International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Acute myeloid leukemia (AML) is a hematological malignancy characterized by the aberrant proliferation of hematopoietic stem cells and is the most prevalent form of acute leukemia in adults, accounting for ~80% of cases in this group (1,2). Despite the continuous development of medical technology, the 5-year survival rate of patients with AML is 32% (with the rate as high as 50% in younger patients and lower than 10% in those over 60 years old) and the prognosis remains poor (3–5). Although traditional cytotoxic chemotherapy has been the foundation of AML treatment for the past 50 years, researchers are investigating therapeutic approaches aimed at enhancing patient survival (6,7).
Transcription is a complex biological event involving the interaction of transcription factors, RNA polymerase II and transcriptional cofactors with DNA regulatory elements. This process is key for maintaining essential cell functions such as cell proliferation, differentiation, and metabolic homeostasis (8). It converts DNA sequences into translatable mRNA, which regulates protein synthesis. Organisms achieve precise gene expression through a sophisticated transcriptional regulatory network that ensures internal environmental stability and regulates normal cell development (9–11). When transcriptional dysregulation occurs, it can lead to abnormal cell proliferation and differentiation, which ultimately results in tumors (12). Consequently, small-molecule targeted therapies aimed at key proteins in transcriptional regulation have become a focal point of current research (13). Menin protein inhibitors have emerged, with several preclinical studies confirming their notable potential as chromatin regulators (14–16).
The menin protein, encoded by the multiple endocrine neoplasia type 1 gene, functions primarily as a scaffolding protein that modulates gene transcription via interactions with various gene regulators, such as histone methyltransferases and transcription factors including JunD and SMAD (15,17,18). In AML, menin serves a key role in epigenetic regulation by interacting with genes such as lysine methyltransferase 2A (KMT2A) and mutant nucleophosmin 1 (NPM1), thereby facilitating the progression of AML (19,20). Inhibitors of the menin protein may reverse aberrant transcription associated with tumors and restore normal gene expression and cellular function by targeting this essential epigenetic regulator (21). The present review aimed to provide a comprehensive evaluation of the role of menin protein, assess the data supporting its mechanisms in KMT2A/NPM1 mutations and discuss the pharmacological characteristics and clinical challenges associated with current inhibitors, such as functional disparities of menin in various cellular contexts and optimization pathways for combinatorial therapeutic strategies.
AML is categorized into subtypes based on genetic and transcriptomic characteristics, one of which is defined by the homeobox (HOX) gene upregulation and primarily includes NPM1-mutant (NPM1-mut) and MLL-rearranged leukemia (22). MLL proteins (MLL1 and MLL2) belong to the histone H3 position 4 lysine (H3K4) methyltransferase family, which is essential for the maintenance of high HOX gene expression, and activate myeloid ecotropic viral integration site 1 (MEIS1) through direct epigenetic regulation (16). Notably, MLL fusion proteins contain specific Su(var)3-9, Enhancer-of-zeste, Trithorax) structural domains, whereas the crystal structure of menin proteins exhibits a rectangular conformation, which allows for the formation of deep binding pockets that bind specifically to the N-terminal fragment of MLL fusion proteins (23,24). This interaction designates menin protein as an oncogenic cofactor that facilitates histone H3 lysine trimethylation at position 4 (H3K4me3) via its engagement with MLL, subsequently enhancing HOX/MEIS1 gene expression and precipitating leukemia (25). Utilizing a structure-based drug design approach targeting the menin-MLL interaction interface, Krivtsov et al (15) developed a highly selective small molecule inhibitor, VTP50469, which displaces menin from the fusion protein and prevents the recruitment of MLL to target genes. Both cell and animal studies (immunodeficient NSG mice xenotransplanted with human KMT2A-r leukemia cells) have demonstrated notable anti-leukemic efficacy (15,26). While preclinical data indicate that the small molecule inhibitors exhibit potential anti-leukemic efficacy, the extensive menin-MLL binding interface reveals that the crystal structure of VTP50469 occupies a subregion of the menin-MLL interface (15,27). This indicates the necessity for potentially higher inhibitor concentrations to achieve complete menin-MLL disruption, consistent with the typical dose-response association of partial protein-protein interaction inhibitors (28,29). This technical barrier may represent a notable impediment in the translation of menin-MLL-targeted therapies from laboratory research to clinical application.
The sirtuin family comprises seven highly conserved members (SIRT1-SIRT7) that share evolutionary conservation, all of which possess highly conserved catalytic structural domains, differing markedly only at their N- and C-terminus (30,31). This structure enables each member to exhibit NAD+-dependent deacetylase and ADP-ribosyltransferase activity (31). As components of histone deacetylases, SIRTs serve a key role in cell proliferation, survival, maintenance of genomic stability and metabolic regulation by modulating gene expression and chromatin dynamics. A direct interaction between the menin protein and SIRT1 has been identified (30). Menin binds directly to the deacetylase structural domain of SIRT1 through its C-terminal domain, which forms a functional complex that regulates epigenetic processes. In mouse hepatocytes, SIRT1 modulates CD36 gene expression and intracellular triglyceride accumulation via histone deacetylation, a process that is contingent upon the involvement of the menin protein (32). Furthermore, in hepatocellular carcinoma cells, menin enhances NF-κB (p65) deacetylation by recruiting SIRT1, which underscores its key role in the SIRT regulatory network (33). Numerous studies have demonstrated that SIRT1 impacts apoptosis and inflammatory activation by modulating the NF-κB pathway (34–36). For example, in murine models, upregulation of SIRT1 promotes B lymphocyte proliferation, inhibits apoptosis and advances inflammatory responses by suppressing the NF-κB pathway (37,38). Therefore, it is hypothesized that in acute leukemia, the menin-SIRT interaction may also influence apoptosis and inflammatory activation via the NF-κB pathway, potentially inhibiting or promoting acute leukemia (39).
The human genome encodes a total of 11 PRMTs, which regulate cell signaling networks by modifying arginine residues in both histone and non-histone proteins (40). As important epigenetic regulators in eukaryotes, all members of the PRMT family contain highly conserved S-adenosylmethionine (SAM)-dependent methyltransferase structural domains that facilitate the transfer of methyl groups from SAM to the nitrogen atom of substrate arginine residues (41,42). Menin protein can recruit PRMT5 to the growth arrest-specific protein 1 (GAS1) gene locus, inhibiting GAS1 gene expression by promoting the symmetric dimethylation of histone H4 at position 3 (H4R3me2) (43). However, in MLL, the interaction between menin protein and PRMT5 leads to decreased levels of H4R3me2 and fails to effectively inhibit GAS1 expression (44,45). This suggests that the regulation of PRMT by menin protein may be context-dependent (43). In MEN1 tumor syndrome and mixed-lineage leukemia (MLL), the regulation of GAS1 by the menin-PRMT5 complex exhibits opposite outcomes, suggesting an oncogenic isoform of the menin-PRMT5 complex that reverses epigenetic manifestations by altering the conformation of the complex (43,44).
SUV39H1 is a histone H3 lysine 9 methyltransferase (H3K9). Menin protein enhances trimethylation of H3K9 in the promoter regions of target genes by interacting with SUV39H1, thereby silencing transcription of associated genes (24,46). In a previous study on IL-6 gene regulation, Song et al (47) detected the enrichment of menin protein and SUV39H1 around the IL-6 gene using chromatin immunoprecipitation (ChIP). The aforementioned study reported that menin protein and SUV39H1 are specifically recruited to the auxiliary region of the IL-6 promoter and the recruitment of SUV39H1 decreases with the depletion of menin protein, which suggests menin protein may serve a key role in the regulation of SUV39H1. Further analysis indicated that menin protein and SUV39H1 may regulate IL-6 gene expression at the protein level through H3K9 methylation (47). Numerous preclinical and clinical studies have confirmed that IL-6 serves a notable role in the development of acute leukemia (48–50). Anti-IL-6 antibodies, such as cetuximab, have potential therapeutic value in the treatment of various malignancies, including acute leukemia, either alone or in combination with chemotherapy regimens (51). However, the pathogenesis of leukemia involves a complex regulatory network of multiple signaling pathways - such as JAK/STAT, NF-κB and Wnt/β-catenin - which limits the therapeutic efficacy of targeting IL-6 alone (52–54). Following blocking IL-6 signaling, leukemia cells sustain their survival and proliferative capacity by activating alternative pathways (for example, Janus kinase/STAT), which markedly reduces the clinical benefit of single-agent therapy. These direct epigenetic regulatory interactions mediated by menin are summarized in Table I.
Agarwalet al (18) demonstrated, via a yeast two-hybrid assay using a galactose-responsive transcription factor 4 (Gal4) DNA-binding domain fusion of full-length menin protein, that JunD, a member of the activator protein 1 family, interacts with menin protein. This interaction is contingent upon the structural domains located at the N-terminal end of menin protein. The aforementioned study identified the N-terminal region of JunD binding to menin protein as the menin binding sequence (JunDMBL). Notably, although the binding modes of JunDMBL and the MLL binding sequence exhibit similarities, they exert opposite effects on transcriptional regulation (55). Menin-JunD complexes may interfere with JNK-mediated phosphorylation of JunD and c-Jun, thereby inhibiting the Ras signaling pathway (56,57). However, the specific target genes regulated by the menin-JunD complex remain unclear and further research is warranted to explore its potential application in leukemia treatment (55).
SMAD proteins are key downstream effector molecules in the TGF-β signaling pathway and serve a direct role in TGF-β signaling and gene transcriptional regulation (58). As transcriptional regulators, menin protein binds SMAD proteins, thereby indirectly influencing the activity of the TGF-β signaling pathway and regulating the transcription of target genes. In pituitary secretory tumor cell lines, menin protein markedly enhances the binding specificity of SMAD3 to DNA sequences via interaction with SMAD3 (59,60). More importantly, during osteoblast differentiation and maturation, menin protein forms complexes with SMAD1, SMAD5 and the key regulator of osteogenesis Runt-related transcription factor 2 to promote the differentiation of mesenchymal stem cells into osteoblasts (59,61). While the interaction of menin protein with SMAD1, SMAD3 and SMAD5 has been demonstrated (59), the specific molecular mechanisms underlying their synergistic activation of transcription warrant further investigation.
As a key proto-oncogene, the dysregulated expression of Myc is closely associated with >50% of malignant tumorigenesis (62). Myc serves primarily as a transcription factor through the classical enhancer-box (E-box) region (63). Wu et al (64) demonstrated that Myc forms a regulatory complex with positive transcription elongation factor b (P-TEEb) within the E-box region to activate transcription, with the menin protein being a key factor in this process. Menin regulates Myc-mediated transcriptional activity by influencing the transcriptional elongation regulator P-TEEb. In the KMT2A rearrangement (KMT2A-r) AML model, the expression of Myc and its characteristic genes is markedly suppressed in leukemia cells following treatment with a menin inhibitor (65). Zhou et al (65) found a notable positive correlation between menin protein levels and Myc expression, which suggests Myc may serve as a common target of menin protein. Based on these findings, co-targeting menin protein and Myc may represent a novel therapeutic strategy for leukemia treatment in future.
The FOX family is an evolutionarily conserved group of transcription factors, all of which possess the distinctive forkhead DNA-binding structural domain and are key for cell proliferation and differentiation (66). Previous studies have indicated that menin protein engages with several members of the FOX family, such as FOXG1, FOXA1 and FOXO1 (67–69). In FOXG1-associated encephalopathy, menin protein influences α-thalassemia X-linked mental retardation protein-mediated FOXG1 transcription via modulation of the FOXG1 transcripts (67). Bonnavion et al (70) established that menin protein interacts with FOXA2, which influences its trans-auto reactivation ability and serves a role in the control of FOXA2 expression in adult pancreatic α-cells.
The inaugural member of the Wnt family was identified in 1982 and research has consistently validated the essential function of the Wnt/β-catenin signaling pathway in embryonic development and tissue regeneration (71–73), Dysregulation of this system results in numerous illnesses, such as colorectal and gastric cancer (74–77). Menin protein serves as a reciprocal partner of β-catenin proteins, the principal effector molecules of this signaling pathway, and can facilitate their nuclear translocation. ChIP and chromosome conformation capture (3C) studies demonstrated that the menin protein augments the association of β-catenin proteins with the Myc promoter (78–80). The mechanism of action of menin protein inhibitors in the treatment of AML may partially arise from their suppression of the Wnt/β-catenin protein signaling pathway (81–83).
Nuclear receptors are a class of receptor proteins located in the nucleus, which include the androgen receptor (AR), estrogen, thyroid hormone, glucocorticoid, retinoid X, peroxisome proliferator-activated, liver X and retinoic acid receptors (84). These receptors not only serve as biosensors to regulate key cellular activities such as proliferation, differentiation, and apoptosis but also directly bind to DNA to exert transcriptional regulation (85,86). The interactions between nuclear receptors and tumor growth have attracted notable research regarding their role in tumor progression (87–89). Luo et al (90) identified that the menin protein exhibits distinct pro-oncogenic actions in androgen receptor-dependent prostate cancer cells by modulating AR transcription and its target genes. Based on the presence of nuclear receptor interaction sites in the amino acid sequence of the menin protein, menin serves as an important coactivator of nuclear receptor-mediated transcription (68,90).
The NF-κB family comprises five members: Rel or c-Rel, RelA or p65, RelB, NF-κB1 or p50 and NF-κB2 or p52. Each member exists in dimeric form and possesses a Rel homology domain (91). The pro-carcinogenic role of NF-κB is prevalent in hepatocellular carcinoma (92). Previous studies have demonstrated that menin protein engages with NF-κB and suppresses p65-mediated transcriptional activation via the recruitment of SIRT1 (33,93). Another previous study revealed that the degree of menin-NF-κB interaction changes the production of cell cycle protein D1, a downstream signaling molecule of NF-κB that governs the G1/S phase transition and affects cell proliferation (55). Irregularities in this mechanism may result in tumorigenesis.
The menin-MLL complex activates the expression of the HOX/MEIS1 gene cluster by mediating aberrant H3K4me3, a well-established axis of epigenetic regulation implicated in KMT2A-r and NPM1-mut leukemia (Fig. 1 (17,94). This mechanism was first elucidated by Yokoyama et al (94), who demonstrated that blocking the interaction between menin and the methyltransferase KMT2A markedly decreases leukemia incidence in mice (95). Further research has confirmed the key role of menin protein in NPM1-mut leukemia (96). Specifically, menin protein contributes to KMT2A-r leukemia by binding to fusion proteins and to NPM1-mut leukemias via aberrant nucleoplasmic transport pathways (97). The role of the menin protein in KMT2A-rearranged leukemia and NPM1-mutant leukemia cause the dysregulated expression of the HOX and MEIS1 genes (98,99). Menin inhibitors effectively suppress the abnormal proliferation and differentiation of leukemia cells by specifically disrupting the interaction between menin and KMT2A or NPM1, while simultaneously downregulating the expression of HOX and MEIS1, thereby demonstrating notable therapeutic potential (21).
Patients with acute leukemia characterized by KMT2A-r, previously referred to as MLL, exhibit a long-term survival rate <60% and poor prognosis across all age demographics (100,101). This leukemia variant exhibits a high prevalence among infants and children, accounting for >70% of new acute lymphoblastic leukemia (ALL) diagnoses in infants, is marked by notable aggressiveness, frequent relapse, substantial medication resistance and presents considerable challenges for therapeutic management as a high-risk genetic subtype (101).
KMT2A-r may lead to aberrant expression of the HOX gene and its DNA-binding cofactor MEIS1, which may inhibit hematological development and precipitate leukemia (21,102). Although no specific medications have been approved for KMT2A-r leukemia, preclinical studies have identified the chromatin regulatory protein menin as a promising therapeutic target (14,103,104). In KMT2A-driven leukemia, all KMT2A fusion proteins contain menin-binding sequences, with menin protein serving as a key cofactor that facilitates the interaction between the KMT2A protein complex and the HOX gene promoter. In a study using the KMT2A-mut leukemia model (105), it was demonstrated that the inhibition of menin protein markedly reduces the transcript levels of HOX and MEIS1, thereby reversing the leukemogenesis process.
Through the examination of gene expression in pediatric and adult patients with primary AML, researchers discovered that NPM1-mut leukemia exhibits notable similarities to the KMT2A-r subtype and NPM1 is associated with the HOX/MEIS1 gene cluster (106). NPM1-mut is among the most prevalent genetic alterations in AML, affecting ~30% of the total patient population (107). These mutations, primarily located in the terminal exons of the NPM1 gene, enhance nuclear export signaling activity and impair nucleolus localization signaling, which results in the dysregulated expression of the HOXA/B and MEIS1 genes (108). NPM1 may serve as a transcriptional amplifier of gene expression, potentially constituting a notable factor in the development of AML (109). Dillon et al (110) identified microscopic residual lesions in patients with AML through pre-transplantation DNA sequencing of hematopoietic stem cells, which revealed that patients with NPM1-mut AML experience a significantly higher recurrence rate and shorter survival compared with those without NPM1-mut.
The persistence of NPM1-mut AML in an undifferentiated state is attributed to the HOX-associated pathway, which underscores the therapeutic potential of targeting this pathway. Menin protein serves as a cofactor to promote H3K4me3 through interactions with MLL, thereby modulating the expression of HOX and MEIS1 genes. This mechanism highlights the feasibility of targeting menin protein for therapeutic interventions (96). In vivo experiments have demonstrated that inhibitors of menin protein exhibit notable anti-leukemic activity in NPM1-mut leukemia (20,21,111). Previous studies conducted in NPM1-mutant leukemia models have indicated that treatment with the menin inhibitor VTP50469, a precursor drug to revumenib, leads to downregulation of oncogenic cofactors such as MEIS1 and a marked reduction in the self-renewal capacity of leukemic stem cells (15,104).
Understanding of the menin formation mechanism in leukemia, alongside advancements in high-throughput screening techniques and structural biology, facilitate creation of highly selective small chemical inhibitors (112). Based on the efficacy of menin inhibitors in KMT2A-r and NPM1-mut leukemia, a growing array of menin inhibitors (such as revumenib and ziftomenib) exhibiting enhanced pharmacological efficacy against these AML subtypes has recently been introduced into clinical practice (21,113,114).
A total of seven menin inhibitors are undergoing different phases of clinical development for acute myeloid leukemia, with numerous candidates in development (Table II). The leading candidate is revumenib, which demonstrated a promising safety and effectiveness profile in a phase I open-label, dose-escalation and extension study (AUGMENT-101) assessing the menin inhibitor revumenib for KMT2A-r leukemia treatment (21,113). The occurrence of grade ≤3 treatment-related side events was minimal in treated patients, with asymptomatic QT interval prolongation being the sole dose-limiting effect. Revumenib achieved an overall remission rate of ≤53% and a complete remission rate of ≤30% with partial hematological recovery (98,113). In the subsequent phase II trial (AUGMENT-101), a total of 94 patients with KMT2A-r acute leukemia (comprising 78 patients with AML, 14 with ALL and two with an indeterminate subtype) received menin inhibitors, which achieved an overall remission rate of 63.2% (of which, 68.2% exhibited no measurable residual disease; unpublished data). In 57 patients with assessable efficacy, the combined complete response and complete response with incomplete hematological recovery rate reached 22.8% (113). Grade ≥3 adverse events included neutropenia (37.2%), differentiation syndrome (16%) and QT interval prolongation (13.8%). Most of these events were controllable and transitory and menin inhibitors had a predictable safety profile.
The combination therapy of menin inhibitors with other targeted leukemia medications has potential due to the promising efficacy and safety profile of menin inhibitor monotherapy in the treatment of acute leukemia. Miao et al (115) administered a menin inhibitor and kinase inhibitor to NUP98-r leukemia samples, which demonstrated that the combination therapy outperformed monotherapy, with a combination index between 0.12 and 0.65, as determined by the Chou-Talalay method, which indicated notable synergistic effects. The combinatorial therapy induced a more pronounced decrease in both the quantity and size of leukemic blasts in NUP98-r leukemia specimens and was more effective in promoting cell proliferation arrest and differentiation. Furthermore, the combination of Brahma-related gene 1/Brahma inhibitors with menin inhibitors for acute leukemia treatment has demonstrated notable preclinical efficacy, resulting in a more substantial reduction in leukemia burden and extended survival duration in mice compared with monotherapy (116). A clinical trial is currently examining menin inhibitors in conjunction with azacitidine/vincristine for the treatment of acute leukemia, specifically evaluating JNJ-75276617 with AML-targeted treatments (trial no. NCT05453903) (117). The combined regimen inhibits the immune evasion of acute leukemia cells following monotherapy and diminishes relapse in treatment-resistant leukemia (117).
Common adverse effects of menin inhibitors include gastrointestinal reactions, QT interval prolongation, cytopenia and differentiation syndromes, however, these adverse effects are within manageable limits and menin inhibitors are generally safe (113,118). Furthermore, menin inhibitors are not associated with notable off-target toxicity, which has enabled the clinical scale-up of menin inhibitors and their emergence as a potential option for long-term therapy (112). However, recent data have also demonstrated that certain patients have menin mutations that prevent binding of the inhibitor and thus mediate clinical resistance, which leads to clinical relapse; therefore, resistance to menin inhibitors remains a challenge (21,119).
Although menin inhibitors have demonstrated significant efficacy in KMT2A-r and NPM1-mut leukemia, their clinical application faces key challenges such as drug resistance and optimization of combination strategies (21,98). Future research should focus on overcoming resistance mediated by menin protein mutations, including the development of allosteric inhibitors and combination with epigenetic regulatory drugs to block compensatory pathways. In terms of combination therapy, it is necessary to optimize combination regimens with other drugs based on synergy indices (such as the Chou-Talalay model) and explore the potential advantages of sequential therapy. Furthermore, the indication scope of menin inhibitors should be expanded to include other subtypes dependent on the HOX/MEIS1 pathway, such as NUP98-r leukemia and predictive biomarkers based on HOX gene expression profiles or menin-MLL complex activity should be developed to screen beneficiary populations. From a technical perspective, structural biology and artificial intelligence-assisted design should be leveraged to develop high-affinity inhibitors and targeted delivery systems should be developed to enhance efficacy and safety. In-depth studies of the menin protein regulatory network may reveal its functional heterogeneity in different cell environments and provide a theoretical basis for dual-targeting strategies (such as menin-Myc co-inhibition). With the advancement of multidisciplinary collaboration, menin inhibitors may become a key therapy for specific leukemia subtypes and provide novel directions for epigenetically targeted therapy.
Not applicable.
The present study was supported by the National Natural Science Foundation of China (grant no. 81600129), Zhejiang Provincial Natural Science Foundation of China (grant nos. LY21H080001 and BY22H205675), the Medical and Health Research Project of Zhejiang Province (grant nos. WKJ-ZJ-2444 and 2022KY944) and Zhejiang Provincial Traditional Chinese Medicine Science and Technology Project (grant no. 2022ZB276).
Not applicable.
HZ conceived, designed and supervised the study and edited the manuscript. JB wrote the manuscript. Data authentication is not applicable. Both authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Pollyea DA, Bixby D, Perl A, Bhatt VR, Altman JK, Appelbaum FR, de Lima M, Fathi AT, Foran JM, Gojo I, et al: NCCN guidelines insights: Acute myeloid leukemia, version 2.2021. J Natl Compr Canc Netw. 19:16–27. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
De Kouchkovsky I and Abdul-Hay M: Acute myeloid leukemia: A comprehensive review and 2016 update. Blood Cancer J. 6:e4412016. View Article : Google Scholar : PubMed/NCBI | |
|
Shimony S, Stahl M and Stone RM: Acute myeloid leukemia: 2025 update on diagnosis, risk-stratification, and management. Am J Hematol. 100:860–891. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Sasaki K, Ravandi F, Kadia TM, DiNardo CD, Short NJ, Borthakur G, Jabbour E and Kantarjian HM: De novo acute myeloid leukemia: A population-based study of outcome in the United States based on the surveillance, epidemiology, and end results (SEER) database, 1980 to 2017. Cancer. 127:2049–2061. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Fruchtman H, Avigan ZM, Waksal JA, Brennan N and Mascarenhas JO: Management of isocitrate dehydrogenase 1/2 mutated acute myeloid leukemia. Leukemia. 38:927–935. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Short NJ, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD and Daver N: Advances in the treatment of acute myeloid leukemia: New drugs and new challenges. Cancer Discov. 10:506–525. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Carter JL, Hege K, Yang J, Kalpage HA, Su Y, Edwards H, Hüttemann M, Taub JW and Ge Y: Targeting multiple signaling pathways: The new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther. 5:2882020. View Article : Google Scholar : PubMed/NCBI | |
|
Pei H, Guo W, Peng Y, Xiong H and Chen Y: Targeting key proteins involved in transcriptional regulation for cancer therapy: Current strategies and future prospective. Med Res Rev. 42:1607–1660. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Milano L, Gautam A and Caldecott KW: DNA damage and transcription stress. Mol Cell. 84:70–79. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Morgan MP, Finnegan E and Das S: The role of transcription factors in the acquisition of the four latest proposed hallmarks of cancer and corresponding enabling characteristics. Semin Cancer Biol. 86:1203–1215. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Vervoort SJ, Devlin JR, Kwiatkowski N, Teng M, Gray NS and Johnstone RW: Targeting transcription cycles in cancer. Nat Rev Cancer. 22:5–24. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Layden HM, Johnson AE and Hiebert SW: Chemical-genetics refines transcription factor regulatory circuits. Trends Cancer. 10:65–75. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kathman SG, Koo SJ, Lindsey GL, Her HL, Blue SM, Li H, Jaensch S, Remsberg JR, Ahn K, Yeo GW, et al: Remodeling oncogenic transcriptomes by small molecules targeting NONO. Nat Chem Biol. 19:825–836. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Appel LM, Benedum J, Engl M, Platzer S, Schleiffer A, Strobl X and Slade D: SPOC domain proteins in health and disease. Genes Dev. 37:140–170. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Krivtsov AV, Evans K, Gadrey JY, Eschle BK, Hatton C, Uckelmann HJ, Ross KN, Perner F, Olsen SN, Pritchard T, et al: A Menin-MLL inhibitor induces specific chromatin changes and eradicates disease in models of MLL-rearranged leukemia. Cancer Cell. 36:660–673.e11. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kühn MW, Song E, Feng Z, Sinha A, Chen CW, Deshpande AJ, Cusan M, Farnoud N, Mupo A, Grove C, et al: Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia. Cancer Discov. 6:1166–1181. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Cuglievan B, Kantarjian H, Rubnitz JE, Cooper TM, Zwaan CM, Pollard JA, DiNardo CD, Kadia TM, Guest E, Short NJ, et al: Menin inhibitors in pediatric acute leukemia: A comprehensive review and recommendations to accelerate progress in collaboration with adult leukemia and the international community. Leukemia. 38:2073–2084. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Agarwal SK, Guru SC, Heppner C, Erdos MR, Collins RM, Park SY, Saggar S, Chandrasekharappa SC, Collins FS, Spiegel AM, et al: Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription. Cell. 96:143–152. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Huang J, Gurung B, Wan B, Matkar S, Veniaminova NA, Wan K, Merchant JL, Hua X and Lei M: The same pocket in menin binds both MLL and JUND but has opposite effects on transcription. Nature. 482:542–546. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Fiskus W, Mill CP, Birdwell C, Davis JA, Das K, Boettcher S, Kadia TM, DiNardo CD, Takahashi K, Loghavi S, et al: Targeting of epigenetic co-dependencies enhances anti-AML efficacy of Menin inhibitor in AML with MLL1-r or mutant NPM1. Blood Cancer J. 13:532023. View Article : Google Scholar : PubMed/NCBI | |
|
Issa GC, Aldoss I, DiPersio J, Cuglievan B, Stone R, Arellano M, Thirman MJ, Patel MR, Dickens DS, Shenoy S, et al: The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature. 615:920–924. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Gundry MC, Goodell MA and Brunetti L: It's all about MEis: Menin-MLL inhibition eradicates NPM1-Mutated and MLL-rearranged acute leukemias in mice. Cancer Cell. 37:267–269. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Borkin D, He S, Miao H, Kempinska K, Pollock J, Chase J, Purohit T, Malik B, Zhao T, Wang J, et al: Pharmacologic inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell. 27:589–602. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Z, Ma J and Hua X: Epigenetic regulation by the menin pathway. Endocr Relat Cancer. 24:T147–T159. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yokoyama A and Cleary ML: Menin critically links MLL proteins with LEDGF on cancer-associated target genes. Cancer Cell. 14:36–46. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Adriaanse FRS, Schneider P, Arentsen-Peters STCJM, Fonseca AMND, Stutterheim J, Pieters R, Zwaan CM and Stam RW: Distinct responses to menin inhibition and synergy with DOT1L inhibition in KMT2A-rearranged acute lymphoblastic and myeloid leukemia. Int J Mol Sci. 25:60202024. View Article : Google Scholar : PubMed/NCBI | |
|
Kurmasheva RT, Bandyopadhyay A, Favours E, Pozo VD, Ghilu S, Phelps DA, McGeehan GM, Erickson SW, Smith MA and Houghton PJ: Evaluation of VTP-50469, a menin-MLL1 inhibitor, against Ewing sarcoma xenograft models by the pediatric preclinical testing consortium. Pediatr Blood Cancer. 67:e282842020. View Article : Google Scholar : PubMed/NCBI | |
|
Nicholls SJ, Nissen SE, Fleming C, Urva S, Suico J, Berg PH, Linnebjerg H, Ruotolo G, Turner PK and Michael LF: Muvalaplin, an oral small molecule inhibitor of lipoprotein(a) formation: A randomized clinical trial. JAMA. 330:1042–1053. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Groenland SL, Martínez-Chávez A, van Dongen MGJ, Beijnen JH, Schinkel AH, Huitema ADR and Steeghs N: Clinical pharmacokinetics and pharmacodynamics of the cyclin-dependent kinase 4 and 6 inhibitors palbociclib, ribociclib, and abemaciclib. Clin Pharmacokinet. 59:1501–1520. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ianni A, Kumari P, Tarighi S, Braun T and Vaquero A: SIRT7: A novel molecular target for personalized cancer treatment? Oncogene. 43:993–1006. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Goes JVC, Carvalho LG, de Oliveira RTG, Melo MML, Novaes LAC, Moreno DA, Gonçalves PG, Montefusco-Pereira CV, Pinheiro RF and Ribeiro Junior HL: Role of sirtuins in the pathobiology of onco-hematological diseases: A PROSPERO-registered study and in silico analysis. Cancers (Basel). 14:46112022. View Article : Google Scholar : PubMed/NCBI | |
|
Cao Y, Xue Y, Xue L, Jiang X, Wang X, Zhang Z, Yang J, Lu J, Zhang C, Wang W and Ning G: Hepatic menin recruits SIRT1 to control liver steatosis through histone deacetylation. J Hepatol. 59:1299–1306. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gang D, Hongwei H, Hedai L, Ming Z, Qian H and Zhijun L: The tumor suppressor protein menin inhibits NF-κB-mediated transactivation through recruitment of Sirt1 in hepatocellular carcinoma. Mol Biol Rep. 40:2461–2466. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Hernández-Jiménez M, Hurtado O, Cuartero MI, Ballesteros I, Moraga A, Pradillo JM, McBurney MW, Lizasoain I and Moro MA: Silent information regulator 1 protects the brain against cerebral ischemic damage. Stroke. 44:2333–2337. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Teng Y, Huang Y, Yu H, Wu C, Yan Q, Wang Y, Yang M, Xie H, Wu T, Yang H and Zou J: Nimbolide targeting SIRT1 mitigates intervertebral disc degeneration by reprogramming cholesterol metabolism and inhibiting inflammatory signaling. Acta Pharm Sin B. 13:2269–2280. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao X, Li M, Lu Y, Wang M, Xiao J, Xie Q, He X and Shuai S: Sirt1 inhibits macrophage polarization and inflammation in gouty arthritis by inhibiting the MAPK/NF-κB/AP-1 pathway and activating the Nrf2/HO-1 pathway. Inflamm Res. 73:1173–1184. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Yan C, Xin M, Han L, Zhang Y and Sun M: Sirtuin 1 (Sirt1) overexpression in BaF3 cells contributes to cell proliferation promotion, apoptosis resistance and pro-inflammatory cytokine production. Med Sci Monit. 23:1477–1482. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kotas ME, Gorecki MC and Gillum MP: Sirtuin-1 is a nutrient-dependent modulator of inflammation. Adipocyte. 2:113–118. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Wu QJ, Zhang TN, Chen HH, Yu XF, Lv JL, Liu YY, Liu YS, Zheng G, Zhao JQ, Wei YF, et al: The sirtuin family in health and disease. Signal Transduct Target Ther. 7:4022022. View Article : Google Scholar : PubMed/NCBI | |
|
Alinari L, Mahasenan KV, Yan F, Karkhanis V, Chung JH, Smith EM, Quinion C, Smith PL, Kim L, Patton JT, et al: Selective inhibition of protein arginine methyltransferase 5 blocks initiation and maintenance of B-cell transformation. Blood. 125:2530–2543. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Peng J, Ni B, Li D, Cheng B and Yang R: Overview of the PRMT6 modulators in cancer treatment: Current progress and emerged opportunity. Eur J Med Chem. 279:1168572024. View Article : Google Scholar : PubMed/NCBI | |
|
Qian K, Hu H, Xu H and Zheng YG: Detection of PRMT1 inhibitors with stopped flow fluorescence. Signal Transduct Target Ther. 3:62018. View Article : Google Scholar : PubMed/NCBI | |
|
Abe Y and Tanaka N: Fine-Tuning of GLI activity through arginine methylation: Its mechanisms and function. Cells. 9:19732020. View Article : Google Scholar : PubMed/NCBI | |
|
Gurung B, Feng Z, Iwamoto DV, Thiel A, Jin G, Fan CM, Ng JM, Curran T and Hua X: Menin epigenetically represses Hedgehog signaling in MEN1 tumor syndrome. Cancer Res. 73:2650–2658. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Kim H and Ronai ZA: PRMT5 function and targeting in cancer. Cell Stress. 4:199–215. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Padeken J, Methot SP and Gasser SM: Establishment of H3K9-methylated heterochromatin and its functions in tissue differentiation and maintenance. Nat Rev Mol Cell Biol. 23:623–640. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Song TY, Lim J, Kim B, Han JW, Youn HD and Cho EJ: The role of tumor suppressor menin in IL-6 regulation in mouse islet tumor cells. Biochem Biophys Res Commun. 51:308–313. 2014. View Article : Google Scholar | |
|
Mei Y, Ren K, Liu Y, Ma A, Xia Z, Han X, Li E, Tariq H, Bao H, Xie X, et al: Bone marrow-confined IL-6 signaling mediates the progression of myelodysplastic syndromes to acute myeloid leukemia. J Clin Invest. 132:e1526732022. View Article : Google Scholar : PubMed/NCBI | |
|
Burger R: Impact of interleukin-6 in hematological malignancies. Transfus Med Hemother. 40:336–343. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Kaser EC, Zhao L, D'Mello KP, Zhu Z, Xiao H, Wakefield MR, Bai Q and Fang Y: The role of various interleukins in acute myeloid leukemia. Med Oncol. 38:552021. View Article : Google Scholar : PubMed/NCBI | |
|
Yao X, Huang J, Zhong H, Shen N, Faggioni R, Fung M and Yao Y: Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol Ther. 141:125–139. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Qin R, Wang T, He W, Wei W, Liu S, Gao M and Huang Z: Jak2/STAT6/c-Myc pathway is vital to the pathogenicity of Philadelphia-positive acute lymphoblastic leukemia caused by P190(BCR-ABL). Cell Commun Signal. 21:272023. View Article : Google Scholar : PubMed/NCBI | |
|
Di Francesco B, Verzella D, Capece D, Vecchiotti D, Di Vito Nolfi M, Flati I, Cornice J, Di Padova M, Angelucci A, Alesse E and Zazzeroni F: NF-κB: A druggable target in acute myeloid leukemia. Cancers (Basel). 14:35572022. View Article : Google Scholar : PubMed/NCBI | |
|
Láinez-González D, Alonso-Aguado AB and Alonso-Dominguez JM: Understanding the Wnt signaling pathway in acute myeloid leukemia stem cells: A feasible key against relapses. Biology (Basel). 12:6832023.PubMed/NCBI | |
|
Liu P, Shi C, Qiu L, Shang D, Lu Z, Tu Z and Liu H: Menin signaling and therapeutic targeting in breast cancer. Curr Probl Cancer. 51:1011182024. View Article : Google Scholar : PubMed/NCBI | |
|
Paneni F, Osto E, Costantino S, Mateescu B, Briand S, Coppolino G, Perna E, Mocharla P, Akhmedov A, Kubant R, et al: Deletion of the activated protein-1 transcription factor JunD induces oxidative stress and accelerates age-related endothelial dysfunction. Circulation. 127:1229–1240. e1–e21. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gallo A, Cuozzo C, Esposito I, Maggiolini M, Bonofiglio D, Vivacqua A, Garramone M, Weiss C, Bohmann D and Musti AM: Menin uncouples Elk-1, JunD and c-Jun phosphorylation from MAP kinase activation. Oncogene. 21:6434–6445. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Dockray GJ: Keeping neuroendocrine cells in check: Roles for TGFbeta, Smads, and menin? Gut. 52:1237–1239. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Hendy GN, Kaji H, Sowa H, Lebrun JJ and Canaff L: Menin and TGF-beta superfamily member signaling via the Smad pathway in pituitary, parathyroid and osteoblast. Horm Metab Res. 37:375–379. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Matkar S, Thiel A and Hua X: Menin: A scaffold protein that controls gene expression and cell signaling. Trends Biochem Sci. 38:394–402. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Sowa H, Kaji H, Hendy GN, Canaff L, Komori T, Sugimoto T and Chihara K: Menin is required for bone morphogenetic protein 2- and transforming growth factor beta-regulated osteoblastic differentiation through interaction with Smads and Runx2. J Biol Chem. 279:40267–40275. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Liu H and Qing G: Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduct Target Ther. 3:52018. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, Wong E, Orlov YL, Zhang W, Jiang J, et al: Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell. 133:1106–1117. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Wu G, Yuan M, Shen S, Ma X, Fang J, Zhu L, Sun L, Liu Z, He X, Huang D, et al: Menin enhances c-Myc-mediated transcription to promote cancer progression. Nat Commun. 8:152782017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Zhang L, Aryal S, Veasey V, Tajik A, Restelli C, Moreira S, Zhang P, Zhang Y, Hope KJ, et al: Epigenetic regulation of noncanonical menin targets modulates menin inhibitor response in acute myeloid leukemia. Blood. 144:2018–2032. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tsai JW, Cejas P, Wang DK, Patel S, Wu DW, Arounleut P, Wei X, Zhou N, Syamala S, Dubois FPB, et al: FOXR2 is an epigenetically regulated pan-cancer oncogene that activates ETS transcriptional circuits. Cancer Res. 82:2980–3001. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhuang K, Leng L, Su X, Wang S, Su Y, Chen Y, Yuan Z, Zi L, Li J, Xie W, et al: Menin deficiency induces autism-like behaviors by regulating foxg1 transcription and participates in foxg1-related encephalopathy. Adv Sci (Weinh). 11:e23079532024. View Article : Google Scholar : PubMed/NCBI | |
|
Dreijerink KMA, Groner AC, Vos ESM, Font-Tello A, Gu L, Chi D, Chi D, Reyes J, Cook J, Lim E, et al: Enhancer-mediated oncogenic function of the menin tumor suppressor in breast cancer. Cell Rep. 18:2359–2372. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Z, Shi D, Tu Y, Tian J, Zhang W, Xing B, Wang J, Liu S, Lou J, Gustafsson JÅ, et al: Human proislet peptide promotes pancreatic progenitor cells to ameliorate diabetes through FOXO1/menin-mediated epigenetic regulation. Diabetes. 67:1345–1355. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Bonnavion R, Teinturier R, Gherardi S, Leteurtre E, Yu R, Cordier-Bussat M, Du R, Pattou F, Vantyghem MC, Bertolino P, et al: Foxa2, a novel protein partner of the tumour suppressor menin, is deregulated in mouse and human MEN1 glucagonomas. J Pathol. 242:90–101. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Nusse R and Clevers H: Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169:985–999. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Bonnet C, Brahmbhatt A, Deng SX and Zheng JJ: Wnt signaling activation: Targets and therapeutic opportunities for stem cell therapy and regenerative medicine. RSC Chem Biol. 2:1144–1157. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Steinhart Z and Angers S: Wnt signaling in development and tissue homeostasis. Development. 145:dev1465892018. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, Zhou Z, Shu G and Yin G: Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther. 7:32022. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang Z, Wang Y, Ma X, Song S, He Y, Zhou J, Feng L, Yang S, Wu Y, Yu B, et al: Targeting the NOTCH2/ADAM10/TCF7L2 Axis-mediated transcriptional regulation of Wnt pathway suppresses tumor growth and enhances chemosensitivity in colorectal cancer. Adv Sci (Weinh). 12:e24057582025. View Article : Google Scholar : PubMed/NCBI | |
|
Hao J, Liu C, Gu Z, Yang X, Lan X and Guo X: Dysregulation of Wnt/β-catenin signaling contributes to intestinal inflammation through regulation of group 3 innate lymphoid cells. Nat Commun. 15:28202024. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Q, Nie F, Gan L, Wei X, Liu P, Liu H, Zhang K, Fang Z, Wang H and Fang N: Tripartite motif 31 drives gastric cancer cell proliferation and invasion through activating the Wnt/β-catenin pathway by regulating Axin1 protein stability. Sci Rep. 13:200992023. View Article : Google Scholar : PubMed/NCBI | |
|
Luo Y, Vlaeminck-Guillem V, Baron S, Dallel S, Zhang CX and Le Romancer M: MEN1 silencing aggravates tumorigenic potential of AR-independent prostate cancer cells through nuclear translocation and activation of JunD and β-catenin. J Exp Clin Cancer Res. 40:2702021. View Article : Google Scholar : PubMed/NCBI | |
|
Hagège H, Klous P, Braem C, Splinter E, Dekker J, Cathala G, de Laat W and Forné T: Quantitative analysis of chromosome conformation capture assays (3C-qPCR). Nat Protoc. 2:1722–1733. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Sancho A, Li S, Paul T, Zhang F, Aguilo F, Vashisht A, Balasubramaniyan N, Leleiko NS, Suchy FJ, Wohlschlegel JA, et al: CHD6 regulates the topological arrangement of the CFTR locus. Hum Mol Genet. 24:2724–2732. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Krivtsov AV, Sinha AU, North TE, Goessling W, Feng Z, Zon LI and Armstrong SA: The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science. 327:1650–1653. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Wagstaff M, Coke B, Hodgkiss GR and Morgan RG: Targeting β-catenin in acute myeloid leukaemia: Past present, and future perspectives. Biosci Rep. 42:2022. View Article : Google Scholar : PubMed/NCBI | |
|
Khan I, Eklund EE and Gartel AL: Therapeutic vulnerabilities of transcription factors in AML. Mol Cancer Ther. 20:229–237. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Font-Díaz J, Jiménez-Panizo A, Caelles C, Vivanco MD, Pérez P, Aranda A, Estébanez-Perpiñá E, Castrillo A, Ricote M and Valledor AF: Nuclear receptors: Lipid and hormone sensors with essential roles in the control of cancer development. Semin Cancer Biol. 73:58–75. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Z, Gimple RC, Zhou N, Zhao L, Gustafsson J and Zhou S: Targeting nuclear receptors for cancer therapy: Premises, promises, and challenges. Trends Cancer. 7:541–556. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lian F, Wang Y, Xiao Y, Wu X, Xu H, Liang L and Yang X: Activated farnesoid X receptor attenuates apoptosis and liver injury in autoimmune hepatitis. Mol Med Rep. 12:5821–5827. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Huangyang P, Wang Y, Xue L, Devericks E, Nguyen HG, Yu X, Oses-Prieto JA, Burlingame AL, Miglani S, et al: ERα is an RNA-binding protein sustaining tumor cell survival and drug resistance. Cell. 184:5215–5229.e17. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Manickasamy MK, Jayaprakash S, Girisa S, Kumar A, Lam HY, Okina E, Eng H, Alqahtani MS, Abbas M, Sethi G, et al: Delineating the role of nuclear receptors in colorectal cancer, a focused review. Discov Oncol. 15:412024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Xie J, Cai S, Wang Q, Feng Z, Li Y, Lu JJ, Chen W and Ye Z: Elevated expression of nuclear receptor-binding SET domain 3 promotes pancreatic cancer cell growth. Cell Death Dis. 12:9132021. View Article : Google Scholar : PubMed/NCBI | |
|
Luo Y, Vlaeminck-Guillem V, Teinturier R, Abou Ziki R, Bertolino P, Le Romancer M and Zhang CX: The scaffold protein menin is essential for activating the MYC locus and MYC-mediated androgen receptor transcription in androgen receptor-dependent prostate cancer cells. Cancer Commun (Lond). 41:1427–1430. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang T, Ma C, Zhang Z, Zhang H and Hu H: NF-κB signaling in inflammation and cancer. MedComm (2020). 2:618–653. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
He G and Karin M: NF-κB and STAT3 - key players in liver inflammation and cancer. Cell Res. 21:159–168. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA and Mayo MW: Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 23:2369–2380. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Yokoyama A, Somervaille TC, Smith KS, Rozenblatt-Rosen O, Meyerson M and Cleary ML: The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell. 123:207–218. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Mullard A: FDA approves first biparatopic antibody therapy. Nat Rev Drug Discov. 24:72025. View Article : Google Scholar | |
|
Falini B, Brunetti L, Sportoletti P and Martelli MP: NPM1-mutated acute myeloid leukemia: From bench to bedside. Blood. 136:1707–1721. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Falini B, Gjertsen BT and Andresen V: The acidic stretch and the C-terminal nuclear export signal motif of NPM1 mutant: Are they druggable in AML? Leukemia. 37:2173–2175. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Perner F, Stein EM, Wenge DV, Singh S, Kim J, Apazidis A, Rahnamoun H, Anand D, Marinaccio C, Hatton C, et al: MEN1 mutations mediate clinical resistance to menin inhibition. Nature. 615:913–919. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Brunetti L, Gundry MC, Sorcini D, Guzman AG, Huang YH, Ramabadran R, Gionfriddo I, Mezzasoma F, Milano F, Nabet B, et al: Mutant NPM1 maintains the leukemic state through HOX expression. Cancer Cell. 34:499–512.e9. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Krivtsov AV and Armstrong SA: MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 7:823–833. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Forgione MO, McClure BJ, Eadie LN, Yeung DT and White DL: KMT2A rearranged acute lymphoblastic leukaemia: Unravelling the genomic complexity and heterogeneity of this high-risk disease. Cancer Lett. 469:410–418. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Thorsteinsdottir U, Kroon E, Jerome L, Blasi F and Sauvageau G: Defining roles for HOX and MEIS1 genes in induction of acute myeloid leukemia. Mol Cell Biol. 21:224–234. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Grembecka J, He S, Shi A, Purohit T, Muntean AG, Sorenson RJ, Showalter HD, Murai MJ, Belcher AM, Hartley T, et al: Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat Chem Biol. 8:277–284. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Uckelmann HJ, Kim SM, Wong EM, Hatton C, Giovinazzo H, Gadrey JY, Krivtsov AV, Rücker FG, Döhner K, McGeehan GM, et al: Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science. 367:586–590. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Di Fazio P: Targeting menin: A promising therapeutic strategy for susceptible acute leukemia subtypes. Signal Transduct Target Ther. 8:3842023. View Article : Google Scholar : PubMed/NCBI | |
|
Nadiminti KVG, Sahasrabudhe KD and Liu H: Menin inhibitors for the treatment of acute myeloid leukemia: Challenges and opportunities ahead. J Hematol Oncol. 17:1132024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang R, Xu P, Chang LL, Zhang SZ and Zhu HH: Targeted therapy in NPM1-mutated AML: Knowns and unknowns. Front Oncol. 12:9726062022. View Article : Google Scholar : PubMed/NCBI | |
|
Uckelmann HJ, Haarer EL, Takeda R, Wong EM, Hatton C, Marinaccio C, Perner F, Rajput M, Antonissen NJC, Wen Y, et al: Mutant NPM1 directly regulates oncogenic transcription in acute myeloid leukemia. Cancer Discov. 13:746–765. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang XQD, Fan D, Han Q, Liu Y, Miao H, Wang X, Li Q, Chen D, Gore H, Himadewi P, et al: Mutant NPM1 hijacks transcriptional hubs to maintain pathogenic gene programs in acute myeloid leukemia. Cancer Discov. 13:724–745. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dillon LW, Gui G, Page KM, Ravindra N, Wong ZC, Andrew G, Mukherjee D, Zeger SL, El Chaer F, Spellman S, et al: DNA sequencing to detect residual disease in adults with acute myeloid leukemia prior to hematopoietic cell transplant. JAMA. 329:745–755. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Mill CP, Fiskus W, Das K, Davis JA, Birdwell CE, Kadia TM, DiNardo CD, Daver N, Takahashi K, Sasaki K, et al: Causal linkage of presence of mutant NPM1 to efficacy of novel therapeutic agents against AML cells with mutant NPM1. Leukemia. 37:1336–1348. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Huls G, Woolthuis CM and Schuringa JJ: Menin inhibitors in the treatment of acute myeloid leukemia. Blood. 145:561–566. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Issa GC, Aldoss I, Thirman MJ, DiPersio J, Arellano M, Blachly JS, Mannis GN, Perl A, Dickens DS, McMahon CM, et al: Menin inhibition with revumenib for KMT2A-Rearranged relapsed or refractory acute leukemia (AUGMENT-101). J Clin Oncol. 43:75–84. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang ES, Issa GC, Erba HP, Altman JK, Montesinos P, DeBotton S, Walter RB, Pettit K, Savona MR, Shah MV, et al: Ziftomenib in relapsed or refractory acute myeloid leukaemia (KOMET-001): A multicentre, open-label, multi-cohort, phase 1 trial. Lancet Oncol. 25:1310–1324. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Miao H, Chen D, Ropa J, Purohit T, Kim E, Sulis ML, Ferrando A, Cierpicki T and Grembecka J: Combination of menin and kinase inhibitors as an effective treatment for leukemia with NUP98 translocations. Leukemia. 38:1674–1687. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Fiskus W, Piel J, Collins M, Hentemann M, Cuglievan B, Mill CP, Birdwell CE, Das K, Davis JA, Hou H, et al: BRG1/BRM inhibitor targets AML stem cells and exerts superior preclinical efficacy combined with BET or menin inhibitor. Blood. 143:2059–2072. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kwon MC, Thuring JW, Querolle O, Dai X, Verhulst T, Pande V, Marien A, Goffin D, Wenge DV, Yue H, et al: Preclinical efficacy of the potent, selective menin-KMT2A inhibitor JNJ-75276617 (bleximenib) in KMT2A- and NPM1-altered leukemias. Blood. 144:1206–1220. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
An ZY and Zhang XH: Menin inhibitors for acute myeloid leukemia: latest updates from the 2023 ASH Annual Meeting. J Hematol Oncol. 17:522024. View Article : Google Scholar : PubMed/NCBI | |
|
Heikamp EB, Henrich JA, Perner F, Wong EM, Hatton C, Wen Y, Barwe SP, Gopalakrishnapillai A, Xu H, Uckelmann HJ, et al: The menin-MLL1 interaction is a molecular dependency in NUP98-rearranged AML. Blood. 139:894–906. 2022. View Article : Google Scholar : PubMed/NCBI |