Fact-checked by Grok 2 weeks ago

Myc

The Myc family of proto-oncogenes encodes a group of basic helix-loop-helix (bHLH-LZ) transcription factors that function as master regulators of , controlling essential cellular processes such as , , , and . These proteins, including the primary members c-Myc (encoded by MYC), N-Myc (encoded by MYCN), and L-Myc (encoded by MYCL), heterodimerize with the partner protein Max to bind DNA sequences (CANNTG) and activate transcription of thousands of target genes, amplifying overall transcriptional output in a context-dependent manner. In normal , Myc proteins are tightly regulated and play critical roles in embryonic , tissue , and immune responses, with expression levels fluctuating in response to mitogenic signals. Myc deregulation, through diverse mechanisms, occurs in over 70% of human cancers. Dysregulation of genes, often through genomic , chromosomal translocations, or upstream signaling alterations, transforms them into potent s that drive tumorigenesis across a wide spectrum of human cancers. For instance, of family genes occurs in approximately 28% of tumors analyzed in , while MYC translocation—such as the t(8;14) rearrangement in —is a hallmark of certain lymphomas, promoting including sustained proliferation, evasion of , metabolic reprogramming, and immune suppression via upregulation of checkpoints like PD-L1. N-Myc is particularly prevalent in aggressive pediatric cancers like , where it correlates with poor prognosis, while L-Myc contributes to progression. The oncogenic potential of stems from its ability to override cellular safeguards, leading to oncogene addiction in tumors, where sustained high expression becomes essential for survival; consequently, therapeutic strategies targeting or its downstream pathways, including recent clinical trials for direct inhibitors as of 2025, hold promise for cancer intervention.

Discovery and Nomenclature

Initial Discovery

The avian myelocytomatosis virus strain MC29, responsible for inducing myelocytomas and other tumors in chickens, was first isolated in 1964 from a diseased bird in Bulgaria. In the mid-1970s, researchers began characterizing the transforming potential of MC29, identifying its oncogene v-myc in 1977 through biochemical analyses that revealed specific protein products and nucleic acid sequences unique to the virus. This discovery highlighted v-myc as a key driver of the virus's broad tumorigenic effects across various cell types, including myeloid and non-myeloid lineages. Building on the general proto-oncogene concept established by J. Michael Bishop and Harold E. Varmus in 1976 using the src oncogene of Rous sarcoma virus—which earned them the 1989 Nobel Prize in Physiology or Medicine—the cellular homolog of v-myc, termed c-Myc, was identified in 1979 when nucleotide sequences related to the viral gene were detected in DNA and RNA from uninfected vertebrate cells, including those from chickens and humans. This finding linked c-Myc to normal cellular processes while suggesting its potential for oncogenic activation. In 1982, the human c-Myc gene was cloned and characterized, revealing its location on chromosome 8 and its involvement in reciprocal translocations with immunoglobulin loci in Burkitt lymphoma cells, such as t(8;14). Early experiments in the further connected Myc to cell transformation; for instance, transfection of c-Myc into primary rat embryo fibroblasts in 1983 showed it could cooperate with activated to induce full tumorigenic conversion, including immortalization and anchorage-independent growth, underscoring its proto-oncogenic role. These assays established that Myc overexpression alone promotes limited but requires additional factors for complete .

Myc Family and Isoforms

The Myc gene family comprises three primary members in humans: c-Myc (encoded by the MYC gene), N-Myc (encoded by MYCN, first identified in 1983 through amplification in tumors), and L-Myc (encoded by MYCL, discovered in 1986 in cell lines). These proteins are all members of the basic helix-loop-helix (bHLH-LZ) family of transcription factors, sharing structural and functional similarities that enable them to regulate in diverse cellular contexts. The genes are located at distinct chromosomal positions: MYC at 8q24.21, MYCN at 2p24.3, and MYCL at 1p34.2. Isoforms of these genes arise primarily through alternative promoter usage and splicing events, generating variants with potentially distinct regulatory roles. For c-Myc, transcription can initiate from multiple promoters (, P1, P2, and P3), leading to isoforms such as c-Myc1 and c-Myc2, which differ in their N-terminal extensions and sites. Additionally, the short isoform c-MycS, produced by that skips 2, lacks the first 100 of the N-terminal and is transiently expressed during periods of rapid . Similar mechanisms apply to N-Myc and L-Myc, though their isoform diversity is less extensively characterized; for instance, N-Myc variants influence developmental timing in neural tissues. Expression patterns vary across family members: c-Myc is ubiquitously expressed in proliferating cells throughout development and adulthood, whereas N-Myc shows tissue-restricted expression, predominantly in neural and neuroendocrine lineages during embryogenesis. L-Myc exhibits more limited expression, such as in and reproductive tissues. The family demonstrates strong evolutionary conservation, reflecting its fundamental role in cellular growth and proliferation across species. In , the single ortholog dMyc (encoded by the diminutive gene) shares key biochemical properties with vertebrate Myc proteins, including dimerization with Max homologs and regulation of ribosomal biogenesis genes, and mutations in dMyc affect organismal size and . Elements of the Myc-Max regulatory network trace back to premetazoan ancestors, with distant homologs and conserved motifs identifiable in unicellular eukaryotes like , underscoring the ancient origins of Myc-mediated transcription.

Molecular Structure

Gene Organization

The MYC gene, encoding the c-Myc proto-oncogene, is organized into three exons spanning approximately 7.5 kilobases (kb) of genomic DNA on chromosome 8q24.21, with the coding sequence of roughly 1.4 kb primarily distributed across exons 2 and 3, while exon 1 is non-coding and contributes to the 5' untranslated region. The promoter regions of include two major initiation sites, P1 and P2, located within exon 1 and separated by about 160 base pairs, enabling alternative transcription start sites that influence isoform production. The P2 promoter contains a essential for basal transcription and an binding site that modulates activity through cell cycle-dependent regulation. Additional regulatory elements include a CpG island in the promoter region, which remains hypomethylated to facilitate accessibility, and distal enhancers in the 8q24 locus that are responsive to complexes, such as those involving BPTF, to control transcriptional activation. This organization is conserved across the Myc family, with MYCN exhibiting a similar three- structure, including a non-coding 1 and comparable exon-intron boundaries that support analogous . The genomic context of at 8q24 contributes to its frequent amplification in cancers, as this region's structural features, including long-range enhancers and proximity to other oncogenes, promote copy number gains that drive overexpression.

Protein Domains and Architecture

The full-length c-Myc protein comprises 439 and has a calculated molecular weight of approximately 49 . This features a modular architecture with distinct domains that facilitate its roles in . The N-terminal region contains the (TAD), spanning residues 1–143, which is rich in acidic and residues and interacts with components of the transcriptional machinery. Within the TAD lie the conserved Myc box regions, including Myc box I (MBI, residues 45–63) and Myc box II (MBII, residues 128–143), which are essential for protein stability and interactions with regulatory factors. The central portion of c-Myc includes less structured regions, while the C-terminal basic helix-loop-helix leucine zipper (bHLH-LZ) domain (residues 353–439) mediates heterodimerization with Max and specific DNA binding to E-box motifs (CANNTG). The bHLH subdomain provides DNA contact through a basic region, and the HLH-LZ facilitates dimerization via helical structures. Structural insights from X-ray crystallography of the c-Myc:Max heterodimer bound to DNA reveal that the bHLH-LZ domain adopts an extended alpha-helical conformation, with the basic region inserting into the DNA major groove for sequence-specific recognition. Complementary nuclear magnetic resonance (NMR) studies of the isolated bHLH-LZ domain highlight its intrinsic disorder in the monomeric state, transitioning to ordered helices upon dimerization. These structural features underscore the domain's role in enabling precise transcriptional control. In the Myc family, isoforms exhibit variations in domain architecture; for instance, N-Myc possesses an extended TAD with additional acidic sequences beyond residue 143, enhancing its potential in neural contexts. Conversely, L-Myc lacks the conserved MBI region, resulting in a more divergent N-terminal and altered compared to c-Myc and N-Myc.

Post-Translational Variants

Post-translational modifications play a critical role in regulating the stability, activity, and function of the protein, with the N-terminal region, including Myc box I (MBI), serving as a primary site for many of these alterations. at specific residues within MBI, such as Ser62 and Thr58, is mediated by s including extracellular signal-regulated (ERK) and kinase 3β (GSK3β). at Ser62 by ERK promotes Myc stabilization by inhibiting ubiquitin-mediated , while subsequent dephosphorylation of Ser62 followed by Thr58 by GSK3β generates a phosphodegron that facilitates binding to the ubiquitin FBW7, leading to proteasomal via the ubiquitin-proteasome pathway. Ubiquitination and sumoylation further fine-tune Myc's short , typically 20-30 minutes in cycling cells, by targeting it for and modulating its transcriptional output. Polyubiquitination occurs primarily on residues following Thr58 , with FBW7 acting as the key to promote rapid turnover through the 26S ; additional ubiquitination by HUWE1 supports Myc's role in transcriptional elongation. Sumoylation, facilitated by the E2-conjugating enzyme UBC9 and E3 PIAS1, conjugates small ubiquitin-like modifier () proteins to Myc, enhancing its while also influencing RNA polymerase I transcription and preventing inhibitory sumoylation of CDK9 to boost P-TEFb activity. Acetylation represents another key modification that enhances 's transcriptional activity. The coactivators p300 and CBP acetylate at multiple residues, particularly in the C-terminal region, which promotes an open conformation, stabilizes Myc-Max heterodimers, and recruits additional transcriptional machinery to promoters. A distinct post-translational variant arises from proteolytic cleavage under cellular stress conditions, such as nutrient deprivation or , producing Myc-nick, an approximately 40 kDa N-terminal fragment (residues 1-298) generated by calpain-mediated at a site between Myc boxes III and IV. Lacking the C-terminal nuclear localization signal and basic helix-loop-helix domain, Myc-nick localizes to the and exhibits independent functions distinct from full-length , including induction of α-tubulin acetylation via recruitment of GCN5 to , promotion of through fascin expression and Cdc42 activation, and attenuation of to enhance survival during stress or exposure.

Biological Functions

Transcriptional Regulation

Myc functions primarily as a that regulates through its ability to form heterodimers with Max, enabling sequence-specific binding to motifs (5'-CACGTG-3') in the promoter and enhancer regions of target genes. This binding is facilitated by the basic helix-loop-helix (bHLH-LZ) domain of Myc, which allows the heterodimer to recognize and interact with DNA. Genome-wide (ChIP) analyses, including ChIP-chip studies, have revealed that Myc-Max complexes occupy the regulatory elements of approximately 15% of genes in certain cell types, such as Burkitt's lymphoma cells, highlighting Myc's broad influence on the . Myc exhibits a dual role in , acting as both an activator and repressor. In its activating capacity, Myc recruits complexes, such as TIP60 and GCN5, to target sites, promoting and H4 to open structure. Additionally, Myc facilitates the recruitment and phosphorylation of (Pol II), enhancing promoter-proximal pausing release and elongation of transcription. For repression, Myc interacts with the Miz-1 at initiator elements of specific target genes, displacing coactivators and thereby inhibiting transcription of genes involved in growth arrest, such as CDKN1A (encoding p21). ChIP-seq studies further demonstrate that Myc preferentially targets super-enhancers—clusters of enhancers marked by high levels of histone acetylation and occupancy—amplifying the expression of genes critical for cell identity and . Quantitatively, in cells with elevated levels, such as tumor cells, Myc acts as a transcriptional , increasing output from already active promoters by an average of 2- to 4-fold without substantially activating silent genes. Recent studies as of have refined this model, highlighting Myc's additional roles in enhancer rewiring and increasing transcription burst duration to orchestrate context-dependent gene programs. This amplification model underscores Myc's role in fine-tuning global transcription rather than solely initiating new programs.

Control of Cellular Metabolism and Proliferation

Myc exerts profound control over cellular metabolism and proliferation primarily through its role as a transcriptional regulator, activating a network of target genes that coordinate biosynthetic processes essential for cell growth. By binding to E-box sequences in promoter regions, Myc drives the expression of genes involved in nucleotide, lipid, and protein synthesis, thereby linking metabolic flux to proliferative capacity. This orchestration ensures that rapidly dividing cells maintain adequate biomass accumulation while avoiding metabolic bottlenecks. A central mechanism by which promotes is the upregulation of ribosomal biogenesis, which supports increased protein synthesis demands. Myc activates transcription of (rDNA) by , enhancing nucleolar function and rRNA production, while simultaneously inducing ribosomal protein genes through II-dependent mechanisms. For instance, Myc directly binds to promoters of nucleolar proteins such as nucleolin and fibrillarin, facilitating nucleolar and assembly. This coordinated regulation is critical for sustaining high translational rates in proliferating cells, as evidenced in studies showing that Myc depletion impairs rDNA transcription and reduces nucleolar size. Myc also reprograms cellular to favor anabolic pathways, redirecting nutrients toward production and generation. It induces aerobic , known as the Warburg effect, by transcriptionally activating key enzymes such as hexokinase 2 (HK2) and (LDHA), which accelerate glucose uptake and conversion to even in oxygen-rich conditions. Concurrently, enhances to support and ; it upregulates the transporter SLC1A5 to increase uptake and glutaminase (GLS) to promote glutaminolysis, thereby replenishing cycle intermediates for biosynthetic needs. These shifts enable proliferating cells to generate precursors for macromolecules, with serving as a versatile nitrogen and carbon donor. In terms of cell cycle progression, facilitates the by activating and CDK4 expression, which phosphorylate the () to release transcription factors and drive . This promotes continuous cycling and suppresses programs, preventing cells from exiting the proliferative state; for example, represses genes like p15^INK4B^ and p21^CIP1^ that enforce quiescence or . 's influence extends to balancing , where it sensitizes cells to death via upregulation of ARF, which inhibits and stabilizes to trigger pro-apoptotic pathways. However, in certain contexts, can promote survival by repressing targets or activating anti-apoptotic genes, maintaining a delicate that favors over when nutrients are abundant.

Regulation and Interactions

Upstream Regulatory Pathways

The expression of Myc is tightly controlled by several upstream signaling pathways that integrate extracellular cues to regulate its transcriptional activation. The Wnt/β-catenin pathway induces Myc transcription through the binding of β-catenin/TCF complexes to specific Wnt-responsive elements (WREs) in the Myc promoter and upstream enhancers, promoting in contexts such as intestinal and embryonic development. Similarly, the directly activates Myc expression via Notch intracellular domain (NICD)-mediated recruitment of co-activators to the Myc promoter, as demonstrated in T-cell development and leukemogenesis models. The Hedgehog pathway, particularly through Sonic Hedgehog (Shh), upregulates Myc (including N-Myc isoforms) by Gli transcription factors binding to the Myc locus, driving neural progenitor proliferation during cerebellar development. Growth factor signaling pathways further modulate at post-transcriptional levels to enhance its stability and activity. The PI3K/AKT pathway stabilizes protein by inhibiting GSK3β, preventing phosphorylation at threonine 58 and subsequent degradation, thereby amplifying levels in response to mitogenic stimuli like insulin or IGF-1. In parallel, the MAPK/ERK cascade phosphorylates protein at serine 62 (S62), which prevents its ubiquitination and degradation while promoting its transcriptional activity, as observed in Ras-driven proliferative responses. Myc expression is also governed by intricate feedback loops that maintain . Myc undergoes negative autoregulation by binding to its own promoter as a Myc-Max complex, recruiting repressors to suppress further transcription and prevent excessive accumulation. Additionally, the /ARF pathway provides a suppressive feedback mechanism, where ARF sequesters Myc to inhibit its transactivation potential, and directly represses Myc promoter activity, counteracting Myc-induced hyperproliferation. Epigenetic modifications at the locus fine-tune its accessibility to transcription factors. , the catalytic subunit of the Polycomb Repressive Complex 2 (PRC2), deposits repressive marks near the promoter in certain cellular contexts, limiting expression to prevent uncontrolled growth, although this regulation can be context-dependent in and .

Protein Interaction Networks

The protein engages in a of interactions that modulate its transcriptional activity, , and cellular functions. Central to this network is the heterodimerization of with Max, which enables sequence-specific DNA binding to motifs (CACGTG) and subsequent activation. This interaction occurs via the basic helix-loop-helix (bHLH-LZ) domain in the of both proteins, forming a stable complex essential for 's role as a transcriptional amplifier. In contrast, the Mad and Mxi1 proteins compete with for Max binding, forming or Mxi1/Max heterodimers that repress transcription at similar sites by recruiting complexes, thereby antagonizing 's activating effects during processes like . Myc recruits co-activator complexes to enhance accessibility and transcription elongation. A key example is the interaction with the TRRAP-containing STAGA complex, which includes the GCN5 ; Myc's N-terminal (TAD) binds directly to both TRRAP and GCN5, facilitating and H4 at target promoters to promote . This recruitment is critical for Myc's transformative potential, as disruption of the Myc-TRRAP interaction impairs oncogenic activity. Additional interactors link Myc to cell cycle and mitotic regulation. Myc binds p107, a pocket protein related to Rb, through its N-terminal domain, leading to inhibition of Myc's transactivation and modulation of cell cycle progression. Similarly, Aurora B kinase interacts with and phosphorylates Myc at serine 67, stabilizing the protein and promoting its accumulation during mitosis, which supports Myc-driven proliferation in cancer cells. Recent proteomics studies have mapped Myc's interactome, identifying approximately 300-400 high-confidence binding partners through approaches like BioID proximity labeling and affinity purification-mass spectrometry, revealing context-specific hubs such as those involved in RNA polymerase recruitment and chromatin remodeling. These networks highlight Myc's role as a hub integrator, with interactions often mediated by its N-terminal domains for co-activator binding and the C-terminal bHLH-LZ for dimerization partners. Emerging findings also implicate Myc's interactome in regulating immune checkpoint expression, including influencers of PD-L1 transcription via co-activator complexes, underscoring its broader impact on tumor-immune dynamics.

Roles in Development and Stem Cells

Involvement in Embryonic Development

The Myc family of transcription factors plays critical roles in embryonic development, with distinct members contributing to proliferation, organogenesis, and patterning across species. More recently, Myc has been shown to be essential for immediate embryonic genome activation (iEGA) in one-cell stage mouse embryos, where its inhibition leads to developmental arrest by failing to activate approximately 95% of upregulated genes. In mice, homozygous null mutation of c-Myc results in embryonic lethality between embryonic days 9.5 and 10.5 (E9.5-E10.5), characterized by reduced embryo size, delayed development, and widespread cellular proliferation defects that impair gastrulation and extraembryonic tissue formation. These phenotypes arise from c-Myc's essential function in promoting cell cycle progression and metabolic adaptations necessary for rapid embryonic growth, with heterozygous females exhibiting reduced fertility due to preimplantation embryonic resorption in approximately 14% of cases. N-Myc exhibits more specialized functions during mid-gestation , with homozygous knockout mice succumbing around E11.5 and displaying severe defects in multiple s, including failure of neural tube closure leading to exencephaly and open neural folds in the . These mutants also show impaired heart looping, reduced branching, and abnormal liver and gut , underscoring N-Myc's role in coordinating progenitor expansion and specification during embryogenesis. In contrast, L-Myc expression is prominent in developing epithelium and skin structures such as hair follicles during late embryogenesis, suggesting contributions to epithelial and differentiation, though homozygous null mice are viable and fertile with no overt morphological abnormalities. Myc family members further influence embryonic patterning, particularly in limb bud , where conditional inactivation of N-Myc in mice leads to uniformly smaller skeletal elements and due to diminished mesenchymal proliferation in the early limb bud. This indicates N-Myc's involvement in scaling limb structures through integration with signaling pathways that regulate outgrowth and digit formation. Comparative studies in reveal conserved functions, as morpholino-mediated knockdown of the mych (c-myc homolog) gene produces embryos with shortened body length, fewer somites, and craniofacial defects, reflecting impaired axial elongation and neural crest-derived tissue .

Maintenance and Reprogramming of Stem Cells

In embryonic () cells, c- plays an essential role in maintaining self-renewal and pluripotency by regulating a distinct transcriptional module that supports , , and protein synthesis, thereby enabling the core pluripotency network involving Oct4 and Nanog. Specifically, c- interacts with the NuA4 complex to promote histone acetylation at target genes, which is critical for ES cell identity; its knockdown leads to loss of self-renewal and morphological changes indicative of . Although c-'s targets show minimal direct overlap with Oct4 and Nanog binding sites, its activity sustains the metabolic demands required for the pluripotency network's function. Endogenous Myc genes are indispensable for these processes, as their deletion impairs pluripotency markers and self-renewal capacity in cells. A landmark discovery in stem cell reprogramming involved c-Myc as one of four transcription factors—Oct3/4, Sox2, Klf4, and c-Myc—collectively known as the Yamanaka factors, which enable the conversion of somatic cells into induced pluripotent stem (iPS) cells. In 2006, Takahashi and Yamanaka demonstrated that retroviral introduction of these factors into mouse embryonic or adult fibroblasts generated iPS cells at low efficiency (about 0.01-0.1% of transduced cells forming colonies), which expressed ES cell markers like Oct3/4 and Nanog, formed teratomas with derivatives of all three germ layers, and contributed to chimeric mice with germline transmission. c-Myc enhances reprogramming efficiency by driving proliferative and metabolic changes that facilitate epigenetic resetting, though its omission reduces but does not abolish iPS generation. Myc expression exhibits heterogeneity across stem cell states, with higher levels in proliferative populations and lower levels in quiescent ones, reflecting its role in balancing self-renewal and . In hematopoietic stem cells (s), c-Myc is lowly expressed in quiescent long-term HSCs but upregulated (approximately 2.3-fold) in short-term HSCs and multipotent progenitors during and . Conditional c-Myc deletion leads to HSC accumulation due to blocked without altering quiescence or rates, underscoring its promotion of lineage commitment over self-renewal maintenance. Similarly, in neural and cells, Myc inhibition induces a quiescent state, while elevated Myc drives exit from quiescence to support active self-renewal.

Pathological Roles

Oncogenic Mechanisms

Myc exerts oncogenic effects primarily through its aberrant activation, which disrupts normal cellular and promotes tumorigenesis via multiple interconnected pathways. A key aspect of Myc's oncogenic behavior is governed by a of expression levels, where low-level suffices to drive ectopic proliferation and cellular growth without immediate toxicity, while higher thresholds enable more aggressive outputs such as evasion of and induction of genomic instability. This dosage-dependent mechanism ensures that Myc amplification beyond physiological levels tips the balance toward neoplastic transformation, as demonstrated in genetic models where endogenous Myc expression exceeding a critical threshold initiates tumorigenesis. In addition to proliferative signaling, reprograms cellular to support the bioenergetic demands of rapidly dividing tumor cells, prominently amplifying the Warburg effect through upregulation of enzymes and glucose transporters. This shift toward aerobic not only provides biosynthetic precursors for tumor growth but also creates an acidic microenvironment that favors and . By directly transactivating genes involved in production and , sustains this metabolic hallmark, distinguishing it from normal cells and enabling adaptation to hypoxic tumor conditions. Myc further contributes to oncogenesis by facilitating immune evasion, particularly through suppression of machinery, including downregulation of class I (MHC-I) molecules on tumor cell surfaces. This reduction impairs recognition and cytotoxic killing by + T cells, allowing Myc-driven tumors to escape adaptive immune surveillance. Mechanisms such as the Myc-SUMO axis have been implicated in this process, where post-translational modifications stabilize repressors of MHC-I expression, thereby enhancing tumor . Recent studies from 2024-2025 have positioned as a " of its own ," wherein it not only induces transcriptional and replicative to fuel tumor progression but also mitigates these stresses to preserve cellular viability and intratumoral heterogeneity. This dual role allows to maintain a dynamic, adaptable tumor resilient to therapeutic pressures, underscoring its in sustaining oncogenic without catastrophic collapse.

Genetic Alterations Including Rearrangements

Genetic alterations in the family genes, including amplifications, chromosomal rearrangements, and point mutations, contribute to activation across multiple cancer types, with c-MYC at the 8q24 locus being the most frequently affected. These changes often lead to overexpression or stabilization of Myc proteins, driving tumorigenesis in diverse malignancies. Amplifications involving the 8q24 region, which harbors the c-MYC gene, are prevalent in solid tumors such as and cancers, occurring in approximately 20-50% of cases depending on stage and subtype. In , 8q24 gains have been documented in up to 48% of primary tumors, correlating with aggressive disease features. Similarly, in , c-MYC amplification is observed in a substantial proportion of primary lesions and rises to about 37% in metastatic disease, underscoring its role in progression. Amplification of MYCN, located on 2p24, is a defining genetic event in , present in 20-30% of cases and strongly linked to adverse outcomes, including overall survival rates below 50%. Chromosomal rearrangements, particularly translocations, are characteristic of hematologic malignancies and involve juxtaposition of Myc genes to immunoglobulin loci or other enhancers. The t(8;14)(q24;q32) translocation, fusing c-MYC to the immunoglobulin heavy chain (IGH) locus, occurs in roughly 75% of Burkitt lymphomas, resulting in constitutive Myc activation under immunoglobulin regulatory control. Recent analyses from 2023-2025 have further elucidated MYC rearrangements in diffuse large B-cell lymphoma (DLBCL) subtypes, where non-immunoglobulin partner genes facilitate enhancer hijacking by super-enhancers, promoting aberrant expression in 10-15% of aggressive cases. Point mutations in genes are relatively uncommon compared to copy number changes but can profoundly impact protein stability. A prototypical example is the T58A substitution in c-MYC, which disrupts phosphorylation-mediated degradation and has been detected in (T-ALL), enhancing leukemogenic potential in preclinical models.

Clinical and Therapeutic Aspects

Diagnostic and Prognostic Applications

In clinical diagnostics, (IHC) and (FISH) assays are essential for detecting Myc alterations to inform staging and prognosis. In , high c-Myc nuclear staining by IHC, often indicating overexpression, correlates with aggressive disease features such as larger tumor size, nodal metastasis, and reduced survival rates. further identifies , which is present in up to 16% of cases and independently predicts poor outcomes, particularly in hormone receptor-negative tumors. Prognostic biomarkers involving are integrated into staging systems for specific malignancies. In , MYCN amplification detected by is a critical in the International Neuroblastoma Staging System (INSS), classifying amplified tumors—even in low-stage disease—as high-risk with significantly lower event-free survival (approximately 30-50% at 5 years) and overall survival compared to non-amplified cases. Additionally, Myc scores from liquid biopsies, including or plasma c-Myc levels, provide non-invasive monitoring; elevated levels in patients are associated with advanced stages, involvement, and inferior . Myc-high expression strongly correlates with aggressive cancer subtypes. In triple-negative breast cancer, MYC amplification and overexpression occur at higher rates (up to 40%) than in other subtypes, driving immune evasion, metabolic reprogramming, and poorer with reduced relapse-free . Similarly, in , Myc overexpression by IHC identifies an aggressive variant, particularly the activated B-cell subtype, linked to treatment resistance and 5-year overall rates below 50%. Recent developments have incorporated (AI) to enhance Myc analysis in . AI-driven models analyze hematoxylin and eosin-stained whole-slide images to predict rearrangements in with an AUC of 0.81, enabling faster risk stratification without routine FISH and improving prognostic precision in resource-limited settings.

Emerging Therapeutic Strategies

Indirect inhibitors of , such as bromodomain inhibitors, target the bromodomain and extra-terminal () proteins like to disrupt Myc enhancer activity and reduce its transcriptional output in cancer cells. Compounds like and OTX015 have demonstrated preclinical efficacy by downregulating Myc expression in models of pediatric sarcomas and , leading to antitumor effects. OTX015 and related have been evaluated in Phase I and II clinical trials for hematologic malignancies and solid tumors, showing partial responses in Myc-overexpressing lymphomas with manageable toxicity profiles. Direct targeting strategies aim to inhibit Myc-Max heterodimerization, a critical step for Myc's oncogenic activity. Stapled peptides, such as IDP-121, are designed to bind and disrupt the , reducing Myc stability and transcriptional activity in and other Myc-driven cancers; preclinical studies indicate potent inhibition of cell proliferation without significant off-target effects. Similarly, OMO-103, a stabilized mini-protein derived from the Omomyc inhibitor, directly blocks -Max binding; in a 2024 Phase I trial across advanced solid tumors, it achieved a 49% reduction in tumor volume at best response in responsive patients, with a favorable safety profile dominated by mild infusion reactions. Combination therapies enhance inhibition by addressing resistance mechanisms and synergizing with metabolic or degradative pathways. Recent 2025 studies on demonstrate that pairing Myc inhibitors like MYCi975 with metformin exploits impaired mitochondrial in Myc-addicted cells, resulting in enhanced tumor regression and reduced viability compared to monotherapy, due to metformin's amplification of bioenergetic stress. Proteolysis-targeting chimeras (PROTACs) offer a degradation-focused approach, recruiting ligases to ubiquitinate and degrade Myc; novel Myc-specific PROTACs have shown bimodal degradation kinetics in preclinical models, leading to sustained suppression of Myc levels and in cells. Therapeutic challenges in targeting include compensation by family members like N-Myc and L-Myc, which can restore oncogenic signaling upon single-family inhibition. Advances from 2024-2025 emphasize multi-dimensional strategies, such as biomarker-guided combinations that simultaneously target Myc isoforms and downstream pathways to mitigate redundancy and improve durability of response. Additionally, Myc-driven upregulation of promotes immune evasion, and emerging therapies modulating the Myc-PD-L1 axis—through Myc inhibitors that downregulate PD-L1 expression—enhance antitumor immunity when combined with checkpoint blockade, showing synergistic effects in preclinical immune-competent models.

Experimental Models

Animal Models of Myc Dysregulation

Animal models have been instrumental in elucidating the roles of dysregulation in tumorigenesis, particularly through transgenic and conditional approaches that allow spatial and temporal control of Myc expression or deletion. The Eμ-Myc transgenic model, developed in the 1980s, represents a seminal example where the c-Myc gene is overexpressed under the control of the enhancer (Eμ), leading to rapid development of pre-B cell lymphomas in nearly all mice, with 90% succumbing within five months. These tumors exhibit aggressive , thymic involvement, and frequent leukemic dissemination, recapitulating key features of human and providing a platform to study Myc-driven lymphomagenesis and potential therapeutic interventions. To address the embryonic lethality of global Myc knockout and enable tissue-specific studies, conditional knockout models using the Cre-loxP system have been widely adopted. In one such model, floxed c- alleles (c-Myc^{fl/fl}) are crossed with Mx1-Cre transgenic mice, allowing inducible deletion in the liver upon poly(I:C) administration; this revealed that c-Myc is dispensable for during regeneration but essential for regulating cell size and . Similar Cre-loxP strategies have demonstrated that c-Myc is essential for sustaining tumor maintenance in Myc-driven lymphomas without affecting normal B-cell development. For , N-Myc models often involve amplification or overexpression in neural crest-derived cells. A Cre-conditional N-Myc (LSL-MYCN) model crossed with Dbh-iCre drivers induces tumors in 76% of mice, originating from or adrenal glands with histopathological and genomic features akin to high-risk neuroblastoma, including 17q gain. Xenograft approaches using MYCN-amplified neuroblastoma cell lines implanted subcutaneously or orthotopically in immunodeficient mice further mimic amplification-driven tumor growth, vascularization, and metastasis, facilitating preclinical testing of targeted therapies. Orthotopic models of dysregulation in (HCC) typically employ hepatocyte-specific overexpression via tetracycline-inducible systems, such as LAP-tTA/tet-O-Myc mice, where doxycycline withdrawal triggers c-Myc activation, resulting in rapid and HCC formation within weeks, often with intrahepatic dissemination. Hydrodynamic tail-vein injection of plasmids encoding , combined with other oncogenes like , generates autochthonous tumors that preserve the liver microenvironment, enabling studies of Myc's cooperative oncogenic effects and tumor-stroma interactions. Recent advancements include humanized mouse models for MYC-rearranged lymphomas, such as those engrafting human hematopoietic stem cells transduced with MYC and BCL2 into immunodeficient strains, which develop "double-hit" lymphomas resembling aggressive human disease and support evaluation of immunotherapies like CAR-T cells. In 2023, extensions of these models demonstrated enhanced efficacy of CD20-targeted immunotherapy combined with BCL2 inhibitors in MYC/BCL2-coexpressing lymphomas, underscoring their utility for testing combination regimens in a human immune context.

In Vitro and Cellular Models

In vitro and cellular models provide controlled platforms to investigate 's roles in cellular , proliferation, and oncogenesis, enabling precise genetic manipulations and . Immortalized cell lines, such as Rat-1 , serve as foundational systems for assays, where deregulated Myc expression induces morphological changes, anchorage-independent growth, and loss of contact inhibition, recapitulating key hallmarks of oncogenic conversion. For example, of v-myc or rearranged c-myc oncogenes into Rat-1 cells results in focus formation and enhanced tumorigenic potential upon subcutaneous injection into nude mice, establishing Myc as a potent driver of independent of cooperating oncogenes in certain contexts. These assays have been widely adopted to evaluate Myc variants, including N-myc and L-myc, which similarly elicit neoplastic phenotypes in Rat-1 derivatives. Myc-null mouse embryonic fibroblasts (MEFs) complement these models by facilitating rescue experiments that delineate Myc-dependent pathways. Derived from conditional Myc knockout mice, these cells exhibit severely impaired proliferation and G1/S transition defects upon Myc ablation, which can be restored by re-expression of Myc or its downstream targets like cyclin D2 or E2F, underscoring Myc's essential role in cell cycle regulation. Such systems have enabled genetic screens to identify Myc collaborators, revealing that Myc hypomorphism or null status slows growth but can be partially rescued by genes promoting ribosomal biogenesis or metabolism, thus highlighting Myc's integration with core cellular processes. In cancer contexts, CRISPR/Cas9-mediated Myc knockout in established lines, such as lymphoma or prostate cancer cells, triggers replicative senescence characterized by β-galactosidase activity, p16^INK4a upregulation, and irreversible growth arrest, demonstrating Myc's necessity for immortalization and evasion of senescence barriers. These edited lines have been pivotal in high-throughput CRISPR screens for Myc synthetic lethals, identifying vulnerabilities like BET bromodomain inhibition that exacerbate senescence in Myc-dependent tumors. Three-dimensional organoid cultures offer advanced modeling of tissue-specific functions, particularly in epithelial cancers. In intestinal s derived from Apc-mutant mice or human adenomas, Myc overexpression drives expansion and progression through the adenoma-carcinoma sequence by enhancing Wnt/β-catenin signaling and translational output, leading to invasive structures with glandular disorganization and breaches. These Myc-driven s faithfully recapitulate histopathological features of colorectal tumorigenesis, including increased markers like , and support high-throughput drug screening for pathway inhibitors. More recently, in 2025, 3D models have emerged for studying Myc's regulation of cancer s (CSCs), where Myc inhibitors like SMYD3 antagonists disrupt self-renewal and formation in colorectal CSC-enriched cultures, reducing activity and tumor initiation potential without affecting bulk tumor cells. These s enable evaluation of Myc-targeted therapies in hypoxia-mimicking environments, revealing enhanced CSC sensitivity to indirect Myc suppression via epigenetic modulators. Overall, these cellular systems facilitate scalable, mechanistic insights into Myc dysregulation while bridging to more complex models.

References

  1. [1]
    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.Missing: sources | Show results with:sources
  2. [2]
    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.Missing: sources | Show results with:sources
  3. [3]
    The effects of MYC on tumor immunity and immunotherapy - Nature
    Mar 25, 2023 · MYC was the first proto-oncogene that was found to be amplified in tumor cells, and its overexpression is involved in tumor-related processes.
  4. [4]
    Myc family of cellular oncogenes - PubMed - NIH
    The myc family of cellular oncogenes contains three well-defined members: c-myc, N-myc and L-myc. Additional structural and functional evidence now suggests ...
  5. [5]
    Avian Leukosis Virus - an overview | ScienceDirect Topics
    Strain MC29 avian acute leukemia virus was isolated in 1964 in Bulgaria, and causes myelocytomatosis, endotheliomas, liver and kidney carcinomas, sarcomas, and ...
  6. [6]
    The Nobel Prize in Physiology or Medicine 1989 - Press release
    Michael Bishop and Harold Varmus used an oncogenic retrovirus to identify the growth-controlling oncogenes in normal cells. In 1976 they published the ...Missing: avian myelocytomatosis
  7. [7]
    Cloning and characterization of different human sequences ... - PNAS
    We have studied the genomic organization of human cellular sequences (c-myc) homologous to the transforming gene (v-myc) of avian myelocytomatosis virus (MC29).
  8. [8]
    Targeting oncogenic Myc as a strategy for cancer treatment - Nature
    Feb 23, 2018 · The MYC oncogene family consists of three members, C-MYC, MYCN, and MYCL, which encode c-Myc, N-Myc, and L-Myc, respectively. The Myc ...
  9. [9]
    MYC Deregulation in Primary Human Cancers - MDPI
    May 25, 2017 · The MYC gene exists within a “gene desert” region on chromosome 8q24, located within a 2 Mb stretch of the genome that is notable for the ...Myc Deregulation In Primary... · 2. Myc Amplification · 4. Myc Protein Stability<|control11|><|separator|>
  10. [10]
    Changes in MYCN expression in human neuroblastoma cell lines ...
    Apr 4, 2013 · The human MYCN gene is normally located on the short arm of chromosome 2 (2p24). The term 2p24 gain is suggested as a descriptive generic term ...
  11. [11]
    MYCL Gene - GeneCards | MYCL Protein | MYCL Antibody
    The DNA sequence and biological annotation of human chromosome 1. Nature Gregory SG … Prigmore E (PMID:16710414), Protein 4 , Gene 3, 2006, 82. Abstract: The ...
  12. [12]
    Isoform-Directed Control of c-Myc Functions - PubMed Central - NIH
    For instance, transcription can be initiated by binding to one of four alternative promoters (P0, P1, P2, and P3), the last of which is located between ...
  13. [13]
    Human c-Myc Isoforms Differentially Regulate Cell Growth and ...
    In particular, compared to c-Myc2, c-Myc1 harbors a short N-terminal extension while the c-MycS isoform lacks the first 100 amino acids (aa) of the ...
  14. [14]
    The N-myc Oncogene: Maximizing its Targets, Regulation, and ...
    This review outlines N-myc's contribution to normal development and oncogenic progression. In addition, it highlights relevant transcriptional targets and ...
  15. [15]
    MYC function and regulation in physiological perspective - Frontiers
    MYC, a key member of the Myc-proto-oncogene family, is a universal transcription amplifier that regulates almost every physiological process in a cell.
  16. [16]
    Drosophila Myc is oncogenic in mammalian cells and plays ... - PNAS
    The identification of a Myc family member in Drosophila melanogaster and the demonstration of conservation of its biochemical and biological properties ...
  17. [17]
    Premetazoan Ancestry of the Myc–Max Network - Oxford Academic
    May 13, 2011 · Human and mouse c-Myc and D. melanogaster dMyc are the best studied of the Myc homologs, and the conservation of function between these homologs ...
  18. [18]
    MYC (MYC proto-oncogene, bHLH transcription factor)
    Aug 1, 2017 · MYC is broadly expressed during embryogenesis and in tissue with high proliferative capacity such as skin epidermis and gut.<|control11|><|separator|>
  19. [19]
    The Analysis of Structure of c-Myc Gene Transcript
    The c-myc gene has four promoter sites designated as P0, P1, P2 and P3 (Figure 1). The P3 is located near 3' end of intron 1. They are independently regulated ...
  20. [20]
    The chromatin structure of the dual c-myc promoter P1/P2 ... - PubMed
    Jun 8, 2001 · ... E2F results in transcriptional repression without affecting polymerase recruitment, and the TATA box is required for polymerase recruitment.
  21. [21]
    The Chromatin Structure of the Dual c-myc Promoter P1/P2 Is ...
    In addition to the TATA box, two further elements have been identified required for P2 activity: a ME1a1 site at position −40 bp and a E2F-binding site at −58 ...
  22. [22]
    Nuclear factor E2F mediates basic transcription and trans-activation ...
    Abstract. Transcription from one of the two initiation sites, P1 and P2, of the dual human MYC promoter seems to be essential in all proliferating cells.
  23. [23]
    Insights into the Links between MYC and 3D Chromatin Structure ...
    Myelocytomatosis proto-oncogene (c-MYC) is one of the scarce protein-coding genes located in human chromosomal band 8q24, a region known as gene desert (1). The ...Chromatin Remodeling Through... · Ctcf Elements · Myc's Upstream Enhancers
  24. [24]
    BPTF is required for c-MYC transcriptional activity and in vivo ...
    Jan 5, 2016 · ... MYC requires BPTF to activate transcription at low-affinity targets genes. BPTF is required for c-MYC-induced chromatin remodelling. To ...
  25. [25]
    Entry - *164840 - MYCN PROTOONCOGENE, bHLH ... - OMIM
    Gene Structure. Stanton et al. (1986) determined that the MYCN gene is GC rich and contains 3 exons. Exon 1 is noncoding and contributes to a long 5-prime ...
  26. [26]
    The MYCN Oncogene - IntechOpen
    All three tumour-associated MYC genes have the same characteristic three-exon structure with the major polypeptide open reading frame residing in the second and ...
  27. [27]
  28. [28]
    Long-range enhancers on 8q24 regulate c-Myc - PNAS
    Jan 26, 2010 · It has been speculated that these 8q24 genetic variant(s) might affect Myc expression by altering its regulation or amplification status. Here, ...Missing: context | Show results with:context
  29. [29]
    Integrated Genomic Analysis of the 8q24 Amplification in ...
    Chromosome 8q24 is the most commonly amplified region across multiple cancer types, and the typical length of the amplification suggests that it may target ...
  30. [30]
    Chromosome 8q24 amplification associated with human ... - Nature
    Oct 18, 2024 · We predict that as carcinomas progress, chromosome 8q24 is amplified leading to high MYC levels that leads to ZEB1 expression and MIZ1 repression driving cells ...
  31. [31]
    myc Boxes, Which Are Conserved in myc Family Proteins, Are ... - NIH
    MBI and MBII in human c-myc are located between amino acids 45 and 65 and amino acids 128 to 144, respectively.Missing: MB | Show results with:MB
  32. [32]
    Crystal Structures and Nuclear Magnetic Resonance Studies of the ...
    Jul 1, 2019 · The only available crystal structure is that of a c-MYC:MAX complex artificially tethered by an engineered disulfide linker and prebound to DNA.
  33. [33]
    Mammalian MYC Proteins and Cancer - Tansey - Wiley Online Library
    Feb 2, 2014 · Indeed, in just a short period of time in the late 1970s the candidate v-myc gene was identified [13, 14], shown to be distinct from the src ...
  34. [34]
    The human myc gene family: structure and activity of L-myc and an L ...
    While the putative L-myc gene product is of a smaller size when compared to the c- and N-myc proteins, the relative positions of certain conserved residues ...Missing: differences | Show results with:differences
  35. [35]
  36. [36]
    An Overview of MYC and Its Interactome - PMC - PubMed Central
    The history of MYC parallels the discovery of other major oncogenes in ... c-myc (referred to here as MYC) (Roussel et al. 1979; Sheiness and Bishop ...
  37. [37]
    A global transcriptional regulatory role for c-Myc in Burkitt's ... - NIH
    We find that c-Myc together with its heterodimeric partner, Max, occupy >15% of gene promoters tested in these cancer cells. The DNA binding of c-Myc and Max ...
  38. [38]
    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.
  39. [39]
    TRRAP and GCN5 Are Used by c-Myc to Activate RNA Polymerase ...
    Activation of RNA polymerase (pol) II transcription by c-Myc generally involves recruitment of histone acetyltransferases and acetylation of histones H3 and H4.
  40. [40]
    Transcription-independent functions of MYC: regulation of ...
    Aug 13, 2008 · MYC promotes recruitment of the transcription factor TFIIH kinase to promoters and RNA polymerase (pol) II phosphorylation. Increased RNA pol II ...
  41. [41]
    Transcriptional Repression: The Dark Side of Myc - PMC
    Both factors stimulate transcription when bound to DNA in the absence of Myc, and binding of Myc interferes with transcriptional activation by both factors.
  42. [42]
    Super enhancer inhibitors suppress MYC driven transcriptional ...
    Apr 4, 2018 · Since it has been reported that MYC amplifies transcription by preferentially targeting super enhancer genes, we compared the MYC target sites ...
  43. [43]
    Transcriptional Amplification in Tumor Cells with Elevated c-Myc - NIH
    Sep 28, 2012 · c-Myc is a basic helix loop helix (bHLH) transcription factor that forms a heterodimer with Max and binds to E-box sequences near the core ...
  44. [44]
    Regulation of cancer cell metabolism: oncogenic MYC in the driver's ...
    Jul 10, 2020 · Oncogenic levels of MYC regulates almost every aspect of cellular metabolism, a recently revisited hallmark of cancer development.
  45. [45]
    MYC regulates ribosome biogenesis and mitochondrial gene ... - eLife
    Jan 8, 2021 · MYC regulates ribosome biogenesis through controlling and coordinating the transcription of ribosomal DNA, ribosomal protein genes, and ...
  46. [46]
    A role for c‐Myc in the regulation of ribosomal RNA processing
    c‐Myc, which is over‐expressed in many malignancies, induces ribosome biogenesis and protein synthesis by up‐regulating ribosomal and nucleolar proteins, ...
  47. [47]
    MYC-Induced Cancer Cell Energy Metabolism and Therapeutic ...
    Nov 2, 2009 · Myc and HIF-1 are depicted to regulate (dotted lines) genes involved in glucose metabolism (glucose transporter Glut1, HK2, PKM2, LDHA, and PDK1) ...
  48. [48]
    c-Myc suppression of miR-23a/b enhances mitochondrial ... - Nature
    Feb 15, 2009 · c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Ping Gao,; Irina Tchernyshyov, ...
  49. [49]
    Inhibition of cell differentiation: a critical mechanism for MYC ...
    Apr 15, 2009 · To explain the anti-differentiation effects of MYC, it has been argued that MYC impairs differentiation by preventing exit from the cell cycle.
  50. [50]
    Expression and activity of L-Myc in normal mouse development - PMC
    The lack of an apparent phenotype associated with L-Myc deficiency indicates that L-Myc is dispensable for gross morphological development and argues ...Missing: knockout | Show results with:knockout
  51. [51]
    The Mych Gene Is Required for Neural Crest Survival during ...
    We isolated the zebrafish mych (myc homologue) gene. Knockdown of mych leads to severe defects in craniofacial development and in certain other tissues ...
  52. [52]
  53. [53]
    myc maintains embryonic stem cell pluripotency and self-renewal
    Aug 5, 2010 · This study demonstrates that endogenous myc genes are essential for mESC pluripotency and self-renewal as well as providing the first evidence that myc genes ...
  54. [54]
  55. [55]
    c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation
    ### Summary: c-Myc Regulation of Hematopoietic Stem Cell Self-Renewal and Differentiation
  56. [56]
    c-Myc regulates neural stem cell quiescence and activation by ...
    Aug 25, 2021 · c-Myc regulates neural stem cell quiescence and activation by coordinating the cell cycle and mitochondrial remodeling.
  57. [57]
    Quiescence enables unrestricted cell fate in naive embryonic stem ...
    Feb 26, 2024 · Naive mouse embryonic stem cells (ESCs) can enter quiescence spontaneously or upon inhibition of MYC or fatty acid oxidation, mimicking ...<|separator|>
  58. [58]
    Myc inhibition is effective against glioma and reveals a role ... - Nature
    Aug 18, 2014 · Myc is a bHLHZip transcription factor, causally implicated in most human cancers. Myc controls diverse cellular functions, including cell growth ...
  59. [59]
    Distinct Thresholds Govern Myc's Biological Output In Vivo - Cell Press
    Here we show that distinct threshold levels of Myc govern its output in vivo: low levels of deregulated Myc are competent to drive ectopic proliferation of ...
  60. [60]
    A tumorigenesis threshold for endogenous Myc revealed by dosage ...
    Jul 31, 2025 · This "amplifier" model suggests that MYC must exceed an expression threshold to become oncogenic. We designed a genetic test of this model, ...
  61. [61]
    c-Myc and Cancer Metabolism - AACR Journals
    Oct 14, 2012 · Through the upregulation of these genes, c-Myc contributes directly to the Warburg effect (aerobic glycolysis) and the ability of transformed ...
  62. [62]
    Immune evasion: An imperative and consequence of MYC ...
    Indeed, downregulation of MHC‐I on the cell surface has been reported in a wide range of cancers and was shown to often correlate with a decreased number of ...
  63. [63]
    The Role of MYC in Tumor Immune Microenvironment Regulation
    Furthermore, MYC upregulates PD-L1 expression in tumors, directly inhibiting T cell activation and promoting immune evasion through PD-1/PD-L1 interactions [101] ...
  64. [64]
    MYC: The Guardian of Its Own Chaos - PMC - PubMed Central
    Jun 9, 2025 · MYC is an oncogene that drives tumorigenesis through enhanced transcription and replication. Paradoxically, MYC induces transcriptional and replicative stress.Missing: heterogeneity | Show results with:heterogeneity
  65. [65]
    MYC functions are specific in biological subtypes of breast cancer ...
    Mar 8, 2016 · High MYC and c-MYC expression was significantly associated with poor prognostic factors, including grade and basal-like BCs. In luminal A ...
  66. [66]
    MYC overexpression with its prognostic and clinicopathological ...
    Our results showed that MYC overexpression was associated with worse prognosis and high risk of breast cancer, especially in patients with negative hormone ...Missing: staining | Show results with:staining
  67. [67]
    MYC and Breast Cancer - PMC - PubMed Central - NIH
    Further studies have confirmed the correlation between MYC amplification and poor prognosis. In particular, MYC amplification appears to be a strong prognostic ...
  68. [68]
    Significance of MYCN Amplification in International Neuroblastoma ...
    Patients with MYCN-amplified, low-stage tumors had less favorable event-free survival (EFS) and overall survival (OS) than did patients with nonamplified tumors ...
  69. [69]
    Neuroblastoma Treatment (PDQ®) - NCI - National Cancer Institute
    Apr 28, 2025 · Infants younger than 12 months with INSS stage 4 disease and MYCN amplification are categorized as high risk and have a 5-year EFS rate of 37% ...
  70. [70]
    Plasma C-MYC Level Manifesting As An Indicator in Progression of ...
    Results: Plasma C-MYC level was significantly higher in patients with breast cancer than that in the controls, which associated with clinical stages, lymph node ...
  71. [71]
    Cell-free DNA detected by “liquid biopsy” as a potential prognostic ...
    This retrospective study evaluated the prognostic role of CF-DNA quantity and integrity of HER2, MYC, BCAS1 and PI3KCA, which are frequently altered in BC. We ...
  72. [72]
    MYC promotes immune-suppression in triple-negative breast cancer ...
    Nov 2, 2022 · Here we show a strong correlation between MYC expression and loss of immune signatures in human TNBC. In mouse models of TNBC proficient or ...
  73. [73]
    Indirect targeting of MYC and direct targeting in combination with ...
    MYC amplification is disproportionally elevated in triple-negative breast cancer (TNBC) compared to other subtypes of breast cancer. Indeed, MYC has long ...
  74. [74]
    MYC -driven aggressive B-cell lymphomas - Oncotarget
    Nov 17, 2015 · Virtually all lymphomas with MYC deregulation are of B-cell lineage and include Burkitt lymphoma (BL), diffuse large B-cell lymphoma (DLBCL), B- ...
  75. [75]
    Prognostic significances of overexpression MYC and/or BCL2 in R ...
    Apr 19, 2018 · MYC/BCL2 protein coexpression contributes to the inferior survival of activated B-cell subtype of diffuse large B-cell lymphoma and demonstrates ...
  76. [76]
    Artificial Intelligence, Lymphoid Neoplasms, and Prediction of MYC ...
    Apr 9, 2024 · A neural network predicted MYC, BCL2, and BCL6 with high accuracy using a pan-cancer panel of 758 genes of immuno-oncology and translational research.1. Introduction · 3. Results · 3.1. Neural Networks
  77. [77]
    BRD4 PROTAC Inhibits Basal-Like Breast Cancer via KLF5
    Aug 20, 2024 · To date, BRD4-targeted BET inhibitors (BETis) such as JQ1 and OTX015 are undergoing clinical trials. c-MYC is a well-known oncogenic ...
  78. [78]
    Inhibition of bromodomain and extra-terminal motif (BET) proteins in ...
    Oct 27, 2025 · BET inhibitors (BETi), especially those targeting BRD4, show promising preclinical activity against pediatric sarcomas by disrupting ...Keynote (green) · Introduction · Es
  79. [79]
    Bromodomain proteins as potential therapeutic targets for B-cell non ...
    Nov 23, 2024 · This study aimed to collect the most recent evidences and summarize possibility on whether BRD proteins can serve as therapeutic targets for B-NHL.
  80. [80]
    MYC as a Target for Cancer Treatment: from Undruggable to ...
    Aug 15, 2025 · Other anti-MYC inhibitors undergoing evaluation in clinical trials include the stapled peptide IDP-121 (IDP Pharma) which acts by disrupting MYC ...<|separator|>
  81. [81]
    Abstract 2471: IDP-121, a first in class staple peptide targeting c-MYC
    Jul 1, 2021 · Here we describe IDP-121, a stapled peptide specifically designed to target c-MYC protein. A library based on c, l, n-MYC and MAX druggable regions was ...
  82. [82]
    MYC targeting by OMO-103 in solid tumors: a phase 1 trial - Nature
    Feb 6, 2024 · Here we present results from a phase 1 study of OMO-103 in advanced solid tumors, established to examine safety and tolerability as primary outcomes and ...
  83. [83]
    MYC targeting by OMO-103 in solid tumors: a phase 1 trial - PubMed
    Feb 6, 2024 · Here we present results from a phase 1 study of OMO-103 in advanced ... tumors showed a 49% reduction in total tumor volume at best response.Missing: nanobody | Show results with:nanobody
  84. [84]
    Impaired mitochondrial metabolism is a critical cancer vulnerability ...
    Jul 16, 2025 · Our results suggest that well-tolerated agents, such as metformin, could be combined with MYC inhibitors to achieve maximal clinical efficacy.
  85. [85]
    MYC-Targeting PROTACs Lead to Bimodal Degradation and N ...
    Mar 27, 2025 · We rationalized that proteolysis targeting chimeras (PROTACs), which might accomplish the targeted degradation of MYC, would achieve more potent ...Introduction · Results and Discussion · Supporting Information · Author Information
  86. [86]
    Proteolysis‐targeting chimeras in cancer therapy - NIH
    Oct 18, 2025 · By inducing c‐MYC degradation, these PROTACs offer a promising strategy for many cancers in which c‐MYC plays a critical role in tumorigenesis ...
  87. [87]
    Rethinking MYC inhibition: a multi-dimensional approach to ...
    Apr 29, 2025 · In this clinical trial, OMO-103 demonstrated not only a robust safety profile but also encouraging pharmacokinetics, effectively achieving tumor ...
  88. [88]
    Beyond the Limit: MYC Mediates Tumor Immune Escape - MDPI
    In this study, we explore some novel potential mechanisms through which MYC facilitates the immune evasion of tumor cells, alongside a combined therapeutic ...
  89. [89]
    The E mu-myc transgenic mouse. A model for high ... - PubMed - NIH
    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 the first 5 mo of ...Missing: seminal paper
  90. [90]
    c-Myc regulates cell size and ploidy but is not essential for ... - PNAS
    Germ-line inactivation of c-myc leads to multiple abnormalities and death at day 9-10 of embryonic development (23). Here, we used an inducible conditional ...Missing: lethal | Show results with:lethal
  91. [91]
    A Cre-conditional MYCN-driven neuroblastoma mouse model as an ...
    Sep 1, 2014 · We here report on the generation of transgenic mice with Cre-conditional induction of MYCN in dopamine β-hydroxylase-expressing cells, termed LSL-MYCN;Dbh-iCre.Results · Jq1 Treatment Induces... · Human Neuroblastoma...Missing: sources | Show results with:sources
  92. [92]
    Mouse Models of Oncoimmunology in Hepatocellular Carcinoma
    For example, it is possible to induce MYC overexpression in the hepatocytes of LAP-tTA/tet-O-hMYC mice by removing doxycycline treatment from the drinking water ...
  93. [93]
    Hydrodynamic Transfection for Generation of Novel Mouse Models ...
    We review a novel approach for stable gene expression in mouse hepatocytes by hydrodynamic injection in combination with Sleeping Beauty–mediated somatic ...
  94. [94]
    Rapid generation of human B-cell lymphomas via combined ...
    Here, we describe the development of humanized mouse model of MYC/BCL2-driven “double-hit” lymphoma. By engrafting human hematopoietic stem cells transduced ...Missing: 2024 immunotherapy
  95. [95]
    Venetoclax improves CD20 immunotherapy in a mouse model of ...
    Feb 28, 2023 · Venetoclax treatment demonstrated specific killing of MYC + /BCL2 + lymphoma cells by licensing their intrinsically primed apoptosis.Flow Cytometry And Cell... · Rna Sequencing And Analysis · Myc/bcl2 Co-Expression Is...<|control11|><|separator|>
  96. [96]
    Neoplastic transformation by the human gene N-myc - PubMed
    Here we show that N-myc can also act alone to elicit neoplastic growth of an established line of rat fibroblasts (Rat-1). We used recombinant DNA vectors to ...
  97. [97]
    L-myc cooperates with ras to transform primary rat embryo fibroblasts
    We tested the biologic activity of L-myc by using an expression vector containing a cDNA clone coding for the major open reading frame in the 3.9-kilobase mRNA ...Missing: assay | Show results with:assay
  98. [98]
    A genetic screen to identify genes that rescue the slow growth ...
    In this study, we have attempted to identify genes that rescue the slow growth phenotype of c-myc null cells through introduction of a series of potent cell ...Missing: embryonic experiments key paper
  99. [99]
    Myc-enhanced expression of Cul1 promotes ubiquitin-dependent ...
    Most important, addition of in vitro synthesized Cul1 protein to the extracts from c-myc null MEFs or density-arrested. Myc–ER IMR90 cells rescued the ability ...
  100. [100]
    Cellular senescence is an important mechanism of tumor regression ...
    Aug 7, 2007 · Here we report that the suppression of the c-Myc (MYC) oncogene induces cellular senescence in diverse tumor types including lymphoma, osteosarcoma, and ...Missing: CRISPR | Show results with:CRISPR
  101. [101]
    [PDF] Identification of MYC synthetic lethal genes and networks - bioRxiv
    Apr 25, 2024 · Colorectal tumor cells with amplified MYC are more sensitive to SLC25A1 disruption by CRISPR. CRISPR screening and copy number data ...
  102. [102]
    PAF-Myc-Controlled Cell Stemness Is Required for Intestinal ...
    Mar 12, 2018 · In mouse models, PAF knockout inhibits Apc inactivation-driven intestinal tumorigenesis with reduced tumor cell stemness and suppressed Wnt/b- ...
  103. [103]
    Tailoring a novel colorectal cancer stem cell-targeted therapy by ...
    Jun 30, 2025 · Our findings identify SMYD3 as a promising therapeutic target acting directly on c-MYC, with potential implications for countering CRC-SC proliferation and ...
  104. [104]
    [PDF] RNA Methylation dynamics regulates cancer stemness through ...
    Jul 25, 2025 · RNA Methylation modulates Cancer Stem Cell Maintenance in 3D spheroidal cultures of TNBC: The role of m6A modification in stem cell fate ...