AML are blood cancers involving rapid growth and accumulation of abnormal myeloid cells in the BM, PB, and extramedullary sites [
1]. The consequent BM failure causes cytopenia and related clinical features, such as anemia, increased infection susceptibility, and bleeding tendencies. About the onset and the previous patient’s clinical history, AML can arise as primary (newly diagnosed, ND-AML), therapy-related (t-AML), and secondary (s-AML) categories [
1,
4,
20]. In most cases, AML in older patients emerges alongside previous hematologic malignancies [
20,
21], such as myeloproliferative disorders (MPNs) [
20], myelodysplastic neoplasms (MDS) [
22] and potentially predisposing and long-lasting premalignant conditions, such as clonal hematopoiesis (CH), which is prevalent in individuals over 70 [
23]. AMLs arise from BM from leukemia stem cells (LCSs) [
24], deriving by the malignant transformation of hematopoietic stem cells (HCSs) by acquired genetic mutations [
2], resulting in altered cell self-renewal mechanisms, uncontrolled proliferation, and impaired apoptosis [
25] of the leukemic clone. Therefore, accumulating acquired somatic mutations, interfering with the normal development of immature HSCs, drives the leukemic process. In this regard, CH-related genes (
ASXL1, TET2, SRSF2, and
DNMT3A) are most frequently mutated in AML older patients [
2,
23]. In most cases, they can be considered the first hit on the path to malignant transformation and appear to be a relatively early event in leukemogenesis. In contrast, other genomic abnormalities, including mutations in
FLT3, NRAS, and RUNX1 [
2,
26], tend to be acquired later during leukemia development. Acquired genetic mutations that characterize AMLs can be categorized into several pathways [
2,
26]. Indeed, activating mutations involving genes that sustain cell proliferation and survival, such as
FLT3, NRAS, KRAS, and c-KIT, lead to the uncontrolled growth of immortalized LSCs. In addition, mutations in genes related to DNA repair (e.g.,
TP53) [
27,
28], cohesin complex (e.g., RAD21), and spliceosome machinery (e.g.,
SF3B1, SRSF2, U2AF1, and
ZRSR2) can contribute to leukemogenesis and disease progression [
2]. Furthermore, other mutations, such as transcription factors (e.g.,
CEBPA and
RUNX1) and epigenetic regulators (e.g.,
TET2, IDH1/2, DNMT3A, ASXL1, and
EZH2) affect genes involved in hematopoietic differentiation [
2]. Again, mutations in genes encoding epigenetic modifiers, such as histone modification and DNA methylation, play a critical role in the pathogenesis of AML, leading to aberrant epigenetic patterns, altered gene expression profiles, and the arrest of the HCSs differentiation [
2,
26]. In s-AML, the most common driver mutation in patients with prior MPNs is
JAK2 V617F, accounting for 98% of polycythemia vera and 55–60% of essential thrombocythemia and myelofibrosis cases [
20]. In contrast, in MDS, the most frequent mutations affect members of the spliceosome, such as
SF3B1, SRSF2, U2AF1, and
ZRSR2, as well as genes involved in DNA methylation and chromatin remodeling, such as
TET2, DNMT3A, IDH1/2, and
ASXL1 [
22]. Before progressing to s-AML, patients acquire mutations in other genes, particularly in spliceosome members, such as
SF3B1, called “short-term” mutations that lead to rapid malignant transformation. In contrast, mutations in genes such as
TP53 and
ATM are “long-term” as they develop over many years, but they also indicate a poor prognosis upon transformation [
2,
20]. Mutational profile separated two s-AML groups: those with
FLT3, PTPN11, WT1, IDH1, NPM1, IDH2, and
NRAS mutations, conferring a low-risk of progressive disease, whereas the presence of other gene mutations, such as
TP53, GATA2, KRAS, RUNX1,
STAG2, ASXL1, ZRSR2, and
TET2, are predictive of a high risk of leukemic evolution [
20].In addition, several non-genomic facilitating factors of leukemic pathways [
2], such as the epigenetic state [
2], immunological alterations, cytokine release dysregulations [
29,
30,
31,
32], and oxidative stress [
33], can impair the physiological state of HSCs. Indeed, an altered inflamed and immunosuppressive BM microenvironment [
30,
31,
32] can support LSCs survival and proliferation. In addition, dysregulated interactions of LSCc with stromal cells and altered cytokine signaling lead to disease progression and therapy resistance [
30]. In addition, leukemic cells often show genotype-specific metabolic changes, which lead to alterations in epigenetic and functional factors, ultimately resulting in the upregulation or facilitation of oncogenic pathways [
34,
35]. Furthermore, different types of AML exhibit diverse and highly adaptable energy metabolism, which may have therapeutic implications [
36]. In addition, profiling bioactive molecules such as sphingolipids represents a predictive tool for AML. These molecules have differential activities in regulating cell proliferation, differentiation, apoptosis, and immune cell activation and have implications in AML pathogenesis and therapeutic resistance, thus potentially exerting a significant value for understanding and treating AML [
37]. Transcriptomic, proteomic, and phosphoproteomic data are valuable for understanding the underlying pathophysiology of AML beyond mutations, allowing these biological findings for the identification of four proteogenomic subtypes and the detection of specific drug response patterns [
38]. Furthermore, an emerging understanding of the role of tumoral angiogenesis and the impact of endothelial cell subsets in shaping BM niches have been reported [
39]. Moreover, it is relevant to note that AML cells can use different strategies to avoid ferroptosis cell death, controlled by three main cellular processes: iron metabolism, oxidative stress, and lipid metabolism [
40]. Additionally, transcription factors, such as
HOXA9, are overexpressed in approximately 70% of AML cases with poor prognosis, increased chemoresistance, and higher relapse rates [
41]. Lastly, genetic polymorphisms in DNA excision repair systems are essential in maintaining genomic integrity and stability [
42]. Therefore, the pathogenesis of AML involves a complex interplay of genetic mutations, epigenetic alterations, and disrupted cellular signaling pathways that induce the transformation of normal HSCs into LSCs with clonal and uncontrolled self-renewal properties, thus driving the disease progression [
2]. Understanding the molecular mechanisms underlying AML pathogenesis [
2] is essential for developing targeted therapies (
Table 3) [
43,
44,
45,
46,
47,
48,
49,
50] for specific biological targets [
43]. Notably, changes in
BCL-2 protein expression can promote cell survival or trigger apoptosis.
BCL-2, a member of this protein family, is located in the cytoplasm and controls apoptosis by capturing proapoptotic proteins. Thus, it prevents mitochondrial membrane permeabilization and cytochrome C release, which activates apoptosomes [
51].
BCL-2 is crucial for the survival and growth of leukemic cells. Specific compounds, such as venetoclax [
43,
51,
52], can target its abnormal activity. However, the most significant genetic changes observed in approximately 30% of AML cases involve mutations in the FMS-like tyrosine kinase 3 (
FLT-3) gene [
53,
55]. These mutations are found in CD34+ HSCs and regulate the early stages of blood cell formation.
FLT-3 belongs to a group of receptors known as class III receptor tyrosine kinases, which also include
PDGFR and
c-KIT. When a specific molecule binds to
FLT-3 at the cell membrane, it forms pairs, activating the cytoplasmic tyrosine kinase domain (
TKD), which, in turn, leads to signaling through various pathways such as
PIK3A, RAS, and
MAPK/ERK. The two main types of
FLT3 mutations are internal tandem duplications (
ITD) and
TKD point mutations. Both mutations result in continuous activation of the
FLT-3 receptor and uncontrolled growth of LSCs. At the AML onset, patients with
FLT-3 mutations present with increased blasts in BM and PB and higher white blood cell counts. In addition, patients with these mutations also tend to have shorter progression-free survival (PFS) and OS. In particular,
FLT-3 ITD mutations significantly affect the complexity of disease biology and prognosis [
53,
54,
55]. Other genetic abnormalities are potential targets for specific drugs [
43,
44,
56,
57]. For example, mutations in the active sites of isocitrate dehydrogenase 1 (
IDH1) and
IDH2, reported at frequencies of 6–16% and 8–19% for
IDH1 and
IDH2 are significant in AML [
43,
44,
56]. These enzymes convert isocitrate to α-ketoglutarate, producing nicotinamide adenine dinucleotide phosphate (NADH). Therefore, these gene mutations reduce the conversion of isocitrate to α-ketoglutarate and decrease NADPH-dependent conversion of α-ketoglutarate to 2-hydroxyglutarate (2-HG). In turn, the accumulation of 2-HG in cells competitively inhibits α-ketoglutarate-dependent processes, affecting cytosine 5-hydroxymethylation of DNA and leading to hypermethylation patterns in
IDH-mutant LCSs [
43,
57]. Furthermore, high levels of 2-HG inhibit cytochrome C oxidase, making cells more susceptible to apoptosis when
BCL-2 is inhibited [
58]. For AMLs characterized by mutations in specific genes such as lysine methyltransferase 2a (
KMT2A, also known as
MLL1) [
59] and nucleophosmin (
NPM1) [
60,
61], new compounds have been developed, such as Menin inhibitors [
62,
63,
64,
65,
66] which are in clinical investigations.
KMT2A, located on chromosome 11q23, is a DNA-binding protein essential for regular cellular growth. The interaction with some proteins, such as Menin, which regulates gene expression through histone methylation, influences its DNA binding. Abnormalities in the
KMT2A gene occur in 70–80% of cases of infant leukemia. They are rare in older AML patients [
43,
59] but are common in those with t-AML [
20,
59], particularly if they have received topoisomerase II inhibitors.
NPM1 is a nuclear chaperone protein that exerts several cellular functions, including ribosomal synthesis, stress response, and genomic stability [
61,
62]. In adult AML,
NPM1 is one of the most commonly mutated genes, occurring in 20–30% of cases [
43,
61,
62]. The
NPM1 gene encodes a multifunctional protein prevalently located in the nucleoli and shuttles between the nuclear and cytoplasmic compartments. Importantly,
NPM1-mutated AML cells exhibit abnormal cytoplasmic localization of mutant
NPM1c due to the loss of a nucleolar localization signal and gain of a nuclear export signal (NES) at the C-terminus. This NES interacts with the nucleus cell exporter Exportin-1 (
XPO1), causing accumulation of the
NPM1 mutant in the cytoplasm [
60,
61]. The overexpression of the HOX gene [
40], similar to
KMT2A rearranged AML, guides the
NPM-1 mutated AML development. Notably, overlapping features between t-
NPM1 and de novo
NPM1 AMLs suggest they can represent a single disease entity [
67]. Generally,
NPM1 mutations indicate a favorable prognosis without other genetic alterations, and the growth of
NPM1-mutated AML is responsive to Menin inhibitors [
62,
63,
64,
65,
66]. Additionally, nucleoporin 98 (
NUP98) [
67], a gene located on chromosome 11p15, is involved in nuclear membrane transport and acts as a transcription factor in the nucleoplasm. In approximately 1–2% of adult AML patients,
NUP98 fuses with one of over 30 different partners, contributing to the development of leukemia.
NUP98 fusions are usually associated with poor prognosis and may lead to resistance to chemotherapy [
66]. Like
MLL fusion and
NPM1 mutations,
NUP98 fusion proteins bind to chromatin near
HOX genes, causing their overexpression through various mechanisms, including altered DNA methylation and acetylation. The binding of these fusion genes to chromatin depends on both
MLL and Menin. In preclinical studies, leukemic cells with
NUP98 fusions responded to Menin inhibition [
62,
63,
64,
65,
66]. Again, the AML cell surface expresses some target proteins [
68], such as CD123 [
69,
70], CD33, CD37, and CD47 [
71]. Notably, CD123 is a primary diagnostic marker of a rare and aggressive hematodermic neoplasm, such as blastic plasmacytoid dendritic cell neoplasm (BPDCN) [
69,
70] which was included among AML in updated WHO-5 [
10] and new ICC [
11]. Again, the smoothened transmembrane protein mediates Hedgehog signaling [
72] and represents a therapeutic target for an available therapeutic compound [
68]. Thus, advances in genomic profiling and molecular characterization have enabled the development of innovative treatment approaches for patients with AML, particularly older and frail individuals unsuitable for ICT and allogeneic SCT [
13,
14].