Open access peer-reviewed chapter - ONLINE FIRST

MYC: Master Regulator of Cell Death and Tumor Progression

Written By

Lucia Capasso, Donato Mele, Fatima Fayyaz, Lucia Altucci and Angela Nebbioso

Submitted: 05 December 2024 Reviewed: 13 December 2024 Published: 14 March 2025

DOI: 10.5772/intechopen.1008763

Cell Death Regulation in Pathology IntechOpen
Cell Death Regulation in Pathology Edited by Vincenzo Carafa

From the Edited Volume

Cell Death Regulation in Pathology [Working Title]

Dr. Vincenzo Carafa

Chapter metrics overview

12 Chapter Downloads

View Full Metrics

Abstract

MYC gene has become one of the most investigated oncogenes for regulating programmed cell death and tumor growth. MYC is a transcription factor that regulates the expression of numerous genes involved in critical cellular processes, such as metabolism, stress response, and proliferation. However, its dysfunction, often caused by gene amplifications or translocations, makes it a potent oncogenic driver, contributing to uncontrolled growth, angiogenesis, invasiveness, and metastasis. Paradoxically, MYC can promote both tumor cell survival and elimination through the activation of apoptotic mechanisms, creating a delicate balance between cell survival and death. This chapter explores the dual role of MYC as a regulator of cell life and death, analyzing the molecular mechanisms that determine its activity in different biological contexts. The main apoptotic pathways controlled by MYC, its contribution to tumor plasticity, and its interactions with other oncogenes and tumor suppressors will be discussed. Finally, emerging therapeutic strategies aimed at targeting MYC or its regulatory networks will be reviewed, along with the challenges of translating this knowledge into clinical interventions. A thorough understanding of MYC biology is crucial to develop innovative therapies and improve the treatment of aggressive and resistant tumors.

Keywords

  • MYC
  • cell death
  • tumorigenesis
  • signaling pathways
  • therapeutic target

1. Introduction

MYC gene, which locates on chromosome 8q24.21, is responsible for codifying the transcription factor MYC [1, 2]. MYC represents one of the more widely and influential oncogenes in cellular and tumor biology, because of its involvement in a multitude of vital processes [2]. The codified protein belongs to the bHLH-LZ (basic helix-loop-helix leucine zipper) family of transcription factors, including also N-MYC and L-MYC. It regulates the expression of an extraordinary number of genes, estimated to be approximately 15% of the human genome [3]. MYC activates or represses genes involved in vital activities such as cell proliferation, protein synthesis, energy metabolism, DNA repair, and the cell cycle by building complexes with the MAX protein and binding to certain DNA regions known as E-boxes [4]. Under physiological conditions, MYC acts as a mediator of cellular responses to external stimuli, such as growth factors and mitogenic signals (Figure 1) [5, 6]. This function allows cells to adapt their growth and division to the needs of the surrounding tissue, maintaining a dynamic balance between proliferation and differentiation [5]. However, when MYC expression or activity is dysregulated, as frequently occurs in tumors, a cascade of pathological events occurs that favor uncontrolled growth and aberrant survival of cells (Figure 1) [7]. One of the many characteristics of MYC is its dual role in programmed cell death or apoptosis [8]. Under physiological conditions, MYC promotes cell survival and proliferation, but under stress conditions it can activate potent apoptotic pathways that lead to the elimination of damaged cells (Figure 1). This dual role makes MYC not only a growth promoter, but also a guardian against uncontrolled proliferation [9, 10]. At the molecular level, MYC activates apoptotic pathways by interacting with key proteins, such as p53, a major tumor suppressor [11]. Under conditions of cellular stress, excessive MYC activity can induce the expression of pro-apoptotic genes, such as BCL-2-associated X protein (BAX) and p53 upregulated modulator of apoptosis (PUMA), while inhibiting anti-apoptotic proteins, such as B-cell lymphoma 2 (BCL-2). This delicate balance represents an intrinsic safety strategy: if MYC activity exceeds a critical threshold, cells are induced to die to avoid pathological proliferation. However, in tumors, this mechanism is frequently bypassed [12, 13]. Mutations or alterations in apoptotic pathways, such as the loss of p53, allow tumor cells to use the proliferative activity of MYC without suffering its apoptotic consequences [14]. MYC is deregulated in the vast majority of tumors, indeed its overexpression, caused by several genetic alterations, such as gene amplifications, chromosomal translocations, or aberrant transcriptional activation, is associated with the uncontrolled proliferation of tumor cells [5, 15]. MYC promotes tumor progression through uncontrolled cell proliferation, activating genes involved in the synthesis of nucleotides and proteins, allowing tumor cells to replicate in an uncontrolled manner, or promoting the consumption of glucose, an essential element for tumor growth [16]. Furthermore, its hyperactivation makes cells insensitive to antiproliferative drugs, also promoting the production of pro-angiogenic factors, such as vascular endothelial growth (VEGF), which support the development of new blood vessels [17]. These properties make MYC a central element in tumor progression, contributing not only to the primary growth of the tumor but also to the promotion of metastases and therapy resistance. The importance of MYC in tumorigenesis makes it a target of great interest for oncology research, but being a transcription factor and not having enzymatic pockets accessible to conventional drugs, its therapeutic targeting is extremely complex. However, there are innovative approaches, such as epigenetic therapies, that are opening new avenues for MYC targeting. In fact, direct inhibition of MYC could stop tumor growth and overcome resistance to existing treatments.

Figure 1.

Cellular processes regulated by MYC. MYC regulates several biological processes and their upregulation, in conditions of stress, or genetic alterations can cause the deregulation of genes involved in several pathologies.

Advertisement

2. MYC structure and function

MYC, a proto-oncogene encoding a protein crucial in gene regulation, is considered one of the most important regulators of cell biology. It is a transcription factor that, by binding to specific DNA sequences called E-box: 5'-CACGTG-3′, activates or represses a lot of genes involved in different cellular processes [18]. MYC was originally identified in avian oncogenic viruses as a viral oncogene and named v-MYC. Its corresponding c-MYC was later recognized as essential for cell growth and proliferation in both normal and pathological conditions [19]. The MYC family consists of c-MYC, which plays a fundamental role in the regulation of the cell cycle and metabolism. It is the most studied and significantly expressed member of the family [20]; N-MYC, frequently amplified in neuroblastic tumors. It is the member mainly expressed in nervous tissues during embryonic development; L-MYC, involved in the regulation of cell proliferation in small cell lung cancer [21]. It is the member of the family whose functions are less understood than the others. The MYC genes, found in complex organisms, play a crucial role as a transcriptional regulator necessary for fundamental biological processes [22]. The three members of the MYC family exhibit significant similarities in regions that serve as docking points for various cofactors. These cofactors influence both the activity and stability of MYC, contributing to its oncogenic potential [23]. The conserved pattern known as the b-HLH-LZ motif, which is found in the C-terminal region of these proteins, is made up of a basic region (b), a helix-loop-helix (HLH), and a leucine zipper (LZ) domain. As seen in Figure 2, the b-region makes it easier to bind specific DNA sequences, whereas the HLH and LZ domains allow dimerization [24]. These motifs are crucial to create DNA-binding domain trough by MYC/MAX heterodimers [25, 26, 27]. Six domains known as MYC homology boxes (MBs) make up the N-terminal region of MYC proteins. The transactivation domain (TAD) contains the residues MB0 (residues 16–33), MBI (residues 45–65), and MBII (residues 128–144) (Figure 2). These domains are essential for preserving protein stability, allowing interactions between proteins, and regulating the transcriptional activation or repression of target genes [25, 28, 29]. MB0 interacts with transcription elongation factors and contributes to tumor progression, while MBI plays a role in regulating MYC degradation via the ubiquitin-proteasome pathway [29, 30]. MBII, the better characterized region within TAD, is essential for key MYC properties, including specific DNA binding, autoregulation, and transcriptional activity [28]. Transcription of MYC-bound genes can be activated through the recruitment of histone acetyltransferase (HAT) complexes by MBII binding to transformation/transcription domain-associated protein (TRRAP) [29, 31, 32]. In the central region of MYC, there are three additional conserved domains: MBIIIa, MBIIIb, and MBIV (Figure 2). MBIIIa interacts with histone deacetylases (HDACs) and is involved in gene repression mediated by MYC, while MBIV contributes to DNA binding, though its precise mechanism remains unclear [33]. MBIIIb interacts with WD repeat-containing protein 5 (WDR5), a protein having WD-40 repeats, in order to connect MYC to chromatin and control the expression of genes involved in protein synthesis [34, 35]. Mutational analyses have revealed that mutations in MBI and MBII significantly impair MYC amplification, whereas mutations in MBIII enhance it. These data indicate that each MB domain interacts with distinct protein partners, affecting chromatin remodeling and transcription throughout the transcriptional cycle [36].

Figure 2.

Domain structure of MYC and MAX proteins. MYC and MAX domains. The N-terminal region of MYC engages with several interacting partners, while the C-terminal region associates with MAX through the b-HLH-LZ motif. The resulting MYC/MAX heterodimer recognizes and binds E-box sequences within the DNA of target genes.

The structure of MYC is fundamental for its function as a transcriptional regulator and for the precise control of numerous cellular processes. In fact, MYC promotes the entry of cells into the S phase, regulating the cell cycle and activating genes, such as cyclin D, cyclin E, and cyclin-dependent kinase-4 (CDK4), essential for cell cycle progression, and simultaneously repressing cell cycle inhibitors, such as p21 and p27, ensuring continued proliferation [37, 38]. Furthermore, MYC modulates cellular metabolism to support the high energetic and biosynthetic demands of proliferation. It activates glycolysis enzymes, glutamine metabolism, and lipid and nucleotide biosynthesis, adapting the metabolism of tumor cells to the so-called Warburg effect [39]. MYC, under normal conditions, maintains cellular homeostasis, coordinating cell survival through the activation of proliferative pathways and promoting apoptosis after significant stresses, such as irreparable DNA damage [8]. Indeed, it can induce the activation of pro-apoptotic genes such as BAX and PUMA, while repressing anti-apoptotic genes such as BCL-2. However, in tumors, its regulation is subverted [5, 8, 40]. MYC overexpression, and in particular its hyperactivity, alters the correct balance between proliferation and apoptosis by enhancing growth signals and reducing sensitivity to programmed cell death signals [41, 42]. This “oncogenic addiction” makes MYC a crucial therapeutic target for the development of selective drugs.

Advertisement

3. MYC regulation between cell death and survival

Cell death is the irreversible arrest of cellular activity and an important physiological process in all organisms. It plays a crucial role in embryonic development, organ maintenance, and immune regulation [43]. In recent years, a greater understanding of the mechanism of programmed cell death or apoptosis has been gained and some key genes in this process have been identified such as MYC [44], which has a crucial role in the regulation of cell death through its ability to influence various pro-apoptotic pathways (BAX, PUMA, and NOXA (phorbol-12-myristate-13-acetate-induced protein 1) and anti-apoptotic pathways (BCL-2) [8, 45]. MYC regulates apoptosis through intrinsic and extrinsic death regulators: in particular, it stimulates the transcription of the intrinsic regulator BAX, which accumulates on the outer mitochondrial membrane facilitating its permeabilization [8, 46, 47]. This event triggers the movement of cytochrome c into the cytoplasm, promoting the formation of the apoptosome and subsequently the activation of caspases, triggering a cascade of events that culminate in DNA fragmentation and cell death [48, 49, 50]. MYC also has an involvement in the expression of two intrinsic targets: PUMA and NOXA, which neutralize anti-apoptotic proteins such as BCL-2 and B-cell lymphoma-extra large (BCL-XL), amplifying the apoptotic effect [51]. This function is enhanced by the ability of MYC to cooperate with p53, stabilizing and enhancing its transcriptional activity [52]. However, this balance can be disrupted in tumors, in which p53 is mutated or inactive, reducing the effectiveness of MYC-mediated apoptosis and promoting the survival of malignant cells [53, 54]. MYC also regulates the suppression of BCL-2, which plays a key role in protecting mitochondria from permeabilization. MYC-mediated suppression of BCL-2 controls the balance between pro- and anti-apoptotic factors, inducing programmed cell death in the absence of survival signals [55, 56]. In tumors, this balance is dysregulated, with an increase in the persistence of tumor cells and a reduction in their responsiveness to apoptotic stimuli [57]. Despite the complex dual role of MYC in cell growth and cell survival, in response to tumor microenvironment stimuli, the activation or inactivation of certain genes involved in these processes, aberrant MYC expression often exerts a dominant effect by suppressing apoptosis allowing cells to survive and proliferate in an uncontrolled manner (Figure 3) [58, 59, 60]. The paradoxical aspect of MYC biology is fully manifested in the concept of oncogene addiction, where tumor cells exploit its hyperactivity to sustain uncontrolled growth. Recent studies have reported the impact of MYC on several survival pathways, including the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway [61]. MYC, by positively regulating AKT signaling, indirectly promotes cell survival and inhibits apoptotic processes. In addition, AKT activation leads to the phosphorylation and inhibition of pro-apoptotic proteins, such as BCL-2-associated agonist of cell death (BAD), preventing cell death [62]. It has been reported that MYC enhances the anti-apoptotic gene expression, such as BCL-2 and BCL-XL. These proteins block the mitochondrial outer membrane permeabilization (MOMP), a critical step in the intrinsic apoptotic pathway [63]. By inhibiting MOMP, MYC effectively prevents the release of cytochrome c and the activation of caspases, which are key components of apoptosis [64]. MYC can suppress the pro-apoptotic genes’ transcription, such as BAX, thus preventing apoptosis and promoting cell proliferation, even under stress conditions [65]. Dysregulation of MYC can affect the appropriate function of p53, which has an essential function in the apoptotic process. MYC-driven cells often find mechanisms to inactivate or bypass p53 signaling [66, 67]. For example, MYC can increase the levels of murine double minute (MDM2), a negative regulator of p53, leading to a decrease in p53-mediated apoptosis. This is essential to allow MYC-overexpressing cells to proliferate, despite DNA damage or other stressors that would normally activate p53 [68]. Dysregulation of MYC then leads to rapid cellular proliferation, which can overwhelm normal DNA repair mechanisms and increase genomic instability, leading to the accumulation of mutations that can further inhibit apoptosis pathways, strengthening the pro-survival role of MYC [69].

Figure 3.

Differences between apoptotic signaling pathways in normal and malignant cells. In the first representation (left), MYC is shown in normal cells and in the presence of a normal level of growth factor that led to normal cell proliferation without altering p53; in the center of the figure, when myc is associated with a limited level of growth factor, there is an alteration of the p53 level that causes apoptosis; in the last part of the image (right), there is the involvement of MYC in malignant cells, associated with the activation of some genes such as BCL-2 and nuclear factor kappa B (NF-κB) and the absence of growth factor, p53 is inactivated and the final result is cellular transformation.

Advertisement

4. Role of MYC in regulating apoptosis

MYC plays a crucial role in the control of multiple cellular processes, including proliferation, metabolism, and programmed cell death [70]. Although, MYC is primarily known for its ability to promote cell growth and proliferation, it also acts as a potent sensitizer of apoptotic signals [71]. This dual role of MYC is particularly relevant in the context of cancer, where the balance between proliferation and apoptosis determines the fate of transformed cells [72]. MYC is able to regulate both the intrinsic (mitochondrial) and extrinsic (death receptor-mediated) pathways of apoptosis [73]. The intrinsic pathway is characterized by the permeabilization of the mitochondrial membrane and the release of cytochrome c into the cytoplasm, an event that activates a caspase cascade, culminating in the degradation of cellular components [74]. MYC stimulates the expression of pro-apoptotic genes, such as BAX, BID, and PUMA, which promote the opening of mitochondrial pores and the release of cytochrome c, activating caspase-9 and, subsequently, executioner caspases such as caspase-3 and caspase-7 [75]. However, in tumor cells, this pro-apoptotic activity of MYC is often counteracted by the upregulation of anti-apoptotic proteins, such as BCL-2 and BCL-XL, which stabilize the mitochondrial membrane and prevent the initiation of the apoptotic cascade [76, 77]. In tumor contexts, MYC hyperactivity is associated with increased replicative stress and the production of reactive oxygen species (ROS), which can cause DNA damage and activate apoptosis [78]. However, mutations in tumor suppressor genes, such as p53, allow tumor cells to survive, despite high levels of stress. Indeed, p53 mutations, frequently observed in tumors, impair the ability of cells to respond to cellular damage by apoptosis [79, 80]. MYC modulates the balance between pro- and anti-apoptotic members of the BCL-2 family, both in a p53-dependent and -independent manner. Studies in Eμ-MYC transgenic mouse models of lymphoma have shown that MYC can regulate the expression of proteins such as BCL-2 and BCL-XL. In more than half of murine lymphomas, the levels of these anti-apoptotic proteins are elevated, even in the absence of functional p53, suggesting a p53-independent mechanism [76]. At the same time, MYC is able to indirectly suppress BCL-XL expression by reducing its RNA and protein levels. Such repression requires a new protein synthesis, suggesting an indirect transcriptional regulatory mechanism [77]. In parallel, MYC stimulates the expression of pro-apoptotic members such as BAX and BAK, which are essential for the activation of the mitochondrial apoptotic pathway. Indeed, MYC overexpression is associated with an increase in BAX expression at the transcriptional level, enhancing its functional activation [81]. The extrinsic apoptosis pathway is activated by the interaction between death receptors, such as FAS and TRAIL receptor (TRAIL-R, tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor), and their respective ligands. MYC is known to sensitize cells to these apoptotic signals by stimulating the expression of death receptors [82]. Activation of receptors such as FAS leads to the formation of the Death-Inducing Signaling Complex (DISC) and activation of caspase-8, which can act directly on executioner caspases or interact with the intrinsic pathway through the cleavage of BH3 interacting-domain death agonist (BID), an activator of mitochondrial permeabilization [83]. However, in tumors, the extrinsic apoptosis pathway is often deactivated by mechanisms that reduce the expression of death receptors or increase the levels of inhibitors, such as cellular FLICE-like inhibitory protein (c-FLIP), which block caspase-8 activation [84]. Furthermore, MYC counteracts survival signals associated with the death receptor pathway, such as activation of NF-κB, a known pro-survival factor [85]. Understanding the role of MYC in regulating apoptosis has important implications for the development of anticancer therapies. Drugs targeting anti-apoptotic proteins, such as venetoclax, a BCL-2 inhibitor, can reactivate the mitochondrial apoptotic cascade in tumor cells with overactive MYC, especially in hematological malignancies [86]. Similarly, restoring p53 function by compounds such as nutlin-3 could potentiate MYC-induced apoptosis [87]. Another promising strategy is the activation of the extrinsic apoptosis pathway by TRAIL ligands or agonist antibodies against death receptors [88]. These approaches exploit the predisposition of tumor cells with overactive MYC to succumb to apoptotic signals, offering therapeutic opportunities to treat aggressive tumors that are resistant to conventional therapies.

Advertisement

5. MYC in tumor progression

Cancer is a multifaceted process that needs the occurrence of multiple genetic alterations and the acquisition of distinct biological characteristics [89]. Notably, MYC activation is involved in multiple genetic deregulation and in the development of specific biological characteristics, such as proliferation, cell survival, metabolic alterations, angiogenesis, metastasis, and relapse [90]. Early in vitro investigations of MYC demonstrated its transformative potential when combined with other oncogenes in embryonic fibroblasts [91]. These findings demonstrated the ability of MYC to independently induce tumors in multiple tissues, while revealing its dependence on additional genetic alterations for complete tumorigenic activity in vivo [92]. For example, breast cancer induced by the transgenic expression of MYC developed K-Ras mutations, resulting in more aggressive tumors [93]. Similarly, induction of MYC in normal cells activates checkpoints such as Arf or p53, resulting in cell cycle arrest or apoptosis. In MYC-induced transgenic lymphomas, however, the proper functioning of Arf and p53 is subverted [94]. MYC controls the expression of different genes that influence cell growth, survival, and metabolism and directly impact tumor biology and the tumor microenvironment [95]. The role of MYC extends to modulating immune responses and other extrinsic factors that shape cancer progression [25]. Initially, MYC members were thought to play a universal role in human cancers, both hematological and solid, with L-MYC thought to be linked only to small cell lung cancer and N-MYC to neuroblastoma [96]. However, N-MYC and L-MYC seem to be involved in the occurrence of other cancers [97]. Thus, when examining the entire MYC family, it is clear that genetic activation of at least one of its members is a recurrent event in the majority of human cancers [5]. Dysregulation of MYC and related pathways is among the most widespread in human cancers. MYC hyperactivation in cancer occurs through epigenetic, genomic, and post-translational mechanisms, including amplifications, translocations, and upstream regulatory changes of its gene [98]. A pan-cancer analysis of 33 different human cancers, as reported in The Cancer Genome Atlas dataset, found that in 28% of tumors all members of the MYC family are amplified (Figure 4) [99]. These amplifications can cause MYC overexpression, either directly or indirectly, by activating genes involved in the MYC pathway [100]. MYC undergoes genetic amplification in several solid tumors, such as breast and liver cancers, and is often implicated in chromosomal translocations observed in B-cell and T-cell leukemias and lymphomas [101]. Retroviruses can enhance MYC expression through various mechanisms, including the insertion of enhancers upstream of the MYC gene and the activation of several oncogenic pathways such as SRC and Notch or loss of tumor suppressors such as adenomatous polyposis coli (APC) and transforming growth factor-beta (TGF-β) [102]. A lot of post-translational modifications determine MYC protein stability [103]. In normal conditions, a fine balance of phosphorylation events and proteasomal degradation determines the short half-life of MYC [104]. In tumors characterized by MYC overexpression, there is a notable increase in phospho-serine 62 (P-S62-MYC) levels and a corresponding decrease in phospho-threonine 58 (P-T58-MYC) levels. These alterations contribute to enhanced MYC stability and activity [105]. Rat sarcoma virus (RAS)-mitogen-activated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) signaling mitogenic pathway increases the levels of P-S62, destabilizing MYC. MYC T58-related mutations can cause constitutive phosphorylation of S62. Tumors can also decrease the expression of the serine/threonine protein phosphatase 2A (PP2A), that dephosphorylating P-S62 leads to an accumulated amount of MYC [106]. In addition, peptidyl-prolyl cis-trans isomerase, NIMA-interacting 1 (PIN1), a peptidyl-prolyl cis-trans isomerase (PPIase), can contribute to MYC regulation in several tumors, such as uterine cancer (18%), colon cancer (16%), and cervical cancer (13%), through deletion, mutation, or epigenetic changes. These events can enhance tumor growth by increasing MYC levels [107]. Although MYC plays a central role in tumorigenesis, its activation alone is typically insufficient to convert normal cells into malignant ones [108]. Apoptosis, senescence, or cell cycle arrest are physiological processes that frequently impair MYC carcinogenic function. MYC dysregulation can induce p53 activation [109], cyclin-dependent kinase inhibitor 2A (CDKN2A) upregulation [110], or BCL-2 modulation [111, 112]. An additionally protective mechanism against tumorigenesis is the shortening of telomeres during cell division cycles, which limits proliferation [113]. However, MYC can promote cellular immortality by stimulating telomerase activity, through the regulation of telomerase reverse transcriptase [114]. These multiple levels of MYC regulation and dysfunction underscore its critical role in driving cancer progression.

Figure 4.

Body map showing MYC expression in TPM (transcripts per million) different in tumor (left) and normal (right) human system.

Advertisement

6. MYC, a therapeutic target

Targeting MYC has demonstrated the ability to induce tumor regression through both direct and indirect effects, but it remains a therapeutic challenge due to MYC’s disordered structure and lack of a specific binding site or defined enzymatic activity [115]. Decades of research have highlighted several strategies to inhibit MYC activity that are emerging in preclinical models and clinical research [116]. Direct strategies include silencing MYC gene expression, disrupting MYC protein synthesis, or promoting its degradation via the proteasome (Figure 5) [117]. The MYC/MAX interaction is critical for the oncogenic function of MYC, and many approaches target this interaction [118]. For example, small compounds, such as MYCi361 and MYCi975, disrupt the MYC-MAX interface and promote phosphorylation of MYC on threonine-58, promoting proteasomal degradation of MYC [119]. Another study reported that KI-MS2-008 stabilizes MAX-MAX homodimers, reducing the levels of MYC and its transcriptional targets and suppressing tumor growth [120]. ME47, a minimalist small hybrid protein (MHP), disrupts the interaction with MAX by suppressing the transcriptional activity of MYC. These results suggest that MHPs offer an alternative therapeutic targeting method for transcription factors involved in human diseases, including cancer [121]. EN4, on the other hand, binds to an intrinsically disordered region of MYC (cysteine 171), reduces the thermal stability of MYC and MAX, inhibits MYC transcriptional activity, and suppresses carcinogenesis [122]. Another promising approach is to decrease MYC biosynthesis or enhance its degradation. Inhibitors targeting the PI3K-AKT-mTOR (mammalian target of rapamycin) signaling pathway have been shown to reduce MYC translation and lower MYC levels in tumors in mouse models [123]. Additionally, Silvestrol, an inhibitor of the translation initiator eukaryotic initiation factor-4A (eIF4A), suppresses MYC translation and prevents tumor growth in colon cancer mouse models [124]. Furthermore, Aurora kinase inhibitors can promote degradation of MYC protein and specifically reduce its overexpression in tumor cells without altering physiological expression of MYC [125]. Recently, substantial advancements have been achieved in developing specific protein degraders or chimeras, designed to directly induce the proteolysis of MYC (proteolysis targeting chimeras (PROTACs)) [126]. These approaches utilize the ubiquitin-proteasome system to degrade MYC, using bifunctional molecules with two components: a ligand that specifically binds to MYC and a ligand that targets an E3 ubiquitin ligase, such as cereblon or von Hippel-Lindau (VHL) [127]. Another potential strategy to reduce MYC levels is to target the stability of its messenger RNA (mRNA). Antisense oligonucleotides (ASOs), such as MYC-ASO, have been shown to reduce tumor development and stimulate antitumor immune responses in mouse models [128]. Another approach is to suppress MYC transcription. Bromodomain and extra-terminal motif (BET) inhibitors, epigenetic regulators, including IZCZ-3, that stabilize the G-quadruplex structure in the MYC promoter region, have shown antitumor activity [129]. Indirect targeting strategies exploit synthetic lethal interactions, targeting vulnerabilities in MYC-driven cells (Figure 5). For example, inhibiting MYC transcriptional activity through genetic approaches, such as the use of OmoMYC, a dominant negative allele that competes with MYC/MAX for chromatin binding, reducing MYC/MAX occupancy at its targets and modulating gene expression in tumors with high MYC oncogenic activity [130]. Epigenetic modifiers, such as histone deacetylase inhibitor (HDACi) and histone methyltransferase (Enhancer of Zeste 2 Polycomb Repressive Complex 2 Subunit (EZH2)) inhibitors, have been used to treat MYC-induced malignancies. The combination of HDACi and EZH2 reduced MYC levels and activated immune transcriptional pathways, enhancing T-cell-mediated antitumor responses through C-C motif chemokine ligand 5 (CCL5) expression [131, 132]. Recent studies have shown that the use of epigenetic treatments can reduce MYC, reversing immune evasion. Similarly, MYC can cause cancer by regulating chromatin remodeling via miR17-92. Anti-miR-17 oligonucleotides inhibited tumor progression in a liver cancer mouse model [133]. Research continues to develop innovative approaches to target MYC, exploiting both its molecular dependencies and interactions with the immune system. These advances offer new perspectives for the treatment of MYC-induced malignancies.

Figure 5.

Effects of several inhibitors on MYC levels. Direct effect (left) through some inhibitors such as IZCZ3 and EN4 that led to antitumor activity and suppression of carcinogenesis; on the right of the figure is shown the indirect effect of some epigenetic inhibitors and several others causing a “positive” modulation of MYC in tumor.

Advertisement

7. Emerging strategies for targeting MYC in cancer therapy

Targeting MYC in cancer therapy is a promising but challenging frontier due to its critical role in cellular processes and its classification as a “difficult-to-drug” molecule [134]. Future directions to overcome these challenges and exploit MYC as a therapeutic target include several innovative approaches [135]. Studies are underway to design small molecules that disrupt MYC-MAX dimerization, which is essential for MYC transcriptional activity [136]. Additionally, PROTACs are being explored to selectively degrade MYC protein by exploiting the ubiquitin-proteasome system [137]. Clinical trials of OmoMYV a MYC inhibitory peptide, are expanding, along with investigations of its derivatives and other direct MYC inhibitors [138]. Tumors that overexpress MYC have specific vulnerabilities, such as dependence on cyclin-dependent kinases (CDKs), DNA repair enzymes (checkpoint kinase 1 (CHK1), ataxia-telangiectasia-mutated-and-Rad3-related kinase (ATR)), and anti-apoptotic proteins such as myeloid cell leukemia 1 (MCL1). Combining MYC inhibition with drugs that target these synthetic lethal pathways promises to improve therapeutic efficacy and mitigate resistance [5, 139]. HDACi are being explored to regulate MYC-driven transcriptional programs [140, 141]. Similarly, BET inhibitors, which block MYC transcription by interfering with chromatin regulators, offer another promising avenue for treatment [142]. Techniques such as antisense RNA are being developed to directly silence overexpressed MYC mRNA [143]. In addition, MYC inhibition is being combined with immune checkpoint inhibitors to exploit MYC’s role in immune evasion [144]. These multifaceted approaches, individually and in combination, aim to address the complexity of MYC targeting and offer hope for more effective and personalized cancer therapies in the future.

Advertisement

8. Conclusions

MYC plays a pivotal and multifaceted role in tumorigenesis and cancer progression, regulating numerous cellular processes, such as the cell cycle, metabolism, and transcription [145]. It functions as a key determinant of the balance between cell survival and cell death through its transcriptional regulation of a wide range of genes [146]. This dual role makes MYC a critical factor in carcinogenesis. When functioning correctly, MYC can drive cells toward apoptosis, acting as a safeguard against uncontrolled proliferation [147]. However, dysregulation of MYC, often resulting from mutations or concurrent alterations in other oncogenes or tumor suppressor genes, can circumvent apoptosis and promote unchecked cell growth [148].

Despite its critical involvement in cancer, targeting MYC directly poses significant challenges. Its lack of enzymatic pockets and its pervasive overexpression in tumors make it a difficult therapeutic target [108]. Nevertheless, substantial progress has been made in developing strategies to inhibit MYC activity. However, clinical treatments that effectively target MYC remain elusive [149]. An understanding of the mechanisms by which MYC governs tumorigenesis versus apoptosis is essential [103]. Such insights could refine therapeutic approaches, enabling the precise targeting of MYC in cancer cells while minimizing adverse effects.

Advertisement

Acknowledgments

This study was supported by PRIN2022, PNRR P2022CKMPW project, PRIN2020 2020CW39SJ project, and PNRR-MAD-2022-12376672.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Acronyms and abbreviations

ASO

antisense oligonucleotides

bHLH-LZ

basic helix-loop-helix leucine zipper

CDK

cyclin-dependent kinases

DISC

death-inducing signaling complex

EZH2

Enhancer of zeste 2 polycomb repressive complex 2 subunit

HAT

histone acetyltransferase

HDACs

histone deacetylases

HDACi

histone deacetylase inhibitor

MBs

MYC homology boxes

MHP

minimalist small hybrid protein

MOMP

mitochondrial outer membrane permeabilization

PPIase

peptidyl-prolyl cis-trans isomerase

PROTAC

proteolysis targeting chimera

P-S62-MYC

phospho-serine 62

P-T58-MYC

phospho-threonine 58

ROS

reactive oxygen species

TRRAP

transformation/transcription domain-associated protein

VEGF

vascular endothelial growth factor

References

  1. 1. Jha RK, Kouzine F, Levens D. MYC function and regulation in physiological perspective. Frontiers in Cell and Development Biology. 2023;11:1268275
  2. 2. Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. Correction to: MYC: A multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. Journal of Hematology and Oncology. 2021;14(1):135
  3. 3. Stine ZE, Walton ZE, Altman BJ, Hsieh AL, Dang CV. MYC, metabolism, and cancer. Cancer Discovery. 2015;5(10):1024-1039
  4. 4. Scafuro M, Capasso L, Carafa V, Altucci L, Nebbioso A. Gene transactivation and transrepression in MYC-driven cancers. International Journal of Molecular Sciences. 2021;22(7):3458
  5. 5. Dhanasekaran R, Deutzmann A, Mahauad-Fernandez WD, Hansen AS, Gouw AM, Felsher DW. The MYC oncogene - The grand orchestrator of cancer growth and immune evasion. Nature Reviews. Clinical Oncology. 2022;19(1):23-36
  6. 6. Spencer CA, Groudine M. Control of c-myc regulation in normal and neoplastic cells. Advances in Cancer Research. 1991;56:1-48
  7. 7. Lourenco C, Resetca D, Redel C, Lin P, MacDonald AS, Ciaccio R, et al. MYC protein interactors in gene transcription and cancer. Nature Reviews. Cancer. 2021;21(9):579-591
  8. 8. McMahon SB. MYC and the control of apoptosis. Cold Spring Harbor Perspectives in Medicine. 2014;4(7):a014407
  9. 9. Conacci-Sorrell M, Ngouenet C, Anderson S, Brabletz T, Eisenman RN. Stress-induced cleavage of Myc promotes cancer cell survival. Genes and Development. 2014;28(7):689-707
  10. 10. Edwards-Hicks J, Su H, Mangolini M, Yoneten KK, Wills J, Rodriguez-Blanco G, et al. MYC sensitises cells to apoptosis by driving energetic demand. Nature Communications. 2022;13(1):4674
  11. 11. Chen J. The cell-cycle arrest and apoptotic functions of p53 in tumor initiation and progression. Cold Spring Harbor Perspectives in Medicine. 2016;6(3):a026104
  12. 12. Fairlie WD, Lee EF. Co-operativity between MYC and BCL-2 pro-survival proteins in cancer. International Journal of Molecular Sciences. 2021;22(6):2841
  13. 13. Sharma A, Boise LH, Shanmugam M. Cancer metabolism and the evasion of apoptotic cell death. Cancers (Basel). 2019;11(8):1144
  14. 14. Pflaum J, Schlosser S, Müller M. p53 family and cellular stress responses in cancer. Frontiers in Oncology. 2014;4:285
  15. 15. Kumari A, Folk WP, Sakamuro D. The dual roles of MYC in genomic instability and cancer chemoresistance. Genes (Basel). 2017;8(6):158
  16. 16. Wahlström T, Henriksson MA. Impact of MYC in regulation of tumor cell metabolism. Biochimica et Biophysica Acta. 2015;1849(5):563-569
  17. 17. Baudino TA, McKay C, Pendeville-Samain H, Nilsson JA, Maclean KH, White EL, et al. c-Myc is essential for vasculogenesis and angiogenesis during development and tumor progression. Genes and Development. 2002;16(19):2530-2543
  18. 18. Esain-Garcia I, Kirchner A, Melidis L, Tavares RCA, Dhir S, Simeone A, et al. G-quadruplex DNA structure is a positive regulator of MYC transcription. Proceedings of the National Academy of Sciences of the United States of America. 2024;121(7):e2320240121
  19. 19. Wang C, Zhang J, Yin J, Gan Y, Xu S, Gu Y, et al. Alternative approaches to target Myc for cancer treatment. Signal Transduction and Targeted Therapy. 2021;6(1):117
  20. 20. Gnanaprakasam JN, Wang R. MYC in regulating immunity: Metabolism and beyond. Genes (Basel). 2017;8(3):88
  21. 21. Ruiz-Pérez MV, Henley AB, Arsenian-Henriksson M. The MYCN protein in health and disease. Genes (Basel). 2017;8(4):113
  22. 22. Yap CS, Peterson AL, Castellani G, Sedivy JM, Neretti N. Kinetic profiling of the c-Myc transcriptome and bioinformatic analysis of repressed gene promoters. Cell Cycle. 2011;10(13):2184-2196
  23. 23. Carabet LA, Rennie PS, Cherkasov A. Therapeutic inhibition of Myc in cancer. Structural bases and computer-aided drug discovery approaches. International Journal of Molecular Sciences. 2018;20(1):120
  24. 24. Chen H, Liu H, Qing G. Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduction and Targeted Therapy. 2018;3:5
  25. 25. Conacci-Sorrell M, McFerrin L, Eisenman RN. An overview of MYC and its interactome. Cold Spring Harbor Perspectives in Medicine. 2014;4(1):a014357
  26. 26. Adhikary S, Eilers M. Transcriptional regulation and transformation by Myc proteins. Nature Reviews. Molecular Cell Biology. 2005;6(8):635-645
  27. 27. Allevato M, Bolotin E, Grossman M, Mane-Padros D, Sladek FM, Martinez E. Sequence-specific DNA binding by MYC/MAX to low-affinity non-E-box motifs. PLoS One. 2017;12(7):e0180147
  28. 28. Flinn EM, Busch CM, Wright AP. Myc boxes, which are conserved in myc family proteins, are signals for protein degradation via the proteasome. Molecular and Cellular Biology. 1998;18(10):5961-5969
  29. 29. Kalkat M, Resetca D, Lourenco C, Chan PK, Wei Y, Shiah YJ, et al. MYC protein Interactome profiling reveals functionally distinct regions that cooperate to drive tumorigenesis. Molecular Cell. 2018;72(5):836-48.e7
  30. 30. Nair SK, Burley SK. X-ray structures of Myc-max and mad-max recognizing DNA. Molecular bases of regulation by proto-oncogenic transcription factors. Cell. 2003;112(2):193-205
  31. 31. McMahon SB, Van Buskirk HA, Dugan KA, Copeland TD, Cole MD. The novel ATM-related protein TRRAP is an essential cofactor for the c-Myc and E2F oncoproteins. Cell. 1998;94(3):363-374
  32. 32. Brough DE, Hofmann TJ, Ellwood KB, Townley RA, Cole MD. An essential domain of the c-myc protein interacts with a nuclear factor that is also required for E1A-mediated transformation. Molecular and Cellular Biology. 1995;15(3):1536-1544
  33. 33. Cowling VH, Chandriani S, Whitfield ML, Cole MD. A conserved Myc protein domain, MBIV, regulates DNA binding, apoptosis, transformation, and G2 arrest. Molecular and Cellular Biology. 2006;26(11):4226-4239
  34. 34. Thomas LR, Wang Q, Grieb BC, Phan J, Foshage AM, Sun Q, et al. Interaction with WDR5 promotes target gene recognition and tumorigenesis by MYC. Molecular Cell. 2015;58(3):440-452
  35. 35. Thomas LR, Adams CM, Wang J, Weissmiller AM, Creighton J, Lorey SL, et al. Interaction of the oncoprotein transcription factor MYC with its chromatin cofactor WDR5 is essential for tumor maintenance. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(50):25260-25268
  36. 36. Nie Z, Guo C, Das SK, Chow CC, Batchelor E, Simons SSJ, et al. Dissecting transcriptional amplification by MYC. eLife. 2020;9:e52483
  37. 37. Bretones G, Delgado MD, León J. Myc and cell cycle control. Biochimica et Biophysica Acta. 2015;1849(5):506-516
  38. 38. García-Gutiérrez L, Delgado MD, León J. MYC oncogene contributions to release of cell cycle brakes. Genes (Basel). 2019;10(3):244
  39. 39. Jing Z, Liu Q, He X, Jia Z, Xu Z, Yang B, et al. NCAPD3 enhances Warburg effect through c-myc and E2F1 and promotes the occurrence and progression of colorectal cancer. Journal of Experimental and Clinical Cancer Research. 2022;41(1):198
  40. 40. Kirkin V, Joos S, Zörnig M. The role of Bcl-2 family members in tumorigenesis. Biochimica et Biophysica Acta. 2004;1644(2-3):229-249
  41. 41. Campone M, Noël B, Couriaud C, Grau M, Guillemin Y, Gautier F, et al. c-Myc dependent expression of pro-apoptotic Bim renders HER2-overexpressing breast cancer cells dependent on anti-apoptotic Mcl-1. Molecular Cancer. 2011;10:110
  42. 42. Pelengaris S, Rudolph B, Littlewood T. Action of Myc in vivo - Proliferation and apoptosis. Current Opinion in Genetics and Development. 2000;10(1):100-105
  43. 43. Shen S, Shao Y, Li C. Different types of cell death and their shift in shaping disease. Cell Death Discovery. 2023;9(1):284
  44. 44. Wang C, Tai Y, Lisanti MP, Liao DJ. c-Myc induction of programmed cell death may contribute to carcinogenesis: A perspective inspired by several concepts of chemical carcinogenesis. Cancer Biology and Therapy. 2011;11(7):615-626
  45. 45. Juin P, Hunt A, Littlewood T, Griffiths B, Swigart LB, Korsmeyer S, et al. c-Myc functionally cooperates with Bax to induce apoptosis. Molecular and Cellular Biology. 2002;22(17):6158-6169
  46. 46. Cao X, Bennett RL, May WS. c-Myc and caspase-2 are involved in activating Bax during cytotoxic drug-induced apoptosis. The Journal of Biological Chemistry. 2008;283(21):14490-14496
  47. 47. Jan R, Chaudhry GE. Understanding apoptosis and apoptotic pathways targeted cancer therapeutics. Advanced Pharmaceutical Bulletin. 2019;9(2):205-218
  48. 48. Wang C, Youle RJ. The role of mitochondria in apoptosis*. Annual Review of Genetics. 2009;43:95-118
  49. 49. Prokhorova EA, Zamaraev AV, Kopeina GS, Zhivotovsky B, Lavrik IN. Role of the nucleus in apoptosis: Signaling and execution. Cellular and Molecular Life Sciences. 2015;72(23):4593-4612
  50. 50. Jin CY, Moon DO, Choi YH, Lee JD, Kim GY. Bcl-2 and caspase-3 are major regulators in Agaricus blazei-induced human leukemic U937 cell apoptosis through dephoshorylation of Akt. Biological and Pharmaceutical Bulletin. 2007;30(8):1432-1437
  51. 51. Singh R, Letai A, Sarosiek K. Regulation of apoptosis in health and disease: The balancing act of BCL-2 family proteins. Nature Reviews. Molecular Cell Biology. 2019;20(3):175-193
  52. 52. Boone DN, Qi Y, Li Z, Hann SR. Egr1 mediates p53-independent c-Myc-induced apoptosis via a noncanonical ARF-dependent transcriptional mechanism. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(2):632-637
  53. 53. Hientz K, Mohr A, Bhakta-Guha D, Efferth T. The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget. 2017;8(5):8921-8946
  54. 54. Wang H, Guo M, Wei H, Chen Y. Targeting p53 pathways: Mechanisms, structures, and advances in therapy. Signal Transduction and Targeted Therapy. 2023;8(1):92
  55. 55. Zinkel S, Gross A, Yang E. BCL2 family in DNA damage and cell cycle control. Cell Death and Differentiation. 2006;13(8):1351-1359
  56. 56. Shen Y, White E. p53-dependent apoptosis pathways. Advances in Cancer Research. 2001;82:55-84
  57. 57. Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 2007;26(9):1324-1337
  58. 58. Gabay M, Li Y, Felsher DW. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harbor Perspectives in Medicine. 2014;4(6):a014241
  59. 59. Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. MYC: A multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. Journal of Hematology and Oncology. 2021;14(1):121
  60. 60. Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, et al. Tumor initiation and early tumorigenesis: Molecular mechanisms and interventional targets. Signal Transduction and Targeted Therapy. 2024;9(1):149
  61. 61. Harrington CT, Sotillo E, Dang CV, Thomas-Tikhonenko A. Tilting MYC toward cancer cell death. Trends Cancer. 2021;7(11):982-994
  62. 62. Rascio F, Spadaccino F, Rocchetti MT, Castellano G, Stallone G, Netti GS, et al. The pathogenic role of PI3K/AKT pathway in cancer onset and drug resistance. An Updated Review. Cancers (Basel). 2021;13(16):3949
  63. 63. Qian S, Wei Z, Yang W, Huang J, Yang Y, Wang J. The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Frontiers in Oncology. 2022;12:985363
  64. 64. Boice A, Bouchier-Hayes L. Targeting apoptotic caspases in cancer. Biochimica Et Biophysica Acta-Molecular Cell Research. 2020;1867(6):118688
  65. 65. Soucie EL, Annis MG, Sedivy J, Filmus J, Leber B, Andrews DW, et al. Myc potentiates apoptosis by stimulating Bax activity at the mitochondria. Molecular and Cellular Biology. 2001;21(14):4725-4736
  66. 66. Feng YC, Liu XY, Teng L, Ji Q, Wu Y, Li JM, et al. c-Myc inactivation of p53 through the pan-cancer lncRNA MILIP drives cancer pathogenesis. Nature Communications. 2020;11(1):4980
  67. 67. Engeland K. Cell cycle regulation: p53-p21-RB signaling. Cell Death and Differentiation. 2022;29(5):946-960
  68. 68. Ho JS, Ma W, Mao DY, Benchimol S. p53-dependent transcriptional repression of c-myc is required for G1 cell cycle arrest. Molecular and Cellular Biology. 2005;25(17):7423-7431
  69. 69. Kuzyk A, Mai S. c-MYC-induced genomic instability. Cold Spring Harbor Perspectives in Medicine. 2014;4(4):a014373
  70. 70. Dong Y, Tu R, Liu H, Qing G. Regulation of cancer cell metabolism: Oncogenic MYC in the driver's seat. Signal Transduction and Targeted Therapy. 2020;5(1):124
  71. 71. Di Giacomo S, Sollazzo M, Paglia S, Grifoni D. MYC, Cell competition, and cell death in cancer: The inseparable triad. Genes (Basel). 2017;8(4):120
  72. 72. Chan KI, Zhang S, Li G, Xu Y, Cui L, Wang Y, et al. MYC oncogene: A druggable target for treating cancers with natural products. Aging and Disease. 2024;15(2):640-697
  73. 73. Koren E, Fuchs Y. Modes of regulated cell death in cancer. Cancer Discovery. 2021;11(2):245-265
  74. 74. Kashyap D, Garg VK, Goel N. Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. Advances in Protein Chemistry and Structural Biology. 2021;125:73-120
  75. 75. Stevens M, Oltean S. Modulation of the apoptosis gene Bcl-x function through alternative splicing. Frontiers in Genetics. 2019;10:804
  76. 76. Eischen CM, Woo D, Roussel MF, Cleveland JL. Apoptosis triggered by Myc-induced suppression of Bcl-X(L) or Bcl-2 is bypassed during lymphomagenesis. Molecular and Cellular Biology. 2001;21(15):5063-5070
  77. 77. Eischen CM, Packham G, Nip J, Fee BE, Hiebert SW, Zambetti GP, et al. Bcl-2 is an apoptotic target suppressed by both c-Myc and E2F-1. Oncogene. 2001;20(48):6983-6993
  78. 78. Vafa O, Wade M, Kern S, Beeche M, Pandita TK, Hampton GM, et al. c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: A mechanism for oncogene-induced genetic instability. Molecular Cell. 2002;9(5):1031-1044
  79. 79. Hoffman B, Liebermann DA. The proto-oncogene c-myc and apoptosis. Oncogene. 1998;17(25):3351-3357
  80. 80. Nilsson JA, Cleveland JL. Myc pathways provoking cell suicide and cancer. Oncogene. 2003;22(56):9007-9021
  81. 81. Annis MG, Soucie EL, Dlugosz PJ, Cruz-Aguado JA, Penn LZ, Leber B, et al. Bax forms multispanning monomers that oligomerize to permeabilize membranes during apoptosis. The EMBO Journal. 2005;24(12):2096-2103
  82. 82. Indran IR, Tufo G, Pervaiz S, Brenner C. Recent advances in apoptosis, mitochondria and drug resistance in cancer cells. Biochimica et Biophysica Acta. 2011;1807(6):735-745
  83. 83. Hueber AO, Zörnig M, Lyon D, Suda T, Nagata S, Evan GI. Requirement for the CD95 receptor-ligand pathway in c-Myc-induced apoptosis. Science. 1997;278(5341):1305-1309
  84. 84. Ricci MS, Jin Z, Dews M, Yu D, Thomas-Tikhonenko A, Dicker DT, et al. Direct repression of FLIP expression by c-myc is a major determinant of TRAIL sensitivity. Molecular and Cellular Biology. 2004;24(19):8541-8555
  85. 85. Klefstrom J, Arighi E, Littlewood T, Jäättelä M, Saksela E, Evan GI, et al. Induction of TNF-sensitive cellular phenotype by c-Myc involves p53 and impaired NF-kappaB activation. The EMBO Journal. 1997;16(24):7382-7392
  86. 86. Kang MH, Reynolds CP. Bcl-2 inhibitors: Targeting mitochondrial apoptotic pathways in cancer therapy. Clinical Cancer Research. 2009;15(4):1126-1132
  87. 87. Miyachi M, Kakazu N, Yagyu S, Katsumi Y, Tsubai-Shimizu S, Kikuchi K, et al. Restoration of p53 pathway by nutlin-3 induces cell cycle arrest and apoptosis in human rhabdomyosarcoma cells. Clinical Cancer Research. 2009;15(12):4077-4084
  88. 88. Pimentel JM, Zhou JY, Wu GS. The role of TRAIL in apoptosis and immunosurveillance in cancer. Cancers (Basel). 2023;15(10):2752
  89. 89. Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, et al. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduction and Targeted Therapy. 2024;9(1):270
  90. 90. Casacuberta-Serra S, Soucek L. Myc and ras, the bonnie and clyde of immune evasion. Translational Cancer Research. 2018;7(Suppl. 4):S457-S4s9
  91. 91. Melnik S, Werth N, Boeuf S, Hahn EM, Gotterbarm T, Anton M, et al. Impact of c-MYC expression on proliferation, differentiation, and risk of neoplastic transformation of human mesenchymal stromal cells. Stem Cell Research and Therapy. 2019;10(1):73
  92. 92. Chesi M, Robbiani DF, Sebag M, Chng WJ, Affer M, Tiedemann R, et al. AID-dependent activation of a MYC transgene induces multiple myeloma in a conditional mouse model of post-germinal center malignancies. Cancer Cell. 2008;13(2):167-180
  93. 93. Wang C, Lisanti MP, Liao DJ. Reviewing once more the c-myc and Ras collaboration: Converging at the cyclin D1-CDK4 complex and challenging basic concepts of cancer biology. Cell Cycle. 2011;10(1):57-67
  94. 94. D’Cruz CM, Gunther EJ, Boxer RB, Hartman JL, Sintasath L, Moody SE, et al. c-MYC induces mammary tumorigenesis by means of a preferred pathway involving spontaneous Kras2 mutations. Nature Medicine. 2001;7(2):235-239
  95. 95. Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nature Reviews. Cancer. 2008;8(12):976-990
  96. 96. Jinbo T, Iwamura Y, Kaneko M, Sawaguchi S. Coamplification of the L-myc and N-myc oncogenes in a neuroblastoma cell line. Japanese Journal of Cancer Research. 1989;80(4):299-301
  97. 97. Kalkat M, De Melo J, Hickman KA, Lourenco C, Redel C, Resetca D, et al. MYC deregulation in primary human cancers. Genes (Basel). 2017;8(6):151
  98. 98. Lancho O, Herranz D. The MYC enhancer-ome: Long-range transcriptional regulation of MYC in cancer. Trends Cancer. 2018;4(12):810-822
  99. 99. Turner KM, Deshpande V, Beyter D, Koga T, Rusert J, Lee C, et al. Extrachromosomal oncogene amplification drives tumour evolution and genetic heterogeneity. Nature. 2017;543(7643):122-125
  100. 100. Schaub FX, Dhankani V, Berger AC, Trivedi M, Richardson AB, Shaw R, et al. Pan-cancer alterations of the MYC oncogene and its proximal network across the cancer genome atlas. Cell Systems. 2018;6(3):282-300.e2
  101. 101. Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899-905
  102. 102. Dudley JP, Mertz JA, Rajan L, Lozano M, Broussard DR. What retroviruses teach us about the involvement of c-Myc in leukemias and lymphomas. Leukemia. 2002;16(6):1086-1098
  103. 103. Duffy MJ, O'Grady S, Tang M, Crown J. MYC as a target for cancer treatment. Cancer Treatment Reviews. 2021;94:102154
  104. 104. Chen GS, Chen SY, Liu ST, Hsieh CC, Lee SP, Huang SM. Stabilization of the c-Myc protein via the modulation of threonine 58 and serine 62 phosphorylation by the disulfiram/copper complex in oral cancer cells. International Journal of Molecular Sciences. 2022;23(16):9137
  105. 105. Sears R, Nuckolls F, Haura E, Taya Y, Tamai K, Nevins JR. Multiple ras-dependent phosphorylation pathways regulate Myc protein stability. Genes and Development. 2000;14(19):2501-2514
  106. 106. Farrell AS, Joly MM, Allen-Petersen BL, Worth PJ, Lanciault C, Sauer D, et al. MYC regulates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma associated with poor outcome and chemoresistance. Nature Communications. 2017;8(1):1728
  107. 107. King B, Trimarchi T, Reavie L, Xu L, Mullenders J, Ntziachristos P, et al. The ubiquitin ligase FBXW7 modulates leukemia-initiating cell activity by regulating MYC stability. Cell. 2013;153(7):1552-1566
  108. 108. Madden SK, de Araujo AD, Gerhardt M, Fairlie DP, Mason JM. Taking the Myc out of cancer: Toward therapeutic strategies to directly inhibit c-Myc. Molecular Cancer. 2021;20(1):3
  109. 109. Bailey ST, Smith AM, Kardos J, Wobker SE, Wilson HL, Krishnan B, et al. MYC activation cooperates with Vhl and Ink4a/Arf loss to induce clear cell renal cell carcinoma. Nature Communications. 2017;8:15770
  110. 110. Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature. 1988;335(6189):440-442
  111. 111. Teitz T, Wei T, Valentine MB, Vanin EF, Grenet J, Valentine VA, et al. Caspase 8 is deleted or silenced preferentially in childhood neuroblastomas with amplification of MYCN. Nature Medicine. 2000;6(5):529-535
  112. 112. Wu KJ, Grandori C, Amacker M, Simon-Vermot N, Polack A, Lingner J, et al. Direct activation of TERT transcription by c-MYC. Nature Genetics. 1999;21(2):220-224
  113. 113. Okamoto K, Seimiya H. Revisiting telomere shortening in cancer. Cells. 2019;8:2
  114. 114. Hofmann JW, Zhao X, De Cecco M, Peterson AL, Pagliaroli L, Manivannan J, et al. Reduced expression of MYC increases longevity and enhances healthspan. Cell. 2015;160(3):477-488
  115. 115. Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduction and Targeted Therapy. 2020;5(1):8
  116. 116. McKeown MR, Bradner JE. Therapeutic strategies to inhibit MYC. Cold Spring Harbor Perspectives in Medicine. 2014;4(10):a014266
  117. 117. Allen-Petersen BL, Sears RC. Mission possible: Advances in MYC therapeutic targeting in cancer. BioDrugs. 2019;33(5):539-553
  118. 118. Tu WB, Helander S, Pilstål R, Hickman KA, Lourenco C, Jurisica I, et al. Myc and its interactors take shape. Biochimica et Biophysica Acta. 2015;1849(5):469-483
  119. 119. Han H, Jain AD, Truica MI, Izquierdo-Ferrer J, Anker JF, Lysy B, et al. Small-molecule MYC inhibitors suppress tumor growth and enhance immunotherapy. Cancer Cell. 2019;36(5):483-97.e15
  120. 120. Struntz NB, Chen A, Deutzmann A, Wilson RM, Stefan E, Evans HL, et al. Stabilization of the max homodimer with a small molecule attenuates Myc-driven transcription. Cell Chemical Biology. 2019;26(5):711-23.e14
  121. 121. Lustig LC, Dingar D, Tu WB, Lourenco C, Kalkat M, Inamoto I, et al. Inhibiting MYC binding to the E-box DNA motif by ME47 decreases tumour xenograft growth. Oncogene. 2017;36(49):6830-6837
  122. 122. Boike L, Cioffi AG, Majewski FC, Co J, Henning NJ, Jones MD, et al. Discovery of a functional covalent ligand targeting an intrinsically disordered cysteine within MYC. Cell Chemical Biology. 2021;28(1):4-13.e7
  123. 123. Pourdehnad M, Truitt ML, Siddiqi IN, Ducker GS, Shokat KM, Ruggero D. Myc and mTOR converge on a common node in protein synthesis control that confers synthetic lethality in Myc-driven cancers. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(29):11988-11993
  124. 124. Wiegering A, Uthe FW, Jamieson T, Ruoss Y, Hüttenrauch M, Küspert M, et al. Targeting translation initiation bypasses signaling crosstalk mechanisms that maintain high MYC levels in colorectal cancer. Cancer Discovery. 2015;5(7):768-781
  125. 125. Gustafson WC, Meyerowitz JG, Nekritz EA, Chen J, Benes C, Charron E, et al. Drugging MYCN through an allosteric transition in Aurora kinase a. Cancer Cell. 2014;26(3):414-427
  126. 126. Winter GE, Buckley DL, Paulk J, Roberts JM, Souza A, Dhe-Paganon S, et al. Drug development. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science. 2015;348(6241):1376-1381
  127. 127. Bondeson DP, Mares A, Smith IE, Ko E, Campos S, Miah AH, et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nature Chemical Biology. 2015;11(8):611-617
  128. 128. Dhanasekaran R, Park J, Yevtodiyenko A, Bellovin DI, Adam SJ, Kd AR, et al. MYC ASO impedes tumorigenesis and elicits oncogene addiction in autochthonous transgenic mouse models of HCC and RCC. Molecular Therapy—Nucleic Acids. 2020;21:850-859
  129. 129. Felsenstein KM, Saunders LB, Simmons JK, Leon E, Calabrese DR, Zhang S, et al. Small molecule microarrays enable the identification of a selective, quadruplex-binding inhibitor of MYC expression. ACS Chemical Biology. 2016;11(1):139-148
  130. 130. Jung LA, Gebhardt A, Koelmel W, Ade CP, Walz S, Kuper J, et al. OmoMYC blunts promoter invasion by oncogenic MYC to inhibit gene expression characteristic of MYC-dependent tumors. Oncogene. 2017;36(14):1911-1924
  131. 131. Tanizaki Y, Sudo M, Kitani H, Kawauchi K, Mifune T, Takeyama H, et al. Characteristic of cell components in bronchoalveolar lavage fluid (BALF) in patients with bronchial asthma classified by clinical symptoms. Arerugī. 1990;39(2 Pt 1):75-81
  132. 132. Topper MJ, Vaz M, Chiappinelli KB, DeStefano Shields CE, Niknafs N, Yen RC, et al. Epigenetic therapy ties MYC depletion to reversing immune evasion and treating lung cancer. Cell. 2017;171(6):1284-300.e21
  133. 133. Dhanasekaran R, Gabay-Ryan M, Baylot V, Lai I, Mosley A, Huang X, et al. Anti-miR-17 therapy delays tumorigenesis in MYC-driven hepatocellular carcinoma (HCC). Oncotarget. 2018;9(5):5517-5528
  134. 134. Xie X, Yu T, Li X, Zhang N, Foster LJ, Peng C, et al. Recent advances in targeting the "undruggable" proteins: From drug discovery to clinical trials. Signal Transduction and Targeted Therapy. 2023;8(1):335
  135. 135. Donati G, Amati B. MYC and therapy resistance in cancer: Risks and opportunities. Molecular Oncology. 2022;16(21):3828-3854
  136. 136. Lu X, Vogt PK, Boger DL, Lunec J. Disruption of the MYC transcriptional function by a small-molecule antagonist of MYC/MAX dimerization. Oncology Reports. 2008;19(3):825-830
  137. 137. Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: The past is prologue. Nature Reviews. Drug Discovery. 2022;21(3):181-200
  138. 138. Martínez-Martín S, Soucek L. MYC inhibitors in multiple myeloma. Cancer Drug Resistance. 2021;4(4):842-865
  139. 139. Kciuk M, Gielecińska A, Mujwar S, Mojzych M, Kontek R. Cyclin-dependent kinase synthetic lethality partners in DNA damage response. International Journal of Molecular Sciences. 2022;23(7):3555
  140. 140. Nebbioso A, Carafa V, Conte M, Tambaro FP, Abbondanza C, Martens J, et al. c-Myc modulation and acetylation is a key HDAC inhibitor target in cancer. Clinical Cancer Research. 2017;23(10):2542-2555
  141. 141. Vollmer J, Ecker J, Hielscher T, Valinciute G, Ridinger J, Jamaladdin N, et al. Class I HDAC inhibition reduces DNA damage repair capacity of MYC-amplified medulloblastoma cells. Journal of Neuro-Oncology. 2023;164(3):617-632
  142. 142. Rathert P, Roth M, Neumann T, Muerdter F, Roe JS, Muhar M, et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature. 2015;525(7570):543-547
  143. 143. Gill T, Wang H, Bandaru R, Lawlor M, Lu C, Nieman LT, et al. Selective targeting of MYC mRNA by stabilized antisense oligonucleotides. Oncogene. 2021;40(47):6527-6539
  144. 144. Doha ZO, Sears RC. Unraveling MYC's role in orchestrating tumor intrinsic and tumor microenvironment interactions driving tumorigenesis and drug resistance. Pathophysiology. 2023;30(3):400-419
  145. 145. Miller DM, Thomas SD, Islam A, Muench D, Sedoris K. c-Myc and cancer metabolism. Clinical Cancer Research. 2012;18(20):5546-5553
  146. 146. Fulda S, Gorman AM, Hori O, Samali A. Cellular stress responses: Cell survival and cell death. International Journal of Cell Biology. 2010;2010:214074
  147. 147. Luoto KR, Meng AX, Wasylishen AR, Zhao H, Coackley CL, Penn LZ, et al. Tumor cell kill by c-MYC depletion: Role of MYC-regulated genes that control DNA double-strand break repair. Cancer Research. 2010;70(21):8748-8759
  148. 148. Dang CV. c-Myc target genes involved in cell growth, apoptosis, and metabolism. Molecular and Cellular Biology. 1999;19(1):1-11
  149. 149. Llombart V, Mansour MR. Therapeutic targeting of "undruggable" MYC. eBioMedicine. 2022;75:103756

Written By

Lucia Capasso, Donato Mele, Fatima Fayyaz, Lucia Altucci and Angela Nebbioso

Submitted: 05 December 2024 Reviewed: 13 December 2024 Published: 14 March 2025