Abstract
In the last few decades, the increasing human life expectancy has led to the inflation of the elderly population and consequently the escalation of age‐related disorders. Biological aging has been associated with the accumulation of somatic mutations in the Hematopoietic Stem Cell (HSC) compartment, providing a fitness advantage to the HSCs leading to clonal hematopoiesis, that includes non‐malignant and malignant conditions (i.e. Clonal Hematopoiesis of Indeterminate Potential, Myelodysplastic Syndrome and Acute Myeloid Leukemia). The Janus Kinase‐Signal Transducer and Activator of Transcription (JAK–STAT) pathway is a key player in both normal and malignant hematopoiesis. STATs, particularly STAT3 and STAT5, are greatly implicated in normal hematopoiesis, immunity, inflammation, leukemia, and aging. Here, the pleiotropic functions of JAK–STAT pathway in age‐associated hematopoietic defects and of STAT3 and STAT5 in normal hematopoiesis, leukemia, and inflammaging are reviewed. Even though great progress has been made in deciphering the role of STATs, further research is required to provide a deeper understanding of the molecular mechanisms of leukemogenesis, as well as novel biomarkers and therapeutic targets for improved management of age‐related disorders.
Keywords: aging, AML, leukemia, MDS, myeloid malignancy, signal transducers and activators of transcription, STAT3, STAT5
The increase in the aging population has resulted in the rise of age‐related disorders, including MDS and AML. The JAK–STAT pathway is essential for several cellular functions in the HSPCs. Deregulation of the STAT3/5 signaling may occur at several pathway stages (indicated by exclamation points) and has been associated with MDS and AML development through various mechanisms. Thus, STAT3/5 factors constitute ideal targets for developing improved therapeutic management strategies.
Abbreviations
ALL, acute lymphocytic leukemia
AML, acute myeloid leukemia
CCD, coiled‐coil domain
CHIP, clonal hematopoiesis of indeterminate potential
CLL, chronic lymphocytic leukemia
CML, chronic myeloid leukemia
CMML, chronic myelomonocytic leukemia
DBD, DNA‐binding domain
DNA, deoxyribonucleic acid
ELN, European leukemia net
FDA, US Food and Drug Administration
HSC, hematopoietic stem cell
HSPCs, hematopoietic stem and progenitor cells
ICC, International Consensus Classification
IFN‐γ, interferon γ
IgE, immunoglobulin E
IgK, immunoglobulin kappa
IL2, interleukin 2
IL6, interleukin 6
Il6ra, interleukin 6 receptor alpha
IL7, interleukin 7
ITD, internal tandem duplication
JAK–STAT, Janus Kinase/signal transducer and activator of transcription
KSL, c‐Kit+ Sca‐1+ lineage−
LD, linker domain
LSC, leukemic stem cell
MDS, myelodysplastic syndrome
miRNA, microRNA
mRNA, messenger RNA
NGS, next‐generation sequencing
NIH, National Institutes of Health
NTD, N‐terminal domain
PIAS, protein inhibitors of activated
PROTAC, proteolysis‐targeting chimera
pSTAT, phosphorylated STAT
PTPs, protein tyrosine phosphatases
R/R, relapsed/refractory
RNA, ribonucleic acid
SH2, Src‐homology 2
SOCS, suppressor of cytokine signaling
TAD, carboxy‐terminal transactivation domain
TCA, tricarboxylic acid cycle
Teff, effector T cell
TFH, T follicular helper cell
TH, T helper cell
sTNF‐RII, soluble tumor necrosis factor receptor II
TPO, thrombopoietin
Treg, regulatory T cell
TYK, tyrosine kinase
UK, United Kingdom
US, United States
uSTAT, unphosphorylated STAT
VISTA, V‐domain Ig suppressor of T‐cell activation
WHO, World Health Organization
y.o., years old
Aging: current situation and why it matters
Significant scientific and technological developments have improved health and life conditions, increasing human lifespan. Median age continues to increase, yet there is a debate in the scientific community regarding a potential upper limit of 125 years in the life expectancy of humankind [1, 2, 3]. According to WHO, by 2050 the population over 60 years old (y.o.) will double worldwide, reaching 2.1 billion [4]. In Europe, the aging of the population has been an observed demographic trend in the last few decades, with the older population (considered 65 y.o. and over), accounting for 21.3% in 2023, expected to reach 32.5% by 2100 [5]. Although we are going through the WHO's Decade of Healthy Aging aiming to support age‐friendly healthcare and communities, and to promote aging‐associated research [6], several challenges linked to age‐related diseases and person‐oriented care remain to be addressed.
Aging has been connected to or considered a risk factor for several ailments, including neurodegenerative [7], cardiovascular [8], and several hematological disorders both malignant, such as myelodysplastic syndromes, leukemias, myelomas, lymphomas, and non‐malignant, such as anemia [9, 10, 11]. Hematologic cancers are considered age‐related diseases and are characterized by increased mortality rate in the elderly population. Additionally, they account for one of the most serious clinical and scientific challenges that despite the immense scientific progress, remain to be decoded.
Namely, leukemia is a malignant progressive disease defined by the overproduction of abnormal hematopoietic cells. Ιt is contemplated as an age‐related disorder as it is more prevalent in older patients, with a median age of diagnosis at 67 y.o. According to NIH data, 59 610 new cases of leukemia and 23 710 deaths have been estimated in 2023, while 490 875 patients were living with leukemia in the US in 2020 [12].
Leukemia has been classified regarding severity, as acute or chronic and based on the cellular origin, as myelogenous or lymphocytic. Hence, there are four broad leukemia categories: acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), and chronic myelogenous leukemia (CML). AML remains the most common acute leukemia, accounting for approximately one‐third of the estimated leukemia cases in 2023 worldwide, with only a 31.7% 5‐year relative survival rate [13]. Additionally, a significant increase in myeloid leukemia incidents has been observed in the US since 2011, with AML deaths rising continuously in the past 20 years [14].
Clinical management of leukemia represents a major challenge for the European Healthcare System, considering that despite the extensive advancements, leukemia cases continue to increase alongside the aging population. Moreover, the economic burden of leukemia treatment remains substantial, with AML treatment costs amounting to a mean of $25 243 per patient per month over the follow‐up period and incremental cost‐effectiveness ratio (ratio of the cost difference to the difference in effectiveness) ranging from $50 000 to $150 000 per patient in the US and $20 000 to $30 000 in the UK [15, 16]. Therefore, the above‐indicated facts stipulate the prominent need for improved and personalized therapeutic approaches for age‐related hematopoietic defects. Although remarkable strides have been achieved through extensive research in this field, further research is required to unveil the underlying molecular mechanism of leukemogenesis.
Age‐associated defects in hematopoiesis: CHIP, MDS, and AML
Biological aging has been associated with several cellular and systemic defects, at least to some extent, due to the accumulation of somatic mutations in the Hematopoietic Stem Cell (HSC) compartment. As a result, mutations provide a fitness advantage to the mutated HSCs, leading to the expansion of identical mutated cells, called clones, giving rise to clonal hematopoiesis [17].
A subset of elderly with clonal hematopoiesis (10–40%), progresses to Clonal Hematopoiesis of Indeterminate Potential (CHIP), which is identified by the presence of cancer‐associated mutated clones in >4% of enucleated blood cells, but with no existing neoplastic features. CHIP is characterized by an increased risk of development of hematologic malignancies, including MDS and AML after the incorporation of additional mutations [17, 18].
MDS is a very heterogeneous group of clonal hematopoietic disorders, which are mainly characterized by ineffective hematopoiesis, dysplasia and the accumulation of immature blood cells (Fig. 1). Next‐generation sequencing (NGS) has allowed the extensive characterization of the molecular pathology of MDS through the determination of driver mutations [19]. The characterized driver mutations have been involved in fundamental cellular processes, namely RNA splicing, DNA modification, chromatin regulation, and cell signaling, whereas the number of the oncogenic aberrations in combination with the subclonal mutations are indicative of disease progression [19]. Although the aforementioned NGS studies have provided a great tool for prognosis and clinical management, further investigation is needed to elucidate the mechanisms underlying disease progression. Various additional factors, including the differential contribution of the identified mutations, chromosomal abnormalities, and the MDS subtype, might be of great importance.
Fig. 1.
The leukemic transformation from MDS to AML. The term MDS refers to a heterogeneous group of clonal hematopoietic disorders, characterized by impaired differentiation and hematopoietic dysfunctions, resulting in reduced erythrocytes, white blood cells and platelets. MDS commonly progresses to AML due to the accumulation of genomic aberrations that promote unrestrained proliferation. AML constitutes a very diverse group of aggressive hematological malignancies characterized by the accumulation of immature myeloblasts in the bone marrow and the peripheral blood.
MDS is commonly referred to as a pre‐leukemic stage due to the increased risk of transformation to AML, a highly aggressive type of leukemia. A two‐hit model had been initially proposed as the underlying mechanism in AML transformation that denotes the accumulation of Class I mutations (i.e. in FLT3, c‐KIT, etc.) providing the advantage of excessive proliferation and Class II mutations (i.e. in CEBPA) blocking the normal hematopoietic differentiation [20, 21]. Additional mutations associated with the promotion of epigenetic alterations (i.e mutations in TET2, DNMT3A, and ASXL1) and mutations at the tumor suppressors level (i.e. TP53 mutations) have been also identified and can cooperatively lead to leukemia development [22, 23, 24]. However, more recent studies have provided contradicting evidence, proposing an alternative model of non‐linear parallel clonal evolution with distinctive subclones within stem cell compartments leading to MDS and AML progression [25]. This indicates the importance for the investigation of the higher subclonal complexity of the distinct stem cell compartments in MDS to AML transformation, which challenges the belief that the increased clonal complexity arises in the blast compartment [25].
AML is the most common acute leukemia in adults, and it has been defined by uncontrolled clonal expansion of undifferentiated myeloid precursors in the bone marrow and peripheral blood, leading to ineffective hematopoiesis and bone marrow failure. The molecular heterogeneity of the disease, the complex pathways involved in the establishment and maintenance of AML and the vigorous clonal landscape render AML a complex dynamic disease. Enduring progress has been made in profiling the genomic scenery in AML. Gene mutations associated with leukemia have been cataloged and categorized by the Cancer Genome Atlas initiative and at least one potential driver mutation has been identified per case [26]. Potent mutational association in AML pathogenesis has been revealed resulting in the determination of three additional driver mutation sub‐categories, namely chromatin‐spliceosome, TP53‐aneuploidy, and IDH2 R172 mutation, as well as in the mapping of clonal trajectories, providing novel insights into the role of clonal complexity through disease progression [27, 28].
In 2022, the 5th edition of the WHO Classification of Hematolymphoid Tumors, the European Leukemia Net (ELN) 2022 risk classification, and a new classification system, the International Consensus Classification (ICC) of Myeloid Neoplasms and Acute Leukemias, have been published [29, 30, 31]. All the aforementioned classification systems have highlighted the significance of the incorporation of molecular analysis in everyday clinical practice and have emphasized the need for personalized therapeutic approaches, arising from the deeper understanding of the pathophysiology underlying AML complexity and heterogeneity, as well as the multiple novel therapeutic agents developed in the last decade. Although exceptional progress has been made, the rudimental mechanisms of leukemic transformation have not been elucidated yet and further research is needed to provide extended knowledge of the molecular pathways governing disease progression for more efficient patient stratification and treatment.
JAK–STAT signaling: a pleiotropic pathway
The Janus Kinase‐Signal Transducer and Activator of Transcription (JAK–STAT) signaling pathway has been of major significance for cellular function. Since its discovery more than 30 years ago [32, 33, 34], JAK–STAT represents one of the most conserved pathways that mediates signal transduction from the extracellular environment to the nucleus and leads to the modification of transcription. Activation of the pathway is induced by a plethora of molecules including cytokines, growth factors, and hormones, and regulates several important cellular functions, such as proliferation, differentiation, migration, and cell survival depending on the stimuli from the environment, thus providing a direct communication diode between the extracellular niche and the nucleus [35].
The JAK kinase family is composed of four members, JAK1, JAK2, JAK3, and TYK2 [36, 37, 38, 39]. STAT protein family in mammals includes seven members, STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6, all of which contain a conserved common structure, that includes the N‐terminal domain, the coiled‐coil domain, the Src‐homology 2 domain that is important for STAT dimerization, the linker domain, the DNA‐binding domain and the transactivation domain [40, 41, 42, 43] (Fig. 2). The pathway has been first discovered within the context of research for interferons, which are major players in anti‐tumor immune response [44, 45] and interleukin‐6 (IL6) signaling [46, 47]. Since its discovery, thorough research has uncovered its activation by over 50 cytokines, resulting in its involvement in many cellular functions, including cell cycle progression, development, proliferation, differentiation, survival, and apoptosis through ligand‐mediated signal transduction [35, 48, 49, 50, 51].
Fig. 2.
Structure of STAT protein family. The STAT family comprises seven highly similar members, STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B and STAT6 that share a structural topological similarity and are organized in six highly conserved structural domains. The schematic illustration shows the structural domains of the STAT proteins, including the N‐terminal domain (NTD), followed by the coiled‐coil (CCD), the DNA‐binding (DBD), the linker (LD), the Src‐homology 2 (SH2) and the carboxy‐terminal transactivation (TAD) domains. The NTD and SH2 domains are involved in protein–protein interactions and dimerization, the CCD is associated with nuclear localization, and the DBD mediates the DNA binding on specific sequences. Finally, the TAD C‐terminal domain includes essential tyrosine and serine residues for phosphorylation. The total number of amino acids constituting each STAT member is shown.
JAK–STAT canonical activation is achieved by the binding of an extracellular ligand to its transmembrane receptor that is noncovalently linked to JAKs, which undergo autophosphorylation and mediate tyrosine phosphorylation of the receptors acting as docking sites for the recruitment of STATs [32, 52] (Fig. 3). Subsequently, STATs are phosphorylated by JAKs at a single tyrosine residue in the C‐terminus, resulting in STAT homo‐ or/and hetero‐dimerization and translocation to the nucleus, where they bind to their target genes and other genomic positions, and regulate transcription. STATs are then dephosphorylated and translocated to the cytoplasm. Negative regulation of the JAK–STAT signaling pathway is mainly achieved by the Protein Inhibitors of Activated STATs (PIASs), the Protein Tyrosine Phosphatases (PTPs), and the Suppressors of Cytokine Signaling (SOCS) proteins. PIASs inhibit STAT phosphorylation and translocation, PTPs control STAT dephosphorylation, while SOCS are regulated by STATs and inhibit the JAK–STAT signaling via various mechanisms, creating a negative feedback loop for STAT activation [53, 54, 55]. Hence, the JAK–STAT pathway provides a simple yet sophisticated, tightly regulated and highly precise system, as different ligands result in the activation of specific STATs, which consequently regulate divergent pathways.
Fig. 3.
Canonical JAK–STAT pathway. The JAK–STAT pathway constitutes a highly conserved system that arbitrates the communication between the nucleus and the extracellular stimuli prompting necessary transcriptional changes and controlling important cellular functions including differentiation, proliferation, and survival. Several ligands, including cytokines, hormones, and growth factors, bind to their transmembrane receptor leading to JAK engagement that mediates the phosphorylation of JAKs and the creation of the docking site for STATs. Specific STAT proteins are recruited and activated by Tyrosine residue phosphorylation mediated by JAKs creating active dimers. Subsequently, activated STAT dimers translocate into the nucleus, where they either induce or inhibit transcription of their target genes through various direct or indirect mechanisms. The main negative regulators of the JAK–STAT pathways are PIASs, PTPs and SOCS proteins. The first two categories are implicated in STAT dephosphorylation, while SOCS are important JAK–STAT inhibitors and known STAT targets, creating a negative feedback loop for the regulation of the STAT function.
Recent studies have added further complexity to the role of STAT proteins by shedding light on novel aspects of non‐canonical STAT function. In contrast to the canonical JAK–STAT pathway, in which unphosphorylated STATs (uSTATs) are considered inactive, recent data have revealed that nuclear colocalization of uSTAT5 with CTCF in the absence of thrombopoietin (TPO), has repressed a megakaryocytic transcriptional program, identifying a novel cytokine‐dependent mechanism for megakaryocytic differentiation [56]. Moreover, STAT localization has been shown to be of great importance. Cytoplasmic localization of STATs has been typically considered to be equivalent to the inactive state, however active STAT5 has exhibited distinct effects in the cytoplasm related to the PI3K‐AKT pathway activation and leukemogenesis. Specifically, protein–protein interaction between STAT5 and p85 (the regulatory subunit of PI3K), mediated by the scaffolding protein GAB2, has been essential for the activation of PI3K‐AKT pathway [57]. In accordance to these findings, constitutively activated STAT5 localized in the cytoplasm in leukemia patient samples have led to PI3K‐AKT pathway activation, assigning an additional role to STAT5 as a cytoplasmic signaling effector molecules [58, 59].
In connection with aging, the JAK–STAT pathway might exhibit different and sometimes opposite functions depending on several factors including the organism, the tissue, the developmental stage, etc. Reduced JAK–STAT signaling has led to early aging‐associated deficiencies in cytokinesis of Drosophila stem cells, through the control of F‐actin, which is implicated in the germline stem cell cytokinesis, leading to abscission failure [60]. Also, reduced JAK–STAT signaling and cellular levels of pSTAT in leukocytes have been associated with increased physical weakness of the elderly [61]. However, age‐related increased JAK–STAT signaling has led to the promotion of senescence in tendon stem and progenitor cells during tendon aging. This effect had been reversed after JAK2 or STAT3 inhibition [62].
As mentioned, irregular cell signaling is positioned on the basis of the pathophysiology of the age‐related hematopoietic defects, such as MDS and AML. Amongst the most common mutations in AML and several other hematologic malignancies, numerous activating mutations in the JAK–STAT signaling pathway have been identified. STATs and particularly, aberrant activation of STAT3, STAT5A, and STAT5B have been implicated in aging and the development of numerous hematopoietic malignancies and solid cancers including MDS and AML, but their role has not been fully delineated yet [63].
Here we are reviewing the role of the STATs, and more specifically STAT3 and STAT5 factors in leukemic transformation to provide further insights into the mechanistic role of STATs in the process of age‐related leukemogenesis.
STAT3 and STAT5 in normal hematopoiesis and leukemia
Amongst the STAT family, STAT3 and STAT5 possess crucial roles in normal hematopoiesis and their abnormal function leads to leukemogenesis. Both STAT3 and STAT5 genes are located on human chromosome 17. STAT3 has four splicing isoforms, STAT3α, STAT3β, STAT3γ, and STAT3δ [64], while STAT5 comprises two highly related proteins, encoded by two separate genes (STAT5A and STAT5B) that are major regulators with pleiotropic roles in normal and pathologic hematopoiesis, including leukemia [65, 66] (Fig. 3). Despite the genomic proximity and the high peptide sequence similarity (more than 90%), STAT5A and STAT5B appear to have distinct roles [67, 68, 69]. However, the respective contribution of STAT3, STAT5A and STAT5B proteins, as regulators in the HSCs and leukemia, has not been fully elucidated.
STAT3 and STAT5 in normal hematopoiesis
SΤΑΤ3 has been an important factor at several levels for the development, as well as the maintenance and the regulation of a plethora of biological processes, including normal hematopoiesis. In greater detail, STAT3 has been essential in early embryonic development, and its ablation has led to early embryonic lethality in mice [70]. Specifically, IL6‐mediated STAT3 activation has prompted an anti‐apoptotic benefit through the high expression of BCL‐2 in human CD34+ cells [71]. Additionally, STAT3 has been important for Hematopoietic Stem and Progenitor Cells (HSPCs) sustainability and lineage balanced hematopoiesis. Conditional Stat3 inhibition in mice has resulted in a reduction of the Lin−Sca1+c‐Kit+CD150+CD48− HSPC subset, myeloid skewing, and increased DNA damage in HSPCs. This phenotype has been rescued by concomitant Ube2n deletion [72], which encodes the E2 ubiquitin‐conjugating enzyme UBC13 implicated in proinflammatory signaling and NF‐kB activation. Ube2n transcription has been suppressed by STAT3, influencing NF‐kB activity. These findings imply that STAT3 holds a protective role in bone homeostasis and control of inflammation [72, 73]. Regarding STAT5, its role in hematopoiesis has been even more extensively researched than STAT3, but its function has not been fully elucidated yet, due to its high complexity and the different levels of control. However, several studies have provided significant insights into STAT5 function. Specifically, STAT5 deficiency in HSCs has led to impaired long‐term repopulating activity of the HSCs in the bone marrow of lethally irradiated mice [74], while its activation has been determined as a major regulator of the cytokine‐induced HSC repopulating ability, but not for the self‐renewal capacity [75]. This effect could not be fully attributed to the lack of TPO responsiveness upstream STAT5, implying that STAT5 has been activated through different pathways to stimulate differentiation and it is essential at several steps of hematopoiesis [76]. Further research findings have indicated that STAT5 has been necessary in the perpetuation of transcriptional heterogeneity of HSPCs, acting as a moderator for clonal balance. Thus reduction of STAT5 activation during aging might be linked to increased clonality observed in the elderly and development of leukemias [77].
Moreover, STAT5 has been involved in differentiation and quiescence of HSCs through multiple mechanisms. STAT5 has been identified as a moderator in both primitive and definitive erythropoiesis and has exhibited inhibitory features in early embryonic erythropoiesis in xenopus models [78]. Besides, HSC quiescence in steady‐state hematopoiesis has been preserved through STAT5 signaling and has maintained the hematopoietic reservoir [79]. Accordingly, constitutive STAT5 activation and increased levels of BCLXL expression has led to induction of endogenous erythroid colony formation in human‐derived primary cells [80]. Further studies have provided evidence about the critical role of the STAT5 cellular level that has influenced HSC lineage commitment. Specifically, reduced STAT5 signaling in CD34+ cells has stimulated megakaryocytic growth, whilst STAT5 activation has promoted erythropoiesis [81]. Also, other evidence has suggested that the absence of Stat5 expression in adult bone marrow and fetal liver, resulted in decreased lymphomyeloid repopulating activity in mice [82]. Further investigation has indicated that STAT5 has been essential for the lymphomyeloid equilibrium and the repopulating capacity in hematopoiesis and has also underlined the significance specifically of the STAT5 N‐terminal domain in HSCs [83]. Moreover, STAT5 has been essential for myeloid differentiation through the regulation of Bclx resulting in promotion of survival and proliferation of the myeloid differentiating progenitors [84]. Another mechanism that implicates STAT5 in myeloid differentiation has been uncovered through the STAT5‐mediated CEBPA downregulation that has prompted self‐renewal and impaired myelopoiesis in human HSPCs [85].
Furthermore, STAT5 has been associated with stem cell proliferation through its regulation by microRNAs (miRNAs). Specifically, miR‐193b upregulation has restricted uncontrolled HSCs expansion through a STAT5‐mediating mechanism upon self‐renewal via the promotion of TPO‐MPL‐STAT5 signaling. This mechanism has provided negative feedback for unrestricted HSCs proliferation [86].
In addition, STATs interact with other proteins creating complexes to directly or indirectly activate or repress the transcription of target genes depending on the interacting partners, the binding affinity, the underlying motifs, and the genomic localization of the binding [87]. This differential regulation through STAT3 and STAT5 factors is at least partially achieved through the interaction with other proteins and/or transcription factors allowing broad functionality for STATs, with distinctive roles depending on the cell type, the differentiation stage, and the cellular status in healthy and malignant conditions.
The role of STAT3 and STAT5 in leukemia
Several factors, including upstream regulation, activation, mutations, protein stability, and post‐translational modifications, can lead to either hyperactivation or inactivation of STAT3 and STAT5A/B that result in various cancers, suggesting that the quote of Ancient Greeks ‘Metron Ariston’, which means moderation is best, is the necessary approach when considering STAT‐mediated signaling (Fig. 4).
Fig. 4.
JAK–STAT effects in Leukemia. The JAK–STAT pathway has been involved in several essential cellular functions including survival, apoptosis, proliferation, and differentiation. Additionally, biological processes such as inflammation and aging strongly affect hematopoiesis, through the dysregulation of bone marrow niche and clonal hematopoiesis, which have been mediated by JAK–STAT signaling. Impairment in the JAK–STAT pathway, mostly involving constitutive activation of STAT3 and STAT5 has been implicated in leukemogenesis and increased relapse rates, through numerous molecular mechanisms resulting in blockade of differentiation, and induced survival and proliferation. This indicates the necessity to delineate the role of STAT3 and STAT5 in leukemia development to allow design of novel therapeutic strategies.
STAT3 has been extensively researched in terms of its role in MDS and AML and multiple mechanisms have been uncovered, commonly being appointed an oncogenic character [88]. However, controversial evidence has also been presented, supporting STAT3 tumor suppressing activity in various cancers [89, 90].
In addition, STAT5, as already stated, is an essential regulator for several important cellular functions such as cell proliferation, differentiation, and survival. Ostensively divergent STAT5 signaling has been associated with numerous malignancies, including MDS and AML. Aberrant STAT3 and STAT5A/B signaling has been associated with leukemogenesis. Although several mechanisms and signaling pathways in MDS and AML implicating STAT3 and STAT5 activation and deregulation have been unveiled, the molecular pathogenesis has not been fully elucidated yet.
Concerning STAT3, it has been associated with negative disease outcome in AML via various mechanisms. Specifically, its activation has been connected with poor disease outcome in AML, through increased levels of LAPTM4B that interacts with RPS9 causing increased protein stability of RPS9, thus enhancing AML cell progression [91]. Moreover, expression of the immune checkpoint molecule, V‐domain Ig suppressor of T‐cell activation (VISTA) has been transcriptionally controlled by STAT3 via direct binding. Hence, constitutive STAT3 has caused increased VISTA expression in AML cells, which has been linked to unfavorable disease prediction [92].
Additionally, STAT3 pathway has been implicated in important cellular functions in AML cells including proliferation, apoptosis, and survival. For example, STAT3 has regulated autophagy downstream of the oncogenic KIT D816V mutation in AML cells resulting in increased proliferation and survival [93]. Besides, suppressed STAT3 expression by RNA interference in HL‐60 AML cells, has restrained proliferation and promoted apoptosis, assigning an oncogenic feature to STAT3 [94]. An additional mechanism has been identified, implicating STAT3 in MDS and AML immune resistance. In detail, maturation of myeloid cells has led to STAT3‐mediated atypical IFN‐γ signaling and PD‐1 ligands (PD‐L1 and PD‐L2) upregulation, thus increasing the responsiveness of MDS and AML cells to IFN‐γ. However, MDS and AML cells have utilized STAT3 pathway and PD‐1 ligands to surpass the immune response caused by IFN‐γ and resulted in secondary immune resistance [95].
Concerning STAT5, it plays a significant role in the survival and proliferation of leukemia. In greater detail, STAT5 has been involved in the long‐term preservation of both normal and leukemic HSPCs [96], while its activation in CD34− KSL cells has led to an increase in the multipotent progenitors population and has promoted HSC self‐renewal ex vivo [97]. On the other hand, STAT3 has been non‐essential for the HSC maintenance in vivo, and has stimulated differentiation in vitro providing a better understanding of the distinct functions of STAT3 and STAT5 in normal hematopoiesis and leukemogenesis [97].
An additional level of control for STATs lies in the activation via phosphorylation of specific tyrosine residues, which when perturbed can lead to aberrant activation of STATs that has been strongly associated with leukemias. Interestingly, STAT5A activation through serine phosphorylation can be critical for leukemogenesis. For instance, STAT5A phosphorylation on Ser725 and Ser779 residues in human leukemic cell lines and patient‐derived primary samples has been essential for hematopoietic cell transformation [98].
The complexity of the pleiotropic function of STATs is, at least to some extent, owed to the large number of signaling molecules that lead to their activation. In accordance with the described role of STATs thus far in leukemia, knockdown of SPATS2L, a poor prognosis marker in AML, has led to decreased JAK2, STAT3, and STAT5 protein levels and increased apoptosis and cell growth suppression, highlighting the significance of JAK–STAT signaling in AML [99]. However, abnormally low STAT3 and STAT5 activation has been linked to dismal diagnosis through expression of inflammation‐associated genes in pediatric AML [100].
Another mechanism responsible for irregular STAT5 signaling has been identified involving a constitutively activated STAT5A mutant that formed enhanced levels of stable tetramers. STAT5 tetramers have accumulated in excess to dimers in human leukemias and have been linked to leukemogenesis [101]. The tetrameric STAT5 has been demonstrated to possess the ability of recruiting the chromatin modifier EZH2, leading to inhibition of gene expression of the IgK locus. However, this repressive role has not been identified in STAT5 dimers [102]. This further supports recent findings that provide an epigenetic regulator role to STAT3 and STAT5 through the control and recruitment of epigenetic factors to specific gene loci [103]. Specifically, STAT5A and STAT5B have been shown to induce the expression of the Dpf3 gene that encodes for an epigenetic factor, through direct binding to its promoter causing increased expression that has been linked to leukemia [104]. Additionally, DNMT3A expression, an epigenetic modifier that is commonly mutated in both MDS and AML, has been positively controlled by STAT5A, leading to epigenetic inhibition of tumor‐suppressor genes in CD34+CD38− AML cells [105].
To further investigate the role of STAT3, the different STAT3 isoforms in leukemogenesis have been investigated, but their function has not been delineated yet. Nonetheless, there is evidence for distinct roles of STAT3α and STAT3β isoforms in hematologic malignancies. Specifically, it has been recently revealed that the α and β splicing isoform ratio has been significant in AML, with the higher STAT3β/α mRNA ratio having been identified as a prediction marker for favorable disease outcome [106]. Additionally, STAT3β has been characterized as a tumor suppressor in an AML transgenic mouse model, leading to decreased leukemia progression [106]. Accordingly, for STAT5 there is increasing evidence about the distinct role of STAT5A and STAT5B in leukemia, with STAT5B possibly holding a more dominant role. Importantly, STAT5B has been identified to control self‐renewal in both normal HSCs and leukemic stem cells (LSCs), through the STAT5B/CD9 axis [107]. Further evidence provides significant insights about the distinct function of STAT5A and STAT5B in MDS and AML, highlighting the significance of distinguishing between the two STAT5 factors [108].
Furthermore, STAT3 and STAT5 mediate the effects of key mutations in genes such as FLT3 and SF3B1 that have been associated with AML. FLT3 activating mutations are amongst the most common genetic aberrations in AML [26] and they result in the induction of STAT target genes [109, 110]. Several direct and indirect mechanisms involving the interplay between STAT5 and FLT3 mutations have been determined in AML. The FLT3 D835/I836 mutations have led to constitutive activation of STAT5 in pediatric ALL with hyperdiploidy and infant ALL with MLL rearrangements [111]. Additionally, internal tandem duplications (ITD) in FLT3 gene occur in approximately 25% of AML patients [112, 113]. The FLT3‐ITD mutation constitutively activated STAT5, leading to upregulated expression of the crucial factor for survival MCL‐1, providing a survival advantage in LSCs, irrespective of the wild‐type FLT3 signaling [114]. Also, in FLT3‐ITD AML, CDC25A, a phosphatase important for proliferation and differentiation, is directly regulated by STAT5 through the STAT5/miR‐16 axis [115]. Furthermore, STAT5 signaling in FLT3‐ITD mutated cells has caused increased PIM kinase expression and mTORC1/MCL1 signaling, which has served as a resistance mechanism for the PI3K/AKT pathway inhibitors [116]. Further evidence supports the involvement of STAT5 signaling in resistance mechanisms, provided that FLT3‐ITD mutations have caused CXCR4 upregulation in AML patients, involving also the signaling components STAT5 and PΙΜ1 that have been associated with therapy resistance and relapse [117].
Nevertheless, the inactivation of STAT5 by a type I tyrosine kinase inhibitor (gilteritinib), has resulted in the diminishment of FLT3‐ITD/D835 mutant cells through reduction of the expression of the downstream target of STAT5, BCL2A1, preventing treatment resistance [118]. Moreover, STAT5 aberrant activation downstream of FLT3‐ITD, has resulted in induced PIM kinases expression and protection of the mTORC1/4EBP1/S6K/MCL1 signaling axis providing drug resistance to the PI3K/AKT inhibitors. Hence, suppression of the STAT5/PIM signaling axis has resulted in the reinforcement of the cytotoxic effects of proteasome inhibitors in FLT3‐ITD AML cells, providing more efficient treatment [119]. STAT5 has been also shown to play a crucial role in the resistance mechanism to IDH inhibitors through differentiation blockade in leukemic cells, signifying the necessity for targeting STAT5 signaling selectively, in combination with IDH inhibition, for more efficient treatment [120].
In addition, SF3B1 mutations are commonly present in AML and they may be linked to a MDS pre‐phase. SF3B1 is frequently mutated in MDS or MDS/myeloproliferative neoplasms (MPN), being linked to a favorable and slow disease progression [121]. Recent studies indicate that STAT3 has been essential for HSPCs in homozygous Sf3b1 mutant zebrafish. Heterozygosity of SF3B1 has been examined, due to its prevalence in hematologic malignancies, resulting in augmented sensitivity to STAT3 inhibition in zebrafish, mouse, and human HSPCs. STAT3 inhibition has increased aberrant splicing in SF3B1 mutant cells, which has provided a selective target in hematologic malignancies [122].
Also, recent findings have increasingly associated STAT3 with the function of miRNAs in AML. In the last decade, it has been shown that miRNAs are involved in leukemogenesis, either as tumor suppressors or as oncogenes, and can constitute precious targets for biomarker development and/or novel therapies [123, 124]. Namely, miR‐548ac delivered by AML cell‐derived extracellular vesicles to CD34+ HSCs have induced TRIM28 expression and downstream activation of STAT3 that resulted in impeded hematopoiesis [125]. In addition, miR‐4532, delivered into CD34+ HSCs via AML cell‐released exosomes, has suppressed normal HSC hematopoiesis through the stimulation of the LDOC1‐mediated STAT3 signaling pathway [126]. Moreover, the STAT3 pathway has been activated by AML cell‐derived extracellular vesicles containing miR‐1246 through the direct regulation of the tumor‐suppressor gene LRIG1, hence leading to increased LSC survival [127]. Collectively, these data suggest that miRNAs derived by AML cells activate STAT3 to promote leukemia and inhibit normal hematopoiesis.
To add another level of complexity, an important role has been attributed to STAT3 regarding cellular metabolism in AML. Recent studies have revealed that STAT3 controls oxidative phosphorylation through numerous mechanisms, a procedure that has been the metabolic pathway of choice for LSCs, and is essential for their proliferation and survival [128]. Upon further research, it has been discovered that STAT3 has mediated oxidative phosphorylation in LSCs through the expression of MYC, which has successively regulated SLC1A5, an important factor for glutamine transport in LSCs. Consequently, SLC1A5 inhibition has resulted in decreased glutamine, glutathione, and TCA cycle metabolites, thus truncating the TCA cycle and repressing oxidative phosphorylation [129]. This provides a direct connection between STAT3 and metabolism in leukemia [129].
Finally, these findings confirm the functional involvement of mutated/activated STAT3 and STAT5 in hematologic malignancies, as well as their distinct roles in each condition. Several studies have identified target genes regulated by STATs and protein complexes of STATs in normal hematopoiesis and leukemia [87, 104]. Although such technological and scientific developments have contributed to vast progress in comprehending the pathways and the processes involved in leukemogenesis, further research is needed. The complexity and diversity of STAT mechanisms in leukemia render the development of efficient therapeutic agents very challenging, requiring deeper investigation to fully depict the molecular mechanisms through which STAT3, STAT5A/B, and their target genes are implicated in leukemogenesis, as well as their crosstalk with other interacting partners and their effects in transcription.
Aging, immunity and inflammaging: the role of STAT3 and STAT5, and their association with leukemogenesis
Leukemia incidents including AML are augmented in the elderly population. Biological aging is accompanied by several cellular changes including accumulation of somatic mutations, epigenetic alterations, stem cell malfunction, cellular senescence, etc. that have been associated with leukemogenesis [130]. However, the exact mechanisms that connect the aging process with leukemia have not been elucidated.
Aging has been strongly correlated with changes in the innate and adaptive immune system [131, 132], possibly creating a connection between aging and leukemogenesis. The JAK–STAT pathway might be a significant link between immunity and aging and thus leukemia development. STATs have been implicated in immunity through various mechanisms [133]. STAT5 has been widely involved in lymphoid development. In particular, STAT5 has been shown to control B‐cell development, albeit it has not been essential for B‐cell maturation and Ig production [134]. STAT5 has been shown to interact with LSD1 and HDAC3 in pro‐B cells regulating STAT5‐dependent transcription, signifying the involvement of STAT5 in multi‐protein complexes that modify chromatin in B‐cell development [87]. Additionally, JAK–STAT5 has been involved in the early development of B cells, being regulated by COX1‐derived thromboxane A2 that is important for the immune effects [135]. Regarding T cells, both STAT5A and STAT5B have controlled cell cycle and proliferation through IL2 signaling, which revealed a functional overlap between the two factors, provided that defects in T‐cell development have not been observed in deficient mice of STAT5A or STAT5B alone [136]. Furthermore, the interplay between mTOR and STAT5 signaling influences the mutual differentiation of effective and regulatory T cells (Teff and Treg cells), leading to different proliferation activity. This finding offers novel insights into the regulation of the Treg‐Teff equilibrium [137]. In Tregs maturation, STAT5 has been bound directly to Foxp3, being necessary for its induction, whereas STAT3 has been necessary for the reduction of FOXP3 through IL6 signaling, presenting opposite roles via differential control of FOXP3 [138]. Additionally, Stat5 expression has been required for mast cell survival, development, and IgE‐induced activation, the latter partly through post‐translational cytokine regulation [139, 140]. Recent studies provide evidence for the existence of interaction between STATs and the Ikaros family, which includes five highly homologous members IKZF1‐5 and has been strongly linked to immunity and lymphocyte development [141, 142]. Research on the Ikaros transcription factor family in T helper (TH) cells has indicated that there is possible cooperation between STATs and IKZFs [143]. Specifically, in T Follicular Helper (TFH) cells STAT3 has been shown to interact with Aiolos (IKZF3), creating a complex for the upregulation of the expression of Bcl6 and TFH cells‐associated cytokine receptor Il6ra [144]. However, in the same cell type the IL2/STAT5 signaling has hindered the Aiolos/STAT3 pathway by dual inhibition of Aiolos (IKZF3) expression and STAT3 activity [143]. In pre‐B cells, Ikaros (IKZF1) has been shown to directly compete for STAT5 DNA binding for common targets with GGAA motifs, as induced Ikaros activity has resulted in rapid extensive loss of STAT5 genome‐wide binding. This has provided an understanding of the mechanism behind the opposite effects between Ikaros and IL7/STAT5 pathways in B‐cell development [145].
All these studies provide continually extended evidence for STAT and Ikaros (IKZF) family engagement in mutual mechanisms, suggesting that interaction between STATs and IKZF factors may transpire to key players for the control of lymphomyeloid development.
Furthermore, the immune system's response to potentially dangerous stimuli, known as inflammation, is amongst the seven pillars of aging, which also include macromolecular damage, epigenetics, adaptation to stress, proteostasis, stem cell regeneration, and metabolism [146]. The term inflammaging has been used to describe a sterile, chronic, low grade inflammatory state that is linked to cellular senescence, dysregulation of mitochondrial function, autophagy, mitophagy, ubiquitin‐proteasome system, and activation of the DNA damage response mechanisms and dysbiosis (in host microbiota). Inflammaging contributes to a plethora of age‐associated diseases, including hematologic malignancies and cancer [147]. Aging has been associated with increased inflammatory signaling pathways that lead to senescence and conversely, senescent cells promote inflammation that accelerates biological aging, creating a vicious cycle. Indeed, inflammation stimulates HSCs aging that affects several cellular functions, such as differentiation, self‐renewal and energy metabolism [148]. However, the causality between inflammation and aging has not been elucidated yet. The JAK–STAT signaling plays an instrumental role in hematopoiesis, inflammation, and immunity, with nearly all of the immune responses triggered by cytokines being STAT‐dependent [149]. Expectedly, dysregulation of the JAK–STAT pathway has been associated with inflammation, autoimmune diseases, and cancers, including hematologic malignancies such as leukemia [150]. Increased inflammatory signaling in the human AML niche has caused aberrant JAK–STAT signaling in AML blasts and bone marrow stromal cells and consequently, escalation of leukemic proliferation [151].
Augmented STAT1, STAT3 and STAT5 activation has been positively correlated with age, alongside with increased inflammatory signaling‐associated molecules, including IL6 and sTNF‐RII [152]. In addition, STAT3 mitochondrial localization has been enhanced with age in CD4+ T cells leading to increased proinflammatory cytokine production and mitochondrial function creating a possible link between metabolism and inflammaging mediated by STAT signaling [153]. Also, cytokine signaling by both proinflammatory and anti‐inflammatory cytokines has been STAT3‐ and STAT5‐mediated [154, 155]. Hence, the deregulated STAT signaling might be the connecting link between inflammation and aging leading to leukemia development. Furthermore, the activation of STAT3 and STAT5 signaling pathways plays a crucial role in the regulation of innate receptor‐induced inflammatory cytokines, with the level of signaling establishing distinct patterns of regulation. In greater detail, following human monocyte‐derived macrophages stimulation by pattern‐recognition receptors, reduced expression of STAT3, STAT5A, and STAT5B has resulted in a decline in anti‐inflammatory cytokines. Initially, proinflammatory cytokines decline, but then increase, when the expression of STAT3 or STAT5 falls below a certain level [155].
STAT3 function in inflammation has been extensively researched and an anti‐inflammatory effect in HSPCs has been uncovered through transcriptional downregulation of the Ube2n gene, involved in proinflammatory signaling. This has indicated a STAT3‐dependent protection of the hematopoietic system preventing uncontrolled proinflammatory signaling by Ubc13 [72]. However, contrary evidence indicated that chronic inflammation reduced the HSC fitness through the stimulation of the JAK–STAT3 signaling pathway, which was inverted after Stat3 inhibition, in mice with chronic multifocal osteomyelitis [156].
As described in this chapter, STAT3 and STAT5 have been implicated in the regulation of innate immunity and inflammation, two functions that have been strongly associated with aging and age‐related conditions, such as hematopoietic malignancies. Albeit, further research is required to better outline the link between STATs in immunity and inflammation with aging and by extension to leukemogenesis. Elucidation of the role of STAT3 and STAT5 in age‐related leukemogenesis will provide novel targets for therapeutic approaches to improve management and treatment of various leukemias.
JAK–STAT targeted therapies in MDS and AML
The continuously increasing evidence about the significance of deregulated JAK–STAT signaling in myeloid disorders, such as MDS and AML, has augmented the interest in the scientific community about direct or indirect targeting for therapeutic purposes. Several therapeutic biological agents targeting JAK–STAT signaling components have been developed, including antibodies targeting cytokines and/or receptors, JAK and STAT inhibitors. Ruxolitinib is a JAK1 and JAK2 inhibitor approved by the US Food and Drug Administration (FDA) for treating myelofibrosis, polycythemia vera, and steroid‐refractory graft‐versus‐host disease [157, 158, 159, 160]. Fedratinib, a JAK–STAT inhibitor has been presented as a potential therapeutic molecule, with 78% tumor efficacy response in an ex vivo tumor explant AML model for patients who are not responding to cytarabine or azacitidine [161]. Still, several adverse effects have been identified by the use of JAK inhibitors highlighting the urge for enhanced understanding of the JAK–STAT pathway [162].
Although direct STAT inhibitors of clinical grade are not currently available, the use of direct or indirect inhibitors at preclinical level has provided further information about the mechanisms underlying STAT3 and STAT5 roles. For STAT3, chidamide, a potent HDAC inhibitor, has been exhibited to decrease MDS and AML cell survival through SOCS3‐mediated JAK2‐STAT3 inhibition [163]. AZD9150, an antisense STAT3 inhibitor, has enhanced apoptosis, hindered survival in leukemic cell lines and promoted differentiation in primary MDS/AML stem and progenitor cells, further confirming the significance of STAT3 as a leukemia‐promoting factor [164].
The possibilities for STAT3 and STAT5 direct targeting have been extensively reviewed for the development of cancer therapies [165, 166]. Despite the challenges in directly targeting STAT3 and STAT5 factors, the imminent technology of the proteolysis‐targeting chimera (PROTAC) degraders provides a promising strategy to subdue the constraints of conventional small molecule inhibitors. In 2023, the development of AK‐2292, a small‐molecule STAT5 PROTAC degrader, was reported [167]. AK‐2292 has selectively led to the degradation of STAT5A and STAT5B proteins achieving STAT5 inhibition in normal and leukemic cells, and resulted in tumor regression in human chronic myeloid leukemia (CML) xenografts in mice and AML cell lines providing a promising new component for STAT5 inhibition in leukemia and a possible therapeutic agent [167, 168].
Additionally, SD‐36 is a highly selective PROTAC degrader that potently induces STAT3 protein degradation, leading to cell cycle arrest and/or apoptosis in AML cell lines and xenograft models [169, 170].
A small number of direct and indirect STAT3/5 inhibitors have been used in clinical trials for MDS and AML therapy (Table 1). Even though the potential of STAT3 and STAT5 for the development of targeted therapies is continuously emerging and many molecules are currently in preclinical trials, further clinical research is required for their utilization in clinical practice and their establishment as therapeutic agents.
Table 1.
Clinical trials using direct or indirect STAT3 and STAT5 inhibitors. Clinical trials from https://www.clinicaltrials.gov/ for the treatment of MDS and AML, using direct and indirect STAT3 and STAT5 inhibitors are shown (R/R, Relapsed/Refractory).
Inhibitor | Clinical trial ID | Target | Phase | Disease/info in clinicaltrials.gov | Current status |
---|---|---|---|---|---|
Danvatirsen (AZD9150) | NCT05986240 | STAT3 | I | R/R MDS and AML/Selective, high‐affinity, antisense oligonucleotide inhibitor of STAT3 | Recruiting |
Pyrimethamine | NCT03057990 | STAT3 | I | Intermediate/High‐risk MDS R/R after treatment with azanucleosides/STAT3 Inhibitor | Withdrawn |
Pacritinib (SB1518) in combination with decitabine or cytarabine | NCT02532010 |
JAK2 FLT3 |
II | Elderly with AML/Reduces pJAK2, pSTAT3 or pSTAT5 in cell lines | Terminated |
Conclusions
Here the role of aging in hematopoietic defects, the pleiotropic JAK–STAT pathway and the role of STAT3 and STAT5 in normal hematopoiesis, leukemia, immunity and inflammaging have been reviewed. It has been established that the JAK–STAT pathway is a major contributor to several processes in normal hematopoiesis and malignancies. Specifically, STAT3 and STAT5 deregulation has been heavily involved in leukemia development and maintenance through several mechanisms, hence they represent very attractive therapeutic targets.
Despite extensive research in the hematology field that has provided new insights into the JAK–STAT function in hematologic malignancies, the role of STAT3 and STAT5 in leukemia remains largely unclear. This is partly due to the increased complexity of the molecular mechanisms of STAT3 and STAT5 in the molecular pathogenesis of leukemia, as they can act as inducers and repressors militating transcription changes and epigenetic alterations. To develop novel treatments to maximize efficacy and minimize adverse effects, it is necessary to gain a deeper knowledge of the STAT3 and STAT5 function in leukemogenesis. Furthermore, the identification of novel biomarkers is of major importance for leukemia stratification and therapeutic management, to contribute to clinical decisions.
Author contributions
ESF and EK wrote and edited the manuscript. EK supervised manuscript preparation. Both authors contributed to the article and approved the submitted version.
Acknowledgements
This work has been supported by the European Union's Horizon 2020 research and innovation program under the Marie Skłodowska‐Curie grant agreement No. 813091 (ARCH) to EK. The Graphical Abstract and the Figs 1, 2, 3, 4 have been created with BioRender.com. We apologize to all authors whose work has not been cited due to space limitations.
Edited by Barry Halliwell
References
- 1. Dong X, Milholland B and Vijg J (2016) Evidence for a limit to human lifespan. Nature 538, 257–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. de Beer J, Bardoutsos A and Janssen F (2017) Maximum human lifespan may increase to 125 years. Nature 546, E16–E17. [DOI] [PubMed] [Google Scholar]
- 3. Blagosklonny MV (2021) No limit to maximal lifespan in humans: how to beat a 122‐year‐old record. Oncoscience 8, 110–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Ageing and health. Available at: https://www.who.int/news‐room/fact‐sheets/detail/ageing‐and‐health
- 5. Population structure and ageing. Available at: https://ec.europa.eu/eurostat/statistics‐explained/index.php?title=Population_structure_and_ageing
- 6. Rudnicka E, Napierała P, Podfigurna A, Męczekalski B, Smolarczyk R and Grymowicz M (2020) The World Health Organization (WHO) approach to healthy ageing. Maturitas 139, 6–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL and Bohr VA (2019) Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol 15, 565–581. [DOI] [PubMed] [Google Scholar]
- 8. Jaiswal S and Libby P (2020) Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol 17, 137–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bron D, Ades L, Fulop T, Goede V and Stauder R (2015) Aging and blood disorders: new perspectives, new challenges. Haematologica 100, 415–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Stengel A, Baer C, Walter W, Meggendorfer M, Kern W, Haferlach T and Haferlach C (2021) Mutational patterns and their correlation to CHIP‐related mutations and age in hematological malignancies. Blood Adv 5, 4426–4434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Zhang H, Liesveld JL, Calvi LM, Lipe BC, Xing L, Becker MW, Schwarz EM, Yeh S-CA. The roles of bone remodeling in normal hematopoiesis and age-related hematological malignancies. Bone Research. 2023;11:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Leukemia – Cancer Stat Facts SEER . Available at: https://seer.cancer.gov/statfacts/html/leuks.html
- 13. Acute Myeloid Leukemia – Cancer Stat Facts SEER . Available at: https://seer.cancer.gov/statfacts/html/amyl.html
- 14. Hao T, Li‐Talley M, Buck A and Chen W (2019) An emerging trend of rapid increase of leukemia but not all cancers in the aging population in the United States. Sci Rep 9, 12070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Bell JA, Galaznik A, Farrelly E, Blazer M, Murty S, Ogbonnaya A, Eaddy M, Fram RJ, Faller DV and Kota V (2018) Economic burden of elderly patients with acute myeloid leukemia treated in routine clinical care in the United States. Leuk Res 71, 27–33. [DOI] [PubMed] [Google Scholar]
- 16. Bewersdorf JP, Huntington SF and Zeidan AM (2023) Cost‐effectiveness analyses in AML: what have we learned, how should this impact patient care, and what needs to Be done in the future? J Natl Compr Canc Netw 21, 522–528. [DOI] [PubMed] [Google Scholar]
- 17. Jaiswal S and Ebert BL (2019) Clonal hematopoiesis in human aging and disease. Science 366, eaan4673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Busque L, Buscarlet M, Mollica L and Levine RL (2018) Concise review: age‐related clonal hematopoiesis: stem cells tempting the devil. Stem Cells 36, 1287–1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Van Loo P, Yoon CJ, Ellis P, Wedge DC, Pellagatti A et al. (2013) Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122, 3616–3627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Gilliland DG (2002) Molecular genetics of human leukemias: new insights into therapy. Semin Hematol 39, 6–11. [DOI] [PubMed] [Google Scholar]
- 21. Kelly LM and Gilliland DG (2002) Genetics of myeloid leukemias. Annu Rev Genomics Hum Genet 3, 179–198. [DOI] [PubMed] [Google Scholar]
- 22. Shen Y, Zhu Y‐M, Fan X, Shi J‐Y, Wang Q‐R, Yan X‐J, Gu Z‐H, Wang Y‐Y, Chen B, Jiang C‐L et al. (2011) Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood 118, 5593–5603. [DOI] [PubMed] [Google Scholar]
- 23. Takahashi S (2011) Current findings for recurring mutations in acute myeloid leukemia. J Hematol Oncol 4, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Testa U and Pelosi E (2013) The impact of FLT3 mutations on the development of acute myeloid leukemias. Leuk Res Treatment 2013, 275760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Chen J, Kao Y‐R, Sun D, Todorova TI, Reynolds D, Narayanagari S‐R, Montagna C, Will B, Verma A and Steidl U (2019) Myelodysplastic syndrome progression to acute myeloid leukemia at the stem cell level. Nat Med 25, 103–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Cancer Genome Atlas Research Network , Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson AG, Hoadley K, Triche TJ, Laird PW et al. (2013) Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 368, 2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N et al. (2016) Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 374, 2209–2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Miles LA, Bowman RL, Merlinsky TR, Csete IS, Ooi AT, Durruthy‐Durruthy R, Bowman M, Famulare C, Patel MA, Mendez P et al. (2020) Single‐cell mutation analysis of clonal evolution in myeloid malignancies. Nature 587, 477–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Arber DA, Orazi A, Hasserjian RP, Borowitz MJ, Calvo KR, Kvasnicka H‐M, Wang SA, Bagg A, Barbui T, Branford S et al. (2022) International consensus classification of myeloid neoplasms and acute Leukemias: integrating morphologic, clinical, and genomic data. Blood 140, 1200–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Döhner H, Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, Ebert BL, Fenaux P, Godley LA, Hasserjian RP et al. (2022) Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 140, 1345–1377. [DOI] [PubMed] [Google Scholar]
- 31. Khoury JD, Solary E, Abla O, Akkari Y, Alaggio R, Apperley JF, Bejar R, Berti E, Busque L, Chan JKC et al. (2022) The 5th edition of the World Health Organization classification of Haematolymphoid Tumours: myeloid and histiocytic/dendritic neoplasms. Leukemia 36, 1703–1719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Hu X, Li J, Fu M, Zhao X and Wang W (2021) The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther 6, 1–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Schindler C, Shuai K, Prezioso VR and Darnell JE (1992) Interferon‐dependent tyrosine phosphorylation of a latent cytoplasmic transcription factor. Science 257, 809–813. [DOI] [PubMed] [Google Scholar]
- 34. Philips RL, Wang Y, Cheon H, Kanno Y, Gadina M, Sartorelli V, Horvath CM, Darnell JE, Stark GR and O'Shea JJ (2022) The JAK‐STAT pathway at 30: much learned, much more to do. Cell 185, 3857–3876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Harrison DA (2012) The JAK/STAT pathway. Cold Spring Harb Perspect Biol 4, a011205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Firmbach‐Kraft I, Byers M, Shows T, Dalla‐Favera R and Krolewski JJ (1990) tyk2, prototype of a novel class of non‐receptor tyrosine kinase genes. Oncogenesis 5, 1329–1336. [PubMed] [Google Scholar]
- 37. Krolewski JJ, Lee R, Eddy R, Shows TB and Dalla‐Favera R (1990) Identification and chromosomal mapping of new human tyrosine kinase genes. Oncogenesis 5, 277–282. [PubMed] [Google Scholar]
- 38. Wilks AF, Harpur AG, Kurban RR, Ralph SJ, Zürcher G and Ziemiecki A (1991) Two novel protein‐tyrosine kinases, each with a second phosphotransferase‐related catalytic domain, define a new class of protein kinase. Mol Cell Biol 11, 2057–2065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Takahashi T and Shirasawa T (1994) Molecular cloning of rat JAK3, a novel member of the JAK family of protein tyrosine kinases. FEBS Lett 342, 124–128. [DOI] [PubMed] [Google Scholar]
- 40. Ihle JN (1996) STATs: signal transducers and activators of transcription. Cell 84, 331–334. [DOI] [PubMed] [Google Scholar]
- 41. Ihle JN (2001) The Stat family in cytokine signaling. Curr Opin Cell Biol 13, 211–217. [DOI] [PubMed] [Google Scholar]
- 42. Darnell JE (1997) STATs and gene regulation. Science 277, 1630–1635. [DOI] [PubMed] [Google Scholar]
- 43. Lim CP and Cao X (2006) Structure, function, and regulation of STAT proteins. Mol Biosyst 2, 536–550. [DOI] [PubMed] [Google Scholar]
- 44. Darnell JE, Kerr IM and Stark GR (1994) Jak‐STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264, 1415–1421. [DOI] [PubMed] [Google Scholar]
- 45. Bach EA, Tanner JW, Marsters S, Ashkenazi A, Aguet M, Shaw AS and Schreiber RD (1996) Ligand‐induced assembly and activation of the gamma interferon receptor in intact cells. Mol Cell Biol 16, 3214–3221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Lütticken C, Wegenka UM, Yuan J, Buschmann J, Schindler C, Ziemiecki A, Harpur AG, Wilks AF, Yasukawa K and Taga T (1994) Association of transcription factor APRF and protein kinase Jak1 with the interleukin‐6 signal transducer gp130. Science 263, 89–92. [DOI] [PubMed] [Google Scholar]
- 47. Zhong Z, Wen Z and Darnell JE (1994) Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin‐6. Science 264, 95–98. [DOI] [PubMed] [Google Scholar]
- 48. Kiger AA, Jones DL, Schulz C, Rogers MB and Fuller MT (2001) Stem cell self‐renewal specified by JAK‐STAT activation in response to a support cell cue. Science 294, 2542–2545. [DOI] [PubMed] [Google Scholar]
- 49. Xiong H, Zhang Z‐G, Tian X‐Q, Sun D‐F, Liang Q‐C, Zhang Y‐J, Lu R, Chen Y‐X and Fang J‐Y (2008) Inhibition of JAK1, 2/STAT3 signaling induces apoptosis, cell cycle arrest, and reduces tumor cell invasion in colorectal cancer cells. Neoplasia 10, 287–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Morris R, Kershaw NJ and Babon JJ (2018) The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci 27, 1984–2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Borensztejn A, Boissoneau E, Fernandez G, Agnès F and Pret A‐M (2013) JAK/STAT autocontrol of ligand‐producing cell number through apoptosis. Development 140, 195–204. [DOI] [PubMed] [Google Scholar]
- 52. Yamaoka K, Saharinen P, Pesu M, Holt VET, Silvennoinen O and O'Shea JJ (2004) The Janus kinases (Jaks). Genome Biol 5, 253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Böhmer F‐D and Friedrich K (2014) Protein tyrosine phosphatases as wardens of STAT signaling. JAKSTAT 3, e28087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Niu G‐J, Xu J‐D, Yuan W‐J, Sun J‐J, Yang M‐C, He Z‐H, Zhao X‐F and Wang J‐X (2018) Protein inhibitor of activated STAT (PIAS) negatively regulates the JAK/STAT pathway by inhibiting STAT phosphorylation and translocation. Front Immunol 9, 2392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Sobah ML, Liongue C and Ward AC (2021) SOCS proteins in immunity, inflammatory diseases, and immune‐related cancer. Front Med 8, 727987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Park HJ, Li J, Hannah R, Biddie S, Leal‐Cervantes AI, Kirschner K, Flores Santa Cruz D, Sexl V, Göttgens B and Green AR (2016) Cytokine‐induced megakaryocytic differentiation is regulated by genome‐wide loss of a uSTAT transcriptional program. EMBO J 35, 580–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Nyga R, Pecquet C, Harir N, Gu H, Dhennin‐Duthille I, Régnier A, Gouilleux‐Gruart V, Lassoued K and Gouilleux F (2005) Activated STAT5 proteins induce activation of the PI 3‐kinase/Akt and Ras/MAPK pathways via the Gab2 scaffolding adapter. Biochem J 390, 359–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Harir N, Pecquet C, Kerenyi M, Sonneck K, Kovacic B, Nyga R, Brevet M, Dhennin I, Gouilleux‐Gruart V, Beug H et al. (2007) Constitutive activation of Stat5 promotes its cytoplasmic localization and association with PI3‐kinase in myeloid leukemias. Blood 109, 1678–1686. [DOI] [PubMed] [Google Scholar]
- 59. Kornfeld J‐W, Grebien F, Kerenyi MA, Friedbichler K, Kovacic B, Zankl B, Hoelbl A, Nivarti H, Beug H, Sexl V et al. (2008) The different functions of Stat5 and chromatin alteration through Stat5 proteins. Front Biosci 13, 6237–6254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Lenhart KF, Capozzoli B, Warrick GSD and DiNardo S (2019) Diminished Jak/STAT signaling causes early‐onset aging defects in stem cell cytokinesis. Curr Biol 29, 256–267.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Samson LD, Engelfriet P, Verschuren WMM, Picavet HSJ, Ferreira JA, de Zeeuw‐Brouwer M, Buisman A‐M and Boots AMH (2022) Impaired JAK‐STAT pathway signaling in leukocytes of the frail elderly. Immun Ageing 19, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Chen M, Xiao L, Dai G, Lu P, Zhang Y, Li Y, Ni M and Rui Y (2021) Inhibition of JAK‐STAT signaling pathway alleviates age‐related phenotypes in tendon stem/progenitor cells. Front Cell Dev Biol 9, 650250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Fasouli ES and Katsantoni E (2021) JAK‐STAT in early hematopoiesis and leukemia. Front Cell Dev Biol 9, 669363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Hevehan DL, Miller WM and Papoutsakis ET (2002) Differential expression and phosphorylation of distinct STAT3 proteins during granulocytic differentiation. Blood 99, 1627–1637. [DOI] [PubMed] [Google Scholar]
- 65. Schepers H, Wierenga ATJ, Vellenga E and Schuringa JJ (2012) STAT5‐mediated self‐renewal of normal hematopoietic and leukemic stem cells. JAKSTAT 1, 13–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Wang Z and Bunting KD (2013) STAT5 in hematopoietic stem cell biology and transplantation. JAKSTAT 2, e27159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Grimley PM, Dong F and Rui H (1999) Stat5a and Stat5b: fraternal twins of signal transduction and transcriptional activation. Cytokine Growth Factor Rev 10, 131–157. [DOI] [PubMed] [Google Scholar]
- 68. Maurer B, Kollmann S, Pickem J, Hoelbl‐Kovacic A and Sexl V (2019) STAT5A and STAT5B‐twins with different personalities in hematopoiesis and leukemia. Cancers 11, 1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Smith MR, Satter LRF and Vargas‐Hernández A (2023) STAT5b: a master regulator of key biological pathways. Front Immunol 13, 1025373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Takeda K, Noguchi K, Shi W, Tanaka T, Matsumoto M, Yoshida N, Kishimoto T and Akira S (1997) Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc Natl Acad Sci USA 94, 3801–3804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Sepúlveda P, Encabo A, Carbonell‐Uberos F and Miñana MD (2007) BCL‐2 expression is mainly regulated by JAK/STAT3 pathway in human CD34+ hematopoietic cells. Cell Death Differ 14, 378–380. [DOI] [PubMed] [Google Scholar]
- 72. Zhang H, Li HS, Hillmer EJ, Zhao Y, Chrisikos TT, Hu H, Wu X, Thompson EJ, Clise‐Dwyer K, Millerchip KA et al. (2018) Genetic rescue of lineage‐balanced blood cell production reveals a crucial role for STAT3 antiinflammatory activity in hematopoiesis. Proc Natl Acad Sci USA 115, E2311–E2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Zhang H, Hu H, Greeley N, Jin J, Matthews AJ, Ohashi E, Caetano MS, Li HS, Wu X, Mandal PK et al. (2014) STAT3 restrains RANK‐ and TLR4‐mediated signalling by suppressing expression of the E2 ubiquitin‐conjugating enzyme Ubc13. Nat Commun 5, 5798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Snow JW, Abraham N, Ma MC, Abbey NW, Herndier B and Goldsmith MA (2002) STAT5 promotes multilineage hematolymphoid development in vivo through effects on early hematopoietic progenitor cells. Blood 99, 95–101. [DOI] [PubMed] [Google Scholar]
- 75. Bradley HL, Hawley TS and Bunting KD (2002) Cell intrinsic defects in cytokine responsiveness of STAT5‐deficient hematopoietic stem cells. Blood 100, 3983–3989. [DOI] [PubMed] [Google Scholar]
- 76. Bradley HL, Couldrey C and Bunting KD (2004) Hematopoietic‐repopulating defects from STAT5‐deficient bone marrow are not fully accounted for by loss of thrombopoietin responsiveness. Blood 103, 2965–2972. [DOI] [PubMed] [Google Scholar]
- 77. Wang Z and Bunting KD (2017) Stat5 deficiency decreases transcriptional heterogeneity and supports emergence of hematopoietic sub‐populations. Oncotarget 8, 22477–22482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Schmerer M, Torregroza I, Pascal A, Umbhauer M and Evans T (2006) STAT5 acts as a repressor to regulate early embryonic erythropoiesis. Blood 108, 2989–2997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Wang Z, Li G, Tse W and Bunting KD (2009) Conditional deletion of STAT5 in adult mouse hematopoietic stem cells causes loss of quiescence and permits efficient nonablative stem cell replacement. Blood 113, 4856–4865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Garçon L, Rivat C, James C, Lacout C, Camara‐Clayette V, Ugo V, Lecluse Y, Bennaceur‐Griscelli A and Vainchenker W (2006) Constitutive activation of STAT5 and Bcl‐xL overexpression can induce endogenous erythroid colony formation in human primary cells. Blood 108, 1551–1554. [DOI] [PubMed] [Google Scholar]
- 81. Olthof SG, Fatrai S, Drayer AL, Tyl MR, Vellenga E and Schuringa JJ (2008) Downregulation of signal transducer and activator of transcription 5 (STAT5) in CD34+ cells promotes megakaryocytic development, whereas activation of STAT5 drives erythropoiesis. Stem Cells 26, 1732–1742. [DOI] [PubMed] [Google Scholar]
- 82. Bunting KD, Bradley HL, Hawley TS, Moriggl R, Sorrentino BP and Ihle JN (2002) Reduced lymphomyeloid repopulating activity from adult bone marrow and fetal liver of mice lacking expression of STAT5. Blood 99, 479–487. [DOI] [PubMed] [Google Scholar]
- 83. Li G, Wang Z, Zhang Y, Kang Z, Haviernikova E, Cui Y, Hennighausen L, Moriggl R, Wang D, Tse W et al. (2007) STAT5 requires the N‐domain to maintain hematopoietic stem cell repopulating function and appropriate lymphoid‐myeloid lineage output. Exp Hematol 35, 1684–1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Kieslinger M, Woldman I, Moriggl R, Hofmann J, Marine JC, Ihle JN, Beug H and Decker T (2000) Antiapoptotic activity of Stat5 required during terminal stages of myeloid differentiation. Genes Dev 14, 232–244. [PMC free article] [PubMed] [Google Scholar]
- 85. Wierenga ATJ, Schepers H, Moore MAS, Vellenga E and Schuringa JJ (2006) STAT5‐induced self‐renewal and impaired myelopoiesis of human hematopoietic stem/progenitor cells involves down‐modulation of C/EBPalpha. Blood 107, 4326–4333. [DOI] [PubMed] [Google Scholar]
- 86. Haetscher N, Feuermann Y, Wingert S, Rehage M, Thalheimer FB, Weiser C, Bohnenberger H, Jung K, Schroeder T, Serve H et al. (2015) STAT5‐regulated microRNA‐193b controls haematopoietic stem and progenitor cell expansion by modulating cytokine receptor signalling. Nat Commun 6, 8928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Nanou A, Toumpeki C, Lavigne MD, Lazou V, Demmers J, Paparountas T, Thanos D and Katsantoni E (2017) The dual role of LSD1 and HDAC3 in STAT5‐dependent transcription is determined by protein interactions, binding affinities, motifs and genomic positions. Nucleic Acids Res 45, 142–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Levy DE and Inghirami G (2006) STAT3: a multifaceted oncogene. Proc Natl Acad Sci USA 103, 10151–10152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Ecker A, Simma O, Hoelbl A, Kenner L, Beug H, Moriggl R and Sexl V (2009) The dark and the bright side of Stat3: proto‐oncogene and tumor‐suppressor. Front Biosci 14, 2944–2958. [DOI] [PubMed] [Google Scholar]
- 90. Avalle L, Camporeale A, Camperi A and Poli V (2017) STAT3 in cancer: a double edged sword. Cytokine 98, 42–50. [DOI] [PubMed] [Google Scholar]
- 91. Huang Y, Peng M, Qin H, Li Y, Pei L, Liu X and Zhao X (2023) LAPTM4B promotes AML progression through regulating RPS9/STAT3 axis. Cell Signal 106, 110623. [DOI] [PubMed] [Google Scholar]
- 92. Mo J, Deng L, Peng K, Ouyang S, Ding W, Lou L, Lin Z, Zhu J, Li J, Zhang Q et al. (2023) Targeting STAT3‐VISTA axis to suppress tumor aggression and burden in acute myeloid leukemia. J Hematol Oncol 16, 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Larrue C, Heydt Q, Saland E, Boutzen H, Kaoma T, Sarry J‐E, Joffre C and Récher C (2019) Oncogenic KIT mutations induce STAT3‐dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis 8, 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Zhao J, Xu Y, Zong Y, Zhang S, Song Y, Yu K, Li Z, Ji Y, Qiu Y and Chen F (2011) Inhibition of Stat3 expression induces apoptosis and suppresses proliferation in human leukemia HL‐60 cells. Hematology 16, 232–235. [DOI] [PubMed] [Google Scholar]
- 95. Yoyen‐Ermis D, Tunali G, Tavukcuoglu E, Horzum U, Ozkazanc D, Sutlu T, Buyukasik Y and Esendagli G (2019) Myeloid maturation potentiates STAT3‐mediated atypical IFN‐γ signaling and upregulation of PD‐1 ligands in AML and MDS. Sci Rep 9, 11697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Schepers H, van Gosliga D, Wierenga ATJ, Eggen BJL, Schuringa JJ and Vellenga E (2007) STAT5 is required for long‐term maintenance of normal and leukemic human stem/progenitor cells. Blood 110, 2880–2888. [DOI] [PubMed] [Google Scholar]
- 97. Kato Y, Iwama A, Tadokoro Y, Shimoda K, Minoguchi M, Akira S, Tanaka M, Miyajima A, Kitamura T and Nakauchi H (2005) Selective activation of STAT5 unveils its role in stem cell self‐renewal in normal and leukemic hematopoiesis. J Exp Med 202, 169–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Friedbichler K, Kerenyi MA, Kovacic B, Li G, Hoelbl A, Yahiaoui S, Sexl V, Müllner EW, Fajmann S, Cerny‐Reiterer S et al. (2010) Stat5a serine 725 and 779 phosphorylation is a prerequisite for hematopoietic transformation. Blood 116, 1548–1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Li F, Ye W, Yao Y, Wei W, Lin X, Zhuang H, Li C, Li X, Ling Q, Hu C et al. (2023) Spermatogenesis associated serine rich 2 like plays a prognostic factor and therapeutic target in acute myeloid leukemia by regulating the JAK2/STAT3/STAT5 axis. J Transl Med 21, 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Narayanan P, Man T‐K, Gerbing RB, Ries R, Stevens AM, Wang Y‐C, Long X, Gamis AS, Cooper T, Meshinchi S et al. (2021) Aberrantly low STAT3 and STAT5 responses are associated with poor outcome and an inflammatory gene expression signature in pediatric acute myeloid leukemia. Clin Transl Oncol 23, 2141–2154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Moriggl R, Sexl V, Kenner L, Duntsch C, Stangl K, Gingras S, Hoffmeyer A, Bauer A, Piekorz R, Wang D et al. (2005) Stat5 tetramer formation is associated with leukemogenesis. Cancer Cell 7, 87–99. [DOI] [PubMed] [Google Scholar]
- 102. Mandal M, Powers SE, Maienschein‐Cline M, Bartom ET, Hamel KM, Kee BL, Dinner AR and Clark MR (2011) Epigenetic repression of the Igk locus by STAT5‐mediated recruitment of the histone methyltransferase Ezh2. Nat Immunol 12, 1212–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Wingelhofer B, Neubauer HA, Valent P, Han X, Constantinescu SN, Gunning PT, Müller M and Moriggl R (2018) Implications of STAT3 and STAT5 signaling on gene regulation and chromatin remodeling in hematopoietic cancer. Leukemia 32, 1713–1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Theodorou M, Speletas M, Mamara A, Papachristopoulou G, Lazou V, Scorilas A and Katsantoni E (2013) Identification of a STAT5 target gene, Dpf3, provides novel insights in chronic lymphocytic leukemia. PLoS ONE 8, e76155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Takeuchi A, Nishioka C, Ikezoe T, Yang J and Yokoyama A (2015) STAT5A regulates DNMT3A in CD34(+)/CD38(−) AML cells. Leuk Res 39, 897–905. [DOI] [PubMed] [Google Scholar]
- 106. Aigner P, Mizutani T, Horvath J, Eder T, Heber S, Lind K, Just V, Moll HP, Yeroslaviz A, Fischer MJM et al. (2019) STAT3β is a tumor suppressor in acute myeloid leukemia. Blood Adv 3, 1989–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Kollmann S, Grausenburger R, Klampfl T, Prchal‐Murphy M, Bastl K, Pisa H, Knab VM, Brandstoetter T, Doma E, Sperr WR et al. (2021) A STAT5B–CD9 axis determines self‐renewal in hematopoietic and leukemic stem cells. Blood 138, 2347–2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Fasouli ES, Giavi K, Makri C, Bystrykh L, Bouchla A, Athanasiadis E, Vatsellas G, Pappa V and Katsantoni E (2023) Distinct role of STAT3 and STAT5 factors in MDS and AML. Blood 142, 4120. [Google Scholar]
- 109. Kiyoi H, Ohno R, Ueda R, Saito H and Naoe T (2002) Mechanism of constitutive activation of FLT3 with internal tandem duplication in the juxtamembrane domain. Oncogenesis 21, 2555–2563. [DOI] [PubMed] [Google Scholar]
- 110. Mizuki M, Schwable J, Steur C, Choudhary C, Agrawal S, Sargin B, Steffen B, Matsumura I, Kanakura Y, Böhmer FD et al. (2003) Suppression of myeloid transcription factors and induction of STAT response genes by AML‐specific Flt3 mutations. Blood 101, 3164–3173. [DOI] [PubMed] [Google Scholar]
- 111. Taketani T, Taki T, Sugita K, Furuichi Y, Ishii E, Hanada R, Tsuchida M, Sugita K, Ida K and Hayashi Y (2004) FLT3 mutations in the activation loop of tyrosine kinase domain are frequently found in infant ALL with MLL rearrangements and pediatric ALL with hyperdiploidy. Blood 103, 1085–1088. [DOI] [PubMed] [Google Scholar]
- 112. Daver N, Schlenk RF, Russell NH and Levis MJ (2019) Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia 33, 299–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Castaño‐Bonilla T, Alonso‐Dominguez JM, Barragán E, Rodríguez‐Veiga R, Sargas C, Gil C, Chillón C, Vidriales MB, García R, Martínez‐López J et al. (2021) Prognostic significance of FLT3‐ITD length in AML patients treated with intensive regimens. Sci Rep 11, 20745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Yoshimoto G, Miyamoto T, Jabbarzadeh‐Tabrizi S, Iino T, Rocnik JL, Kikushige Y, Mori Y, Shima T, Iwasaki H, Takenaka K et al. (2009) FLT3‐ITD up‐regulates MCL‐1 to promote survival of stem cells in acute myeloid leukemia via FLT3‐ITD‐specific STAT5 activation. Blood 114, 5034–5043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Sueur G, Boutet A, Gotanègre M, Mansat‐De Mas V, Besson A, Manenti S and Bertoli S (2020) STAT5‐dependent regulation of CDC25A by miR‐16 controls proliferation and differentiation in FLT3‐ITD acute myeloid leukemia. Sci Rep 10, 1906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Okada K, Nogami A, Ishida S, Akiyama H, Chen C, Umezawa Y and Miura O (2018) FLT3‐ITD induces expression of Pim kinases through STAT5 to confer resistance to the PI3K/Akt pathway inhibitors on leukemic cells by enhancing the mTORC1/mcl‐1 pathway. Oncotarget 9, 8870–8886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Cao T, Jiang N, Liao H, Shuai X, Su J and Zheng Q (2019) The FLT3‐ITD mutation and the expression of its downstream signaling intermediates STAT5 and Pim‐1 are positively correlated with CXCR4 expression in patients with acute myeloid leukemia. Sci Rep 9, 12209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Yamatani K, Ai T, Saito K, Suzuki K, Hori A, Kinjo S, Ikeo K, Ruvolo V, Zhang W, Mak PY et al. (2022) Inhibition of BCL2A1 by STAT5 inactivation overcomes resistance to targeted therapies of FLT3‐ITD/D835 mutant AML. Transl Oncol 18, 101354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Nogami A, Okada K, Ishida S, Akiyama H, Umezawa Y and Miura O (2019) Inhibition of the STAT5/Pim kinase Axis enhances cytotoxic effects of proteasome inhibitors on FLT3‐ITD‐positive AML cells by cooperatively inhibiting the mTORC1/4EBP1/S6K/mcl‐1 pathway. Transl Oncol 12, 336–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Liu ACH, Cathelin S, Yang Y, Dai DL, Ayyathan DM, Hosseini M, Minden MD, Tierens A and Chan SM (2022) Targeting STAT5 signaling overcomes resistance to IDH inhibitors in acute myeloid leukemia through suppression of Stemness. Cancer Res 82, 4325–4339. [DOI] [PubMed] [Google Scholar]
- 121. Huber S, Haferlach T, Meggendorfer M, Hutter S, Hoermann G, Baer C, Kern W and Haferlach C (2022) SF3B1 mutations in AML are strongly associated with MECOM rearrangements and may be indicative of an MDS pre‐phase. Leukemia 36, 2927–2930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Potts KS, Cameron RC, Metidji A, Ghazale N, Wallace L, Leal‐Cervantes AI, Palumbo R, Barajas JM, Gupta V, Aluri S et al. (2022) Splicing factor deficits render hematopoietic stem and progenitor cells sensitive to STAT3 inhibition. Cell Rep 41, 111825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Li M, Cui X and Guan H (2020) MicroRNAs: pivotal regulators in acute myeloid leukemia. Ann Hematol 99, 399–412. [DOI] [PubMed] [Google Scholar]
- 124. Schotte D, Pieters R and Den Boer ML (2012) MicroRNAs in acute leukemia: from biological players to clinical contributors. Leukemia 26, 1–12. [DOI] [PubMed] [Google Scholar]
- 125. Zhao C, Zhao Y, Zhao J, Meng G, Huang S, Liu Y, Wang S and Qi L (2022) Acute myeloid leukemia cell‐derived extracellular vesicles carrying microRNA‐548ac regulate hematopoietic function via the TRIM28/STAT3 pathway. Cancer Gene Ther 29, 918–929. [DOI] [PubMed] [Google Scholar]
- 126. Zhao C, Du F, Zhao Y, Wang S and Qi L (2019) Acute myeloid leukemia cells secrete microRNA‐4532‐containing exosomes to mediate normal hematopoiesis in hematopoietic stem cells by activating the LDOC1‐dependent STAT3 signaling pathway. Stem Cell Res Ther 10, 384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Chen L, Guo Z, Zhou Y, Ni J, Zhu J, Fan X, Chen X, Liu Y, Li Z and Zhou H (2021) microRNA‐1246‐containing extracellular vesicles from acute myeloid leukemia cells promote the survival of leukemia stem cells via the LRIG1‐meditated STAT3 pathway. Aging 13, 13644–13662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Amaya ML, Inguva A, Jones CL, Krug A, Pei S, Reigan P and Jordan CT (2019) STAT3 plays a critical role in mitochondrial function and survival of primary AML cells. Blood 134, 1275.31698438 [Google Scholar]
- 129. Amaya ML, Inguva A, Pei S, Jones C, Krug A, Ye H, Minhajuddin M, Winters A, Furtek SL, Gamboni F et al. (2022) The STAT3‐MYC axis promotes survival of leukemia stem cells by regulating SLC1A5 and oxidative phosphorylation. Blood 139, 584–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Zjablovskaja P and Florian MC (2020) Acute myeloid leukemia: aging and epigenetics. Cancers 12, 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. DeVeale B, Brummel T and Seroude L (2004) Immunity and aging: the enemy within? Aging Cell 3, 195–208. [DOI] [PubMed] [Google Scholar]
- 132. Gomez CR, Nomellini V, Faunce DE and Kovacs EJ (2008) Innate immunity and aging. Exp Gerontol 43, 718–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Awasthi N, Liongue C and Ward AC (2021) STAT proteins: a kaleidoscope of canonical and non‐canonical functions in immunity and cancer. J Hematol Oncol 14, 198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Dai X, Chen Y, Di L, Podd A, Li G, Bunting KD, Hennighausen L, Wen R and Wang D (2007) Stat5 is essential for early B cell development but not for B cell maturation and function. J Immunol 179, 1068–1079. [DOI] [PubMed] [Google Scholar]
- 135. Yang Q, Shi M, Shen Y, Cao Y, Zuo S, Zuo C, Zhang H, Gabrilovich DI, Yu Y and Zhou J (2014) COX‐1‐derived thromboxane A2 plays an essential role in early B‐cell development via regulation of JAK/STAT5 signaling in mouse. Blood 124, 1610–1621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Moriggl R, Topham DJ, Teglund S, Sexl V, McKay C, Wang D, Hoffmeyer A, van Deursen J, Sangster MY, Bunting KD et al. (1999) Stat5 is required for IL‐2‐induced cell cycle progression of peripheral T cells. Immunity 10, 249–259. [DOI] [PubMed] [Google Scholar]
- 137. Shan J, Feng L, Sun G, Chen P, Zhou Y, Xia M, Li H and Li Y (2015) Interplay between mTOR and STAT5 signaling modulates the balance between regulatory and effective T cells. Immunobiology 220, 510–517. [DOI] [PubMed] [Google Scholar]
- 138. Yao Z, Kanno Y, Kerenyi M, Stephens G, Durant L, Watford WT, Laurence A, Robinson GW, Shevach EM, Moriggl R et al. (2007) Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood 109, 4368–4375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Shelburne CP, McCoy ME, Piekorz R, Sexl V, Roh K‐H, Jacobs‐Helber SM, Gillespie SR, Bailey DP, Mirmonsef P, Mann MN et al. (2003) Stat5 expression is critical for mast cell development and survival. Blood 102, 1290–1297. [DOI] [PubMed] [Google Scholar]
- 140. Barnstein BO, Li G, Wang Z, Kennedy S, Chalfant C, Nakajima H, Bunting KD and Ryan JJ (2006) Stat5 expression is required for IgE‐mediated mast cell function. J Immunol 177, 3421–3426. [DOI] [PubMed] [Google Scholar]
- 141. Fan Y and Lu D (2016) The Ikaros family of zinc‐finger proteins. Acta Pharm Sin B 6, 513–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Heizmann B, Kastner P and Chan S (2018) The Ikaros family in lymphocyte development. Curr Opin Immunol 51, 14–23. [DOI] [PubMed] [Google Scholar]
- 143. Powell M, Read K, Sreekumar B and Oestreich K (2019) Ikaros zinc finger transcription factors: regulators of cytokine signaling pathways and CD4+ T helper cell differentiation. Front Immunol 10, 1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Read KA, Powell MD, Baker CE, Sreekumar BK, Ringel‐Scaia VM, Bachus H, Martin RE, Cooley ID, Allen IC, Ballesteros‐Tato A et al. (2017) Integrated STAT3 and Ikaros zinc finger transcription factor activities regulate Bcl‐6 expression in CD4+ T helper cells. J Immunol 199, 2377–2387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Heizmann B, Le Gras S, Simand C, Marchal P, Chan S and Kastner P (2020) Ikaros antagonizes DNA binding by STAT5 in pre‐B cells. PLoS ONE 15, e0242211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, Epel ES, Franceschi C, Lithgow GJ, Morimoto RI, Pessin JE et al. (2014) Aging: a common driver of chronic diseases and a target for novel interventions. Cell 159, 709–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Franceschi C, Garagnani P, Parini P, Giuliani C and Santoro A (2018) Inflammaging: a new immune–metabolic viewpoint for age‐related diseases. Nat Rev Endocrinol 14, 576–590. [DOI] [PubMed] [Google Scholar]
- 148. Li X, Li C, Zhang W, Wang Y, Qian P and Huang H (2023) Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther 8, 1–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Villarino AV, Kanno Y and O'Shea JJ (2017) Mechanisms and consequences of Jak–STAT signaling in the immune system. Nat Immunol 18, 374–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Loh C‐Y, Arya A, Naema AF, Wong WF, Sethi G and Looi CY (2019) Signal transducer and activator of transcription (STATs) proteins in cancer and inflammation: functions and therapeutic implication. Front Oncol 9, 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Habbel J, Arnold L, Chen Y, Möllmann M, Bruderek K, Brandau S, Dührsen U and Hanoun M (2020) Inflammation‐driven activation of JAK/STAT signaling reversibly accelerates acute myeloid leukemia in vitro. Blood Adv 4, 3000–3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Piber D, Olmstead R, Cho JH‐J, Witarama T, Perez C, Dietz N, Seeman TE, Breen EC, Cole SW and Irwin MR (2019) Inflammaging: age and systemic, cellular, and nuclear inflammatory biology in older adults. J Gerontol A Biol Sci Med Sci 74, 1716–1724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Zukowski E, Sannella M, Rockhold JD, Kalantar GH, Yu J, SantaCruz‐Calvo S, Kuhn MK, Hah N, Ouyang L, Wang T et al. (2023) STAT3 modulates CD4 + T mitochondrial dynamics and function in aging. Aging Cell 22, e13996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Ouyang W, Rutz S, Crellin NK, Valdez PA and Hymowitz SG (2011) Regulation and functions of the IL‐10 family of cytokines in inflammation and disease. Annu Rev Immunol 29, 71–109. [DOI] [PubMed] [Google Scholar]
- 155. Hedl M, Sun R, Huang C and Abraham C (2019) STAT3 and STAT5 signaling thresholds determine distinct regulation for innate‐receptor‐induced inflammatory cytokines and STAT3/STAT5 disease variants modulate these outcomes. J Immunol 203, 3325–3338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Grusanovic S, Danek P, Kuzmina M, Adamcova MK, Burocziova M, Mikyskova R, Vanickova K, Kosanovic S, Pokorna J, Reinis M et al. (2023) Chronic inflammation decreases HSC fitness by activating the druggable Jak/Stat3 signaling pathway. EMBO Rep 24, e54729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Ostojic A, Vrhovac R and Verstovsek S (2011) Ruxolitinib: a new JAK1/2 inhibitor that offers promising options for treatment of myelofibrosis. Future Oncol 7, 1035–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Vainchenker W, Yahmi N, Havelange V, Marty C, Plo I and Constantinescu SN (2023) Recent advances in therapies for primary myelofibrosis. Fac Rev 12, 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Boldrini V, Vannucchi AM and Guglielmelli P (2024) A safety evaluation of ruxolitinib for the treatment of polycythemia vera. Expert Opin Drug Saf 23, 1–7. [DOI] [PubMed] [Google Scholar]
- 160. De Togni E, Cole O and Abboud R (2024) Janus kinase inhibition in the treatment and prevention of graft‐versus‐host disease. Front Immunol 15, 1304065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Babu G, Chaudhuri P, Rajappa M, Biswas M, Sansar B, Rajegowda C, Radhakrishnan A, Advani J, Tewary B, Radhakrishnan P et al. (2021) JAK‐STAT inhibitor as a potential therapeutic opportunity in AML patients resistant to cytarabine and epigenetic therapy. Cancer Biol Ther 22, 66–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Hoisnard L, Lebrun‐Vignes B, Maury S, Mahevas M, El Karoui K, Roy L, Zarour A, Michel M, Cohen JL, Amiot A et al. (2022) Adverse events associated with JAK inhibitors in 126,815 reports from the WHO pharmacovigilance database. Sci Rep 12, 7140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Zhao S, Guo J, Zhao Y, Fei C, Zheng Q, Li X and Chang C (2016) Chidamide, a novel histone deacetylase inhibitor, inhibits the viability of MDS and AML cells by suppressing JAK2/STAT3 signaling. Am J Transl Res 8, 3169–3178. [PMC free article] [PubMed] [Google Scholar]
- 164. Shastri A, Choudhary G, Teixeira M, Gordon‐Mitchell S, Ramachandra N, Bernard L, Bhattacharyya S, Lopez R, Pradhan K, Giricz O et al. (2018) Antisense STAT3 inhibitor decreases viability of myelodysplastic and leukemic stem cells. J Clin Invest 128, 5479–5488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Orlova A, Wagner C, de Araujo ED, Bajusz D, Neubauer HA, Herling M, Gunning PT, Keserű GM and Moriggl R (2019) Direct targeting options for STAT3 and STAT5 in cancer. Cancers 11, 1930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Moser B, Edtmayer S, Witalisz‐Siepracka A and Stoiber D (2021) The ups and downs of STAT inhibition in acute myeloid leukemia. Biomedicines 9, 1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Kaneshige A, Bai L, Wang M, McEachern D, Meagher JL, Xu R, Kirchhoff PD, Wen B, Sun D, Stuckey JA et al. (2023) Discovery of a potent and selective STAT5 PROTAC degrader with strong antitumor activity in vivo in acute myeloid leukemia. J Med Chem 66, 2717–2743. [DOI] [PubMed] [Google Scholar]
- 168. Kaneshige A, Bai L, Wang M, McEachern D, Meagher JL, Xu R, Wang Y, Jiang W, Metwally H, Kirchhoff PD et al. (2023) A selective small‐molecule STAT5 PROTAC degrader capable of achieving tumor regression in vivo. Nat Chem Biol 19, 703–711. [DOI] [PubMed] [Google Scholar]
- 169. Bai L, Zhou H, Xu R, Zhao Y, Chinnaswamy K, McEachern D, Chen J, Yang C‐Y, Liu Z, Wang M et al. (2019) A potent and selective small‐molecule degrader of STAT3 achieves complete tumor regression in vivo. Cancer Cell 36, 498–511.e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Zhou H, Bai L, Xu R, Zhao Y, Chen J, McEachern D, Chinnaswamy K, Wen B, Dai L, Kumar P et al. (2019) Structure‐based discovery of SD‐36 as a potent, selective, and efficacious PROTAC degrader of STAT3 protein. J Med Chem 62, 11280–11300. [DOI] [PMC free article] [PubMed] [Google Scholar]