53, 1023–1026. [Google Scholar]
  184. Lee, E.W.; Garner, C.D. Effects of benzene on DNA strand breaks in vivo versus benzene metabolite-induced DNA strand breaks in vitro in mouse bone marrow cells. Toxicol. Appl. Pharmacol. 1991, 108, 497–508. [Google Scholar] [CrossRef]
  185. Niculescu, R.; Bradford, H.N.; Colman, R.W.; Kalf, G.F. Inhibition of the conversion of pre-interleukins-1-alpha and 1-beta to mature cytokines by p-benzoquinone, a metabolite of benzene. Chem. Biol. Interact. 1995, 98, 211–222. [Google Scholar] [CrossRef]
  186. Nepstad, I.; Hatfield, K.J.; Grønningsæter, I.S.; Reikvam, H. The PI3K-Akt-mTOR Signaling Pathway in Human Acute Myeloid Leukemia (AML) Cells. Int. J. Mol. Sci. 2020, 21, 907. [Google Scholar] [CrossRef] [Green Version]
  187. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the Phosphoinositide 3-kinase Pathway in Cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Thorpe, L.M.; Yuzugullu, H.; Zhao, J.J. PI3K in Cancer: Divergent Roles of Isoforms, Modes of Activation and Therapeutic Targeting. Nat. Rev. Cancer 2015, 15, 7–24. [Google Scholar] [CrossRef] [Green Version]
  189. Hernandez-Aya, L.F.; Gonzalez-Angulo, A.M. Targeting the Phosphatidylinositol 3-Kinase Signaling Pathway in Breast Cancer. Oncologist 2011, 16, 404–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Murray, M.; Zhou, F. Trafficking and Other Regulatory Mechanisms for Organic Anion Transporting Polypeptides and Organic Anion Transporters That Modulate Cellular Drug and Xenobiotic Influx and That Are Dysregulated in Disease. Br. J. Pharmacol. 2017, 174, 1908–1924. [Google Scholar] [CrossRef] [Green Version]
  191. Esteves, F.; Rueff, J.; Kranendonk, M. The Central Role of Cytochrome P450 in Xenobiotic Metabolism—A Brief Review on a Fascinating Enzyme Family. J. Xenobiot. 2021, 11, 94–114. [Google Scholar] [CrossRef]
  192. Roy, P.; Yu, L.J.; Crespi, C.L.; Waxman, D.J. Development of a substrateactivity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. Drug Metab. Dispos. 1999, 27, 655–666. [Google Scholar]
  193. Dong, L.M.; Potter, J.D.; White, E.; Ulrich, C.M.; Cardon, L.R.; Peters, U. Genetic susceptibility to cancer: The role of polymorphisms in candidate genes. JAMA 2008, 299, 2423–2436. [Google Scholar] [CrossRef] [Green Version]
  194. Agundez, J.A. Cytochrome P450 gene polymorphism and cancer. Curr. Drug Metab. 2004, 5, 211–224. [Google Scholar] [CrossRef]
  195. Bozina, N.; Bradamante, V.; Lovrić, M. Genetic polymorphism of metabolic enzymes P450 (CYP) as a susceptibility factor for drug response, toxicity, and cancer risk. Arh. Hig. Rada. Toksikol. 2009, 60, 217–242. [Google Scholar] [CrossRef]
  196. Majumdar, S.; Mondal, B.C.; Ghosh, M.; Dey, S.; Mukhopadhyay, A.; Chandra, S.; Dasgupta, U.B. Association of cytochrome P450, glutathioneS-transferase and N-acetyltransferase 2 gene polymorphisms with incidence of acute myeloid leukemia. Eur. J. Cancer Prev. 2008, 17, 125–132. [Google Scholar] [CrossRef] [PubMed]
  197. Berköz, M.; Yalin, S. Association of CYP2B6 G15631T polymorphism with acute leukemia susceptibility. Leuk. Res. 2009, 33, 919–923. [Google Scholar] [CrossRef]
  198. Yuan, Z.H.; Liu, Q.; Zhang, Y.; Liu, H.X.; Zhao, J.; Zhu, P. CYP2B6 gene single nucleotide polymorphisms and leukemia susceptibility. Ann. Hematol. 2011, 90, 293–299. [Google Scholar] [CrossRef]
  199. Voso, M.T.; Fabiani, E.; D’Alo’, F.; Guidi, F.; Di Ruscio, A.; Sica, S.; Pagano, L.; Greco, M.; Hohaus, S.; Leone, G. Increased risk of acute myeloid leukaemia due to polymorphisms in detoxification and DNA repair enzymes. Ann. Oncol. 2007, 18, 1523–1528. [Google Scholar] [CrossRef] [PubMed]
  200. Guengerich, F.P. Cytochrome P450 2E1 and its roles in disease. Chem. Biol. Interact. 2020, 322, 109056. [Google Scholar] [CrossRef]
  201. Hofmann, M.H.; Blievernicht, J.K.; Klein, K.; Saussele, T.; Schaeffeler, E.; Schwab, M.; Zanger, U.M. Aberrant splicing caused by single nucleotide polymorphism c.516G.T [Q172H], a marker of CYP2B6*6, is responsible for decreased expression and activity of CYP2B6 in liver. J. Pharmacol. Exp. Ther. 2008, 325, 284–292. [Google Scholar] [CrossRef]
  202. Felix, C.A.; Walker, A.H.; Lange, B.J.; Williams, T.M.; Winick, N.J.; Cheung, N.K.; Lovett, B.D.; Nowell, P.C.; Blair, I.A.; Rebbeck, T.R. Association of CYP3A4 genotype with treatment-related leukemia. Proc. Natl. Acad. Sci. USA 1998, 95, 13176–13181. [Google Scholar] [CrossRef] [Green Version]
  203. Rebbeck, T.R.; Jaffe, J.M.; Walker, A.H.; Wein, A.J.; Malkowicz, S.B. Modification of clinical presentation of prostate tumors by a novel genetic variant in CYP3A4. J. Natl. Cancer Inst. 1998, 90, 1225–1229. [Google Scholar] [CrossRef] [Green Version]
  204. Ball, S.E.; Scatina, J.; Kao, J.; Ferron, G.M.; Fruncillo, R.; Mayer, P.; Weinryb, I.; Guida, M.; Hopkins, P.J.; Warner, N.; et al. Population distribution and effects on drug metabolism of a genetic variant in the 5′ promoter region of CYP3A4. Clin. Pharmacol. Ther. 1999, 66, 288–294. [Google Scholar] [CrossRef]
  205. Paris, P.L.; Kupelian, P.A.; Hall, J.M.; Williams, T.L.; Levin, H.; Klein, E.A.; Casey, G.; Witte, J.S. Association between a CYP3A4 genetic variant and clinical presentation in African-American prostate cancer patients. Cancer Epidemiol. Biomark. Prev. 1999, 8, 901–905. [Google Scholar]
  206. Croom, E. Metabolism of Xenobiotics of Human Environments. In Progress in Molecular Biology and Translational Science; Ernest, H., Ed.; Elsevier: Amsterdam, The Netherlands, 2012; Volume 112, pp. 31–88. [Google Scholar]
  207. Juchau, M.R.; Chen, H. Developmental Enzymology. In Handbook of Developmental Neurotoxicology; William, S., Jr., Louis, W.C., Eds.; Elsevier: Amsterdam, The Netherlands, 1998; pp. 321–337. [Google Scholar]
  208. Evans, T.J. Chapter 2—Toxicokinetics and Toxicodynamics. In Small Animal Toxicology, 3rd ed.; Peterson, M.E., Talcott, P.A., Eds.; W.B. Saunders: Saint Louis, MO, USA, 2013; pp. 13–19. [Google Scholar]
  209. Johnson, C.H.; Patterson, A.D.; Idle, J.R.; Gonzalez, F.J. Xenobiotic Metabolomics: Major Impact on the Metabolome. Annu. Rev. Pharmacol. Toxicol. 2012, 52, 37–56. [Google Scholar] [CrossRef] [PubMed]
  210. Clarke, G.; Sandhu, K.V.; Griffin, B.T.; Dinan, T.G.; Cryan, J.F.; Hyland, N.P. Gut reactions: Breaking down xenobiotic-microbiome interactions. Pharmacol. Rev. 2019, 71, 198–224. [Google Scholar] [CrossRef] [Green Version]
  211. Nelson, D.R. Cytochrome P450: Structure, Mechanism, and Biochemistry, 3rd ed Edited by Paul R. Ortiz de Montellano (University of California, San Francisco). J. Am. Chem. Soc. 2005, 127, 12147–12148. [Google Scholar] [CrossRef]
  212. Michalopoulos, G.K. Liver regeneration. J. Cell. Physiol. 2007, 213, 286–300. [Google Scholar] [CrossRef] [PubMed]
  213. Stanley, L.A. Drug Metabolism. In Pharmacognosy; Simone, B., Rupika, D., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 527–545. [Google Scholar]
  214. Jain, R.K.; Kapur, M.; Labana, S.; Lal, B.; Sarma, P.M.; Bhattacharya, D.; Thakur, I.S. Microbial diversity: Application of microorganisms for the biodegradation of xenobiotics. Curr. Sci. 2005, 89, 101–112. [Google Scholar]
  215. Jancova, P.; Siller, M. Phase II Drug Metabolism. In Topics on Drug Metabolism; Paxton, J., Ed.; InTech: London, UK, 2012; pp. 35–60. [Google Scholar]
  216. Gan, J.; Ma, S.; Zhang, D. Non-Cytochrome P450-Mediated Bioactivation and Its Toxicological Relevance. Drug Metab. Rev. 2016, 48, 473–501. [Google Scholar] [CrossRef]
  217. Penner, N.; Woodward, C.; Prakash, C.; Surapaneni, S. Drug metabolizing enzymes and biotransformation reactions. In ADME-Enabling Technologies in Drug Design and Development; Zhang, D., Surapaneni, S., Eds.; John Wiley & Sons, Inc.: New York, NY, USA, 2012; pp. 545–565. [Google Scholar]
  218. Manikandan, P.; Nagini, S. Cytochrome P450 Structure, Function and Clinical Significance: A Review. Curr. Drug Targets 2018, 19, 38–54. [Google Scholar] [CrossRef]
  219. Furge, L.L.; Guengerich, F.P. Cytochrome P450 Enzymes in Drug Metabolism and Chemical Toxicology: An Introduction. Biochem. Mol. Biol. Educ. 2006, 34, 66–74. [Google Scholar] [CrossRef] [PubMed]
  220. Bernhardt, R. Cytochromes P450 as Versatile Biocatalysts. J. Biotechnol. 2006, 124, 128–145. [Google Scholar] [CrossRef]
  221. Grillo, M.P. Bioactivation by Phase-II-Enzyme-Catalyzed Conjugation of Xenobiotics. In Encyclopedia of Drug Metabolism and Interactions; Lyubimov, A.V., Ed.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2012. [Google Scholar]
  222. Bachmann, K. Drug Metabolism. In Pharmacology; Miles, H., William, M., Kenneth, B., Eds.; Elsevier: Amsterdam, The Netherlands, 2009; pp. 131–173. [Google Scholar]
  223. Iyanagi, T. Molecular Mechanism of Phase I and Phase II Drug-Metabolizing Enzymes: Implications for Detoxification. In International Review of Cytology; Elsevier: Amsterdam, The Netherlands, 2007; Volume 260, pp. 35–112. [Google Scholar]
  224. Roy, U.; Barber, P.; Tse-Dinh, Y.C.; Batrakova, E.V.; Mondal, D.; Nair, M. Role of MRP Transporters in Regulating Antimicrobial Drug Inefficacy and Oxidative Stress-Induced Pathogenesis during HIV-1 and TB Infections. Front. Microbiol. 2015, 6, 948. [Google Scholar] [CrossRef] [Green Version]
  225. Döring, B.; Petzinger, E. Phase 0 and Phase III Transport in Various Organs: Combined Concept of Phases in Xenobiotic Transport and Metabolism. Drug Metab. Rev. 2014, 46, 261–282. [Google Scholar] [CrossRef]
  226. Petzinger, E.; Geyer, J. Drug Transporters in Pharmacokinetics. Naunyn. Schmiedebergs Arch. Pharmacol. 2006, 372, 465–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Almazroo, O.A.; Miah, M.K.; Venkataramanan, R. Drug Metabolism in the Liver. Clin. Liver Dis. 2017, 21, 1–20. [Google Scholar] [CrossRef] [PubMed]
  228. Zanger, U.M.; Schwab, M. Cytochrome P450 Enzymes in Drug Metabolism: Regulation of Gene Expression, Enzyme Activities, and Impact of Genetic Variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef]
  229. Frederiks, C.N.; Lam, S.W.; Guchelaar, H.J.; Boven, E. Genetic Polymorphisms and Paclitaxel- or Docetaxel-Induced Toxicities: A Systematic Review. Cancer Treat. Rev. 2015, 41, 935–950. [Google Scholar] [CrossRef]
  230. Annalora, A.J.; Marcus, C.B.; Iversen, P.L. Alternative Splicing in the Cytochrome P450 Superfamily Expands Protein Diversity to Augment Gene Function and Redirect Human Drug Metabolism. Drug Metab. Dispos. 2017, 45, 375–389. [Google Scholar] [CrossRef] [Green Version]
  231. Omura, T. Future Perception in P450 Research. J. Inorg. Biochem. 2018, 186, 264–266. [Google Scholar] [CrossRef]
  232. Hartman, J.H.; Martin, H.C.; Caro, A.A.; Pearce, A.R.; Miller, G.P. Subcellular localization of rat CYP2E1 impacts metabolic efficiency toward common substrates. Toxicology 2015, 338, 47–58. [Google Scholar] [CrossRef] [Green Version]
  233. Hartman, J.H.; Miller, G.P.; Meyer, J.N. Toxicological implications of mitochondrial localization of CYP2E1. Toxicol. Res. 2017, 6, 273–289. [Google Scholar] [CrossRef] [Green Version]
  234. Yamazoe, Y.; Ito, K.; Yoshinari, K. Construction of a CYP2E1-template system for prediction of the metabolism on both site and preference order. Drug Metab. Rev. 2011, 43, 409–439. [Google Scholar] [CrossRef] [PubMed]
  235. Aydin-Sayitoglu, M.; Hatirnaz, O.; Erensoy, N.; Ozbek, U. Role of CYP2D6, CYP1A1, CYP2E1, GSTT1, and GSTM1 genes in the susceptibility to acute leukemias. Am. J. Hematol. 2006, 81, 162–170. [Google Scholar] [CrossRef] [PubMed]
  236. Zhang, J.; Yin, L.H.; Liang, G.Y.; Liu, R.; Fan, K.H.; Pu, Y.P. Detection of CYP2E1, a genetic biomarker of susceptibility to benzene metabolism toxicity in immortal human lymphocytes derived from the Han Chinese population. Biomed. Environ. Sci. 2010, 24, 300–309. [Google Scholar]
  237. Bolufer, P.; Collado, M.; Barragan, E.; Calasanz, M.J.; Colomer, D.; Tormo, M.; González, M.; Brunet, S.; Batlle, M.; Cervera, J. Profile of polymorphisms of drug-metabolising enzymes and the risk of therapy-related leukaemia. Br. J. Haematol. 2007, 136, 590–596. [Google Scholar] [CrossRef]
  238. Han, X.; Zheng, T.; Foss, F.M.; Lan, Q.; Holford, T.R.; Rothman, N.; Ma, S.; Zhang, Y. Genetic polymorphisms in the metabolic pathway and non-Hodgkin lymphoma survival. Am. J. Hematol. 2010, 85, 51–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Ingelman-Sundberg, M.; Ekström, G.; Tindberg, N. Lipid peroxidation dependent on ethanol-inducible cytochrome p-450 from rat liver. Adv. Biosci. 1988, 71, 43–47. [Google Scholar]
  240. Weltman, M.D.; Farrell, G.C.; Hall, P.; Ingelman-Sundberg, M.; Liddle, C. Hepatic cytochrome p450 2e1 is increased in patients with nonalcoholic steatohepatitis. Hepatology 1998, 27, 128–133. [Google Scholar] [CrossRef]
  241. Larson, R.A.; Wang, Y.; Banerjee, M.; Wiemels, J.; Hartford, C.; Beau, M.M.L.; Smith, M.T. Prevalence of the inactivating 609C→T polymorphism in the NAD(P)H: Quinone oxidoreductase (NQO1) gene in patients with primary and therapy-related myeloid leukemia. Blood 1999, 94, 803–807. [Google Scholar] [CrossRef] [PubMed]
  242. Wiemels, J.L.; Pagnamenta, A.; Taylor, G.M.; Eden, O.B.; Alexander, F.E.; Greaves, M.F. A lack of a functional NAD(P)H: Quinone oxidoreductase allele is selectively associated with pediatric leukemias that have MLL fusions. Cancer Res. 1999, 59, 4095–4099. [Google Scholar]
  243. Smith, M.T.; Wang, Y.; Kane, E.; Rollinson, S.; Wiemels, J.L.; Roman, E.; Roddam, P.; Cartwright, R.; Morgan, G. Low NAD(P)H: Quinone oxidoreductase 1 activity is associated with increased risk of acute leukemia in adults. Blood 2001, 97, 1422–1426. [Google Scholar] [CrossRef] [Green Version]
  244. Sun, X.; Zhang, W.; Ramdas, L.; Stivers, D.N.; Jones, D.M.; Kantarjian, H.M.; Estey, E.H.; Vadhan-Raj, S.; Medeiros, L.J.; Bueso-Ramos, C.E. Comparative analysis of genes regulated in acute myelomonocytic leukemia with and without Inv (16)(p13q22) using microarray techniques, real-time PCR, immunohistochemistry, and flow cytometry immunophenotyping. Mod. Pathol. 2007, 20, 811–820. [Google Scholar] [CrossRef] [Green Version]
  245. Keating, G.M. Azacitidine. Drugs 2012, 72, 1111–1136. [Google Scholar] [CrossRef]
  246. Bernstein, I.; Byun, H.M.; Mohrbacher, A.; Douer, D.; Gorospe, G., 3rd; Hergesheimer, J.; Groshen, S.; O’Connell, C.; Yang, A.S. A Phase I biological study of azacitidine (Vidaza™) to determine the optimal dose to inhibit DNA methylation. Epigenetics 2010, 5, 750–757. [Google Scholar] [CrossRef] [Green Version]
  247. Leone, G.; Teofili, L.; Voso, M.T.; Lübbert, M. DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias. Haematologica 2002, 87, 1324–1341. [Google Scholar] [PubMed]
  248. Marcucci, G.; Silverman, L.; Eller, M.; Lintz, L.; Beach, C.L. Bioavailability of azacitidine subcutaneous versus intravenous in patients with the myelodysplastic syndromes. J. Clin. Pharmacol. 2005, 45, 597–602. [Google Scholar] [CrossRef]
  249. Laille, E.; Goel, S.; Mita, A.C.; Gabrail, N.Y.; Kelly, K.; Liu, L.; Songer, S.; Beach, C.L. A phase I study in patients with solid or hematologic malignancies of the dose proportionality of subcutaneous Azacitidine and its pharmacokinetics in patients with severe renal impairment. Pharmacotherapy 2014, 34, 440–451. [Google Scholar] [CrossRef]
  250. Dugan, J.; Pollyea, D. Enasidenib for the treatment of acute myeloid leukemia. Exp. Rev. Clin. Pharmacol. 2018, 11, 755–760. [Google Scholar] [CrossRef]
  251. Dogra, R.; Bhatia, R.; Shankar, R.; Bansal, P.; Rawal, R.K. Enasidenib: First mutant IDH2 inhibitor for the treatment of refractory and relapsed acute myeloid leukemia. Anti-Cancer Agents Med. Chem. 2018, 18, 1936–1951. [Google Scholar] [CrossRef] [PubMed]
  252. Stein, E.M. Enasidenib, a targeted inhibitor of mutant IDH2 proteins for treatment of relapsed or refractory acute myeloid leukemia. Future Oncol. 2018, 14, 23–40. [Google Scholar] [CrossRef] [PubMed]
  253. Abou Dalle, I.; Dinardo, C.D. The role of enasidenib in the treatment of mutant IDH2 acute myeloid leukemia. Ther. Adv. Hematol. 2018, 9, 163–173. [Google Scholar] [CrossRef] [Green Version]
  254. Cheng, Y.; Wang, X.; Tong, Z.; Reyes, J.; Carayannopoulos, L.; Zhou, S.; Li, Y. Assessment of Transporter-Mediated Drug Interactions for Enasidenib Based on a Cocktail Study in Patients With Relapse or Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome. J. Clin. Pharmacol. 2022, 62, 494–504. [Google Scholar] [CrossRef]
  255. Martinelli, G.; Oehler, V.G.; Papayannidis, C.; Courtney, R.; Shaik, M.N.; Zhang, X.; O’Connell, A.; McLachlan, K.R.; Zheng, X.; Radich, J.; et al. Treatment with PF-04449913, an oral smoothened antagonist, in patients with myeloid malignancies: A phase 1 safety and pharmacokinetics study. Lancet Haematol. 2015, 2, e339–e346. [Google Scholar] [CrossRef] [PubMed]
  256. Shaik, M.N.; Hee, B.; Wei, H.; LaBadie, R.R. Evaluation of the effect of rifampin on the pharmacokinetics of the Smoothened inhibitor glasdegib in healthy volunteers. Br. J. Clin. Pharmacol. 2018, 84, 1346–1353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. James, A.J.; Smith, C.C.; Litzow, M.; Perl, A.E.; Altman, J.K.; Shepard, D.; Kadokura, T.; Souda, K.; Patton, M.; Lu, Z.; et al. Pharmacokinetic Profile of Gilteritinib: A Novel FLT-3 Tyrosine Kinase Inhibitor. Clin. Pharmacokinet. 2020, 59, 1273–1290. [Google Scholar] [CrossRef] [Green Version]
  258. Sechaud, R.; Sinclair, K.; Grosch, K.; Ouatas, T.; Pathak, D. Evaluation of drug-drug interactions between midostaurin and strong CYP3A4 inhibitors in patients with FLT-3-mutated acute myeloid leukemia (AML). Cancer Chemother. Pharmacol. 2022, 90, 19–27. [Google Scholar] [CrossRef]
  259. De la Garza-Salazar, F.; Peña-Lozano, S.P.; Gómez-Almaguer, D.; Colunga-Pedraza, P.R. Orbital myeloid sarcoma treated with low-dose venetoclax and a potent cytochrome P450 inhibitor. J. Oncol. Pharm. Pract. 2023, 29, 493–497. [Google Scholar] [CrossRef] [PubMed]
  260. Relias, V.; McBride, A.; Newman, M.J.; Paul, S.; Saneeymehri, S.; Stanislaus, G.; Tobin, J.; Hoang, C.J.; Ryan, J.C.; Galinsky, I. Glasdegib plus low-dose cytarabine for acute myeloid leukemia: Practical considerations from advanced practitioners and pharmacists. J. Oncol. Pharm. Pract. 2021, 27, 658–672. [Google Scholar] [CrossRef] [PubMed]
  261. Shaik, M.N.; LaBadie, R.R.; Rudin, D.; Levin, W.J. Evaluation of the effect of food and ketoconazole on the pharmacokinetics of the smoothened inhibitor PF-04449913 in healthy volunteers. Cancer Chemother. Pharmacol. 2014, 74, 411–418. [Google Scholar] [CrossRef] [PubMed]
  262. Lin, S.; Shaik, N.; Martinelli, G.; Wagner, A.J.; Cortes, J. Population Pharmacokinetics of Glasdegib in Patients With Advanced Hematologic Malignancies and Solid Tumors. J. Clin. Pharmacol. 2020, 60, 605–616. [Google Scholar] [CrossRef] [Green Version]
  263. Ostgard, L.S.; Norgaard, M.; Sengelov, H.; Medeiros, B.C.; Kjeldsen, L.; Overgaard, U.M.; Severinsen, M.T.; Marcher, C.W.; Jensen, M.K.; Norgaard, J.M. Improved outcome in acute myeloid leukemia patients enrolled in clinical trials: A national population-based cohort study of danish intensive chemotherapy patients. Oncotarget 2016, 7, 72044–72056. [Google Scholar] [CrossRef] [Green Version]
  264. Pleyer, L.; Döhner, H.; Dombret, H.; Seymour, J.F.; Schuh, A.C.; Beach, C.; Swern, A.S.; Burgstaller, S.; Stauder, R.; Girschikofsky, M.; et al. Azacitidine for Front-Line Therapy of Patients with AML: Reproducible Efficacy Established by Direct Comparison of International Phase 3 Trial Data with Registry Data from the Austrian Azacitidine Registry of the AGMT Study Group. Int. J. Mol. Sci. 2017, 18, 415. [Google Scholar] [CrossRef] [Green Version]
  265. Tombak, A.; Uçar, M.A.; Akdeniz, A.; Tiftik, E.N.; Şahin, D.G.; Akay, O.M.; Yıldırım, M.; Nevruz, O.; Kis, C.; Gürkan, E.; et al. The Role of Azacitidine in the Treatment of Elderly Patients with Acute Myeloid Leukemia: Results of a Retrospective Multicenter Study. Turk. J. Hematol. 2016, 33, 273–280. [Google Scholar] [CrossRef]
  266. Lyer, S.G.; Stanchina, M.; Bradley, T.J.; Watts, J. Profile of Glasdegib for the Treatment of Newly Diagnosed Acute Myeloid Leukemia (AML): Evidence to Date. Cancer Manag. Res. 2022, 14, 2267–2272. [Google Scholar]
  267. Sievers, E.L.; Larson, R.A.; Stadtmauer, E.A.; Estey, E.; Löwenberg, B.; Dombret, H.; Karanes, C.; Theobald, M.; Bennett, J.M.; Sherman, M.L.; et al. Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. J. Clin. Oncol. 2001, 19, 3244–3254. [Google Scholar] [CrossRef] [PubMed]
  268. Heiblig, M.; Elhamri, M.; Tigaud, I.; Plesa, A.; Barraco, F.; Labussière, H.; Ducastelle, S.; Michallet, M.; Nicolini, F.; Plesa, C.; et al. Treatment with Low-Dose Cytarabine in Elderly Patients (Age 70 Years or Older) with Acute Myeloid Leukemia: A Single Institution Experience. Mediterr. J. Hematol. Infect. Dis. 2016, 8, e2016009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  269. Richard-Carpentier, G.; DiNardo, C.D. Venetoclax for the treatment of newly diagnosed acute myeloid leukemia in patients who are ineligible for intensive chemotherapy. Ther. Adv. Hematol. 2019, 10, 2040620719882822. [Google Scholar] [CrossRef] [Green Version]
  270. Wei, A.H.; Hou, Z.; Fiedler, W.; Lin, T.L.; Walter, R.B.; Enjeti, A.; Tiong, I.S.; Savona, M.; Lee, S.; Chyla, B.; et al. Venetoclax Combined With Low-Dose Cytarabine for Previously Untreated Patients With Acute Myeloid Leukemia: Results From a Phase Ib/II Study. J. Clin. Oncol. 2019, 37, 1277–1284. [Google Scholar] [CrossRef]
  271. Craig, W.J. Health-promoting properties of common herbs. Am. J. Clin. Nutr. 1999, 70 (Suppl. S3), 491S–499S. [Google Scholar] [CrossRef] [Green Version]
  272. Rimm, E.B.; Stampfer, M.J.; Ascherio, A.; Giovannucci, E.; Colditz, G.A.; Willett, W.C. Vitamin E consumption and the risk of coronary heart disease in men. N. Engl. J. Med. 1993, 328, 1450–1456. [Google Scholar] [CrossRef] [Green Version]
  273. Gaziano, J.M.; Manson, J.E.; Branch, L.G.; Colditz, G.A.; Willett, W.C.; Buring, J.E. A prospective study of consumption of carotenoids in fruits and vegetables and decreased cardiovascular mortality in the elderly. Ann. Epidemiol. 1995, 5, 255–260. [Google Scholar] [CrossRef]
  274. Sahyoun, N.R.; Jacques, P.F.; Russell, R.M. Carotenoids, vitamins C and E, and mortality in an elderly population. Am. J. Epidemiol. 1996, 144, 501–511. [Google Scholar] [CrossRef]
  275. Sandoval, C.; Mella, L.; Godoy, K.; Adeli, K.; Farías, J. β-Carotene Increases Activity of Cytochrome P450 2E1 during Ethanol Consumption. Antioxidants 2022, 11, 1033. [Google Scholar] [CrossRef]
  276. Sandoval, C.; Vásquez, B.; Souza-Mello, V.; Adeli, K.; Mandarim-de-Lacerda, C.; del Sol, M. Morphoquantitative effects of oral β-carotene supplementation on liver of C57BL/6 mice exposed to ethanol consumption. Int. J. Clin. Exp. Pathol. 2019, 12, 1713–1722. [Google Scholar] [PubMed]
  277. Sandoval, C.; Vásquez, B.; Vasconcellos, A.; Souza-Mello, V.; Adeli, K.; Mandarim-de-Lacerda, C.; del Sol, M. Oral Supplementation of β-Carotene Benefits the Hepatic Structure and Metabolism in Mice (Mus musculus) Exposed to A Chronic Ethanol Consumption. Sains Malays. 2022, 51, 285–296. [Google Scholar] [CrossRef]
Figure 1. Flow diagram.
Figure 1. Flow diagram.
Ijms 24 06031 g001
Figure 2. Cytogenetic abnormalities and mutations involved in development in acute myeloid leukemia (AML).
Figure 2. Cytogenetic abnormalities and mutations involved in development in acute myeloid leukemia (AML).
Ijms 24 06031 g002
Figure 3. Processes involved in reactive oxygen species production during acute myeloid leukemia cells and their significance in leukemogenesis. Abbreviations: mDNA, mitochondrial DNA; mETC, mitochondrial electron transport chain; NADPH: Nicotinamide adenine dinucleotide phosphate; NOX: NADPH oxidase; XO, xanthine oxidase; XDH, xanthine dehydrogenase.
Figure 3. Processes involved in reactive oxygen species production during acute myeloid leukemia cells and their significance in leukemogenesis. Abbreviations: mDNA, mitochondrial DNA; mETC, mitochondrial electron transport chain; NADPH: Nicotinamide adenine dinucleotide phosphate; NOX: NADPH oxidase; XO, xanthine oxidase; XDH, xanthine dehydrogenase.
Ijms 24 06031 g003
Table 1. Recurrent mutations in acute myeloid leukemia.
Table 1. Recurrent mutations in acute myeloid leukemia.
Functional ClassSpecific Example MutationsReferences
Signaling and kinase pathwaysFLT3, KRAS, NRAS, KIT, PTPN11, and NF1[15]
DNA methylation and chromatin modificationDNMT3A, IDH1, IDH2, TET2, ASXL1, EZH2, and MLL/KMT2A[15,34,35]
NucleophosminNPM1[36,37]
Transcription factorsCEBPA, RUNX1, and GATA2[38,39,40,41,42,43,44,45]
Tumor suppressorsTP53[46,47,48]
Spliceosome complexSRSF2, U2AF1, SF3B1, and ZRSR2[49,50]
Cohesin complexRAD21, STAG1, STAG2, SMC1A, and SMC3[51,52]
Table 2. Classification of acute myeloid leukemia based on the National Comprehensive Cancer Network’s cytogenetic and molecular criteria.
Table 2. Classification of acute myeloid leukemia based on the National Comprehensive Cancer Network’s cytogenetic and molecular criteria.
TypeDiagnostic Criteria
Acute myeloid leukemia with minimal differentiationBlasts are negative (<3%) for MPO and SBB.
Expression of two or more myeloid-associated antigens, such as CD13, CD33, and CD117.
Acute myeloid leukemia without maturation≥3% blasts positive for MPO or SBB and negative for NSE.
Maturing cells of the granulocytic lineage constitute <10% of the nucleated bone marrow cells.
Expression of two or more myeloid-associated antigens, such as MPO, CD13, CD33, and CD117.
Acute myeloid leukemia with maturation≥3% blasts positive for MPO or SBB.
Maturing cells of the granulocytic lineage constitute ≥10% of the nucleated bone marrow cells.
Monocyte lineage cells constitute <20% of bone marrow cells.
Expression of two or more myeloid–associated antigens, such as MPO, CD13, CD33, and CD117.
Acute basophilic leukemiaBlasts and immature/mature basophils with metachromasia on toluidine blue staining.
Blasts are negative for cytochemical MPO, SBB, and NSE.
No expression of strong CD117 equivalent (to exclude mast cell leukemia).
Acute myelomonocytic leukemia≥20% monocytes and their precursors.
≥20% maturing granulocytic cells.
≥3% of blasts positive for MPO.
Acute monocytic leukemia≥80% monocytes and/or their precursors (monoblasts and/or promonocytes).
<20% maturing granulocytic cells.
Blasts and promonocytes expressing at least two monocytic markers including CD11c, CD14, CD36 and CD64, or NSE.
Acute erythroid leukemia≥30% immature erythroid cells (proerythroblasts)
Bone marrow with erythroid predominance, usually ≥80% of cellularity
Acute megakaryoblastic leukemiaBlasts express at least one or more of the platelet glycoproteins: CD41 (glycoprotein llb), CD61 (glycoprotein IIIa), or CD42b (glycoprotein lb)
MPO: myeloperoxidase; NSE: nonspecific esterase–butyrate; SBB: Sudan Black B.
Table 3. Drugs approved for the treatment of relapsed or refractory acute myeloid leukemia and the prognoses after treatment.
Table 3. Drugs approved for the treatment of relapsed or refractory acute myeloid leukemia and the prognoses after treatment.
DrugOne-Year Survival (%)References
AzacitidineComplete remission for patients >60 years old: 61.0%[263]
Azacitidine45.8%[264]
AzacitidineComplete remission, complete remission with incomplete recovery, partial remission in patients >60 years old: 36.2%[265]
EnasidenibComplete remission: 19.3%[261]
EnasidenibComplete remission with incomplete hematologic recovery: 6.8%[261]
EnasidenibPartial remission: 6.3%[261]
EnasidenibMorphologic leukemia free state: 8.0%[261]
GlasdegibPatients with newly diagnosed acute myeloid leukemia: 20.0%[266]
Gemtuzumab ozogamicinComplete remission in patients with a median age of 61 years old: 30%[267]
GilteritinibComplete remission with full or partial hematologic recovery in patients with FLT3 mutations: 34.0%[253]
Low-dose cytarabineComplete remission for patients >70 years old: 7.0%[268]
MidostaurinComplete remission in patients between 18 to 59 years old, and FLT3 mutations: 58.9%[18]
VenetoclaxComplete remission + complete remission with incomplete hematological recovery: 54% (in combination with low-dose cytarabine)[269]
Venetoclax + low-dose cytarabineComplete remission/ Complete remission with incomplete blood count recovery for patients >60 years old: 54.0%[270]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sandoval, C.; Calle, Y.; Godoy, K.; Farías, J. An Updated Overview of the Role of CYP450 during Xenobiotic Metabolization in Regulating the Acute Myeloid Leukemia Microenvironment. Int. J. Mol. Sci. 2023, 24, 6031. https://doi.org/10.3390/ijms24076031

AMA Style

Sandoval C, Calle Y, Godoy K, Farías J. An Updated Overview of the Role of CYP450 during Xenobiotic Metabolization in Regulating the Acute Myeloid Leukemia Microenvironment. International Journal of Molecular Sciences. 2023; 24(7):6031. https://doi.org/10.3390/ijms24076031

Chicago/Turabian Style

Sandoval, Cristian, Yolanda Calle, Karina Godoy, and Jorge Farías. 2023. "An Updated Overview of the Role of CYP450 during Xenobiotic Metabolization in Regulating the Acute Myeloid Leukemia Microenvironment" International Journal of Molecular Sciences 24, no. 7: 6031. https://doi.org/10.3390/ijms24076031

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop