Fact-checked by Grok 2 weeks ago

MYC

MYC is a proto-oncogene that encodes a functioning as a basic helix-loop-helix (bHLH-LZ) , which heterodimerizes with MAX to bind DNA sequences and regulate critical for progression, , , and cellular transformation. Located on chromosome 8q24.21 in humans, is ubiquitously expressed across tissues, with particularly high levels in the and , and belongs to a superfamily including MYCL and MYCN that collectively act as master regulators of cellular and . In normal cellular physiology, integrates diverse signals from pathways such as WNT and to drive processes like , protein synthesis, and metabolic reprogramming, ensuring tightly controlled while preventing uncontrolled growth that could lead to or cell death. Its activity is precisely regulated through transcriptional activation, epigenetic modifications, and post-translational mechanisms, including at serine 62 for stabilization and ubiquitin-mediated degradation via E3 ligases like FBW7. Dysregulation of , often via genomic amplification of or related family members (observed in approximately 28% of tumors across 33 cancer types) or chromosomal translocations (as in and ), transforms it into a potent that promotes including sustained , genomic instability, , and immune evasion through upregulation of and CD47. MYC overexpression is implicated in a wide array of malignancies, serving as a susceptibility locus for cancers such as , , and , and is associated with poor due to its role in tumor progression and resistance to therapy. Despite its central role in tumorigenesis—evidenced by tumor upon MYC inactivation in preclinical models—no direct MYC-targeted therapies are clinically approved as of 2025, though promising strategies include small-molecule inhibitors of the MYC-MAX interaction (e.g., Omomyc derivatives), protein degraders, and synthetic lethal approaches exploiting MYC dependency, such as CDK inhibitors, are under investigation in clinical trials.

Discovery and Molecular Basics

Historical Discovery

The discovery of the proto-oncogene originated from studies of retroviral oncogenesis in the . The avian myelocytomatosis virus strain MC29 (MC29), a replication-defective first isolated in the 1960s but extensively characterized in the , was observed to rapidly induce myelocytomas, carcinomas, and leukemias in infected chickens. In 1977, researchers identified an unusual 110-kDa viral-related polypeptide produced in MC29-transformed avian cells, leading to the recognition of the v-myc as the transforming principle within the viral genome; v-myc was a hybrid gene fusing partial gag sequences with novel myc coding regions responsible for tumorigenesis. This work built on the emerging understanding of retroviral oncogenes, for which J. Michael and received the 1989 in Physiology or Medicine for demonstrating their cellular origins. In 1982, the cellular homolog of v-myc, termed c-myc, was isolated and characterized from normal chicken DNA using techniques, revealing high between the viral and cellular genes and confirming c-myc as a proto-oncogene. Concurrently, the human c-MYC homolog was identified through Southern blotting and cloning experiments on DNA from Burkitt's lymphoma cells, showing that it mapped to and shared structural similarities with v-myc, including conserved exons encoding functional domains. These findings established c-MYC as the cellular counterpart to the viral oncogene, with potential roles in normal cellular growth control. By the early 1980s, c-MYC was directly linked to cancer through cytogenetic and molecular analyses of Burkitt's lymphoma, a B-cell malignancy endemic in certain regions and associated with Epstein-Barr virus. In 1982, studies demonstrated that the characteristic t(8;14)(q24;q32) in these tumors juxtaposed the c-MYC locus on with the (IGH) locus on chromosome 14, leading to deregulation of c-MYC expression via enhancer activation. Variant translocations t(2;8) and t(8;22) involving light chain loci were also identified, consistently placing c-MYC under immunoglobulin transcriptional control and highlighting its role as a driver of lymphomagenesis. These discoveries, achieved through mapping and hybridization probes, marked c-MYC as the first proto-oncogene implicated in a specific chromosomal abnormality.

Gene Structure and Genomic Location

The MYC , also known as c-MYC, is located on the long arm of at the cytogenetic band 8q24.21, with genomic coordinates spanning from 127,735,434 to 127,742,951 on the GRCh38.p14 reference assembly. This positions it in a prone to structural variations, though the itself consists of three s distributed over approximately 7.5 kb of genomic DNA. The first is largely non-coding, while s 2 and 3 encode the functional protein domains, separated by two introns that facilitate alternative processing. Transcription of is primarily regulated by two tandem promoters, P1 and P2, located upstream of 1 and separated by about 150 base pairs. The upstream P1 promoter lacks a and initiates a longer transcript that includes the full 1 sequence, accounting for roughly 25% of total transcripts in normal cells. In contrast, the downstream P2 promoter features a canonical and drives the majority of expression (approximately 75%), producing a shorter transcript with a truncated . These promoters respond to distinct regulatory signals, enabling fine-tuned control of expression during cellular processes. The gene exhibits strong evolutionary conservation, reflecting its fundamental role in cellular . Homologs are present across mammals and , with the v-myc originating from the cellular c-myc in the myelocytomatosis MC29. This conservation extends to more distant eukaryotes, including myc-like genes in invertebrates such as (dMyc) and even basal metazoans like , where myc and max homologs date back over 600 million years. Such broad preservation underscores the ancient origins of the Myc network in eukaryotic . Alternative processing of pre-mRNA and mRNA contributes to isoform diversity, primarily through differential initiation rather than extensive splicing variants. The predominant isoform, c-Myc1, initiates from a CUG codon in 1; c-Myc2 uses an AUG in 2; and the short isoform c-MycS starts from a downstream within 2, yielding a truncated protein lacking most of the N-terminal . This c-MycS form retains proliferative functions but has reduced apoptotic activity compared to full-length isoforms.

Protein Structure and Expression

Protein Domains and Architecture

The c-Myc protein, the prototypical member of the MYC family, is a 439-amino-acid polypeptide with a calculated of approximately 49 . Its architecture is modular, featuring an intrinsically disordered N-terminal (TAD) spanning residues 1–143, which mediates interactions with transcriptional co-activators and the core machinery. Flanking the TAD are two conserved central regions known as Myc boxes I (MBI, residues 45–63) and II (MBII, residues 129–141), which are short motifs enriched in acidic and residues that facilitate protein-protein interactions essential for 's regulatory functions. These Myc boxes are sites of post-translational modifications, including at serine 62 (Ser62) within MBI, which promotes initial stabilization, and 58 (Thr58), also in MBI, which triggers ubiquitin-mediated via recruitment of E3 ligases like FBXW7. The C-terminal region (residues ~353–439) comprises the basic helix-loop-helix leucine zipper (bHLH-LZ) domain, a bipartite motif critical for DNA binding and dimerization. The leucine zipper (LZ) subdomain forms a coiled-coil alpha-helix that enables heterodimerization with the partner protein MAX, while the adjacent basic region (b) and helix-loop-helix (HLH) motifs position the dimer on DNA. This bHLH-LZ architecture allows the MYC-MAX heterodimer to specifically recognize and bind the canonical E-box sequence 5'-CACGTG-3' in target gene promoters, facilitating transcriptional activation. Phosphorylation within the bHLH-LZ, such as at serine 373, can further modulate DNA affinity and stability, though these sites are less central than those in the Myc boxes. Structural studies using (NMR) spectroscopy and have elucidated the bHLH-LZ conformation, revealing predominantly alpha-helical secondary structures in both the basic region and LZ upon dimerization and DNA engagement. For instance, crystal structures of the MYC-MAX bHLH-LZ complex bound to E-box DNA show the basic regions inserting into the major groove as extended helices, with the HLH motifs stabilizing the dimer through hydrophobic packing. NMR analyses of the apo form (without DNA) indicate residual helical propensity in the basic region, suggesting intrinsic structural flexibility that enhances binding versatility. These models highlight how the bHLH-LZ domain's helical architecture underpins MYC's role in dimerization-dependent transcription, distinct from its N-terminal regulatory elements.

Expression Patterns in Tissues

MYC displays dynamic spatiotemporal expression patterns that correlate closely with cellular states during normal and in adult tissues. In embryonic stem cells, MYC is highly expressed, where it maintains pluripotency and supports self-renewal by amplifying the transcription of active genes essential for these processes. During early embryonic stages, MYC levels are initially low but increase significantly during , peaking to drive the expansion of populations and lineage commitment in developing organs such as and intestine. Postnatally, MYC expression is broadly downregulated in most tissues as cells exit the cell cycle and undergo terminal differentiation, thereby restricting its activity to specific renewing compartments. In adult physiology, maintains elevated expression in actively proliferating tissues, including the crypts of the , where it sustains epithelial renewal, and the basal layer of the in , aligning with regions of high activity. Similarly, is prominently expressed in immune cells, such as B and T lymphocytes, where it orchestrates and in response to antigenic stimuli within lymphoid tissues. In contrast, levels are low or undetectable in quiescent or postmitotic differentiated cells, including mature neurons in the and fibers, reflecting its restricted role outside proliferative contexts. These expression patterns have been characterized through various detection methods, including quantitative polymerase chain reaction (qPCR) for mRNA quantification, Western blotting for protein levels, and (IHC) to visualize spatial gradients in embryonic and adult s. For instance, IHC studies on embryos reveal progressive MYC gradients from proliferative zones to differentiating regions during . This distribution underscores MYC's selective association with proliferative states in normal biology.

Biological Functions

Role in Cell Proliferation and Growth

MYC plays a central role in promoting during normal physiological processes by facilitating the transition from the G1 to of the . It achieves this by inducing the expression of D-type cyclins (such as and D2) and cyclin-dependent kinases CDK4 and CDK6, which form active complexes that phosphorylate the (). Phosphorylated Rb releases transcription factors, enabling the activation of genes required for and S-phase entry. This mechanism ensures efficient progression through the in response to growth signals in proliferating tissues like embryonic development and tissue regeneration. In addition to cell cycle control, MYC drives cellular growth by reprogramming metabolism to support biomass accumulation and energy demands. It upregulates glycolytic enzymes, including lactate dehydrogenase A (LDHA), which converts pyruvate to lactate, thereby enhancing aerobic glycolysis even in oxygen-rich environments to generate intermediates for biosynthesis. MYC also promotes glutaminolysis by increasing expression of glutaminase (GLS), which hydrolyzes glutamine to glutamate for entry into the tricarboxylic acid cycle, providing precursors for amino acid and nucleotide production. Furthermore, MYC stimulates nucleotide synthesis pathways by activating genes such as phosphoribosyl pyrophosphate synthetase 2 (PRPS2) and enzymes in the pentose phosphate pathway, ensuring sufficient purine and pyrimidine pools for DNA replication. MYC further supports growth by enhancing ribosome biogenesis and protein synthesis, essential for increasing cellular mass. It directly regulates transcription by RNA polymerases I and III, which produce ribosomal RNA (rRNA) components and 5S rRNA, respectively, leading to elevated ribosome assembly. This amplification of translational capacity boosts overall protein production, coordinating with metabolic shifts to sustain proliferative demands. Quantitative studies in rat fibroblasts demonstrate that restoring MYC expression in myc-null cells reduces the cell doubling time from approximately 45 hours to 24 hours, underscoring its potent impact on proliferation rates.

Transcriptional Activity and Target Genes

MYC functions as a basic helix-loop-helix leucine zipper (bHLH-LZ) that dimerizes with MAX to bind canonical sequences (5'-CACGTG-3') in DNA, thereby regulating a substantial portion of the . -wide studies indicate that MYC binds to approximately 10-15% of all promoters and enhancers in mammalian cells, preferentially occupying sites at active genomic regions rather than initiating transcription . In this capacity, MYC primarily acts as a transcriptional , enhancing the output of genes that are already poised for expression by boosting (Pol II) processivity and elongation rates, particularly in highly transcribed loci. MYC exerts its activating effects through recruitment of co-activator complexes that modify and facilitate transcription initiation and elongation. Notably, MYC interacts with the TIP60 complex, which includes subunits such as TRRAP, p400, TIP48, and TIP49, to promote and H4 at target sites, thereby opening structure for Pol II recruitment and activity. This co-activation mechanism is evident at promoters where MYC binding correlates with increased Pol II occupancy and reduced Pol II pausing, amplifying without requiring strict dependency in some contexts. MYC directly regulates diverse target genes involved in core cellular processes, with effects varying by context. For cell cycle progression, MYC activates genes such as CCND1 (encoding cyclin D1) and CDK4 (cyclin-dependent kinase 4), which drive G1/S transition through retinoblastoma protein phosphorylation. In metabolism, MYC upregulates glycolytic enzymes and transporters, including GLUT1 (glucose transporter 1) and HK2 (hexokinase 2), to enhance glucose uptake and flux, supporting biomass production in proliferating cells. Regarding apoptosis, MYC can suppress pro-apoptotic factors like BIM (BCL2L11), thereby promoting survival under stress conditions, though this repression is often indirect via interference with other regulators. Genome-wide ChIP-seq analyses have revealed that MYC's transcriptional effects are context-dependent, with both and repression observed across thousands of sites. In normal cells, MYC predominantly activates genes associated with and , but in transformed cells with elevated MYC levels, it also represses specific subsets, such as those involved in or immune , through recruitment of repressive complexes like G9a for H3K9 . These studies highlight MYC's , where affinity, local state, and cellular context determine whether a target gene is amplified or silenced.

Regulation Mechanisms

Transcriptional and Epigenetic Control

The transcription of the gene is tightly regulated by multiple signaling pathways that converge on its promoter and distal regulatory elements. In the canonical Wnt/β-catenin pathway, stabilized β-catenin translocates to the and forms complexes with TCF/LEF transcription factors, which bind to Wnt-responsive elements (WREs) upstream and downstream of the promoter to activate transcription. These include a proximal 5' WRE at -889 bp and a distal -335 WRE, which interact via looping to enhance promoter and recruit co-activators. Similarly, signaling activates through the intracellular domain of NOTCH1 (NICD), which binds the RBPJ transcription factor to directly engage the promoter, driving expression in contexts like T-cell leukemia. This interaction forms part of a feed-forward loop where reinforces Notch targets. Growth factor stimulation, such as via serum or insulin, engages the MAPK/ERK pathway, where ERK phosphorylates and promotes degradation of the Myc repressor Mad1 through RSK, thereby derepressing transcription and enabling proliferation. Distal enhancers at the 8q24 locus, spanning approximately 3 Mb around , play a pivotal role in its long-range transcriptional control, often forming super-enhancer complexes that amplify expression in proliferative cells. These enhancers, including the Notch-sensitive +81/+84 region, loop to the promoter via co-activator recruitment, facilitating high-level transcription. , a protein, binds these super-enhancers to stabilize the transcriptional machinery, and its inhibition disrupts looping and reduces levels in cancers like . Such super-enhancers are enriched in multiple cell types, integrating signals from pathways like Wnt and to sustain output. Epigenetic modifications further modulate MYC accessibility, with active histone acetylation marking enhancers for transcription. Histone H3 lysine 27 acetylation (H3K27ac) is prominently enriched at 8q24 super-enhancers and the proximal promoter, correlating with occupancy and elevated expression in proliferating cells. In contrast, during differentiation, such as in granulocytes or primed pluripotent cells, loss of H3K27ac at enhancers represses , contributing to exit. DNA methylation at the promoter also enforces in differentiated states, preventing aberrant reactivation and maintaining lineage commitment. MYC engages in auto-regulatory feedback to fine-tune its expression, notably by directly transcribing the miR-17-92 cluster, which encodes miR-19 to suppress PTEN and amplify PI3K/AKT signaling, thereby indirectly sustaining MYC activity. This loop integrates with upstream signals, ensuring balanced proliferation without excessive .

Post-transcriptional and Degradation Pathways

The stability of MYC mRNA is primarily regulated post-transcriptionally through AU-rich elements () located in its 3' (3'UTR), which serve as binding sites for RNA-binding proteins that either promote or inhibit mRNA decay. The protein HuR binds to these and stabilizes MYC mRNA by preventing its degradation, thereby enhancing MYC expression in proliferating cells. In contrast, AUF1 (also known as hnRNP D) binds to the same and promotes MYC mRNA destabilization and rapid turnover, acting as a key negative regulator of MYC levels. These opposing actions of HuR and AUF1 on shared mRNAs, including MYC, allow for fine-tuned control of transcript in response to cellular signals. MicroRNAs further contribute to post-transcriptional repression of by targeting conserved sites in the 3'UTR, leading to mRNA destabilization or translational inhibition. For instance, members of the let-7 family, such as let-7g, directly bind to the 3'UTR, promoting mRNA degradation and reducing protein levels, which suppresses in contexts like epithelial . Similarly, miR-34a targets the 3'UTR, inducing its decay and downregulating expression to exert tumor-suppressive effects, as observed in endometrial carcinoma cells where miR-34a overexpression decreases alongside other oncogenes. These miRNA interactions exemplify how non-coding RNAs integrate with ARE-mediated control to modulate mRNA abundance. At the protein level, stability is tightly controlled through ubiquitin-proteasome-mediated , with a short of approximately 20-30 minutes in proliferating cells. at 58 (Thr58) within the N-terminal Myc box I serves as a key signal for ubiquitination by the SCF^{Fbw7} complex, which recognizes the phosphorylated site and targets for proteasomal ; mutations at Thr58, such as those in human cancers, impair this process and extend . Conversely, at serine 62 (Ser62) by kinases including Aurora A stabilizes by inhibiting the subsequent Thr58 required for SCF^{Fbw7} recognition, thereby delaying and enhancing oncogenic activity. This dual code within the conserved motif thus balances .

Role in Disease

Involvement in Oncogenesis

MYC plays a central role in oncogenesis by driving uncontrolled , metabolic reprogramming, and evasion of when deregulated. Overexpression of MYC occurs in more than 70% of human cancers, contributing to tumor initiation and progression across diverse malignancies. This deregulation is particularly prevalent in aggressive tumors such as , , and , where elevated MYC levels correlate with poor prognosis and advanced disease stages. For instance, in , MYC amplification is a hallmark of the high-risk Group 3 subtype, promoting rapid tumor growth and . In , MYC rearrangements and overexpression sustain proliferation and survival. The historical association of MYC with arose from its identification as the target of chromosomal translocations in Burkitt's lymphoma. Deregulation of arises through multiple genetic alterations, including , chromosomal translocations, and point mutations that stabilize the protein. Amplification of the locus at 8q24 is common in solid tumors, such as and cancers, where it leads to increased gene dosage and transcriptional hyperactivity. In , amplification is acquired in metastatic lesions and associates with aggressive phenotypes and reduced survival. Similarly, in , 8q24 amplification disrupts signaling and drives tumor progression. Chromosomal translocations, most notably t(8;14)(q24;q32), juxtapose with immunoglobulin heavy chain enhancers in Burkitt's lymphoma, resulting in constitutive expression driven by B-cell-specific promoters. Point mutations, such as the T58A substitution in the MYC Box I domain, impair phosphorylation-dependent ubiquitination and proteasomal degradation, thereby stabilizing the protein and enhancing its oncogenic potential in lymphomas. In transformation models, MYC cooperates with RAS to induce full malignant conversion, bypassing senescence and promoting anchorage-independent growth. This synergy was first demonstrated in primary rat embryo fibroblasts, where co-expression of MYC and activated RAS enabled tumorigenic transformation. MYC also drives genomic instability, facilitating additional mutations that accelerate cancer progression; for example, it upregulates activation-induced cytidine deaminase (AID), leading to DNA double-strand breaks at the MYC locus and IgH, which promote oncogenic translocations in B cells. In vivo, the Eμ-Myc transgenic mouse model recapitulates human Burkitt's lymphoma, with mice developing pre-B-cell lymphomas within months due to MYC overexpression under IgH enhancer control. These tumors exhibit dependency on stromal microenvironment support, as lymphoma cells rapidly apoptose in vitro without dendritic cell-derived survival signals, highlighting the role of tumor-host interactions in sustaining oncogenesis.

Associations with Non-Cancer Diseases

MYC overexpression has been observed in peripheral blood lymphocytes from patients with systemic lupus erythematosus (SLE), correlating with enhanced B-cell proliferation and hyperactivity. In SLE, B lymphocytes exhibit significantly higher c-Myc RNA levels compared to T cells and normal controls, indicating a pathologically activated state that contributes to autoimmune B-cell responses. This dysregulation aligns with broader roles of MYC in immune cell activation, where elevated expression promotes metabolic reprogramming and proliferation in autoimmune contexts. In fibrotic conditions such as (IPF), MYC is upregulated in lung tissues and drives the proliferation and differentiation of pulmonary fibroblasts into . Specifically, C-MYC activates miR-9-5p to suppress TBPL1 expression, thereby enhancing fibroblast proliferation, deposition, and progression in IPF models. Silencing C-MYC or using like 10,058-F4 reduces miR-9-5p levels, upregulates TBPL1, and attenuates , highlighting its pro-fibrotic role. in IPF further amplifies this by derepressing C-MYC through miR-210-mediated repression of its MNT, linking environmental stress to activation. MYC activation in the cardiovascular system shows context-dependent effects, with transient upregulation promoting adaptive cardiac while chronic activation leads to heart failure. Inducible C-Myc expression in adult cardiomyocytes induces myocyte , increases protein synthesis, and reactivates , mimicking pressure-overload responses without initial functional impairment. However, sustained Myc induction triggers cell cycle re-entry, mitochondrial defects (including reduced complex I and III activity), increased , and severe , culminating in ventricular dysfunction and 100% mortality within weeks in mouse models. In infectious diseases, the HIV Tat protein promotes MYC-mediated metabolic reprogramming in T cells through activation of signaling. This activation enhances , , and overall metabolic reprogramming in T cells, promoting and immune dysregulation that supports viral persistence in HIV infection.

Therapeutic and Clinical Aspects

Strategies for Targeting MYC

The development of strategies to target has been challenging due to its intrinsically disordered structure and lack of well-defined binding pockets, often described as "undruggable." Despite this, multiple pharmacological and genetic approaches have emerged to inhibit activity, focusing on disrupting its dimerization, , stability, or expression. These methods aim to suppress MYC-driven oncogenesis while minimizing off-target effects in preclinical models. Direct inhibition of MYC has targeted its basic helix-loop-helix leucine zipper (bHLH-LZ) domain, which is essential for dimerization with MAX and subsequent DNA binding. Small molecules such as KI-MS2-008 bind to the bHLH-LZ region of MAX, stabilizing the MAX homodimer and thereby preventing MYC-MAX heterodimer formation. This compound reduces MYC protein levels and target gene expression in MYC-dependent cancer cell lines, demonstrating preclinical efficacy in suppressing tumor growth in mouse models of T-cell lymphoma. Other small molecules, including 10058-F4 and IIA6B17, similarly disrupt the MYC-MAX interaction by binding to the leucine zipper motif, leading to decreased transcriptional activity in vitro. Indirect pharmacological strategies modulate through upstream regulators or stability pathways. BET bromodomain inhibitors, such as , disrupt the interaction between and acetylated histones at super-enhancers, which are critical for high-level transcription in cancers like . Treatment with preferentially evicts from super-enhancers, resulting in rapid downregulation of expression and selective inhibition of oncogene-driven proliferation in tumor cells. Complementing this, Aurora kinase inhibitors like alisertib (MLN8237) promote degradation by disrupting the stabilizing complex between Aurora A and the transactivation domain, leading to proteasomal breakdown of protein in neuroblastoma and other MYC-amplified cancers. These inhibitors induce conformational changes in Aurora A that release , enhancing its ubiquitination and turnover. Genetic approaches leverage nucleic acid-based tools to reduce expression. Antisense oligonucleotides (), such as conformationally stabilized MYCASOs, hybridize to mRNA, promoting its degradation via RNase H and thereby potently suppressing protein in cancer cell lines. These exhibit sequence-specific inhibition, with preclinical studies showing reduced tumor progression in -overexpressing models. Similarly, () constructs achieve knockdown by , inhibiting stem-like cell characteristics and proliferation in and xenografts, with ongoing exploration in therapeutic vectors for clinical translation. Addressing MYC's undruggability, proteolysis-targeting chimeras (PROTACs) represent an emerging paradigm by recruiting E3 ubiquitin ligases to enhance MYC and proteasomal degradation. MYC-directed PROTACs, such as those linking MYC-binding motifs to ligands, induce bimodal degradation in preclinical settings, bypassing traditional binding pocket requirements and offering potential for broader applicability in MYC-driven malignancies.

Clinical Applications and Challenges

MYC serves as a valuable in clinical . In a subset of high-risk pediatric neuroblastomas without MYCN amplification, MYC overexpression and rare 8q24 amplification contribute to aggressive disease and poor prognosis, identifiable via (FISH) and (IHC). In (DLBCL), elevated expression assessed via IHC is a robust prognostic indicator, with high levels correlating to significantly reduced overall survival rates, independent of other factors like MYC/BCL2 co-expression. Ongoing clinical trials explore direct inhibition as a therapeutic avenue in MYC-driven malignancies. For instance, phase I studies of DCR-MYC, a nanoparticle-formulated RNAi therapeutic targeting MYC mRNA, have demonstrated tolerability and preliminary antitumor activity in patients with advanced solid tumors, including reductions in tumor size, with results reported as of 2025. Similarly, the dominant-negative inhibitor OMO-103 has advanced through phase I dose-escalation and into phase 2 trials in solid tumors as of 2025, including for advanced , showing manageable safety profiles and hints of efficacy in MYC-overexpressing subsets. Combination strategies integrating MYC inhibitors with hold promise for enhancing response in MYC-amplified cancers. Preclinical data support pairing MYC blockade with PD-1 inhibitors, where MYC suppression boosts T-cell infiltration and sensitizes tumors to checkpoint blockade, as evidenced by improved tumor regression in models; early-phase trials are now evaluating such synergies in MYC-driven solid tumors. Despite these advances, translating MYC-targeted therapies to the clinic faces substantial hurdles. MYC's essential role in normal cellular —regulating up to 15% of the —predisposes inhibitors to on-target , including gastrointestinal and hematopoietic adverse effects observed in early trials. Additionally, resistance emerges through bypass mechanisms, such as activation of parallel survival pathways like PI3K/AKT, which sustain tumor growth despite MYC inhibition and necessitate combination regimens to circumvent adaptive responses. These challenges highlight the need for tumor-selective delivery and biomarker-driven patient selection to optimize therapeutic indices.

References

  1. [1]
    MYC MYC proto-oncogene, bHLH transcription factor [ (human)]
    Sep 14, 2025 · This gene is a proto-oncogene and encodes a nuclear phosphoprotein that plays a role in cell cycle progression, apoptosis and cellular transformation.
  2. [2]
    The MYC oncogene — the grand orchestrator of cancer growth ... - NIH
    The MYC proto-oncogenes encode a family of transcription factors that are among the most commonly activated oncoproteins in human neoplasias.
  3. [3]
    A recovered avian myelocytomatosis virus that induces lymphomas ...
    However, MC29 has never been reported to cause lymphoid tumors, the disease associated with activation of the c-myc gene by the insertion of a lymphoid leukosis ...
  4. [4]
    cshperspectmed-MYC-A014357 1..24
    (2014). MYC-ENCODED PROTEINS. In 1977, an avian cell line transformed by the v-myc-containing retrovirus MC29 was found to produce an unusual viral-related ...
  5. [5]
    The Nobel Prize in Physiology or Medicine 1989 - Press release
    Bishop's and Varmus' discovery of the cellular origin of retroviral oncogenes has had an extensive influence on the development of our knowledge about ...
  6. [6]
    Identification and nucleotide sequence of a human locus ... - PubMed
    Feb 24, 1983 · Identification and nucleotide sequence of a human locus homologous to the v-myc oncogene of avian myelocytomatosis virus MC29. Nature. 1983 ...
  7. [7]
    Translocation of the c-myc gene into the immunoglobulin ... - PubMed
    In two t(8;14) human Burkitt cell lines, c-myc appears to have been translocated directly into a DNA restriction fragment that also encodes the immunoglobulin ...
  8. [8]
    A variant translocation places the λ immunoglobulin genes 3 - Nature
    Feb 23, 1984 · Abstract ; Burkitt lymphoma · 15 December 2022 ; Rare t(X;14)(q28;q32) translocation reveals link between MTCP1 and chronic lymphocytic leukemia.Missing: discovery | Show results with:discovery
  9. [9]
    Translocation of the c-myc gene into the immunoglobulin heavy ...
    Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. R Taub, I Kirsch, C ...
  10. [10]
    structure and activity of L-myc and an L-myc pseudogene
    While the untranslated regions of the indi- vidual c o, N-, and L-myc genes are conserved across species, the untranslated regions of each type of myc gene ...
  11. [11]
    MYC (MYC proto-oncogene, bHLH transcription factor)
    Aug 1, 2017 · There are four MYC transcriptional promoters. In normal cells, promoter P2 contributes to approximately 75% of total MYC transcripts while P1 ...
  12. [12]
    Isolation and characterization of c-myc, a cellular homolog ... - PubMed
    We have isolated chicken DNA (c-myc) that is homologous to the oncogene (v-myc) of the avian myelocytomatosis virus MC29 and have compared the structures of the ...
  13. [13]
    Stem cell-specific activation of an ancestral myc protooncogene with ...
    Feb 8, 2010 · Here, we describe the cloning and structural and functional analysis of myc and max genes from Hydra, representing the oldest and most primitive ...
  14. [14]
    Evolutionarily conserved regions of the human c-myc protein ... - NIH
    The myc family of oncogenes contains coding sequences that have been preserved in different species for over 400 million years. This conservation (which ...Missing: across | Show results with:across
  15. [15]
    Human c-Myc Isoforms Differentially Regulate Cell Growth and ...
    To obtain two copies of the c-MycS transgene, we recombined two UAS-c-MycS transgenes on chromosome 2 or 3. ... The truncated c-MycS isoform rescues the growth ...
  16. [16]
    Anti-Myc Tag Antibody, clone 4A6 (mouse monoclonal IgG1) | 05 ...
    ... C-MYC, c-Myc UniProt Number: P01106 · View Products ... Seems to activate the transcription of growth-related genes. SIZE: 439 amino acids; 48804 Da ... Predicted ...
  17. [17]
    Taking the Myc out of cancer: toward therapeutic strategies to ...
    Jan 4, 2021 · c-Myc is a 62 kDa protein of 439 residues and ... Omomyc contains four amino acid substitutions in the leucine zipper domain of c-Myc ...
  18. [18]
    Phosphorylation-dependent degradation of c-Myc is mediated ... - NIH
    However, the turnover of c-Myc is largely dependent on phosphorylation of threonine-58 and serine-62 in MB1, residues that are often mutated in cancer.
  19. [19]
    Genome Recognition by MYC - PMC - NIH
    MYC dimerizes with MAX to bind DNA, with a preference for the E-box consensus CACGTG and several variant motifs. In cells, MYC binds DNA preferentially within ...
  20. [20]
    Structural and Biophysical Insights into the Function of the ... - PMC
    Apr 22, 2020 · Myc is a transcription factor driving growth and proliferation of cells and involved in the majority of human tumors.
  21. [21]
    Crystal Structures and Nuclear Magnetic Resonance Studies of the ...
    Crystal Structures and Nuclear Magnetic Resonance Studies of the Apo Form of the c-MYC:MAX bHLHZip Complex Reveal a Helical Basic Region in the Absence of DNA.Missing: bHLH- | Show results with:bHLH-
  22. [22]
    Crystal Structures and Nuclear Magnetic Resonance Studies of the ...
    Jul 1, 2019 · The N-terminal domain contains the transcription activation domain (TAD) and two highly conserved sequence elements, known as “MYC boxes” (MBI ...Materials and Methods · Results and Discussion · Conclusions · References
  23. [23]
    Control of Vertebrate Development by MYC - PMC - PubMed Central
    After organogenesis commences, MYCN is down-regulated in most if not all ... Expression of cellular oncogenes during embryonic and fetal development of the mouse.
  24. [24]
    myc maintains embryonic stem cell pluripotency and self-renewal
    Aug 5, 2010 · The three main myc family members are involved in fundamental normal cellular processes including proliferation, differentiation, and apoptosis ...
  25. [25]
    Manipulating Myc for reparative regeneration - PMC - PubMed Central
    N-myc expression is significantly downregulated in terminally differentiated adult tissues, whereas c-myc expression persists in some adult tissues such as ...<|separator|>
  26. [26]
    c-Myc Is Required for the Formation of Intestinal Crypts but ...
    c-myc is a known target of the Wnt pathway in intestinal cells (19, 38, 53) and has been shown to alter the expression of cell adhesion genes such as ...
  27. [27]
    Skin epidermis lacking the c-myc gene is resistant to Ras-driven ...
    In the mammalian skin epidermis, c-Myc is expressed in the basal layer and the hair bulb, as well as the bulge region correlating with the proliferative and ...
  28. [28]
    c-Myc controls the development of CD8αα TCRαβ intestinal ...
    Jun 3, 2010 · c-Myc controls the development of CD8αα TCRαβ IELs from thymic precursors by regulating interleukin-15 receptor expression and consequently Bcl-2–dependent ...
  29. [29]
    Making Sense of MYC in Skeletal Muscle: Location, Duration, and ...
    MYC levels are typically low or undetectable in healthy adult skeletal muscle tissue, but MYC transcript and protein is upregulated in muscle fiber nuclei ...
  30. [30]
    Low-level expression of Cmyc in mature neurons - PubMed
    Apr 29, 2025 · Cmyc , a proto-oncogene, is expressed at extremely low levels in mature neurons and is traditionally thought to have no function in these cells.
  31. [31]
    Tissue expression of MYC - Summary - The Human Protein Atlas
    A summary of the overall protein expression pattern across the analyzed normal tissues. ... Muscle tissues. Heart muscle · Skeletal muscle · Smooth muscle ...
  32. [32]
    c-myc protein in normal tissue. Effects of fixation on its ... - PMC - NIH
    The c-myc protein is thought to be a DNA-associated nuclear protein. However, immunohistochemical studies on normal or tumor tissues have shown conflicting ...
  33. [33]
    MYC Oncogene Contributions to Release of Cell Cycle Brakes - MDPI
    MYC has been reported to induce the expression of D-type cyclins and CDK4/6 by repressing these miRNAs. Altogether, MYC induces the formation of cyclin D ...Myc Oncogene Contributions... · 3. Myc And P21 Regulation · 4. Myc And P27 Regulation
  34. [34]
    Division of labour between Myc and G1 cyclins in cell cycle ... - Nature
    Sep 1, 2014 · The potent oncogene, Myc, dramatically affects E2F activity, presumably through modulating G1 cyclins expression as well as cyclin-dependent ...
  35. [35]
    Regulation of cancer cell metabolism: oncogenic MYC in the driver's ...
    Jul 10, 2020 · Even in a single tumor cell, MYC may simultaneously promote both GS and GLS expression, as these two reactions occur at different subcellular ...
  36. [36]
    From the editors - Nature Reviews Molecular Cell Biology
    **Summary of MYC's Role in Ribosome Biogenesis, RNA Pol I/III, and Protein Synthesis**
  37. [37]
    A Functional Screen for Myc-Responsive Genes Reveals Serine ...
    Rat-1 fibroblasts, rat c-myc-null fibroblasts (HO15.19 cells), HO15.19 cells ... The doubling time of the c-myc-null cells decreased from 47.1 ± 1.9 h ...
  38. [38]
    Deep Sequencing of MYC DNA-Binding Sites in Burkitt Lymphoma
    MYC is a transcription factor encoded by the c-MYC gene (thereafter termed MYC) which regulates an estimated 15% of genes in the human genome [1]. ... The typical ...
  39. [39]
    MYC recruits the TIP60 histone acetyltransferase complex to chromatin
    Here, we show that MYC associates with TIP60 and recruits it to chromatin in vivo with four other components of the TIP60 complex: TRRAP, p400, TIP48 and TIP49.
  40. [40]
    Tip60 complex binds to active Pol II promoters and a subset of ...
    Nov 6, 2015 · Our study suggests that the Tip60 complex functions as a global transcriptional co-activator at most active Pol II promoters, co-regulates the ESC-specific c- ...
  41. [41]
    c-Myc target gene specificity is determined by a post-DNAbinding ...
    First, it is possible that DNA-binding specificity between family members occurs at certain E box elements in vivo. Second, a post-DNA-binding mechanism may ...Missing: source | Show results with:source
  42. [42]
    Identification of CDK4 as a target of c-MYC - PMC - NIH
    Likewise, c-MYC and CDK4 genes can both immortalize primary cells (22, 23). Induction of CDK4 mRNA was detectable as early as 6 h after infection with Ad-Myc ...Missing: source | Show results with:source
  43. [43]
    Genome-wide mapping of Myc binding and gene regulation ... - Nature
    Aug 22, 2011 · The c-myc gene product, Myc, is a transcription factor that can either activate or repress gene expression. Activation occurs through ...
  44. [44]
    Regulation of MYC gene expression by aberrant Wnt/β-catenin ... - NIH
    Aberrant Wnt/β-catenin signaling drives MYC expression in colorectal cancer via β-catenin/TCF complexes binding to WREs, often through long-range chromatin ...
  45. [45]
    A β-catenin/TCF-coordinated chromatin loop at MYC integrates 5
    Dec 29, 2009 · We propose that a distinct chromatin architecture coordinated by β-catenin/TCF-bound WREs accompanies transcriptional activation of MYC gene expression.
  46. [46]
    NOTCH1 directly regulates c-MYC and activates a feed-forward-loop ...
    Using an integrative systems biology approach we show that NOTCH1 controls a feed-forward-loop transcriptional network that promotes cell growth.
  47. [47]
    Activation of PI3K/Akt and MAPK pathways regulates Myc ... - PNAS
    Our findings provide a novel mechanism by which the growth factor signaling pathways cooperate with Myc to promote proliferation and cellular transformation.
  48. [48]
    The MYC enhancer-ome: Long-range transcriptional regulation of ...
    Of note, we will focus on the enhancer regions of MYC identified in the 8q24 locus, not on the translocations that juxtapose MYC with other enhancer regions ...
  49. [49]
    Disruption of BRD4 at H3K27Ac-enriched Enhancer ... - PubMed - NIH
    May 5, 2015 · Here we show that an enhancer proximal to the c-Myc promoter is enriched in H3K27Ac and associated with high occupancy of BRD4, and coincides with a putative c ...
  50. [50]
    Regulation of Myc transcription by an enhancer cluster dedicated to ...
    May 10, 2024 · Here we use genomic rearrangements, transgenesis and targeted mutation to analyse Myc regulation in early mouse embryos and pluripotent stem cells.
  51. [51]
    DNA methylation: an epigenetic mark of cellular memory - Nature
    Apr 28, 2017 · DNA methylation is a stable epigenetic mark that can be inherited through multiple cell divisions. During development and cell differentiation, DNA methylation ...
  52. [52]
    Tumorigenicity of the miR-17-92 cluster distilled
    The miR-17-92 cluster is a direct transcriptional target of c-Myc. miR-17/20a targets E2F1, a regulator of the cell cycle and apoptosis. miR-92 targets BIM, a ...
  53. [53]
    Phosphorylation Regulates c-Myc's Oncogenic Activity in the ...
    Feb 1, 2011 · Phosphorylation of c-Myc at conserved residues serine 62 (S62) and threonine 58 (T58) can regulate c-Myc protein stability in response to ...
  54. [54]
    Alternative approaches to target Myc for cancer treatment - Nature
    Mar 10, 2021 · The deregulation of Myc occurs in >70% of human cancers, and is related to poor prognosis; hence, hyperactivated Myc oncoprotein has been ...
  55. [55]
    MYC Deregulation in Primary Human Cancers - MDPI
    May 25, 2017 · In cancer, the activity of the MYC transcriptional network is frequently deregulated, contributing to the initiation and maintenance of disease.
  56. [56]
    PRMT5 in MYC-Amplified Medulloblastoma - Encyclopedia.pub
    Jan 3, 2024 · MYC amplification or overexpression is most common in Group 3 medulloblastomas and is positively associated with poor clinical outcomes.<|separator|>
  57. [57]
    MYC gene amplification is often acquired in lethal distant breast ...
    Nov 4, 2011 · In breast cancer, amplification of MYC is consistently observed in aggressive forms of disease and correlates with poor prognosis and ...
  58. [58]
    Nuclear MYC protein overexpression is an early alteration in human ...
    Jun 20, 2008 · A region on chromosome 8q24 encompassing the MYC locus is amplified in prostate cancer, but this occurs mostly in advanced disease suggesting ...
  59. [59]
    MYC drives aggressive prostate cancer by disrupting transcriptional ...
    May 13, 2022 · Our findings suggest that MYC overexpression antagonizes the canonical AR transcriptional program and contributes to prostate tumor initiation and progression.<|separator|>
  60. [60]
    c-Myc hot spot mutations in lymphomas result in inefficient ...
    Our results show that proteasome-mediated turnover of c-Myc is substantially impaired in Burkitt's lymphoma cells with mutated Thr58 or other mutations that ...Abstract · Materials and methods · Results · Discussion
  61. [61]
    Tumorigenic conversion of primary embryo fibroblasts requires at ...
    Aug 18, 1983 · Transfection of embryo fibroblasts by a human ras oncogene does not convert them into tumour cells unless the fibroblasts are established ...
  62. [62]
    The E mu-myc transgenic mouse. A model for high ... - PubMed - NIH
    Feb 1, 1988 · Mice transgenic for a c-myc gene driven by the IgH enhancer (E mu-myc) were shown to almost invariably develop lymphomas, 90% succumbing in ...Missing: stromal | Show results with:stromal
  63. [63]
    Dendritic cell-mediated survival signals in Eμ-Myc B-cell lymphoma ...
    Sep 30, 2014 · In vitro, Eμ-Myc lymphoma B cells undergo rapid apoptosis in the absence of stromal support, a property shared with human acute lymphoblastic ...
  64. [64]
    Increased proto-oncogene expression in peripheral blood ... - PubMed
    T and B lymphocytes from these patients were found to have significantly increased expression of c-myc, c-myb, and c-raf RNA when compared with those of normal ...
  65. [65]
    MYC in Regulating Immunity: Metabolism and Beyond - PubMed - NIH
    Feb 25, 2017 · Emerging evidence suggests that MYC family members play essential roles in regulating the development, differentiation and activation of immune cells.
  66. [66]
    C-MYC induces idiopathic pulmonary fibrosis via modulation of miR ...
    Feb 3, 2022 · We sought to pinpoint the potential role of C-MYC in pulmonary fibroblast proliferation in idiopathic pulmonary fibrosis (IPF) and its mechanism ...
  67. [67]
    miR-210 promotes IPF fibroblast proliferation in response to hypoxia
    Aug 15, 2014 · Idiopathic pulmonary fibrosis (IPF) is characterized by the ... c-myc inhibitor, MNT. In situ analysis of IPF lung tissue demonstrates ...
  68. [68]
    Inducible activation of c-Myc in adult myocardium in vivo ... - PubMed
    Activation of Myc in adult myocardium was sufficient to reproduce the characteristic changes in myocyte size, protein synthesis, and cardiac-specific gene ...Missing: failure | Show results with:failure
  69. [69]
    Cell cycle re-entry and mitochondrial defects in myc ... - PubMed
    Our findings indicate that the induction of Myc in cardiomyocytes is sufficient to cause cardiomyopathy and heart failure.
  70. [70]
    Metabolic signaling in T cells | Cell Research - Nature
    Jul 24, 2020 · Myc mRNA and c-Myc protein levels in activated T cells are also ... This axis specifically is the target of the HIV Tat protein, which ...
  71. [71]
    Therapeutic targeting of “undruggable” MYC - PMC - PubMed Central
    KI-MS2-008 reduces the expression of MYC target genes and cancer cell growth in MYC-dependent cell lines and suppresses tumor growth in mouse models of T-cell ...Missing: 062 | Show results with:062
  72. [72]
    MYC: a multipurpose oncogene with prognostic and therapeutic ...
    Aug 9, 2021 · MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and ...
  73. [73]
    Selective Inhibition of Tumor Oncogenes by Disruption of Super ...
    Apr 11, 2013 · Treatment of MM tumor cells with the BET-bromodomain inhibitor JQ1 led to preferential loss of BRD4 at super-enhancers and consequent ...
  74. [74]
    Selective targeting of MYC mRNA by stabilized antisense ...
    MYCASOs are stabilized antisense oligonucleotides designed to target MYC mRNA, using LNA bases, phosphorothioate backbone, and internal DNA bases.Missing: 412 | Show results with:412<|control11|><|separator|>
  75. [75]
    RNAi-Mediated Silencing of Myc Transcription Inhibits Stem-like Cell ...
    This important study offers a preclinical proof of concept to target Myc function in cancer stem-like cells as a general strategy to attack most if not.Abstract · Introduction · Results · Discussion<|control11|><|separator|>
  76. [76]
    MYC-Targeting PROTACs Lead to Bimodal Degradation and N ...
    Mar 27, 2025 · Our studies suggest that PROTACs can promote unconventional events when bringing the ubiquitin–proteasomal–system machinery together with ...Missing: enhancement | Show results with:enhancement
  77. [77]
    The role of cytogenetics and molecular diagnostics in the diagnosis ...
    High-level amplification of MYC (8q24) is a consistent hallmark of ... The current risk stratification scheme used by the Children's Oncology Group (COG) ...
  78. [78]
    MYC/BCL2 protein coexpression contributes to the inferior survival ...
    May 16, 2013 · DLBCL patients with MYC/BCL2 coexpression demonstrate inferior prognosis and high-risk gene expression signatures. Diffuse large B-cell lymphoma ...
  79. [79]
    Research progress on non-protein-targeted drugs for cancer therapy
    Mar 14, 2023 · This paper summarizes the research progress of the non-protein target drugs, mainly RNA-based drugs in cancer treatment in recent years.
  80. [80]
    MYC targeting by OMO-103 in solid tumors: a phase 1 trial - PMC
    In this first-in-human phase 1 dose-escalation trial of OMO-103, a MYC inhibitor, in patients with solid tumors, treatment was safe and showed preliminary ...Missing: antisense | Show results with:antisense
  81. [81]
    Combinatorial immunotherapies overcome MYC-driven immune ...
    Jun 27, 2022 · MYC-elevated tumors are less sensitive to anti-PD-L1 in preclinical models. To understand how MYC might alter response to immune checkpoint ...
  82. [82]
    MYC and therapy resistance in cancer: risks and opportunities - PMC
    An example of bypass resistance is the activation of PI3K signaling by amplification of the MET oncogene in lung cancer resistant to EGFR inhibitor [57].