The p53 tumor suppressor is activated in response to cellular stresses to induce cell-cycle arrest, cellular senescence, and apoptosis. The p53 gene is inactivated by mutations in more than 50% of human tumors. In addition, tumor cells dampen p53 activities via overexpression of p53-negative regulators, in particular 2 structurally related proteins, Mdm2 and Mdm4. And yet, Mdm2 and Mdm4 possess p53-independent activities, which also contribute to tumor formation and progression. Given that Mdm2 and Mdm4 inhibit p53 activities to promote tumor development, small molecules and peptides were developed to abrogate the inhibition of p53 by Mdm proteins. Antitumor activities of these molecules have already been confirmed in preclinical studies and early-phase clinical trials. These research endeavors and clinical advances constitute the main focus of this review. Clin Cancer Res; 19(1); 34–41. ©2012 AACR.

p53, the most frequently inactivated gene in human cancers, is mutated in more than half of human tumors. In response to various extra- and intracellular stresses including but not limited to oncogene activation, DNA damage, and hypoxia, p53 functions as a transcriptional factor and transactivates a set of genes engaged in multiple cellular processes such as cell-cycle arrest, cellular senescence, energy metabolism, and apoptosis. p53 signaling is also alternatively inactivated by high levels of p53 inhibitors. Suppression of p53 activities is largely attributed to 2 major negative regulators of p53, Mdm2 and Mdm4 (reviewed in ref. 1). Different mechanisms increase the levels of Mdm2 and/or Mdm4 in a large number of tumors that carry wild-type p53 alleles. These observations suggest that upregulation of Mdm2 and/or Mdm4 serves as an alternate means of inactivating the p53 pathway, further highlighting that silencing p53 signaling is a more common event in tumorigenesis than previously thought.

In this review, we will discuss the regulation of p53 by the Mdm proteins, p53-independent oncogenic functions of Mdm2 and Mdm4, as well as potential pharmaceuticals targeting Mdm2 and Mdm4 in cancer treatment.

The murine double minute 2 (Mdm2) was first identified as one of the genes that were amplified on double minute chromosomes in transformed mouse NIH-3T3 fibroblasts (2). Early studies showed that Mdm2 could bind the p53 transactivation domain and repress its transcriptional activity (3, 4). In addition, Mdm2 functions as an E3 ubiquitin ligase, mediates ubiquitination of p53, and targets it for degradation (reviewed in ref. 1). By exploiting the beauty of genetic models, 2 research groups independently showed that biallelic deletion of p53 rescued the embryonic lethal phenotypes in Mdm2-deficient embryos (5, 6). Deletion of Mdm2 in specific types of cells such as neuronal progenitors and cardiomyocytes also leads to p53-dependent embryonic lethal phenotypes (7–9). These in vitro and in vivo data together suggest an essential function of Mdm2 as a negative regulator of p53 in numerous cell types.

Mdm4 was originally discovered as a p53-interacting protein through screening of a mouse embryo cDNA expression library (10). The Mdm4 and Mdm2 proteins are very similar at the primary structural level (10, 11). The p53-binding domains at the N-termini of Mdm4 and Mdm2 show very high structural and functional similarities (10). The p53-binding domain of Mdm4, like that of Mdm2, interacts with the transactivation domain of p53 to represses its transcriptional activity (10). Another prominent conserved domain between Mdm4 and Mdm2 is the RING-finger domain at the C-termini of both proteins (10, 11). The RING domain of Mdm2 is responsible for its E3 ubiquitin ligase function (12); however, the Mdm4 RING domain lacks E3 ligase activity (reviewed in ref. 1). Mice with Mdm4 RING domain alterations recently revealed that interaction of Mdm2 with Mdm4 through this RING domain is required for modulating p53 activities in embryonic stages but dispensable for Mdm2 and p53 stabilization in the adult mouse (13, 14).

Loss of Mdm4 in mice also results in embryonic lethality, which is completely rescued by concomitant p53 loss (15–17). These data indicate that Mdm4 negatively regulates p53 activity in vivo. Deletion of Mdm4 in mouse erythroid progenitors, embryonic neuronal progenitors, cardiomyocytes, and intestinal epithelia leads to distinct pathologic phenotypes, compared with that of Mdm2 (18–22). Deletion of both Mdm2 and Mdm4 in mouse embryonic neuronal progenitors results in a more severe phenotype compared with deletion of either gene alone (8). All Mdm loss-of-function phenotypes are rescued by deletion of p53 (7, 8, 18–20). Together, these data suggest that Mdm2 and Mdm4 function in a nonoverlapping manner to suppress p53 activities in vivo. Intriguingly, overexpression of Mdm2 also rescues the Mdm4−/− embryonic lethality (23), implying that high levels of Mdm2 can compensate for loss of Mdm4, possibly through inhibition of p53 activities.

Although the major function of Mdm2 is to suppress p53 activities, emerging evidence has identified p53-independent roles of Mdm2 in tumor formation and progression. Early studies showed that p53−/− mice carrying an Mdm2 transgene developed a higher percentage of sarcomas compared with p53−/− mice (24), supporting the p53-independent functions of Mdm2 in tumorigenesis. p53-independent activities of Mdm2 include regulation of genomic instability, apoptosis, and metastasis.

Elevated genomic instability is a hallmark of cancers (reviewed in ref. 25). Ectopic expression of Mdm2 in murine mammary epithelial cells and murine embryonic fibroblasts (MEF) increases ploidy and chromosome/chromatid breaks regardless of p53 status (reviewed in ref. 26), suggesting that Mdm2 promotes genomic instability independently of p53. Mdm2 induces genomic instability likely through inhibiting DNA damage repair and suppressing cell-cycle arrest. The repair of DNA double-strand breaks (DSB) caused by ionizing radiation (IR) requires the Mre11 complex, composed of Mre11, Rad50, and Nbs1 (27). Mdm2 directly binds and colocalizes with Nbs1 at DNA damage sites after IR treatment, and in turn inhibits DNA DSB repair (ref. 28; Fig. 1A). The p53-independent functions of Mdm2 in suppressing cell-cycle arrest involve the direct interaction with Retinoblastoma protein (Rb) and E2F transcription factors (Fig. 1A). The Rb protein interacts with several E2Fs, suppresses E2F-mediated transcription of cell-cycle essential genes, and therefore causes cell-cycle arrest especially at the G1–S transition (reviewed in ref. 29). Loss of Rb activity leads to genomic instability and tumorigenesis due to defective cell proliferation and chromosome missegregation (30). Mdm2 compromises Rb-mediated G1 arrest through multiple mechanisms: (i) Mdm2 directly binds Rb protein, suppresses the interaction between Rb and E2F1, and induces activation of E2F1; and (ii) Mdm2 targets Rb for ubiquitin-dependent and -independent degradation (reviewed in ref. 26; Fig. 1A). In addition, Mdm2 also directly binds E2F1 and activates its transcriptional activities (ref. 31; Fig. 1A). Moreover, Mdm2 targets other cell-cycle inhibitors, such as p21 and hnRNP K, for proteasomal degradation, and therefore promotes cell proliferation (reviewed in refs. 22, 26; Fig. 1A). Notably, p21 is involved in maintaining genomic stability (32), and loss of p21 induces aneuploidy, chromosomal aberrations, and accelerated tumor onset in mice (33). These data suggest that degradation of p21 by Mdm2 contributes to genomic aberrations (Fig. 1A). Thus, Mdm2 binds and alters the activities of several proteins involved in DNA repair and transition through the cell-cycle impacting ploidy and other chromosomal abnormalities.

Figure 1.

Schematic representation of p53-independent functions of Mdm2 and Mdm4. A, Mdm2 interacts with multiple factors to affect genomic instability, apoptosis, and metastasis. hnRNP, hnRNP K protein. B, Mdm4 interacts with p21 and E2F1 to regulate cell-cycle arrest and apoptosis.

Figure 1.

Schematic representation of p53-independent functions of Mdm2 and Mdm4. A, Mdm2 interacts with multiple factors to affect genomic instability, apoptosis, and metastasis. hnRNP, hnRNP K protein. B, Mdm4 interacts with p21 and E2F1 to regulate cell-cycle arrest and apoptosis.

Close modal

Additional data indicate a role of Mdm2 in inhibition of apoptosis independently of p53. FOXO3a, a forkhead transcription factor, induces apoptosis and decreases tumorigenicity of breast cancer cells (34), and inverse correlation between Mdm2 and FOXO3a expression exists in human breast cancer samples, because of Mdm2-mediated degradation of FOXO3a (ref. 34; Fig. 1A), indicating that Mdm2 may counteract FOXO3a-mediated apoptosis during tumor development. Unexpectedly, Mdm2 was also reported to suppress apoptosis via a different pathway by upregulating the antiapoptotic protein XIAP through translational regulation (ref. 35; Fig. 1A). Upon DNA damage, cytoplasmic Mdm2 binds the internal ribosome entry site at the XIAP 5′-UTR and enhances the translation of XIAP mRNA, which may confer resistance to radiation-induced apoptosis in tumor cells (35). The mechanism by which Mdm2 regulates this process is unknown.

Additional p53-independent functions of Mdm2 include the ability to promote epithelial-to-mesenchymal transition (EMT) and metastasis (Fig. 1A). The EMT effector TGF-β1 induces Mdm2 transcription via activation of Smad3 in murine mammary epithelial cells (36). Histopathologic analyses of human breast cancers indicate that activation of Smad3 and overexpression of Mdm2 coexist in 65% of late-stage carcinomas and are strongly correlated to metastatic phenotypes in patients with breast cancer (36), suggesting an important role of Mdm2 overexpression in metastasis. A mechanism by which Mdm2 promotes EMT and metastasis is through E-cadherin. Mdm2 binds E-cadherin, which targets it for degradation, thus inducing EMT (ref. 37; Fig. 1A). Overexpression of Mdm2 in breast cancer cells disrupts cell–cell contacts, enhances cell motility, and promotes cell invasiveness (37). High Mdm2 levels are also strongly associated with low E-cadherin levels in human metastatic breast cancer specimens (37). Thus, a series of experiments in breast cancers indicate that high levels of Mdm2 correlate with metastasis.

Data for p53-independent functions of Mdm4 are, by far, more limited. Mdm4 was found to interact with p21 and target it for proteasomal degradation independently of ubiquitination (ref. 38; Fig. 1B). Degradation of p21 mediated by Mdm4 is independent of, but in cooperation with, Mdm2 and leads to abrogation of G1 cell-cycle arrest (38). Surprisingly, loss of Mdm4 in p53-null cells leads to multipolar spindle formation, enhanced loss of chromosomes, elevated proliferation potentials, and increased spontaneous transformation as compared with p53−/− cells (39). In addition, deletion of Mdm4 results in accelerated tumorigenesis in both p53+/− and p53−/− mice (39). As Mdm4 forms heterodimers with Mdm2 (reviewed in ref. 1), loss of Mdm4 may promote interaction of Mdm2 with other proteins such as Rb and p21 and thus enhances tumorigenesis. Moreover, Mdm4 represses E2F1 transactivation by either disrupting E2F1-DNA binding (40) or altering localization of E2F1 transcription complex (ref. 41; Fig. 1B). Overexpression of E2F1 promotes G1–S transition in cells, and is associated with tumorigenesis (42). However, upregulation of E2F1 also triggers both p53-dependent and-independent apoptosis (reviewed in ref. 43). These data together suggest that upregulation of Mdm4 that is present in many human tumors (44), in addition to suppressing p53 activity, may represent a mechanism for tumor cells to overcome apoptosis induced by elevated E2F1 levels.

The functions of Mdm2 and Mdm4 including p53 inhibition and p53-independent activities all suggest important roles in tumorigenesis and tumor progression. High levels of Mdm2 are commonly observed in human cancers including sarcomas, gliomas, hematologic malignancies, melanomas, and carcinomas (reviewed in ref. 45). By analyzing gene amplification and overexpression status of Mdm2 in human soft tissue tumors, Patterson and colleagues (46) showed that high levels of Mdm2 detected by immunohistochemistry occurred in tumors with Mdm2 gene amplification, but were also found in tumors without Mdm2 gene amplification (46). This observation indicates that Mdm2 gene amplification indeed results in its overexpression, but other mechanisms may also confer Mdm2 upregulation. Extensive studies have indicated that upregulation of Mdm2 results from elevated transcription, increased mRNA stability, enhanced translation, and altered posttranslational modification (reviewed in refs. 22, 47). In addition, these mechanisms are associated with upregulation of Mdm2 in tumors.

High levels of Mdm4 are also found in a variety of human cancers. Mdm4 upregulation in malignancies is mostly ascribed to Mdm4 gene amplification (reviewed in ref. 44). Gene amplification and consequent upregulation of Mdm4 are reported in multiple types of human tumors including glioma, soft tissue sarcoma, head and neck squamous carcinoma, retinoblastoma, melanoma, and breast cancer (reviewed in ref. 44). To date, information about transcriptional regulation of Mdm4 remains extremely limited. Stimulation with mitogens, for example, IGF-1, leads to phosphorylation of mitogen-activated protein kinase (MAPK) ERK and thus activates transcriptional factors c-Ets-1 and Elk-1, which bind the Mdm4 promoter and induce Mdm4 expression (48). Human colon tumors that are stained positive for phosphorylated ERK are 2-fold more likely to have high levels of Mdm4 (48), implying a role of Mdm4 overexpression stimulated by the MAPK in tumor development. Mdm4-S, the first Mdm4 transcript variant identified in human and mouse cells, produces an Mdm4 isoform containing the p53-binding domain but missing the negative regulatory sites, which is more stable and oncogenic than full-length Mdm4 (reviewed in ref. 49). Overexpression of Mdm4-S has been found in several types of cancers, and is associated with decreased survival and poor prognosis for patients with cancer (reviewed in refs. 44, 49), indicating a role of Mdm4-S in tumor development.

A number of studies have modeled Mdm2 and Mdm4 overexpression in vivo using genetically engineered mice. Transgenic mice overexpressing Mdm2/Mdm4 either globally or in specific tissues/cell types all develop spontaneous tumors (24, 50–54). A T to G single-nucleotide polymorphism (SNP309) in the human Mdm2 P2 promoter creates an additional binding site for the transcriptional activator Sp1, leads to elevated Mdm2 expression, and attenuates p53 signaling (55). An association between SNP309 and earlier tumor onset is revealed in both patients with hereditary and sporadic cancer (55). A knockin mouse with a humanized Mdm2 intron 1 harboring SNP309 was generated in our laboratory and develops multiple types of malignancies with accelerated onset in both p53 wild-type and p53 mutant backgrounds, providing causal evidence for the role SNP309 in tumorigenesis (54). Approximately 65% of human retinoblastomas undergo gene amplification of Mdm4 and 10% have extra copies of Mdm2, but no p53 mutations or defects downstream to p53 are found in the retinoblastomas caused by Rb loss (56), suggesting that p53 is inactivated in retinoblastomas via overexpression of p53 negative regulators. A very low retinoblastoma penetrance is observed in mice with loss of Rb family genes Rb and p107 in retina (56), but ectopic expression of Mdm4 in retina significantly promotes retinoblastoma development in these mice with Rb and p107 loss (56). In addition, the treatment of mice with p53-activating drugs leads to growth suppression of Mdm4-overepressing retinoblastomas (56), suggesting that p53 remains intact in these tumors. Therefore, this retinoblastoma mouse model recapitulates human defects and emphasizes the causative role of Mdm4 overexpression in retinoblastoma formation. In a recent study, Mdm4 is overexpressed in 65% of human melanoma samples examined, and p53 mutations are rare among these melanoma cases (57). Specific overexpression of Mdm4 in the murine melanocytes enhances tumorigenesis of Nras-induced melanoma in both p53 wild-type and heterozygous backgrounds, whereas knockdown of Mdm4 in human melanoma cells leads to increased cell death but reduced cell proliferation and oncogenic ability (57). However, the p53 status in these mouse melanomas has not been examined. Nevertheless, this melanoma mouse model elucidates the tumorigenic roles of Mdm4 overexpression in melanoma formation in vivo.

Targeting Mdm proteins in cancer therapy

Clearly, Mdm proteins are present at high levels and inhibit p53 activities in many human cancers. Thus, disrupting the Mdm-p53 interaction to reactivate p53 is a valuable therapeutic strategy for tumor treatment.

Several strategies have been used to develop small molecules to block Mdm2–p53 interaction. Using structure-based design and high-throughput screening, several groups independently developed distinct small molecules including Nutlins (58), benzodiazepinediones (59), and MI-63/MI-219 (refs. 60, 61; Fig. 2) that bind Mdm2 in its p53-binding pocket and disrupt Mdm2–p53 interaction (58–61). Treatment with these small molecules leads to elevated p53 protein levels, induction of p53 target gene expression, decrease in cell proliferation, upregulation of apoptosis, and reduced tumorigenic ability in a variety of tumor cell lines carrying wild-type but not mutant p53 (58–61). Notably, no significant weight loss or any other gross abnormalities is observed in the mice undergoing Nutlin-3 or MI-219 treatment (58, 61). Another small molecule, RITA (Fig. 2), binds the p53 N-terminus instead of Mdm2 and blocks its interaction with Mdm2 (62). Treatment of cells with RITA results in similar effects as treatment with Nutlins or MI-219 (62). A novel strategy using stapled peptides is also applied to block interaction between Mdm2 and p53 (63). These small synthetic peptides, locked into certain folded shapes by an all-hydrocarbon crosslink, are optimized for protein interaction and are resistant to proteolysis (63). Stapled peptides SAH-p53s (Fig. 2) compete with p53 for Mdm2 binding, reactivate p53, and lead to apoptosis in cancer cells with high levels of Mdm2 (63). Yang and colleagues showed that HLI98 (Fig. 2), a family of small molecules inhibiting Mdm2, could bind Mdm2 at the C-terminus and compromise its E3 ubiquitin ligase activity (64). HLI98 treatment suppresses degradation of p53 and induces p53-dependent apoptosis in tumor cells (64). A different small molecule, JNJ-26854165 (Fig. 2), reactivates p53 by inhibiting Mdm2 E3 activity and inhibits tumor growth in mouse xenografts modeling multiple types of human cancers (65). As p53-independent G2 arrest was observed in tumor cells upon HLI98 treatment (64), small molecules that bind the Mdm2 RING domain may also inhibit p53-independent functions of Mdm2.

Figure 2.

Schematic representation of the targeting sites of Mdm protein antagonists on Mdm2, Mdm4, and p53. BDP, benzodiazepinedione; MI, MI-63/219; Stapled, SAH-p53 stapled peptides; JNJ, JNJ-26854165; SJ, SJ-172550; p53 binding, p53 binding domain; Acidic, acidic domain; Zn Finger, Zn finger domain; RING, RING domain; TA, transactivation domain; PR, proline-rich domain; DNA binding, DNA binding domain; oligo, oligomerization domain.

Figure 2.

Schematic representation of the targeting sites of Mdm protein antagonists on Mdm2, Mdm4, and p53. BDP, benzodiazepinedione; MI, MI-63/219; Stapled, SAH-p53 stapled peptides; JNJ, JNJ-26854165; SJ, SJ-172550; p53 binding, p53 binding domain; Acidic, acidic domain; Zn Finger, Zn finger domain; RING, RING domain; TA, transactivation domain; PR, proline-rich domain; DNA binding, DNA binding domain; oligo, oligomerization domain.

Close modal

The small-molecule inhibitors of Mdm2 described above show very low affinity for Mdm4 binding. But high levels of Mdm4 attenuate the apoptotic response to the small molecules targeting Mdm2–p53 interaction, for example, Nutlin-3a and MI-219 in tumor cells (57, 61, 66, 67). Given the importance of Mdm4 in inhibition of p53 and its presence at high levels in a variety of human tumors such as retinoblastomas and melanomas, efforts have recently been dedicated to the development of Mdm4 inhibitors. A small molecule, SJ-172550 (Fig. 2), targets p53–Mdm4 binding (68). A benzofuroxan derivative, XI-006, suppresses the transcription of Mdm4 (69). Both induce p53-dependent apoptosis, and act additively with Nutlin-3a in human cancer cell lines, which retain wild-type p53 but overexpress Mdm4 (68, 69). These observations suggest that an inhibitor against both Mdm2 and Mdm4 may promise a greater potential for antitumor activities. A stapled peptide, SAH-p53-8 (Fig. 2), binds both Mdm2 and Mdm4 and disrupts their interaction with p53 (67). Treatment of SAH-p53-8 reduces the viability of wild-type p53-carrying cancer cells that also contain high levels of Mdm2 and/or Mdm4 proteins (67). Of note, wild-type p53-carrying cancer cells with high levels of Mdm4 show minimal response to Nutlin-3 treatment (67). Recently, a dual Mdm2/Mdm4 antagonist, RO-5963 (Fig. 2), was identified that blocks p53 interaction with both Mdm2 and Mdm4 by inducing formation of homo- or heterodimers between Mdm proteins through their p53-binding domains (70). In the p53 wild-type, but not mutant tumor cells, the dual antagonist induces apoptotic activity even in the presence of high levels of Mdm4, whereas Nutlin-3a induces a much weaker apoptotic response (70).

To date, a phase I clinical study of JNJ-26854165 (NCT00676910) to determine safety and dosing in patients with advanced stage or refractory solid tumors has been completed. Although JNJ-26854165 was well tolerated in these patients, no objective responses were observed (71). Several phase I studies of a Nutlin family member, RG7112, in patients with liposarcomas (before debulking surgery), solid tumors, hematologic neoplasms, and soft tissue sarcomas (NCT01143740, NCT01164033, NCT00559533, NCT00623870, and NCT01605526) have been completed or are in progress. Acceptable safety profiles and favorable clinical outcomes including complete remission, induction of apoptosis, and decrease in proliferation were seen in patients treated with RG7112 for leukemias, lymphomas, sarcomas, and other solid tumors (72–76). Further studies are needed to verify the efficacy of this agent in long-term cancer treatment.

Despite these initial outcomes, concerns arise for potential adverse effects of Mdm2/Mdm4 inhibitor treatment of patients with cancer. Genetically restoring p53 activity in mice in the absence of Mdm2 leads to ablation of the classically radiosensitive tissues including bone marrow, spleen, thymus, and intestine, which results in 100% fatality of mice within 5 to 6 days (77). In addition, mice heterozygous for Mdm2 or Mdm4, although normal, are sensitive to low-dose IR treatment (78), suggesting that the haploinsufficiency of Mdm2 and Mdm4 in response to DNA damage is detrimental for the organism. These 2 observations question whether p53 activated by Mdm2/Mdm4 inhibitors will be toxic to normal tissues especially under stressed conditions. Xenograft models indicate that the tumor-suppressing doses of Nutlin-3, MI-219, and SAH-p53-8 are well tolerated in mice, leading to no significant weight loss or any gross abnormalities (58, 61, 67). Moreover, the treatment with Nutlin-3 in combination of the therapeutic DNA-damaging agent topotecan results in regression of retinoblastomas without any obvious side effects in mice (56), implying that Mdm protein inhibitors may not be toxic to normal tissues even when combined with other therapeutic agents in patients. However, these data should be cautiously interpreted, as these preclinical studies in mice were short-term, ranging from 6 to 20 days. It still needs to be examined whether prolonged administration of Mdm inhibitors together with other antitumor agents will be detrimental to patients.

Other studies have shown the interaction of Mdm2 with mutant p53 (79, 80). Disruption of this interaction causes stabilization of mutant p53 (79, 80), which has been shown to have gain-of-function metastatic activities (81). Treatment with Mdm2 inhibitors might lead to unfavorable outcomes in patients with even one allele of mutant p53. Therefore, it is extremely important that Mdm2 inhibitor treatment should be given to patients with tumors carrying only wild-type and not mutant p53. Moreover, Nutlin-3 treatment of SJSA-1 tumor cells (p53 wild-type) leads to selection for p53 mutations and subsequent resistance to Nutlin-3 treatment (82). In addition, selected p53 mutants may lead to more adverse effects, if mutant p53 accumulates and exerts gain-of-function activities in the resistant cells. Thus, combinational therapies and intertreatment evaluation of p53 status should be a standard-of-care for patients with cancer administered Mdm inhibitors. Finally, accumulation of Mdm2 proteins are observed when Mdm2/Mdm4 inhibitors are applied to normal and tumor cells (58–60, 62, 64, 66, 69, 70), and accumulated Mdm2 can execute p53-independent oncogenic functions as discussed in this review. Therefore, administration of Mdm2/Mdm4 inhibitors to patients with cancer might induce oncogenic side effects, which may be evident only after a long latency. Thus, studies with long-term follow-up will be necessary to evaluate the safety of these inhibitors.

Studies of Mdm2 and Mdm4 have long been focused on their roles as the negative regulators of the p53 tumor suppressor. Small molecules and synthetic peptides antagonizing Mdm2 and/or Mdm4 reactivate p53 in preclinical studies and have produced favorable outcomes in patients with cancer in early-phase clinical trials. And yet, accumulating evidence has implicated p53-independent functions of Mdm2 and Mdm4, which accelerate tumorigenesis and tumor progression. However, it remains largely unclear whether inhibiting the binding of Mdm proteins to p53 compromises their p53-independent functions. Therefore, mouse models carrying mutations that will abrogate or enhance the p53-independent functions of Mdm proteins may serve as valuable tools to examine the relationship between these functions and tumor phenotypes. Furthermore, drugs targeting these p53-independent activities of Mdm2 and Mdm4 may potentiate the effectiveness of pharmaceuticals targeting Mdm2 and Mdm4 to reactivate p53 in cancer treatment.

No potential conflicts of interest were disclosed.

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Q. Li

Writing, review, and/or revision of the manuscript: Q. Li, G. Lozano

The authors thank Drs. Shunbin Xiong and Vinod Pant for critical reading of the manuscript.

These studies are supported by NIH grant CA47296 (to G. Lozano) and M. D. Anderson CPRIT training grant and Hearst Foundation Scholarship (to Q. Li).

1.
Marine
JC
,
Francoz
S
,
Maetens
M
,
Wahl
G
,
Toledo
F
,
Lozano
G
. 
Keeping p53 in check: essential and synergistic functions of Mdm2 and Mdm4
.
Cell Death and Differ
2006
;
13
:
927
34
.
2.
Fakharzadeh
SS
,
Trusko
SP
,
George
DL
. 
Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line
.
EMBO J
1991
;
10
:
1565
9
.
3.
Oliner
JD
,
Pietenpol
JA
,
Thiagalingam
S
,
Gyuris
J
,
Kinzler
KW
,
Vogelstein
B
. 
Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53
.
Nature
1993
;
362
:
857
60
.
4.
Momand
J
,
Zambetti
GP
,
Olson
DC
,
George
D
,
Levine
AJ
. 
The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation
.
Cell
1992
;
69
:
1237
45
.
5.
Jones
SN
,
Roe
AE
,
Donehower
LA
,
Bradley
A
. 
Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53
.
Nature
1995
;
378
:
206
8
.
6.
Montes de Oca Luna
R
,
Wagner
DS
,
Lozano
G
. 
Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53
.
Nature
1995
;
378
:
203
6
.
7.
Grier
JD
,
Xiong
S
,
Elizondo-Fraire
AC
,
Parant
JM
,
Lozano
G
. 
Tissue-specific differences of p53 inhibition by Mdm2 and Mdm4
.
Mol Cell Biol
2006
;
26
:
192
8
.
8.
Xiong
S
,
Van Pelt
CS
,
Elizondo-Fraire
AC
,
Liu
G
,
Lozano
G
. 
Synergistic roles of Mdm2 and Mdm4 for p53 inhibition in central nervous system development
.
Proc Natl Acad Sci U S A
2006
;
103
:
3226
31
.
9.
Francoz
S
,
Froment
P
,
Bogaerts
S
,
De Clercq
S
,
Maetens
M
,
Doumont
G
, et al
Mdm4 and Mdm2 cooperate to inhibit p53 activity in proliferating and quiescent cells in vivo
.
Proc Natl Acad Sci U S A
2006
;
103
:
3232
7
.
10.
Shvarts
A
,
Steegenga
WT
,
Riteco
N
,
van Laar
T
,
Dekker
P
,
Bazuine
M
, et al
MDMX: a novel p53-binding protein with some functional properties of MDM2
.
EMBO J
1996
;
15
:
5349
57
.
11.
Shvarts
A
,
Bazuine
M
,
Dekker
P
,
Ramos
YF
,
Steegenga
WT
,
Merckx
G
, et al
Isolation and identification of the human homolog of a new p53-binding protein, Mdmx
.
Genomics
1997
;
43
:
34
42
.
12.
Fang
S
,
Jensen
JP
,
Ludwig
RL
,
Vousden
KH
,
Weissman
AM
. 
Mdm2 is a RING finger-dependent ubiquitin protein ligase for itself and p53
.
J Biol Chem
2000
;
275
:
8945
51
.
13.
Pant
V
,
Xiong
S
,
Iwakuma
T
,
Quintas-Cardama
A
,
Lozano
G
. 
Heterodimerization of Mdm2 and Mdm4 is critical for regulating p53 activity during embryogenesis but dispensable for p53 and Mdm2 stability
.
Proc Natl Acad Sci U S A
2011
;
108
:
11995
2000
.
14.
Huang
L
,
Yan
Z
,
Liao
X
,
Li
Y
,
Yang
J
,
Wang
ZG
, et al
The p53 inhibitors MDM2/MDMX complex is required for control of p53 activity in vivo
.
Proc Natl Acad Sci U S A
2011
;
108
:
12001
6
.
15.
Parant
J
,
Chavez-Reyes
A
,
Little
NA
,
Yan
W
,
Reinke
V
,
Jochemsen
AG
, et al
Rescue of embryonic lethality in Mdm4-null mice by loss of Trp53 suggests a nonoverlapping pathway with MDM2 to regulate p53
.
Nat Genet
2001
;
29
:
92
5
.
16.
Finch
RA
,
Donoviel
DB
,
Potter
D
,
Shi
M
,
Fan
A
,
Freed
DD
, et al
Mdmx is a negative regulator of p53 activity in vivo
.
Cancer Res
2002
;
62
:
3221
5
.
17.
Migliorini
D
,
Lazzerini Denchi
E
,
Danovi
D
,
Jochemsen
A
,
Capillo
M
,
Gobbi
A
, et al
Mdm4 (Mdmx) regulates p53-induced growth arrest and neuronal cell death during early embryonic mouse development
.
Mol Cell Biol
2002
;
22
:
5527
38
.
18.
Xiong
S
,
Van Pelt
CS
,
Elizondo-Fraire
AC
,
Fernandez-Garcia
B
,
Lozano
G
. 
Loss of Mdm4 results in p53-dependent dilated cardiomyopathy
.
Circulation
2007
;
115
:
2925
30
.
19.
Valentin-Vega
YA
,
Okano
H
,
Lozano
G
. 
The intestinal epithelium compensates for p53-mediated cell death and guarantees organismal survival
.
Cell Death Differ
2008
;
15
:
1772
81
.
20.
Valentin-Vega
YA
,
Box
N
,
Terzian
T
,
Lozano
G
. 
Mdm4 loss in the intestinal epithelium leads to compartmentalized cell death but no tissue abnormalities
.
Differentiation
2009
;
77
:
442
9
.
21.
Maetens
M
,
Doumont
G
,
Clercq
SD
,
Francoz
S
,
Froment
P
,
Bellefroid
E
, et al
Distinct roles of Mdm2 and Mdm4 in red cell production
.
Blood
2007
;
109
:
2630
3
.
22.
Marine
J-C
. 
Chapter 3—MDM2 and MDMX in cancer and development
.
In: Michael AD, ed. Current topics in developmental biology
.
Academic Press
USA
; 
2011
.
p.
45
75
.
23.
Steinman
HA
,
Hoover
KM
,
Keeler
ML
,
Sands
AT
,
Jones
SN
. 
Rescue of Mdm4-deficient mice by Mdm2 reveals functional overlap of Mdm2 and Mdm4 in development
.
Oncogene
2005
;
24
:
7935
40
.
24.
Jones
SN
,
Hancock
AR
,
Vogel
H
,
Donehower
LA
,
Bradley
A
. 
Overexpression of Mdm2 in mice reveals a p53-independent role for Mdm2 in tumorigenesis
.
Proc Natl Acad Sci U S A
1998
;
95
:
15608
12
.
25.
Negrini
S
,
Gorgoulis
VG
,
Halazonetis
TD
. 
Genomic instability—an evolving hallmark of cancer
.
Nat Rev Mol Cell Biol
2010
;
11
:
220
8
.
26.
Bouska
A
,
Eischen
CM
. 
Murine double minute 2: p53-independent roads lead to genome instability or death
.
Trends Biochem Sci
2009
;
34
:
279
86
.
27.
Stracker
TH
,
Petrini
JH
. 
The MRE11 complex: starting from the ends
.
Nat Rev Mol Cell Biol
2011
;
12
:
90
103
.
28.
Alt
JR
,
Bouska
A
,
Fernandez
MR
,
Cerny
RL
,
Xiao
H
,
Eischen
CM
. 
Mdm2 binds to Nbs1 at sites of DNA damage and regulates double strand break repair
.
J Biol Chem
2005
;
280
:
18771
81
.
29.
Harbour
JW
,
Dean
DC
. 
The Rb/E2F pathway: expanding roles and emerging paradigms
.
Genes Dev
2000
;
14
:
2393
409
.
30.
Manning
AL
,
Dyson
NJ
. 
pRB, a tumor suppressor with a stabilizing presence
.
Trends Cell Biol
2011
;
21
:
433
41
.
31.
Martin
K
,
Trouche
D
,
Hagemeier
C
,
Sorensen
TS
,
La Thangue
NB
,
Kouzarides
T
. 
Stimulation of E2F1/DP1 transcriptional activity by MDM2 oncoprotein
.
Nature
1995
;
375
:
691
4
.
32.
Begus-Nahrmann
Y
,
Lechel
A
,
Obenauf
AC
,
Nalapareddy
K
,
Peit
E
,
Hoffmann
E
, et al
p53 deletion impairs clearance of chromosomal-instable stem cells in aging telomere-dysfunctional mice
.
Nat Genet
2009
;
41
:
1138
43
.
33.
Barboza
JA
,
Liu
G
,
Ju
Z
,
El-Naggar
AK
,
Lozano
G
. 
p21 delays tumor onset by preservation of chromosomal stability
.
Proc Natl Acad Sci U S A
2006
;
103
:
19842
7
.
34.
Yang
JY
,
Zong
CS
,
Xia
W
,
Yamaguchi
H
,
Ding
Q
,
Xie
X
, et al
ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation
.
Nat Cell Biol
2008
;
10
:
138
48
.
35.
Gu
L
,
Zhu
N
,
Zhang
H
,
Durden
DL
,
Feng
Y
,
Zhou
M
. 
Regulation of XIAP translation and induction by MDM2 following irradiation
.
Cancer Cell
2009
;
15
:
363
75
.
36.
Araki
S
,
Eitel
JA
,
Batuello
CN
,
Bijangi-Vishehsaraei
K
,
Xie
XJ
,
Danielpour
D
, et al
TGF-beta1-induced expression of human Mdm2 correlates with late-stage metastatic breast cancer
.
J Clin Invest
2010
;
120
:
290
302
.
37.
Yang
JY
,
Zong
CS
,
Xia
W
,
Wei
Y
,
Ali-Seyed
M
,
Li
Z
, et al
MDM2 promotes cell motility and invasiveness by regulating E-cadherin degradation
.
Mol Cell Biol
2006
;
26
:
7269
82
.
38.
Jin
Y
,
Zeng
SX
,
Sun
XX
,
Lee
H
,
Blattner
C
,
Xiao
Z
, et al
MDMX promotes proteasomal turnover of p21 at G1 and early S phases independently of, but in cooperation with, MDM2
.
Mol Cell Biol
2008
;
28
:
1218
29
.
39.
Matijasevic
Z
,
Steinman
HA
,
Hoover
K
,
Jones
SN
. 
MdmX promotes bipolar mitosis to suppress transformation and tumorigenesis in p53-deficient cells and mice
.
Mol Cell Biol
2008
;
28
:
1265
73
.
40.
Strachan
GD
,
Jordan-Sciutto
KL
,
Rallapalli
R
,
Tuan
RS
,
Hall
DJ
. 
The E2F-1 transcription factor is negatively regulated by its interaction with the MDMX protein
.
J Cell Biochem
2003
;
88
:
557
68
.
41.
Wunderlich
M
,
Ghosh
M
,
Weghorst
K
,
Berberich
SJ
. 
MdmX represses E2F1 transactivation
.
Cell Cycle
2004
;
3
:
472
8
.
42.
Crosby
ME
,
Almasan
A
. 
Opposing roles of E2Fs in cell proliferation and death
.
Cancer Biol Ther
2004
;
3
:
1208
11
.
43.
Stanelle
J
,
Putzer
BM
. 
E2F1-induced apoptosis: turning killers into therapeutics
.
Trends Mol Med
2006
;
12
:
177
85
.
44.
Markey
MP
. 
Regulation of MDM4
.
Front Biosci
2011
;
16
:
1144
56
.
45.
Onel
K
,
Cordon-Cardo
C
. 
MDM2 and prognosis
.
Mol Cancer Res
2004
;
2
:
1
8
.
46.
Patterson
H
,
Barnes
D
,
Gill
S
,
Spicer
J
,
Fisher
C
,
Thomas
M
, et al
Amplification and over-expression of the MDM2 gene in human soft tissue tumours
.
Sarcoma
1997
;
1
:
17
22
.
47.
Riley
MF
,
Lozano
G
. 
The many faces of MDM2 binding partners
.
Genes Cancer
2012
;
3
:
226
39
.
48.
Gilkes
DM
,
Pan
Y
,
Coppola
D
,
Yeatman
T
,
Reuther
GW
,
Chen
J
. 
Regulation of MDMX expression by mitogenic signaling
.
Mol Cell Biol
2008
;
28
:
1999
2010
.
49.
Mancini
F
,
Di Conza
G
,
Moretti
F
. 
MDM4 (MDMX) and its transcript variants
.
Curr Genomics
2009
;
10
:
42
50
.
50.
Lundgren
K
,
Montes de Oca Luna
R
,
McNeill
YB
,
Emerick
EP
,
Spencer
B
,
Barfield
CR
, et al
Targeted expression of MDM2 uncouples S phase from mitosis and inhibits mammary gland development independent of p53
.
Genes Dev
1997
;
11
:
714
25
.
51.
Ganguli
G
,
Abecassis
J
,
Wasylyk
B
. 
MDM2 induces hyperplasia and premalignant lesions when expressed in the basal layer of the epidermis
.
EMBO J
2000
;
19
:
5135
47
.
52.
Alkhalaf
M
,
Ganguli
G
,
Messaddeq
N
,
Le Meur
M
,
Wasylyk
B
. 
MDM2 overexpression generates a skin phenotype in both wild type and p53 null mice
.
Oncogene
1999
;
18
:
1419
34
.
53.
Xiong
S
,
Pant
V
,
Suh
YA
,
Van Pelt
CS
,
Wang
Y
,
Valentin-Vega
YA
, et al
Spontaneous tumorigenesis in mice overexpressing the p53-negative regulator Mdm4
.
Cancer Res
2010
;
70
:
7148
54
.
54.
Post
SM
,
Quintas-Cardama
A
,
Pant
V
,
Iwakuma
T
,
Hamir
A
,
Jackson
JG
, et al
A high-frequency regulatory polymorphism in the p53 pathway accelerates tumor development
.
Cancer Cell
2010
;
18
:
220
30
.
55.
Bond
GL
,
Hu
W
,
Bond
EE
,
Robins
H
,
Lutzker
SG
,
Arva
NC
, et al
A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans
.
Cell
2004
;
119
:
591
602
.
56.
Laurie
NA
,
Donovan
SL
,
Shih
CS
,
Zhang
J
,
Mills
N
,
Fuller
C
, et al
Inactivation of the p53 pathway in retinoblastoma
.
Nature
2006
;
444
:
61
6
.
57.
Gembarska
A
,
Luciani
F
,
Fedele
C
,
Russell
EA
,
Dewaele
M
,
Villar
S
, et al
MDM4 is a key therapeutic target in cutaneous melanoma
.
Nat Med
2012
;
18
:
1239
47
.
58.
Vassilev
LT
,
Vu
BT
,
Graves
B
,
Carvajal
D
,
Podlaski
F
,
Filipovic
Z
, et al
In vivo activation of the p53 pathway by small-molecule antagonists of MDM2
.
Science
2004
;
303
:
844
8
.
59.
Grasberger
BL
,
Lu
T
,
Schubert
C
,
Parks
DJ
,
Carver
TE
,
Koblish
HK
, et al
Discovery and cocrystal structure of benzodiazepinedione HDM2 antagonists that activate p53 in cells
.
J Med Chem
2005
;
48
:
909
12
.
60.
Ding
K
,
Lu
Y
,
Nikolovska-Coleska
Z
,
Wang
G
,
Qiu
S
,
Shangary
S
, et al
Structure-based design of spiro-oxindoles as potent, specific small-molecule inhibitors of the MDM2-p53 interaction
.
J Med Chem
2006
;
49
:
3432
5
.
61.
Shangary
S
,
Qin
D
,
McEachern
D
,
Liu
M
,
Miller
RS
,
Qiu
S
, et al
Temporal activation of p53 by a specific MDM2 inhibitor is selectively toxic to tumors and leads to complete tumor growth inhibition
.
Proc Natl Acad Sci U S A
2008
;
105
:
3933
8
.
62.
Issaeva
N
,
Bozko
P
,
Enge
M
,
Protopopova
M
,
Verhoef
LG
,
Masucci
M
, et al
Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors
.
Nat Med
2004
;
10
:
1321
8
.
63.
Bernal
F
,
Tyler
AF
,
Korsmeyer
SJ
,
Walensky
LD
,
Verdine
GL
. 
Reactivation of the p53 tumor suppressor pathway by a stapled p53 peptide
.
J Am Chem Soc
2007
;
129
:
2456
7
.
64.
Yang
Y
,
Ludwig
RL
,
Jensen
JP
,
Pierre
SA
,
Medaglia
MV
,
Davydov
IV
, et al
Small molecule inhibitors of HDM2 ubiquitin ligase activity stabilize and activate p53 in cells
.
Cancer Cell
2005
;
7
:
547
59
.
65.
Arts
J
,
Page
M
,
Valckx
A
,
Blattner
C
,
Kulikov
R
,
Floren
W
, et al
JNJ-26854165 - a novel hdm2 antagonist in clinical development showing broad-spectrum preclinical antitumor activity against solid malignancies [abstract]
.
99th AACR Annual Meeting; 2008 Apr 12–16
;
San Diego, CA. Philadelphia, PA
:
AACR; 2008. Abstract nr
. 1592.
66.
Patton
JT
,
Mayo
LD
,
Singhi
AD
,
Gudkov
AV
,
Stark
GR
,
Jackson
MW
. 
Levels of HdmX expression dictate the sensitivity of normal and transformed cells to Nutlin-3
.
Cancer Res
2006
;
66
:
3169
76
.
67.
Bernal
F
,
Wade
M
,
Godes
M
,
Davis
TN
,
Whitehead
DG
,
Kung
AL
, et al
A stapled p53 helix overcomes HDMX-mediated suppression of p53
.
Cancer Cell
2010
;
18
:
411
22
.
68.
Reed
D
,
Shen
Y
,
Shelat
AA
,
Arnold
LA
,
Ferreira
AM
,
Zhu
F
, et al
Identification and characterization of the first small molecule inhibitor of MDMX
.
J Biol Chem
2010
;
285
:
10786
96
.
69.
Wang
H
,
Ma
X
,
Ren
S
,
Buolamwini
JK
,
Yan
C
. 
A small-molecule inhibitor of MDMX activates p53 and induces apoptosis
.
Mol Cancer Ther
2011
;
10
:
69
79
.
70.
Graves
B
,
Thompson
T
,
Xia
M
,
Janson
C
,
Lukacs
C
,
Deo
D
, et al
Activation of the p53 pathway by small-molecule-induced MDM2 and MDMX dimerization
.
Proc Natl Acad Sci U S A
2012
;
109
:
11788
93
.
71.
Tabernero
J
,
Dirix
L
,
Schoffski
P
,
Cervantes
A
,
Capdevila
J
,
Baselga
J
, et al
Phase I pharmacokinetic (PK) and pharmacodynamic (PD) study of HDM-2 antagonist JNJ-26854165 in patients with advanced refractory solid tumors
.
J Clin Oncol
27:15s, 2009 (suppl; abstr 3514)
.
72.
Andreeff
M
,
Kojima
K
,
Padmanabhan
S
,
Strair
R
,
Kirschbaum
M
,
Maslak
PG
, et al
A multi-center, open-label, phase I study of single agent RG7112, a first in class p53-MDM2 antagonist, in patients with relapsed/refractory acute myeloid and lymphoid leukemias (AML/ALL) and refractory chronic lymphocytic leukemia/small cell lymphocytic lymphomas (CLL/SCLL) [abstract]
. In:
Proceedings of the 53rd ASH Annual Meeting and Exposition
;
2010 Dec 10–13; Orlando, FL. Washington, DC: ASH
; 
2010
.
Abstract nr 657
.
73.
Andreeff
M
,
Kojima
K
,
Ruvolo
V
,
Younes
A
,
Wei
W
,
Konopleva
M
, et al
Pharmacodynamic biomarkers in the phase 1 trial of RG7112, a small-molecule MDM2 antagonist in leukemia [abstract]
. In:
Proceedings of the 53rd ASH Annual Meeting and Exposition
;
2011 Dec 10–13; San Diego, CA. Washington, DC: ASH
; 
2011
.
Abstract nr 1545
.
74.
Ray-Coquard
IL
,
Blay
J
,
Italiano
A
,
Cesne
AL
,
Penel
N
,
Zhi
J
, et al
Neoadjuvant MDM2 antagonist RG7112 for well-differentiated and dedifferentiated liposarcomas (WD/DD LPS): A pharmacodynamic (PD) biomarker study
.
J Clin Oncol
29
:
15s, 2011 (suppl; abstr 10007b)
.
75.
Beryozkina
A
,
Nichols
GL
,
Reckner
M
,
Vassilev
LT
,
Rueger
R
,
Jukofsky
L
, et al
Pharmacokinetics (PK) and pharmacodynamics (PD) of RG7112, an oral murine double minute 2 (MDM2) antagonist, in patients with leukemias and solid tumors
.
J Clin Oncol
29:15s, 2011 (suppl; abstr 3039)
.
76.
Kurzrock
R
,
Blay
J-Y
,
Nguyen
BB
,
Wagner
AJ
,
Maki
RG
,
Schwartz
GK
, et al
A phase I study of MDM2 antagonist RG7112 in patients (pts) with relapsed/refractory solid tumors
.
J Clin Oncol
30:15s, 2012 (suppl; abstr e13600)
.
77.
Ringshausen
I
,
O'Shea
CC
,
Finch
AJ
,
Swigart
LB
,
Evan
GI
. 
Mdm2 is critically and continuously required to suppress lethal p53 activity in vivo
.
Cancer Cell
2006
;
10
:
501
14
.
78.
Terzian
T
,
Wang
Y
,
Van Pelt
CS
,
Box
NF
,
Travis
EL
,
Lozano
G
. 
Haploinsufficiency of Mdm2 and Mdm4 in tumorigenesis and development
.
Mol Cell Biol
2007
;
27
:
5479
85
.
79.
Terzian
T
,
Suh
YA
,
Iwakuma
T
,
Post
SM
,
Neumann
M
,
Lang
GA
, et al
The inherent instability of mutant p53 is alleviated by Mdm2 or p16INK4a loss
.
Genes Dev
2008
;
22
:
1337
44
.
80.
Lukashchuk
N
,
Vousden
KH
. 
Ubiquitination and degradation of mutant p53
.
Mol Cell Biol
2007
;
27
:
8284
95
.
81.
Lozano
G
. 
The oncogenic roles of p53 mutants in mouse models
.
Curr Opin Genet Dev
2007
;
17
:
66
70
.
82.
Aziz
MH
,
Shen
H
,
Maki
CG
. 
Acquisition of p53 mutations in response to the non-genotoxic p53 activator Nutlin-3
.
Oncogene
2011
;
30
:
4678
86
.