Abstract
MYC family of oncogenes (MYC, MYCN and MYCL), encodes a basic helix-loop-helix leucine zipper (bHLHLZ) transcriptional regulator that is responsible for moving the cell through the restriction-point. Through the HLHZIP domain, MYC heterodimerizes with the bHLHLZ protein MAX, which enables this MYC-MAX complex to bind to E-box regulatory DNA elements thereby controlling transcription of a large group of genes and their proteins. Translationally, MYC is one of the foremost oncogenic targets and deregulation of expression of MYC family gene/proteins occurs in over half of all human tumors and is recognized as a hallmark of cancer initiation and maintenance. Additionally, unexpected roles for this oncoprotein have been found in cancers that nominally have a non-MYC etiology. Although, MYC is rarely mutated its gain of function in cancer results from overexpression or from amplification. Moreover, MYC is a pleiotropic transcription factor possessing broad pathogenic prominence making it a coveted cancer target. A widely held notion within the biomedical research community is that the reliable modulation of MYC represents a tremendous therapeutic opportunity given its role in directly potentiating oncogenesis. However, the MYC-MAX heterodimer interaction contains a large surface area with a lack of well-defined binding sites creating the perception that targeting of MYC-MAX to be forbidding. Here we discuss the biochemistry behind MYC and MYC-MAX as it relates to cancer progression associated with these transcription factors. We also discuss the notion that MYC should no longer be regarded as undruggable providing examples that a therapeutic window is achievable despite global MYC inhibition.
Graphical Abstract:
INTRODUCTION
The MYC oncogene family encodes three proteins: c-MYC, N-MYC and L-MYC in mammalian cells that are differentially expressed in terms of tissue type and developmental stage.1, 2 While c-MYC is universally overexpressed in rapid proliferation cells, deregulation of N-MYC and L-MYC is restricted to a limited number of carcinomas.3, 4 As one of the most studied multifunctional transcription factors, it is implicated in a variety of physiological and pathological processes including: (1) cell proliferation, (2) differentiation, (3) apoptosis, (4) ribosome biogenesis, (5) protein translation, (6) orchestrating biological events like angiogenesis, (7) metabolism, (8) DNA repair, (9) immune surveillance and (10) stem cell formation.5, 6 Beyond selectively targeting greater than 15% of the human genome, MYC indirectly acts as a global transcription amplifier likely as a combined effect of the non-selective invasion into active promoters and the feedback loop following MYC induced physiological change.7–11 Hence, its deregulation induced by gene amplification, chromosomal translocation, as well as mutation in MYC gene or upstream regulators, promotes malignant transformation resulting in hallmark features of over 50% human cancers.12–15 Studies in transgenic mouse models demonstrated the role of MYC in tumor maintenance, which provides proof of principle of inactivating the oncoprotein as a treatment in reversing tumorigenesis.10, 16, 17
MYC has typically been touted as “undruggable” based upon its nuclear localization, intrinsically disordered structure, protein–protein interaction (PPI) interface and a lack of any cleft like protein pockets.18 Another general concern in the field has been that MYC inhibition would cause deleterious side effects in normal tissues as MYC responds to virtually every signaling pathway in every cell type. Yet, many of these stereotypes were overturned after the success of applying Omomyc, a MYC dominant-negative peptide in murine cancer models, suggesting a transient inhibition of MYC is highly tolerable.19 Moreover, other strategies have also appeared in an attempt to “drug MYC”, among which most promise has been seen in with molecules interfering with MYC-MAX protein/protein interactions as well as deterring MYC-MAX DNA association. Particularly promising has been direct targeting of MYC thereby reducing off-target effects, which in turn can effectively impede MYC function. This perspective aims to summarize the biochemistry behind the MYC/MAX network, MYC small molecule targeting strategies, in vivo evaluation of MYC inhibitors and future prospects for clinical trials.
BIOCHEMISTRY OF MYC
As stated MYC proteins belong to the superfamily of basic helix-loop-helix leucine zipper (bHLHLZ) DNA binding proteins. Within this complex is contained three-domains, including an N-terminal transactivation domain (TAD), a central domain, and a C-terminal basic-helix-loop-helix-leucine zipper (bHLHLZ) domain, this interacting with its essential partner, MYC-associated protein X (MAX) (Figure 1A). The central domain consists of a proline rich PEST segment, a calpain cleavage site (CAPN), which results in cytosolic MYC partial cleavage of the C-terminus, producing “MYC-nick”,20 and a nuclear localization sequence (NLS). Six highly conserved regions dubbed MYC homology boxes (MBs 0–IV) are sequentially distributed across TAD and the central domain, which bind to different MYC interactors and thus enable MYC to regulate distinct enzymatic reactions throughout the transcriptional process (Figure 1B).21
Figure 1.
MYC and its biochemical impact on the cell. (A) Crystal structure of the c-MYC/MAX dimer bound to E-box DNA (PDB ID 1NKP). (B) Structure components of MYC protein and its interaction partners. (C) MYC stability controlled by the ubiquitination-proteasome system. (D) Scheme showing MYC/MAX/MXD network and their roles in counteracting MYC function. Figure created with BioRender.
Proteomic profiling of the MB interactomes have revealed that half of the MYC interactors require one or more MBs for binding wherein two MBs, MB0 and MBII, are universally required for transformation.22 It should be understood that the interactome MB0 accelerates the transcription process by binding to the general transcription factor IIF (TFIIF); it also recruits PP1 phosphatase via cofactors to regulate MYC activity and stability.23 Moreover, MBII participates in chromatin remodeling and its modification brings together acetyltransferase-containing complexes through direct interaction with Transactivation/Transformation-Domain Associated Protein (TRRAP) enabling histone acetylation.24, 25 In addition, particularly prominent interactions occur between acetyltransferases p300, CBP and the WDR5 protein containing histone lysine methyltransferase complex, recruiting at MBIIIb. In sum these sequences of events all play important roles in directing MYC to chromatin modification.26, 27
From a biochemical perspective MBI is noteworthy for stepwise phosphorylation and dephosphorylation at serine 62 (S62) and threonine 58 (T58) residues and is responsible for ubiquitin-proteasome system (UPS) mediated degradation of the MYC protein (Figure 1C).28 Accordingly, MYC protein is expressed at a low level in normal cells with a fast turnover rate through the ubiquitin proteasome pathway involving interaction between Fbxw7 and its phosphorylation sites before polyubiquitination takes place at lysine 48.29, 30 By contrast, MYC binding to Aurora-A, a serine/threonine kinase, antagonizes Fbxw7 mediated degradation.31 Moreover, MBI has direct contact with positive transcription elongation factor b (P-TEFb). P-TEFb then phosphorylates the C-terminal domain (CTD) of RNAPII, and stimulates transcription pause release.32, 33 In addition to stability control, phosphorylation at S62 by mitogen-activated protein kinases (MAPK), which acts as a downstream signal of oncogenic RAS, or cyclin-dependent kinases (CDK) also plays a critical role influencing activity, target gene selection and intracellular localization of MYC.34 Besides aforementioned phosphorylation within MBI, the whole protein is also subjected to extensive post-translational modification spanning not only phosphorylation but acetylation, ubiquitination, sumoylation, and glycosylation.35 We highlight the modifications inside the bHLHLZ region. Here, MYC is found to be a substrate of Pak2, a serine/threonine kinase that participates in the cellular response to stress. In short Pak2 phosphorylates MYC at three sites, (S373 and T400) and thereby interferes with MYC/MAX complex formation, while (T358) restricts DNA recogniiton.36 By contrast acetylation at lysine has no impact on the assembly but potentially competes with other lysine dependent modifications like sumoylation or ubiquitination.37 Moreover, lysine residues (K389, K392, K398 and K430) within the bHLHLZ are found to be promiscuous SUMO acceptors. Toward this end sumoylation initiates concomitant ubiquitination for MYC degradation while also switching the transcriptional subprogram of MYC from activated to repressed or vice versa.38–40
Complementing activation, a number of genes have been identified that are transcriptionally silenced by MYC.41 On the whole, a large fraction is associated with the MYC-interacting zinc finger protein 1 (Miz-1). Here, MYC binding abrogates its individual activation effect on tumor suppressor genes like p15INK4b, a factor for cell cycle arrest.42 Myc also interacts with Sp1/Sp3 to repress the p21 transcription thus affecting the p21-mediated cell cycle checkpoint.43 This dual function is best exemplified in the extensive reprogramming of the miRNA transcriptome concomitantly activating the miR-17–92 cluster while predominantly downregulating other miRs.44 Adding another layer of complexity, proliferation is not the only output of MYC overexpression. MYC also sensitizes cells to apoptosis depending on tissue type and its environment, therefore mutations in these cells that elevate pro-survival signals like BCL2 synergize with MYC in tumorigenesis.45–47 This is compatible with the fact that additional mutagenic events are sometimes necessary to release its tumorigenic potential.48
Despite these biochemical complexities in MYC induced tumor occurrence, all pathways converged to the same point where MYC must dimerize with MAX via the bHLHLZ domain. Formation of this master transcriptional regulator is followed by DNA recognition on a specific sequence termed enhancer box (E-box) with high affinity against a canonical sequence of CACGTG.49 However, for the moment we need only point out that DNA affinity is not sufficient to account for the occupancies of core promoters by the MYC/MAX complexe.50 Moreover, multiple additional transcription factors compete with the same consensus sequence as MYC/MAX, while an active chromatin context is required for MYC/MAX access.51, 52 Accordingly, a model has been proposed wherein protein-protein interactions with resident chromatin proteins, such as WDR5, strongly enhance the promoter affinity and the subsequent sequence recognition leading to stabilization in these binding sites.11 The overall occupancy is determined by the kinetics between stratified apparent promoter affinities and the cellular MYC levels to shape the specific gene expression programs.53 Endogenous MYC levels only allows binding at high affinity promoters representing the fundamental role of MYC in cell growth (ribosome biogenesis and protein synthesis), cell cycle control, energy production, anabolic metabolism and DNA replication.54, 55 It should be further understood that as MYC levels escalate during transformation, more and more distal sites become occupied, a panoply genes become activated or repressed, eventually breaking the homeostasis and completely rewriting the cell’s behavior.
Within the MYC/MAX/MXD network, MAX functions as an essential dimerization partner. Moreover, MAX is constitutively expressed regardless of the cells stage whereas its interacting partners normally have a very short half-life.56 Heterodimers of MAX with MXD family members (MXD 1–4, formally called Mad proteins), Mnt, and Mga, antagonize MYC-dependent cell transformation by competition and transcriptional repression of the same E-box sequences (Figure 1D).57 The transcriptional repression is accomplished by association with Sin3, a corepressor complex that recruits HDAC for histone deacetylation.58, 59 In addition, loss of Mnt function releases the apoptotic activity of MYC mimicking the outcome of MYC aberrantly expressed at high concentrations, which indicates Mnt’s separate pro-survival capability.60 A parallel system centered on another MAX like protein, Mlx, forms a heterodimer with MXD family serving as a potential channel to divert these proteins away from direct participation in the MAX-interacting network.61 Mlx, when bound to nutrient sensing partners like MondoA and CHREBP, regulates glucose and lipid metabolism depending on the availability of multiple metabolites such as glucose and glucose-6-phosphate, and likely forms competition on similar Myc binding sites.62,63 Furthermore, MAX can form transcription inert homodimers acting as a tumor suppressor whereas its alternative spliced isoform delta MAX truncated at the C-terminal loses tumor suppressor activity shifting the equilibrium towards tumorigenesis.64, 65
STRUCTURAL STUDIES
Available crystallographic data of the heterodimer bHLHLZ domain to the CACGTG E-box provides a structural basis for DNA recognition and MYC-MAX interactions.66 Here, basic regions from the N-terminal insert into the E-box aligned DNA major groove making the major contact with DNA sequence, while the two helical segments (H1 and H2/Leucine zipper) allow dimerization with their counterparts. It has been demonstrated that all of the conserved hydrophobic amino acids within H1 and H2 contribute to the dimerization.67 Moreover, it has been shown that extensive hydrophobic and polar interactions between the HLH and LZ regions stabilize both MAX-MAX homodimers and MYC-MAX heterodimers with key residues providing H-bonding between MAX Gln91-Asn92 (monomer numbering), positively charged interactions between Arg-Arg on MYC and the negatively charged interactions found within Glu-Gln on MXD. Critical here is that these charged pairing disfavor MYC/MYC and MXD/MXD homodimer formation.68 In addition particularly important are three conserved residues within the basic region making base-specific contacts with the E-box, the His, Glu, and Arg residues at MAX positions 28, 32, and 35, which have been validated through mutagenesis experiments.69, 70 Strikingly, a recent X-ray structure and nuclear magnetic resonance (NMR) spectroscopy study of the apo form of Omomyc and MYC/MAX provide critical insights into the binding pattern in the absence of DNA. Here, an extensive network of helical structural interactions was determined in the basic region suggesting that the molecular recognition between MYC/MAX and DNA occurs via a conformational selection rather than an induced fit model.71, 72
MYC TARGETING STRATEGIES
Overall, a tremendous volume of medicinal research has been conducted in terms of targeting MYC; with an array of strategies utilized to target this notorious protein. These tactics are graphically described in Figure 2. Noteworthy efforts include the stabilization of the G-quadruplex,73 which consist of guanine rich segments of DNA, typically present in promoter region of c-MYC gene.73 In short, existence of G-quadruplex in promoter region acts as a transcriptional repressor of c-MYC expression. Hence, stabilization of existing G-quadruplex leads to downregulation of the transcription of c-MYC gene and eventually translation of c-MYC protein.73 Importantly clinical trials have been initiated with this approach.74, 75 Another tactic includes of antisense oligonucleotides (ASOs), wherein these synthetic, single stranded nucleotides are used to target RNA. Mechanistically this approach takes advantage of the inhibition of mRNA translation resulting in tumor growth reduction. Efforts here have resulted in ASO’s including INX-3280,76,77 INX-6295,78 and AVI-4126,79 which entered in clinical trials; however, they have recently been abandoned.80,81,82 An additional oligonucleotide agenda involves siRNA, which are well known to regulate gene expression through RNA interference (RNAi).83 These small interfering RNAs are processed by RNA-induced silencing complex and the enzyme named dicer to target RNA.83 In addition an siRNA, named DCR-MYC, reached clinical trials that were later halted due to lack of efficacy.84
Figure 2.
Graphical representation of the strategies to target MYC in a cancer cell.
A more chemistry concentric approach involves targeting/inhibition of the bromodomain (BRD) ultimately leading to downregulation of MYC protein. Although, function of bromodomain is not restricted to MYC gene regulation, many bromodomain extra-terminal domain inhibitors (BETis), are currently in clinical trials for MYC inhibition therapy.84 An additional indirect approach to regulate MYC that is noteworthy involves the inhibition of the holoenzyme protein complex named PP1/PNUTS, which leads to hyperphosphorylation of MYC. Ultimately this phosphorylation event leads to reduced chromatin binding of MYC as well as elevated proteasomal degradation.85
There are other miscellaneous strategies that target MYC through inhibition of its transcription, translation, regulators of MYC stability and its transcriptional cofactors. However, we will focus on direct inhibitors of the MYC-MAX heterodimer or MYC-MAX-DNA complex, encompassing small molecule inhibitors, peptides and miniproteins that have been engaged to inhibit these associations. More specifically, we will feature molecules with known in vitro success that have shown promising in vivo results. Lastly, we will highlight Omomyc, the only MYC-MAX inhibitor that has reached clinical trials.
IN VIVO EVALUATION OF MYC-MAX-DNA INHIBITORS
Cancer animal models have considerably advanced since the introduction of the first xenograft mouse model in the 1950’s. Through decades of cancer research, a better understanding of the advantages and limitations of commonly used animal models have enabled the exploitation of these models to gain insight into the mechanism, tolerability, and efficacy of anticancer immunotherapeutic and pharmacologics.86 Xenograft mouse models, including cell line-derived (CDX) and patient-derived (PDX), provide important tools for preclinical evaluation of anticancer therapeutics. The more simplistic CDX approach relies on the implantation of established human cancer cell lines, often commercially available, into immunodeficient animals providing predictive values. Although inexpensive and suitable for numerous types of studies, CDX models lack tumor heterogeneity and tumor micro-environment often diminishing translational value. Alternatively, the multifaceted and more informative PDX approach involves the engraftment of human derived cells or tumor tissue from a patient’s tumor into immunodeficient or humanized mice. Generally, PDX models can accurately depict the biology, heterogeneity, complexity, and molecular diversity of different cancers and recapitulate the tumor microenvironment providing an invaluable platform for translational cancer research. A complimentary approach to xenograft models is allografts that maintain host species, i.e. mouse cancer cell lines or mouse tumor tissue are implanted into mice. Using a combination of both xenograft and allograft mouse models, the therapeutic potential of numerous small molecule and peptide-based inhibitors targeting the MYC-MAX E-box complex have been evaluated in vivo
Myc-Max Dimerization Disruption.
Many small molecule inhibitors that disrupt MYC-MAX dimerization in vitro have been described in the literature, however only a small number of these compounds have shown in vivo potency against MYC-driven or MYC involved cancers, Figure 3. Some of the earliest MYC inhibitor studies identified two unrelated small molecules, 10058-F4 and 10074-G5, among others that bind distinct regions within the c-MYC bHLHLZ domain and stabilize the monomer, thus interfering with MYC-MAX heterodimerization and its association with DNA targets.87–89 Lack of efficacy in established CDX models, mice bearing human androgen-independent prostate cancer cells or human Burkitt’s lymphoma cells, was attributed to unfavorable pharmacological profiles including rapid metabolism and low tumor penetration.90, 91 Although, some success was later reported using 10058-F4 in MYCN transgenic mice.92 An improved second generation analog of 10074-G5, termed 3jc48-3, displaying greater potency in vitro and increased half-life underwent preliminary testing in a high grade prostate cancer PDX model.93, 94 Albeit a small sample size, 3jc48-3 was well tolerated with evidence of slowing tumor growth reflecting the drugs enhanced properties.
Figure 3.
Structures of non-protein inhibitors of MYC and MYC-MAX as discussed within the text.
Perhaps the first small molecule to display in vivo effectiveness against a MYC-driven human cancer was the novel inhibitor KJ-Pyr-9, discovered through screening efforts of the Janda and Vogt laboratories.95 KJ-Pyr-9 was shown to possess a nanomolar dissociation constant for MYC as well as favorable PK properties in vivo with no signs of acute toxicity. Notably, KJ-Pyr-9 was found to cross the blood-brain barrier (BBB) with higher drug concentrations detected in the brain compared to the blood. This latter observation suggests that MYC-targeted pharmacological interventions may have the potential to treat cancers, such as brain cancers, which are not only shielded by the blood-tumor barrier but also the BBB. KJ-Pyr-9 was put to the test against a highly aggressive, invasive triple negative breast cancer (TNBC) human cell line, MDA-MB-231, commonly used to model late-stage breast cancer. Tumor growth was halted in xenograft mice by penetrating tumor tissue and suppressing MYC’s transcriptional activity demonstrating its antitumor activity.
Another compound identified using MYC-targeted high through put (HTS) screening strategies, dubbed Mycro3, precludes MYC-MAX dimerization and binding to DNA, and is highly selective with great specificity.96, 97 To evaluate the in vivo efficacy of Mycro3, a K-RAS-induced pancreatic ductal adenocarcinoma (PDA) mouse model with MYC-dependence was developed. The relationship between these two “undruggable” oncoproteins (RAS and MYC) in tumorigenesis is very complex, however a cooperative mechanism between them has been proposed to drive cancer development and progression in several cancer types.98 Therefore, the prospect of modulating K-RAS through MYC targeting highlights the tremendous widespread potential of MYC inhibitors. In fact, daily oral administration of Mycro3 to moribund mice resulted in protracted survival and reduced tumor size.99 Although tumors did not completely disappear in this study, lack of tumor ablation was not necessarily unexpected and has been noted in other studies with anti-MYC inhibitors. Mycro3 treatment also significantly attenuated tumor growth in xenograft models of lung and breast cancer, illustrating the broad value of such a therapeutic strategy for the treatment of human cancers, including K-RAS-driven PDA.
The small molecule MYCMI-6 was identified through a cellular HTS and shown to disrupt MYC-MAX dimerization by binding of the MYC/MYCN bHLHLZ domain with no observable interaction with MAX.100 In vivo confirmation was demonstrated wherein MYC-MAX association was significantly reduced in tumor tissue in a MYCN-amplified neuroblastoma xenograft tumor model, correlating with both apoptosis induction and cell proliferation curtailment in tumor areas. MYCMI-7 also identified from this same screen was shown to directly bind MYC/MYCN, induce apoptosis in tumor cells while reducing tumor burden and increasing survival in MYC-driven acute myeloid leukemia, breast cancer and MYCN-amplified neuroblastoma cancer in vivo models.101 An important feature highlighted in these set of studies is the pan-MYC inhibitory activity of MYCMI-6 and −7 involving both MYC and NMYC, findings that are in line with reports involving 10058-F4 vide supra. While MYC has been the main therapeutic target among the MYC family of oncoproteins, NMYC and LMYC have been associated with neuroblastoma, NMYC-driven brain cancer and lung cancer, and are thought to play a role in tumor progression and maintenance. Based on sequence and structural homology between MYC proteins, the development of clinically viable pan-MYC inhibitors appears feasible.
Recently, a MyC-CaP allograft/xenograft mouse prostate model was engaged in a rapid in vivo screening format identifying a set of anti-MYC compounds, including MYCi361 and MYCi975.102 Strategically, this CDX approach took advantage of an engineered MyC-CaP E-box-Luc cell line that afforded live imaging of bioluminescence in tumors, monitoring MYC-transcriptional activity. Following this initial in vivo screen, a series of studies were conducted shedding light on MYCi361’s mechanism of action and revealing its ability to bind MYC but not MAX, impair MYC-MAX dimer binding to DNA, and promote MYC destabilization through modulating phosphorylation at threonine 58 of the MYC protein (p58). MYCi361 treatment of Myc-CaP xenograft mice revealed an increase in Myc p58 levels and a decrease in the Ki67 proliferation marker in tumor tissue along with remodeling of the tumor microenvironment. The latter observation led to combination studies with MYCi361 and an immune checkpoint inhibitor (anti-PDL) demonstrating the potential synergistic effect of combination therapy involving an anti-MYC inhibitor and immune checkpoint blockade. However, MYCi361 toxicity concerns prompted the development of MYCi975, an analog with an enhanced therapeutic index. MYCi975 was well-tolerated, displayed an exceptional PK profile, and showed in vivo anti-tumor activity including synergistic tumor suppression when administered in combination with a PD-L1 antibody, analogous to MYCi361.
A somewhat different MYC targeting approach sought to covalently modify the MYC protein itself by covalent linkage of a small molecule through reactive cysteine residues. EN4 was uncovered by a cysteine-reactive covalent ligand screen and shown to label a single site, cysteine 171, within an intrinsically disordered region of MYC.103 EN4 inhibits MYC-MAX DNA binding, impairs cell proliferation, and diminishes tumor growth in vivo in a 231 MFB breast cancer CDX model. Despite prospective off-targets liabilities and selectivity concerns, EN4 may provide a new avenue towards the treatment of MYC-driven cancers due to the covalent nature of MYC attachment.
Attenuation Of Myc-Max-E-Box DNA Association Through Alternative Mechanisms
Alternative to directly targeting the MYC protein, compounds and mini-proteins that disrupt the MYC-MAX-DNA complex through other mechanisms have been reported to show in vivo efficacy. For example, ME47, an engineered homodimer hybrid consisting of a region of MAX and the HLH domain of transcription factor E47, competes with MYC-MAX for binding to E-box DNA response elements of MYC target genes.104 Inducible expression of this minimalist protein in a breast cancer CDX model delayed tumor growth, reduced cellular proliferation, and extended survival.
KSI-3716, a small molecule inhibitor discovered by HTS efforts, likewise blocks MYC-MAX binding to target gene promoters.105 Delivery of KSI-3716 as an intravesical medication curbed tumor growth with no significant systemic toxicity in orthotopic bladder xenografts.106 KSI-3716’s potential utility in gemcitabine-resistant bladder cancer was also suggested based on results from in vitro resistance studies, although in vivo efficacy has yet to be reported.107
Lastly, an alternative approach explored perturbation of MYC’s transcriptional activity through targeting its binding partner MAX. KI_MS2-008 was shown to stabilize transcriptionally inert MAX-MAX homodimers, resulting in a decrease in both MYC binding at promoters and MYC protein levels.108 The in vivo potency of KI_MS2-008 was assessed in allograft mouse models of MYC-induced T cell acute lymphoblastic leukemia and hepatocellular carcinoma mouse models showing a marked reduction in tumor burden.
REACHING CLINICAL TRIALS AFTER DECADES OF MYC RESEARCH
For decades speculation, often skepticism, has centered around whether MYC targeted therapy would become a viable treatment option for MYC-driven and -associated cancers. Given MYC’s central role in a wide array of fundamental cellular processes, the potential for severe consequences caused by hampering MYC’s function were considered inevitable. The multitude of small molecule and peptide-based MYC inhibitor studies have exposed numerous hurdles that still need to be overcome such as lack of specificity, adverse off-targets effects, low potency and tumor penetration, and subpar PK profiles. However great strides have been made as evidenced by the positive in vivo results presented. Importantly, the notion of combination therapies or drug cocktails including MYC targeting drugs as a key component has gained traction. In fact, preclinical studies have revealed drug synergism between an anti-MYC compound and immune checkpoint inhibitor,102 as well as the mini-protein Omomyc and paclitaxel, vide infra.
Perhaps the best support of a MYC targeting drug becoming a clinically viable cancer treatment option has come from the recent entry of Omomyc (OMO-103) into clinical trials. Omomyc, a first in class MYC inhibitor initially disclosed in 1998, is a dominant negative mutant of the MYC bHLHLZ domain consisting of four mutations that prevent MYC-MAX dimerization.109, 110 Through formation of heterodimers with either MYC or MAX or formation of homodimers, Omomyc prevents MYC-MAX dimerization, inhibits E-box DNA binding, and promotes MYC degradation.109 In early preclinical studies, transgenically-expressed Omomyc showed in vivo efficacy in cancer mouse models without significant side effects, providing proof of concept.110 Possibly one of the most significant findings came later when recombinant Omomyc was shown to have cell penetrating activity.111 Intranasal administration of recombinant Omomyc halted tumor progression in a lung adenocarcinoma model, penetrating both cellular and nuclear membranes.111 Additionally, intravenously dosed Omomyc was shown to be better than paclitaxel at blunting tumor growth in a lung adenocarcinoma xenograft model and combination therapy with both Omomyc and paclitaxel proved to be far superior to each monotherapy alone, resulting in near complete eradication of tumor growth and prolonging survival without observable side effects.111 Overall, these later findings helped solidify Omomyc’s potential as a clinically viable anti-MYC therapeutic catapulting Omomyc into Phase I/II human clinical trials where promising Phase I preliminary results have been reported.
OUTLOOK
The MYC proto-oncogene family has been at the forefront of cancer biology for greater than 30 years, yet it has constantly presented the research community with undeniable biological as well as chemical challenges. For example, MYC is one of the most frequently overexpressed genes in human cancer.112 A major difficulty with targeting MYC driven cancer is that it is also the gatekeeping regulator of cell division.113 As such a multitude of transcriptional programs are under MYC control with distinct effects on the cellular phenotype. Success in this arena has also had to contend with the disordered nature of Myc, which by the physical standards of the time, vide supra, made it a quite complex and challenging drug target to model.114 Further confidence in the modulation of MYC also waned because the MYC-MAX heterodimer has an estimated protein-protein interaction (PPI) surface area of over 3000 angstroms115 and as well documented PPIs lack well-defined pockets or grooves that could be used for high energy binding interactions with a small ligand.68, 116
At the clinical level therapeutic efforts against MYC and MYC-MAX have also stalled due to off-target effects on other oncoproteins.117 While more efforts on oncogene selectivity is warranted new MYC drug candidates will also need to contend with fast metabolism, nonselective distribution after systematic dosing, and tumor penetrability of these small molecule and peptide inhibitors, all of which will undoubtably hold back their clinical feasibility.90, 91 Accordingly, we also envision that it is time to place more weight on the integration of well-designed delivery systems in MYC drug discovery efforts. Studies have shown that cell penetrating peptides and nuclear targeting sequences fused to the peptide inhibitors led to unprecedented enhancement of tumor growth inhibition both in vitro and in vivo.114, 118–120 Indeed, a lipase-labile 10058-F4 prodrug carried by integrin-targeted nanoparticles showed reduced tumor burden in integrin overexpressed multiple myeloma animal model against free drugs.121 It would be interesting to see additional efforts grounded upon nanoparticle platforms making their way to help boost Myc inhibitors in the future.
Apart from all these hurdles, extensive MYC research has generated a profound biomedical arsenal though robust high throughput screening interventions, meticulous computer aided drug design campaigns, screenings of natural products and fastidiously engineered peptides and miniproteins. These efforts have led to the genesis of Omomyc, which has cleared phase I clinical trials with low toxicity and excellent tolerability in patients.122 Finally, while not fully insulated from scattered skeptics and still considerable pessimism, we believe it can now be fully acknowledged that after three decades of chemistry centric imagination, diligence and perseverance that the most deregulated oncogene in human cancer can now be delisted from the “undruggable” list of proteins.
ACKNOWLEDGMENT
Research was supported by the National Institutes of Health grant R01 CA262290 (to KDJ). This work was also supported by the Skaggs Institute for Chemical Biology (KDJ).
Contributor Information
Nishant M. Karadkhelkar, Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States
Mingliang Lin, Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States.
Lisa M. Eubanks, Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States
Kim D. Janda, Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States.
REFERENCES
- 1.Adhikary S; Eilers M, Transcriptional regulation and transformation by MYC proteins. Nat Rev Mol Cell Bio 2005, 6 (8), 635–645. [DOI] [PubMed] [Google Scholar]
- 2.Wolf E; Eilers M, Targeting MYC Proteins for Tumor Therapy. Annu Rev Canc Biol 2020, 4, 61–75. [Google Scholar]
- 3.Rickman DS; Schulte JH; Eilers M, The Expanding World of N-MYC-Driven Tumors. Cancer Discov 2018, 8 (2), 150–163. [DOI] [PubMed] [Google Scholar]
- 4.Bragelmann J; Bohm S; Guthrie MR; Mollaoglu G; Oliver TG; Sos ML, Family matters: How MYC family oncogenes impact small cell lung cancer. Cell Cycle 2017, 16 (16), 1489–1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chen H; Liu HD; Qing GL, Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduct Tar 2018, 3 (1), 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Duffy MJ; O’Grady S; Tang MH; Crown J, MYC as a target for cancer treatment. Cancer Treat Rev 2021, 94, 102154. [DOI] [PubMed] [Google Scholar]
- 7.Fernandez PC; Frank SR; Wang LQ; Schroeder M; Liu SX; Greene J; Cocito A; Amati B, Genomic targets of the human c-Myc protein. Gene Dev 2003, 17 (9), 1115–1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sabo A; Kress TR; Pelizzola M; de Pretis S; Gorski MM; Tesi A; Morelli MJ; Bora P; Doni M; Verrecchia A; Tonelli C; Faga G; Bianchi V; Ronchi A; Low D; Muller H; Guccione E; Campaner S; Amati B, Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis. Nature 2014, 511 (7510), 488–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lin CY; Loven J; Rahl PB; Paranal RM; Burge CB; Bradner JE; Lee TI; Young RA, Transcriptional Amplification in Tumor Cells with Elevated c-Myc. Cell 2012, 151 (1), 56–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Soucek L; Whitfield J; Martins CP; Finch AJ; Murphy DJ; Sodir NM; Karnezis AN; Swigart LB; Nasi S; Evan GI, Modelling Myc inhibition as a cancer therapy. Nature 2008, 455 (7213), 679–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kress TR; Sabo A; Amati B, MYC: connecting selective transcriptional control to global RNA production. Nat Rev Cancer 2015, 15 (10), 593–607. [DOI] [PubMed] [Google Scholar]
- 12.Gabay M; Li YL; Felsher DW, MYC Activation Is a Hallmark of Cancer Initiation and Maintenance. Csh Perspect Med 2014, 4 (6), a014241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kalkat M; De Melo J; Hickman KA; Lourenco C; Redel C; Resetca D; Tamachi A; Tu WB; Penn LZ, MYC Deregulation in Primary Human Cancers. Genes-Basel 2017, 8 (6), 151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lancho O; Herranz D, The MYC Enhancer-ome: Long-Range Transcriptional Regulation of MYC in Cancer. Trends Cancer 2018, 4 (12), 810–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Meyer N; Penn LZ, MYC - TIMELINE Reflecting on 25 years with MYC. Nat Rev Cancer 2008, 8 (12), 976–990. [DOI] [PubMed] [Google Scholar]
- 16.Shachaf CM; Felsher DW, Tumor dormancy and MYC inactivation: Pushing cancer to the brink of normalcy. Cancer Res 2005, 65 (11), 4471–4474. [DOI] [PubMed] [Google Scholar]
- 17.Arvanitis C; Felsher DW, Conditional transgenic models define how MYC initiates and maintains tumorigenesis. Semin Cancer Biol 2006, 16 (4), 313–317. [DOI] [PubMed] [Google Scholar]
- 18.Dang CV; Reddy EP; Shokat KM; Soucek L, Drugging the ‘undruggable’ cancer targets. Nat Rev Cancer 2017, 17 (8), 502–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Soucek L; Whitfield JR; Sodir NM; Masso-Valles D; Serrano E; Karnezis AN; Swigart LB; Evan GI, Inhibition of Myc family proteins eradicates KRas-driven lung cancer in mice. Gene Dev 2013, 27 (5), 504–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Conacci-Sorrell M; Ngouenet C; Eisenman RN, Myc-Nick: A Cytoplasmic Cleavage Product of Myc that Promotes alpha-Tubulin Acetylation and Cell Differentiation. Cell 2010, 142 (3), 480–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhou YH; Gao XM; Yuan M; Yang B; He QJ; Cao J, Targeting Myc Interacting Proteins as a Winding Path in Cancer Therapy. Front Pharmacol 2021, 12, 748852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kalkat M; Resetca D; Lourenco C; Chan PK; Wei Y; Shiah YJ; Vitkin N; Tong YF; Sunnerhagen M; Done SJ; Boutros PC; Raught B; Penn LZ, MYC Protein Interactome Profiling Reveals Functionally Distinct Regions that Cooperate to Drive Tumorigenesis. Mol Cell 2018, 72 (5), 836–848. [DOI] [PubMed] [Google Scholar]
- 23.Wei Y; Redel C; Ahlner A; Lemak A; Johansson-Akhe I; Houliston S; Kenney TMG; Tamachi A; Morad V; Duan SL; Andrews DW; Wallner B; Sunnerhagen M; Arrowsmith CH; Penn LZ, The MYC oncoprotein directly interacts with its chromatin cofactor PNUTS to recruit PP1 phosphatase. Nucleic Acids Res 2022, 50 (6), 3505–3522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zippo A; De Robertis A; Serafini R; Oliviero S, PIM1-dependent phosphorylation of histone H3 at serine 10 is required for MYC-dependent transcriptional activation and oncogenic transformation. Nat Cell Biol 2007, 9 (8), 932–944. [DOI] [PubMed] [Google Scholar]
- 25.McMahon SB; Van Buskirk HA; Dugan KA; Copeland TD; Cole MD, The novel ATM-related protein TRRAP is an essential cofactor for the c-Myc and E2F oncoproteins. Cell 1998, 94 (3), 363–374. [DOI] [PubMed] [Google Scholar]
- 26.Thomas LR; Wang QG; Grieb BC; Phan J; Foshage AM; Sun Q; Olejniczak ET; Clark T; Dey S; Lorey S; Alicie B; Howard GC; Cawthon B; Ess KC; Eischen CM; Zhao ZM; Fesik SW; Tansey WP, Interaction with WDR5 Promotes Target Gene Recognition and Tumorigenesis by MYC. Mol Cell 2015, 58 (3), 440–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Weinert BT; Narita T; Satpathy S; Srinivasan B; Hansen BK; Scholz C; Hamilton WB; Zucconi BE; Wang WW; Liu WSR; Brickman JM; Kesicki EA; Lai A; Bromberg KD; Cole PA; Choudhary C, Time-Resolved Analysis Reveals Rapid Dynamics and Broad Scope of the CBP/p300 Acetylome. Cell 2018, 174 (1), 231–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Welcker M; Orian A; Jin JP; Grim JA; Harper JW; Eisenman RN; Clurman BE, The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. P Natl Acad Sci USA 2004, 101 (24), 9085–9090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Salghetti SE; Kim SY; Tansey WP, Destruction of Myc by ubiquitin-mediated proteolysis: cancer-associated and transforming mutations stabilize Myc. Embo J 1999, 18 (3), 717–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yada M; Hatakeyama S; Kamura T; Nishiyama M; Tsunematsu R; Imaki H; Ishida N; Okumura F; Nakayama K; Nakayama KI, Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7. Embo J 2004, 23 (10), 2116–2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Otto T; Horn S; Brockmann M; Eilers U; Schuttrumpf L; Popov N; Kenney AM; Schulte JH; Beijersbergen R; Christiansen H; Berwanger B; Eilers M, Stabilization of N-Myc Is a Critical Function of Aurora A in Human Neuroblastoma. Cancer Cell 2009, 15 (1), 67–78. [DOI] [PubMed] [Google Scholar]
- 32.Kanazawa S; Soucek L; Evan G; Okamoto T; Peterlin BM, c-Myc recruits P-TEFb for transcription, cellular proliferation and apoptosis. Oncogene 2003, 22 (36), 5707–5711. [DOI] [PubMed] [Google Scholar]
- 33.Rahl PB; Lin CY; Seila AC; Flynn RA; McCuine S; Burge CB; Sharp PA; Young RA, c-Myc Regulates Transcriptional Pause Release. Cell 2010, 141 (3), 432–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hydbring P; Castell A; Larsson LG, MYC Modulation around the CDK2/p27/SKP2 Axis. Genes-Basel 2017, 8 (7), 174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ma ZY; Vosseller K, O-GlcNAc in cancer biology. Amino Acids 2013, 45 (4), 719–733. [DOI] [PubMed] [Google Scholar]
- 36.Huang ZD; Traugh JA; Bishop JM, Negative control of the Myc protein by the stress-responsive kinase Pak2. Mol Cell Biol 2004, 24 (4), 1582–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Vervoorts J; Luscher-Firzlaff J; Luscher B, The ins and outs of MYC regulation by posttranslational mechanisms. J Biol Chem 2006, 281 (46), 34725–34729. [DOI] [PubMed] [Google Scholar]
- 38.Gonzalez-Prieto R; Cuijpers SAG; Kumar R; Hendriks IA; Vertegaal ACO, c-Myc is targeted to the proteasome for degradation in a SUMOylation-dependent manner, regulated by PIAS1, SENP7 and RNF4. Cell Cycle 2015, 14 (12), 1859–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kessler JD; Kahle KT; Sun TT; Meerbrey KL; Schlabach MR; Schmitt EM; Skinner SO; Xu QK; Li MZ; Hartman ZC; Rao M; Yu P; Dominguez-Vidana R; Liang AC; Solimini NL; Bernardi RJ; Yu B; Hsu T; Golding I; Luo J; Osborne CK; Creighton CJ; Hilsenbeck SG; Schiff R; Shaw CA; Elledge SJ; Westbrook TF, A SUMOylation-Dependent Transcriptional Subprogram Is Required for Myc-Driven Tumorigenesis. Science 2012, 335 (6066), 348–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sun XX; Chen YX; Su YL; Wang XY; Chauhan KM; Liang J; Daniel CJ; Sears RC; Dai MS, SUMO protease SENP1 deSUMOylates and stabilizes c-Myc. P Natl Acad Sci USA 2018, 115 (43), 10983–10988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Walz S; Lorenzin F; Morton J; Wiese KE; von Eyss B; Herold S; Rycak L; Dumay-Odelot H; Karim S; Bartkuhn M; Roels F; Wustefeld T; Fischer M; Teichmann M; Zender L; Wei CL; Sansom O; Wolf E; Eilers M, Activation and repression by oncogenic MYC shape tumour-specific gene expression profiles. Nature 2014, 511 (7510), 483–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Staller P; Peukert K; Kiermaier A; Seoane J; Lukas J; Karsunky H; Moroy T; Bartek J; Massague J; Hanel F; Eilers M, Repression of p15(INK4b) expression by Myc through association with Miz-1. Nat Cell Biol 2001, 3 (4), 392–399. [DOI] [PubMed] [Google Scholar]
- 43.Gartel AL; Ye X; Goufman E; Shianov P; Hay N; Najmabadi F; Tyner AL, Myc represses the p21(WAF1/CIP1) promoter and interacts with Sp1/Sp3. Proc Natl Acad Sci U S A 2001, 98 (8), 4510–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chang TC; Yu DN; Lee YS; Wentzel EA; Arking DE; West KM; Dang CV; Thomas-Tikhonenko A; Mendell JT, Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet 2008, 40 (1), 43–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Strasser A; Harris AW; Bath ML; Cory S, Novel Primitive Lymphoid Tumors Induced in Transgenic Mice by Cooperation between Myc and Bcl-2. Nature 1990, 348 (6299), 331–333. [DOI] [PubMed] [Google Scholar]
- 46.Nguyen HV; Vandenberg CJ; Ng AP; Robati MR; Anstee NS; Rimes J; Hawkins ED; Cory S, Development and survival of MYC-driven lymphomas require the MYC antagonist MNT to curb MYC-induced apoptosis. Blood 2020, 135 (13), 1019–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pelengaris S; Rudolph B; Littlewood T, Action of Myc in vivo - proliferation and apoptosis. Curr Opin Genet Dev 2000, 10 (1), 100–105. [DOI] [PubMed] [Google Scholar]
- 48.Dang CV, MYC on the Path to Cancer. Cell 2012, 149 (1), 22–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Blackwell TK; Kretzner L; Blackwood EM; Eisenman RN; Weintraub H, Sequence-Specific DNA-Binding by the C-Myc Protein. Science 1990, 250 (4984), 1149–1151. [DOI] [PubMed] [Google Scholar]
- 50.Nie ZQ; Hu GQ; Wei G; Cui KR; Yamane A; Resch W; Wang RN; Green DR; Tessarollo L; Casellas R; Zhao KJ; Levens D, c-Myc Is a Universal Amplifier of Expressed Genes in Lymphocytes and Embryonic Stem Cells. Cell 2012, 151 (1), 68–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Guccione E; Martinato F; Finocchiaro G; Luzi L; Tizzoni L; Dall’Olio V; Zardo G; Nervi C; Bernard L; Amati B, Myc-binding-site recognition in the human genome is determined by chromatin context. Nat Cell Biol 2006, 8 (7), 764–770. [DOI] [PubMed] [Google Scholar]
- 52.Wolf E; Lin CY; Eilers M; Levens DL, Taming of the beast: shaping Myc-dependent amplification. Trends Cell Biol 2015, 25 (4), 241–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lorenzin F; Benary U; Baluapuri A; Walz S; Jung LA; von Eyss B; Kisker C; Wolf J; Eilers M; Wolf E, Different promoter affinities account for specificity in MYC-dependent gene regulation. eLife 2016, 5, e15161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pelengaris S; Khan M; Evan G, c-MYC: More than just a matter of life and death. Nat Rev Cancer 2002, 2 (10), 764–776. [DOI] [PubMed] [Google Scholar]
- 55.Eilers M; Eisenman RN, Myc’s broad reach. Gene Dev 2008, 22 (20), 2755–2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Blackwood EM; Luscher B; Eisenman RN, Myc and Max Associate Invivo. Gene Dev 1992, 6 (1), 71–80. [DOI] [PubMed] [Google Scholar]
- 57.Grandori C; Cowley SM; James LP; Eisenman RN, The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Bi 2000, 16, 653–699. [DOI] [PubMed] [Google Scholar]
- 58.Kurland JF; Tansey WP, Myc-mediated transcriptional repression by recruitment of histone deacetylase. Cancer Res 2008, 68 (10), 3624–3629. [DOI] [PubMed] [Google Scholar]
- 59.Laherty CD; Yang WM; Sun JM; Davie JR; Seto E; Eisenman RN, Histone deacetylases associated with the mSin3 corepressor mediate Mad transcriptional repression. Cell 1997, 89 (3), 349–356. [DOI] [PubMed] [Google Scholar]
- 60.Link JM; Ota S; Zhou ZQ; Daniel CJ; Sears RC; Hurlin PJ, A critical role for Mnt in Myc-driven T-cell proliferation and oncogenesis. P Natl Acad Sci USA 2012, 109 (48), 19685–19690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hurlin PJ; Huang J, The MAX-interacting transcription factor network. Semin Cancer Biol 2006, 16 (4), 265–274. [DOI] [PubMed] [Google Scholar]
- 62.Havula E; Hietakangas V, Glucose sensing by ChREBP/MondoA-Mlx transcription factors. Semin Cell Dev Biol 2012, 23 (6), 640–7. [DOI] [PubMed] [Google Scholar]
- 63.Diolaiti D; McFerrin L; Carroll PA; Eisenman RN, Functional interactions among members of the MAX and MLX transcriptional network during oncogenesis. Biochim Biophys Acta 2015, 1849 (5), 484–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Makela TP; Koskinen PJ; Vastrik I; Alitalo K, Alternative Forms of Max as Enhancers or Suppressors of Myc-Ras Cotransformation. Science 1992, 256 (5055), 373–377. [DOI] [PubMed] [Google Scholar]
- 65.Maltais L; Montagne M; Bedard M; Tremblay C; Soucek L; Lavigne P, Biophysical characterization of the b-HLH-LZ of Delta Max, an alternatively spliced isoform of Max found in tumor cells: Towards the validation of a tumor suppressor role for the Max homodimers. Plos One 2017, 12 (3), e0174413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Nair SK; Burley SK, X-ray structures of Myc-Max and Mad-Max recognizing DNA: Molecular bases of regulation by proto-oncogenic transcription factors. Cell 2003, 112 (2), 193–205. [DOI] [PubMed] [Google Scholar]
- 67.Davis LJ; Halazonetis TD, Both the Helix-Loop-Helix and the Leucine Zipper Motifs of C-Myc Contribute to Its Dimerization Specificity with Max. Oncogene 1993, 8 (1), 125–132. [PubMed] [Google Scholar]
- 68.Carabet LA; Rennie PS; Cherkasov A, Therapeutic Inhibition of Myc in Cancer. Structural Bases and Computer-Aided Drug Discovery Approaches. Int J Mol Sci 2018, 20 (1), 120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Fisher F; Goding CR, Single Amino-Acid Substitutions Alter Helix Loop Helix Protein Specificity for Bases Flanking the Core Canntg Motif. Embo J 1992, 11 (11), 4103–4109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Dang CV; Dolde C; Gillison ML; Kato GJ, Discrimination between Related DNA Sites by a Single Amino-Acid Residue of Myc-Related Basic Helix Loop Helix Proteins. P Natl Acad Sci USA 1992, 89 (2), 599–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Jung LA; Gebhardt A; Koelmel W; Ade CP; Walz S; Kuper J; von Eyss B; Letschert S; Redel C; d’Artista L; Biankin A; Zender L; Sauer M; Wolf E; Evan G; Kisker C; Eilers M, OmoMYC blunts promoter invasion by oncogenic MYC to inhibit gene expression characteristic of MYC-dependent tumors. Oncogene 2017, 36 (14), 1911–1924. [DOI] [PubMed] [Google Scholar]
- 72.Sammak S; Hamdani N; Gorrec F; Allen MD; Freund SMV; Bycroft M; Zinzalla G, 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. Biochemistry-Us 2019, 58 (29), 3144–3154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chaudhuri R; Bhattacharya S; Dash J; Bhattacharya S, Recent Update on Targeting c-MYC G-Quadruplexes by Small Molecules for Anticancer Therapeutics. J Med Chem 2021, 64 (1), 42–70. [DOI] [PubMed] [Google Scholar]
- 74.Balaratnam S; Schneekloth JS, Transcriptional regulation of MYC through G-quadruplex structures. Annu Rep Med Chem 2020, 54, 361–407. [Google Scholar]
- 75.Aptose Biosciences Inc. Aptose provides update on apto-253 program. https://www.aptose.com/news-media/press-releases/detail/220/aptose-provides-update-on-apto-253-program (accessed 6 January 2023).
- 76.thepharmaletter Inex completes patient enrollment in Phase I INX-3280 trial. https://www.thepharmaletter.com/article/inex-completes-patient-enrollment-in-phase-i-inx-3280-trial (accessed 5 January, 2023).
- 77.Webb, M. S. T. N; Cremese M; Kozlowska H; Blaquiere M; Devine, D. V; Kornbrust, D J, Toxicity and Toxicokinetics of a Phosphorothioate Oligonucleotide Against the C- Myc Oncogene in Cynomolgus Monkeys. Antisense and Nucleic Acid Drug Development 2001, 11 (3), 155–163. [DOI] [PubMed] [Google Scholar]
- 78.BioCentury INXC-6295: Began Phase I/II testing. https://www.biocentury.com/article/113753/inxc-6295-began-phase-i-ii-testing (accessed 5 January, 2023).
- 79.Hudziak RM; Summerton J; Weller DD; Iversen PL, Antiproliferative effects of steric blocking phosphorodiamidate morpholino antisense agents directed against c-myc. Antisense Nucleic Acid Drug Dev 2000, 10 (3), 163–76. [DOI] [PubMed] [Google Scholar]
- 80.springer INXC 6295. https://adisinsight.springer.com/drugs/800010320 (accessed 5 January, 2023).
- 81.springer INX 3280. https://adisinsight.springer.com/drugs/800007051 (accessed 5 January 2023).
- 82.springer AVI 4126. https://adisinsight.springer.com/drugs/800015229 (accessed 5 January 2023).
- 83.Dana H; Chalbatani GM; Mahmoodzadeh H; Karimloo R; Rezaiean O; Moradzadeh A; Mehmandoost N; Moazzen F; Mazraeh A; Marmari V; Ebrahimi M; Rashno MM; Abadi SJ; Gharagouzlo E, Molecular Mechanisms and Biological Functions of siRNA. Int J Biomed Sci 2017, 13 (2), 48–57. [PMC free article] [PubMed] [Google Scholar]
- 84.Whitfield JR; Beaulieu ME; Soucek L, Strategies to Inhibit Myc and Their Clinical Applicability. Front Cell Dev Biol 2017, 5, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Dingar D; Tu WB; Resetca D; Lourenco C; Tamachi A; De Melo J; Houlahan KE; Kalkat M; Chan PK; Boutros PC; Raught B; Penn LZ, MYC dephosphorylation by the PP1/PNUTS phosphatase complex regulates chromatin binding and protein stability. Nat Commun 2018, 9 (1), 3502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Abdolahi S; Ghazvinian Z; Muhammadnejad S; Saleh M; Asadzadeh Aghdaei H; Baghaei K, Patient-derived xenograft (PDX) models, applications and challenges in cancer research. J Transl Med 2022, 20 (1), 206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yin X; Giap C; Lazo JS; Prochownik EV, Low molecular weight inhibitors of Myc-Max interaction and function. Oncogene 2003, 22 (40), 6151–9. [DOI] [PubMed] [Google Scholar]
- 88.Follis AV; Hammoudeh DI; Wang H; Prochownik EV; Metallo SJ, Structural rationale for the coupled binding and unfolding of the c-Myc oncoprotein by small molecules. Chem Biol 2008, 15 (11), 1149–55. [DOI] [PubMed] [Google Scholar]
- 89.Hammoudeh DI; Follis AV; Prochownik EV; Metallo SJ, Multiple independent binding sites for small-molecule inhibitors on the oncoprotein c-Myc. J Am Chem Soc 2009, 131 (21), 7390–401. [DOI] [PubMed] [Google Scholar]
- 90.Clausen DM; Guo J; Parise RA; Beumer JH; Egorin MJ; Lazo JS; Prochownik EV; Eiseman JL, In vitro cytotoxicity and in vivo efficacy, pharmacokinetics, and metabolism of 10074-G5, a novel small-molecule inhibitor of c-Myc/Max dimerization. J Pharmacol Exp Ther 2010, 335 (3), 715–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Guo J; Parise RA; Joseph E; Egorin MJ; Lazo JS; Prochownik EV; Eiseman JL, Efficacy, pharmacokinetics, tisssue distribution, and metabolism of the Myc-Max disruptor, 10058-F4 [Z,E]-5-[4-ethylbenzylidine]-2-thioxothiazolidin-4-one, in mice. Cancer Chemother Pharmacol 2009, 63 (4), 615–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zirath H; Frenzel A; Oliynyk G; Segerström L; Westermark UK; Larsson K; Munksgaard Persson M; Hultenby K; Lehtiö J; Einvik C; Påhlman S; Kogner P; Jakobsson PJ; Henriksson MA, MYC inhibition induces metabolic changes leading to accumulation of lipid droplets in tumor cells. Proc Natl Acad Sci U S A 2013, 110 (25), 10258–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Chauhan J; Wang H; Yap JL; Sabato PE; Hu A; Prochownik EV; Fletcher S, Discovery of methyl 4’-methyl-5-(7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)-[1,1’-biphenyl]-3-carboxylate, an improved small-molecule inhibitor of c-Myc-max dimerization. ChemMedChem 2014, 9 (10), 2274–2285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Shukla S; Fletcher S; Chauhan J; Chalfant V; Riveros C; Mackeyev Y; Singh PK; Krishnan S; Osumi T; Balaji KC, 3JC48–3 (methyl 4’-methyl-5-(7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)-[1,1’-biphenyl]-3-carboxylate) : a novel MYC/MAX dimerization inhibitor reduces prostate cancer growth. Cancer Gene Ther 2022, 29 (11), 1550–1557. [DOI] [PubMed] [Google Scholar]
- 95.Hart JR; Garner AL; Yu J; Ito Y; Sun M; Ueno L; Rhee JK; Baksh MM; Stefan E; Hartl M; Bister K; Vogt PK; Janda KD, Inhibitor of MYC identified in a Krohnke pyridine library. Proc Natl Acad Sci U S A 2014, 111 (34), 12556–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kiessling A; Sperl B; Hollis A; Eick D; Berg T, Selective inhibition of c-Myc/Max dimerization and DNA binding by small molecules. Chem Biol 2006, 13 (7), 745–51. [DOI] [PubMed] [Google Scholar]
- 97.Kiessling A; Wiesinger R; Sperl B; Berg T, Selective inhibition of c-Myc/Max dimerization by a pyrazolo[1,5-a]pyrimidine. ChemMedChem 2007, 2 (5), 627–30. [DOI] [PubMed] [Google Scholar]
- 98.Mahauad-Fernandez WD; Felsher DW, The Myc and Ras Partnership in Cancer: Indistinguishable Alliance or Contextual Relationship? Cancer Res 2020, 80 (18), 3799–3802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Stellas D; Szabolcs M; Koul S; Li Z; Polyzos A; Anagnostopoulos C; Cournia Z; Tamvakopoulos C; Klinakis A; Efstratiadis A, Therapeutic effects of an anti-Myc drug on mouse pancreatic cancer. J Natl Cancer Inst 2014, 106 (12), dju320. [DOI] [PubMed] [Google Scholar]
- 100.Castell A; Yan Q; Fawkner K; Hydbring P; Zhang F; Verschut V; Franco M; Zakaria SM; Bazzar W; Goodwin J; Zinzalla G; Larsson LG, A selective high affinity MYC-binding compound inhibits MYC:MAX interaction and MYC-dependent tumor cell proliferation. Sci Rep 2018, 8 (1), 10064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Castell A; Yan Q; Fawkner K; Bazzar W; Zhang F; Wickstrom M; Alzrigat M; Franco M; Krona C; Cameron DP; Dyberg C; Olsen TK; Verschut V; Schmidt L; Y., L. S; Mahmoud L; Hydbring P; Lehmann S; Baranello L; Nelander S; Johnsen, J. I; Larsson, L. G, MYCMI-7: A Small MYC-Binding Compound that Inhibits MYC: MAX Interaction and Tumor Growth in a MYC-Dependent Manner Cancer Res Commun 2022, 2 (3), 182–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Han H; Jain AD; Truica MI; Izquierdo-Ferrer J; Anker JF; Lysy B; Sagar V; Luan Y; Chalmers ZR; Unno K; Mok H; Vatapalli R; Yoo YA; Rodriguez Y; Kandela I; Parker JB; Chakravarti D; Mishra RK; Schiltz GE; Abdulkadir SA, Small-Molecule MYC Inhibitors Suppress Tumor Growth and Enhance Immunotherapy. Cancer Cell 2019, 36 (5), 483–497.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Boike L; Cioffi AG; Majewski FC; Co J; Henning NJ; Jones MD; Liu G; McKenna JM; Tallarico JA; Schirle M; Nomura DK, Discovery of a Functional Covalent Ligand Targeting an Intrinsically Disordered Cysteine within MYC. Cell Chem Biol 2021, 28 (1), 4–13.e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lustig LC; Dingar D; Tu WB; Lourenco C; Kalkat M; Inamoto I; Ponzielli R; Chan WCW; Shin JA; Penn LZ, Inhibiting MYC binding to the E-box DNA motif by ME47 decreases tumour xenograft growth. Oncogene 2017, 36 (49), 6830–6837. [DOI] [PubMed] [Google Scholar]
- 105.Jeong KC; Ahn KO; Yang CH, Small-molecule inhibitors of c-Myc transcriptional factor suppress proliferation and induce apoptosis of promyelocytic leukemia cell via cell cycle arrest. Mol Biosyst 2010, 6 (8), 1503–9. [DOI] [PubMed] [Google Scholar]
- 106.Jeong KC; Kim KT; Seo HH; Shin SP; Ahn KO; Ji MJ; Park WS; Kim IH; Lee SJ; Seo HK, Intravesical instillation of c-MYC inhibitor KSI-3716 suppresses orthotopic bladder tumor growth. J Urol 2014, 191 (2), 510–8. [DOI] [PubMed] [Google Scholar]
- 107.Seo HK; Ahn KO; Jung NR; Shin JS; Park WS; Lee KH; Lee SJ; Jeong KC, Antitumor activity of the c-Myc inhibitor KSI-3716 in gemcitabine-resistant bladder cancer. Oncotarget 2014, 5 (2), 326–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Struntz NB; Chen A; Deutzmann A; Wilson RM; Stefan E; Evans HL; Ramirez MA; Liang T; Caballero F; Wildschut MHE; Neel DV; Freeman DB; Pop MS; McConkey M; Muller S; Curtin BH; Tseng H; Frombach KR; Butty VL; Levine SS; Feau C; Elmiligy S; Hong JA; Lewis TA; Vetere A; Clemons PA; Malstrom SE; Ebert BL; Lin CY; Felsher DW; Koehler AN, Stabilization of the Max Homodimer with a Small Molecule Attenuates Myc-Driven Transcription. Cell Chem Biol 2019, 26 (5), 711–723.e14. [DOI] [PubMed] [Google Scholar]
- 109.Whitfield JR; Soucek L, The long journey to bring a Myc inhibitor to the clinic. J Cell Biol 2021, 220 (8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Massó-Vallés D; Soucek L, Blocking Myc to Treat Cancer: Reflecting on Two Decades of Omomyc. Cells 2020, 9 (4), 883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Beaulieu ME; Jauset T; Massó-Vallés D; Martínez-Martín S; Rahl P; Maltais L; Zacarias-Fluck MF; Casacuberta-Serra S; Serrano Del Pozo E; Fiore C; Foradada L; Cano VC; Sánchez-Hervás M; Guenther M; Romero Sanz E; Oteo M; Tremblay C; Martín G; Letourneau D; Montagne M; Morcillo Alonso M; Whitfield JR; Lavigne P; Soucek L, Intrinsic cell-penetrating activity propels Omomyc from proof of concept to viable anti-MYC therapy. Sci Transl Med 2019, 11 (484), eaar5012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gu Y; Zhang J; Ma X; Kim BW; Wang H; Li J; Pan Y; Xu Y; Ding L; Yang L; Guo C; Wu X; Wu J; Wu K; Gan X; Li G; Li L; Forman SJ; Chan WC; Xu R; Huang W, Stabilization of the c-Myc Protein by CAMKIIgamma Promotes T Cell Lymphoma. Cancer Cell 2017, 32 (1), 115–128 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Bretones G; Delgado MD; Leon J, Myc and cell cycle control. Biochim Biophys Acta 2015, 1849 (5), 506–16. [DOI] [PubMed] [Google Scholar]
- 114.Bidwell GL 3rd; Davis AN; Raucher D, Targeting a c-Myc inhibitory polypeptide to specific intracellular compartments using cell penetrating peptides. J Control Release 2009, 135 (1), 2–10. [DOI] [PubMed] [Google Scholar]
- 115.Smith MC; Gestwicki JE, Features of protein-protein interactions that translate into potent inhibitors: topology, surface area and affinity. Expert Rev Mol Med 2012, 14, e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Martinez-Martin S; Soucek L, MYC inhibitors in multiple myeloma. Cancer Drug Resist 2021, 4 (4), 842–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Berg T; Cohen SB; Desharnais J; Sonderegger C; Maslyar DJ; Goldberg J; Boger DL; Vogt PK, Small-molecule antagonists of Myc/Max dimerization inhibit Myc-induced transformation of chicken embryo fibroblasts. Proc Natl Acad Sci U S A 2002, 99 (6), 3830–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bidwell GL 3rd; Perkins E; Hughes J; Khan M; James JR; Raucher D, Thermally targeted delivery of a c-Myc inhibitory polypeptide inhibits tumor progression and extends survival in a rat glioma model. Plos One 2013, 8 (1), e55104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ting TA; Chaumet A; Bard FA, Targeting c-Myc with a novel Peptide Nuclear Delivery Device. Sci Rep 2020, 10 (1), 17762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wang E; Sorolla A; Cunningham PT; Bogdawa HM; Beck S; Golden E; Dewhurst RE; Florez L; Cruickshank MN; Hoffmann K; Hopkins RM; Kim J; Woo AJ; Watt PM; Blancafort P, Tumor penetrating peptides inhibiting MYC as a potent targeted therapeutic strategy for triple-negative breast cancers. Oncogene 2019, 38 (1), 140–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Soodgupta D; Pan D; Cui G; Senpan A; Yang X; Lu L; Weilbaecher KN; Prochownik EV; Lanza GM; Tomasson MH, Small Molecule MYC Inhibitor Conjugated to Integrin-Targeted Nanoparticles Extends Survival in a Mouse Model of Disseminated Multiple Myeloma. Molecular Cancer Therapeutics 2015, 14 (6), 1286–1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Garralda E; Moreno V; Alonso G; Corral E; Hernandez-Guerrero T; Ramon J; Doger de Spéville B; Martinez E; Soucek L; Niewel M; Calvo E, Dose escalation study of OMO-103, a first in class Pan-MYC-Inhibitor in patients (pts) with advanced solid tumors. European Journal of Cancer 2022, 174, S5–S6. [Google Scholar]