The p53 tumor suppressor is part of a small family of related proteins that includes two other members, p73 and p63. Interest in the p53 family members, their functions and their complex interactions and regulation, has steadily grown over recent years and does not show signs of waning. p73 is a major determinant of chemosensitivity in humans, and mutant p53 proteins carrying specific polymorphisms can induce drug resistance by inhibiting TAp73. Cooperation between TA (transactivating, proapoptotic, antiproliferative) and ΔN (truncated, antiapoptotic, pro-proliferative) p73 isoforms and among the three family members guarantees equilibrium between proliferation, differentiation, and cell death, thus creating a harmony that is lost in several human cancers. In this article, we review our current knowledge of the role of p73 in cancer chemosensitivity and the real prospect of therapy targeting this molecule. We also draw attention to the crucial role of specific phosphorylation and acetylation events for p73-induced apoptosis and drug chemosensitivity. (Clin Cancer Res 2009;15(21):6495–502)

Translational Relevance

The scientific evidence currently available highlights the crucial role of the p53 family member p73 in chemosensitivity. One important issue that arises from these studies is the need, both in the prognostic consideration of many cancer patients and in the evaluation of therapeutic options, to go beyond the p53 status and include an assessment of the expression of p73 proteins. The development of standardized methods and reagents and the need for reliable and rapid tests to detect p73 proteins in the clinical setting are important objectives for cancer medicine. Real-time PCR, robust immunohistochemistry reagents and methods, and protein microarray techniques will help achieve this goal.

The recent screening by different laboratories of small molecules targeting the p73 pathway and the preclinical observation of successful inhibition of tumor growth by some of them greatly encourage research physicians to start clinical trials, using these new compounds alone or in combination therapies.

The p53 tumor suppressor (1, 2) belongs to a small family of related proteins that includes two other members, p73 and p63, discovered in 1997 (3) and 1998 (47), respectively.

This review, after a brief introduction on evolution within the p53 family, has two primary aims: to focus on the crucial role of p73 in cancer chemosensitivity and to discuss the prospect of cancer treatment targeting this molecule. Despite the translational focus of this review, however, the end purpose of a CCR Review precludes citing all contributions and all contributors, for which we apologize in advance.

Overall structure and sequence homology indicate that p63, p73, and p53 evolved from a common ancestor (8). p73 and p63 share hallmark features that identify p53 across species from Drosophila melanogaster and Caenorhabditis elegans to human: an acidic NH2-terminal transactivation (TA) domain, a highly conserved core DNA-binding domain (DBD), and a COOH-terminal oligomerization domain (Fig. 1). Both p63 and p73 have a sterile α-motif domain implicated in protein-protein interaction. The highest degree of homology is shown within the DBD, where p63 and p73 share approximately 65% amino acidic identity with the DBD of p53 and higher identity (85%) with each other. All three genes are now known to contain a second intronic promoter that controls the expression of NH2-terminally truncated ΔN proteins (ΔNp73, ΔNp63, and Δ133p53; refs. 4, 913). Additional ΔN isoforms are generated by alternative splicing and initiation of translation (Δ40p53, Δex2p73, Δex2/3p73, and ΔN′p73) of mRNAs transcribed from the canonical upstream promoter regions (1315). Δ40p53 and ΔN′p73 are originated by variations of the proline-rich domain. The term ΔTAp73 comprises ΔNp73, Δex2p73, Δex2/3p73, and ΔN′p73 isoforms. Because the lack of acidic NH2-terminal TA domain hampers activation of target genes, TA full-length isoforms (FLp53, TAp73, and TAp63) are functionally distinct from ΔN isoforms, which behave as dominant-negative isoforms and have antiapoptotic and pro-proliferative functions (12, 13). Further modifications induced by alternative splicing of COOH-terminal exons develop numerous different isoforms (α, β, γ, δ, ϵ, θ, ζ, and η) with still incompletely known DNA-binding properties and functions (3, 13, 1517). Interestingly, phylogenetic analysis firmly supports the concept that p53 is not the ancestor gene but a more recent product of evolution than p63 and p73, p63 being the ancestor, followed by p73 and then by p53 (8).

Fig. 1.

p53 family isoforms and gene structure. p53 belongs to a multigene family that also includes p63 and p73. The overall structure and sequence homology are highly conserved from D. melanogaster to human, and consist of an acidic NH2-terminal TA domain, a central highly conserved core DBD, and a COOH-terminal oligomerization domain (OD). Both p63 and p73 have a sterile α-motif (SAM) domain implicated in protein-protein interactions. The highest degree of homology is shown within the DBD, where p63 and p73 share about 65% amino acidic identity with the DBD of p53 and higher identity (85%) with each other. All three genes contain a second intronic promoter (p2) that controls the expression of NH2-terminally truncated ΔN proteins (ΔNp73, ΔNp63, and Δ133p53). Further isoforms, generated by alternative splicing events and alternative initiation of translation, are the Δ40p53 isoform and the ΔTAp73 isoforms Δex2p73, Δex2/3p73, and ΔN′p73. Δ40p53 and ΔN′p73 are originated by variations of the proline-rich domain (PRD). Further modifications induced by alternative splicing of COOH-terminal exons develop numerous different isoforms (α, β, γ, δ, ϵ, θ, ζ, and η) with still incompletely known DNA-binding properties, transcriptional activities, and biological functions.

Fig. 1.

p53 family isoforms and gene structure. p53 belongs to a multigene family that also includes p63 and p73. The overall structure and sequence homology are highly conserved from D. melanogaster to human, and consist of an acidic NH2-terminal TA domain, a central highly conserved core DBD, and a COOH-terminal oligomerization domain (OD). Both p63 and p73 have a sterile α-motif (SAM) domain implicated in protein-protein interactions. The highest degree of homology is shown within the DBD, where p63 and p73 share about 65% amino acidic identity with the DBD of p53 and higher identity (85%) with each other. All three genes contain a second intronic promoter (p2) that controls the expression of NH2-terminally truncated ΔN proteins (ΔNp73, ΔNp63, and Δ133p53). Further isoforms, generated by alternative splicing events and alternative initiation of translation, are the Δ40p53 isoform and the ΔTAp73 isoforms Δex2p73, Δex2/3p73, and ΔN′p73. Δ40p53 and ΔN′p73 are originated by variations of the proline-rich domain (PRD). Further modifications induced by alternative splicing of COOH-terminal exons develop numerous different isoforms (α, β, γ, δ, ϵ, θ, ζ, and η) with still incompletely known DNA-binding properties, transcriptional activities, and biological functions.

Close modal

p73 functions are highly integrated with p53 and p63

An analysis of the phenotype of p73 knockout mice revealed neurologic pheromonal and inflammatory defects, but early death precluded the evaluation of tumor development (9). The investigation by Flores et al. (18) of p73+/− mice underlined the role of p73 as a tumor suppressor. Indeed, full necropsy showed that p73+/− and p63+/− mice developed malignant lesions: 10% of p73+/− mice developed lung adenocarcinoma, whereas 12.5% developed thymic lymphoma and 12.5% hemangiosarcomas. In addition to these malignant lesions, p73+/− mice showed an increase in premalignant lesions such as squamous cell hyperplasia and multiple lung adenoma (18). Double mutant mice for p73 and p63 developed bladder carcinomas, mammary adenocarcinomas, and myelogenous leukemias (18). p73 and/or p63 loss, when accompanied by p53 mutation, generated very aggressive, metastatic phenotypes (18), possibly due to the role of TAp73 and TAp63 in anchorage-independent growth control (19, 20). Recently, Tomasini et al. (21) showed that mice specifically lacking in TAp73 exhibited genomic instability, enhanced aneuploidy, and increased incidence of spontaneous tumors, thus reinforcing the view that the isoform exerts tumor-suppressive functions. Taken together, these results showed that close cooperation between the family members exists and is needed to amplify tumor suppression functions.

Two landmark articles recently showed that p73 is an important determinant of chemosensitivity in humans, and likewise of tumor suppression, the function of which is highly integrated with that of p53 (22, 23).

Irwin et al. found that TAp73 is induced by a large variety of chemotherapeutic agents such as camptothecin, etoposide, cisplatinum, doxorubicin, and Taxol, and that the experimental blocking of the TAp73 function with a dominant-negative mutant or small interfering RNA (siRNA) leads to enhanced chemoresistance of human tumor cells and engineered transformed cells (22) in the presence of wild-type, null, or fully inactive (not gain-of-function) p53. It is noteworthy that for certain chemotherapeutic agents, p73 induction was observed at low, but not high, drug concentrations, suggesting the presence of subtle and specific mechanisms.

A further observation by the authors that mutant p53 could inactivate p73 and that downregulation of mutant p53 by transfection of p53 siRNA could enhance chemosensitivity supports a model wherein mutant p53 can induce chemoresistance through neutralization of TAp73 (22). Accordingly, specific p53 mutants that inhibit p73-dependent apoptosis have been identified, and p53-dependent inhibition of p73 is correlated with clinical drug resistance (23). Moreover, patients affected with head and neck cancers that express a mutated p53 with an arginine polymorphism at position 72 (72R) have a worse response to therapy, with shorter progression-free survival, compared with patients carrying a proline at position 72 (72P). These results have established a link between a specific polymorphism in p53, p73 inhibition, and the individual response to cancer therapy, underlining the potential effect of these factors in the prediction of clinical outcomes.

In a further study on 122 ovarian cancers, the patients with p53 mutations (65.6%) that efficiently inhibited TAp73 presented a significantly shorter overall survival than those with other p53 mutations (P = 0.044), although the p53 polymorphism at codon 72 is less relevant in these ovarian cancer series (24).

Investigations of human cancers, focused on understanding the role of the antiapoptotic and pro-proliferative isoforms of p53 family proteins, have underlined the high integration of the functions.

A study including 35 human cancers (cancers of the ovary, endometrium, cervix, vulva, vagina, breast, kidney, and colon) showed that about 70% of tumors, but not normal tissues, exhibit upregulation of the ΔNp73 isoform (25). The authors have shown that ΔNp73α suppresses the apoptotic function of wild-type p53 and TAp73 by heterocomplex formation and confers drug resistance to wild-type p53–harboring tumor cells. Consequently, suppression of ΔNp73 expression by specific siRNA enhances p53- and TAp73-mediated apoptosis.

Further investigation of 100 ovarian carcinomas revealed that transdominant ΔTAp73 isoforms are frequently upregulated and have a role as epigenetic p53 inhibitors in vivo (26).

ΔNp73 is frequently overexpressed in rhabdomyosarcoma and is essential for tumor progression in vivo (27). p53 is required to induce transcription of the retinoblastoma (RB) gene, whereas p73 and p63 induce the cyclin-dependent kinase inhibitor p57 to maintain RB in an active, hypophosphorylated state. Ablation of these functions by ΔNp73 overexpression or p53 mutations blocks myogenic differentiation and enables cooperating oncogenes to transform myoblasts to tumorigenicity (27).

Moreover, in most head and neck squamous cell carcinomas, the high levels of ΔNp63α are found to represent an essential survival factor for the tumor, due to their ability to suppress TAp73-dependent apoptosis both by direct promoter binding and by physical protein-protein interaction (28). Inhibition of endogenous p63 expression by specific siRNAs leads to the TAp73-dependent and p53-independent upregulation of apoptotic target genes to induce the selective apoptosis of tumor cells that overexpress ΔNp63α, and correlates with sensitivity to radiation and/or chemotherapy (28).

Strikingly, in a subset of breast cancers that do not express estrogen and progesterone receptors, lack Her2 amplification, and make up the majority of BRCA1-associated tumors, p63 can regulate the p73-dependent pathway that controls cisplatin sensitivity in vivo (29). In these tumors, which commonly exhibit mutational inactivation of p53 and coexpress ΔNp73 and TAp73 isoforms, ΔNp63α promotes the survival of breast cancer cells by binding TAp73 and thereby inhibiting its proapoptotic activity (29). The inhibition of p63 expression by specific siRNAs leads to the TAp73-dependent induction of proapoptotic Bcl-2 family members and apoptosis. Significantly, in these cases, the sensitivity to cisplatin is uniquely dependent on TAp73: after exposure to cisplatin, but not to other chemotherapeutic agents, TAp73 dissociates from the ΔNp63α/TAp73 protein complex and transactivates the proapoptotic members of the Bcl-2 family to induce apoptosis (29).

Figure 2 reports the crucial role of ΔNp73, ΔNp63, and mutated p53 in inhibiting TAp73 transactivation of proapoptotic target genes such as Bax, p53 upregulated modulator of apoptosis (PUMA), and p53-regulated apoptosis inducing protein 1 (p53AIP1).

Fig. 2.

Inactivation of TAp73 by ΔNp73, ΔNp63, and mutated p53. Inactivated TAp73 lacks the capability to transactivate proapoptotic target genes such as Bax, PUMA, and p53AIP1.

Fig. 2.

Inactivation of TAp73 by ΔNp73, ΔNp63, and mutated p53. Inactivated TAp73 lacks the capability to transactivate proapoptotic target genes such as Bax, PUMA, and p53AIP1.

Close modal

Several other articles have confirmed the importance of the assessment of p73 isoform expression in the prediction of tumor chemosensitivity and in cancer prognosis by studying different tumor types. Indeed, dysfunctional equilibrium between p73 isoforms has been observed in human thyroid cancer (30), hepatocellular carcinoma (31), neuroblastoma (32), low-grade glioma (33), gynecologic cancers (34), acute myelogenous leukemia (AML; refs. 3538), colon cancer (39), breast cancer (39, 40), and lung cancer (41, 42). In many cases (23, 24, 26, 3134, 39, 42), patients with p73 dysfunction proved to have a worse prognosis than those with normal p73 function.

Mechanisms of p73-induced chemosensitivity

The regulatory mechanisms that control p73 drug response are primarily linked to posttranslational modifications and protein-protein interactions involving both signaling molecules and transcription factors. Indeed, mechanical behavior can be complex and variable, being dependent on the specific drugs and tissues involved.

Three seminal articles have contemporaneously shown the key role of the c-Abl oncogene in the processes of p73 activation. In cells exposed to either chemotherapeutic drugs or ionizing radiations, the tyrosine kinase c-Abl phosphorylates p73 on the tyrosine residue at position 99 and potentiates p73-mediated transactivation and apoptosis (4345). p73 half-life is prolonged and p73 protein levels are increased by cisplatin and by coexpression with c-Abl tyrosine kinase (43). Notably, the interaction between p73 and c-Abl is potentiated by p73 phosphorylation in primary breast cancer cells as well (46), and p38 mitogen-activated protein kinase can participate in mediating this process (47). Furthermore, a robust and translational confirmation of these studies is provided by the observation that c-Abl–dependent phosphorylation of TAp73 is essential for the dissociation of the ΔNp63α/TAp73 protein complex by cisplatin in BRCA1-associated breast cancer (29).

In anaplastic thyroid cancer, c-Abl is excluded by the nucleus, losing its capacity to activate TAp73 (48). This behavior fits with the observation that the Bcr-Abl fusion protein in chronic myelogenous leukemia loses its oncogenic pro-proliferative and antiapoptotic properties when driven off the cytoplasm and entrapped in the nucleus (49). These observations prompt the need for future investigations on the relation between subcellular localization and p53 family protein functions.

c-Abl is also required for TAp73 acetylation by acetyltranferase p300. Indeed, Costanzo et al. (50) have shown that doxorubicin-induced acetylation strongly potentiates the apoptotic functions of TAp73 by enhancing its ability to selectively activate the transcription of proapoptotic target genes but not that of growth arrest genes, thus suggesting that drug-induced TAp73 acetylation can shift the balance between the antiproliferative effect of antitumor drugs toward apoptosis and tumor killing.

Protein-protein interactions with two interesting partners contribute to TAp73 activation and apoptosis in response to cell stress and DNA damage. On treatment with chemotherapeutic drugs, the prolyl isomerase Pin1 interacts with p73 and promotes p73 acetylation by p300 (51). Indeed, c-Abl and p300 require Pin1 to increase p73 stability and transcriptional activity. Furthermore, the WW domain Yes-associated protein (YAP) contributes to the DNA damage–induced accumulation of p73 and acts as a transcriptional coactivator to potentiate p300-mediated acetylation of p73 and apoptosis (52). Tyrosine-phosphorylated YAP1 by c-Abl displays higher affinity to p73 and selectively coactivates p73 proapoptotic target genes (53). Overall, these breakthrough demonstrations underline the role of specific phosphorylation and acetylation events for p73-induced apoptosis and drug chemosensitivity. Significantly, the phosphorylation of YAP2 by tumor suppressor Kpm/Lats2, which is downregulated in poor-prognosis acute lymphoblastic leukemia (54) and breast cancer (55), leads to nuclear accumulation of p73, resulting in induction of its target genes p21 and PUMA (56). Kpm/Lats2 is downregulated in adult T-cell leukemia and natural killer leukemia/lymphoma as well (56), both of which are known to be highly resistant to conventional chemotherapy.

Lunghi et al. determined that arsenic trioxide (ATO; 1 μmol/L), the drug of choice for patients with relapsed acute promyelocytic leukemia (PML) previously exposed to all-trans retinoic acid, targets the p73 proteins to suppress acute leukemia cell growth (35, 36). Indeed, ATO reduces ΔNp73 protein levels (more in K562 than in NB4 cells) and promotes p300-mediated acetylation of endogenous TAp73, thus favoring the apoptosis of the PML NB4 and the Bcr/Abl+ erythroleukemia K562 cell lines that carry inactive p53 (35). Based on the observation that mitogen-activated protein kinases confer an aggressive, apoptosis-resistant phenotype to leukemia cells and that small-molecule inhibitors of the upstream mitogen-activated protein/extracellular signal-regulated kinase (MEK), such as PD98059 (Cell Signaling Technology) or PD184352 (Pfizer), sensitize leukemia cells to drug-induced apoptosis (5759), Lunghi et al. explored, in a preclinical setting, the potential of a combination of MEK inhibitor PD184352 (Pfizer) and ATO in the treatment of AML. PD184352 considerably increased ATO-induced apoptosis in 21 of 25 primary AML blasts of different FAB subtypes, showing synergism in 13 of 21 and addictiveness in 8 of 21 responders (36). At the molecular level, MEK inhibition reduced the levels of dominant-negative ΔNp73 proteins and promoted the accumulation of endogenous TAp73 through its tyrosine phosphorylation and transcriptional activation (35, 36). Thus, the combined treatment of ATO and MEK inhibitor enhanced the affinity of phospho-acetylated TAp73 for the p53AIP1 promoter in vivo, leading to p53AIP1 upregulation and increased apoptosis (35). p53AIP1, a direct transcriptional target and a primary effector of wild-type p53– and TAp73-induced apoptosis (50, 60), is located in the mitochondria, and its overexpression induces massive apoptotic cell death through the dissipation of mitochondrial ΔΨm (60, 61). Anti-TAp73 siRNA strongly decreased apoptosis, whereas anti-ΔNp73 siRNA exerted the opposite effect (35).

In blasts of AML patients with active p53 (about 50% of cases), a high concentration of ATO (2 μmol/L) promoted the accumulation of TAp73 and p53 protein levels, whereas at low concentrations (1 μmol/L), ATO only induced TAp73 (36). TAp73 induction through low doses of ATO is similar to the behavior observed by Irwin et al. (22) for certain chemotherapeutic agents.

Contrary to AML blasts, Lunghi et al. (62) showed that in multiple myeloma cells, p73 cooperates in inducing apoptosis when p53 is active, but in the presence of mutated or inactive p53, the apoptosis of malignant cells may occur without the aid of p73. Shammas et al. (63) discovered that the treatment of INA6 multiple myeloma cells with epigallocatechin-3-gallate is associated with elevated transcript and protein levels of p73 and p63, but not p53, whereas Raab et al. (64) showed that targeting protein kinase C by small-molecule inhibitor enzastaurin accumulates β-catenin, which triggers c-Jun–dependent induction of p73 and apoptosis of multiple myeloma cells. These findings suggest that stimulus and tissue specificity regulate p53 family member reactions, and prompt the need for further investigations of primary tumor cells of various tissues in different physiopathologic conditions.

It is clear that the PML tumor suppressor gene potentiates p73 transcriptional and proapoptotic activities by regulating the ability of p300 acetyltransferase to acetylate and stabilize p73, thus inhibiting its ubiquitin-dependent degradation (52, 65). Because PML targeting by ATO has recently been found to drive leukemia-initiating stem cells of primary chronic myelogenous leukemia patients from a quiescent drug-resistant status to a drug-responsive cell cycle entry, it would be of great interest to investigate the role of p73 in these processes (66). Indeed, a proapoptotic autoregulatory feedback loop between p73-PML-YAP has recently been found to exist, and this could have relevant implications for chemosensitivity as well (67). The cisplatin regulation of this network emphasizes the translational effect of these results (67). Mechanisms of TAp73 activation are depicted in Fig. 3.

Fig. 3.

Mechanisms of TAp73 activation through phospho-acetylation and protein-protein interactions. The tyrosine kinase c-Abl acts as a tumor suppressor by phosphorylating TAp73 in response to chemotherapeutic drugs and DNA damage (29, 4346). The support of p38 mitogen-activated protein kinase is essential to the process of TAp73 regulation by c-Abl (47). At the same time, c-Abl phosphorylates YAP1, thus increasing its affinity to TAp73 (52); PML physically interacts with YAP1, causing PML-mediated sumoylation and stabilization of the molecule (67). The potent transactivation of proapoptotic target genes by TAp73 is only possible after acetylation by the acetyltransferase p300 in a c-Abl–dependent manner (50). The binding of prolyl isomerase Pin1 to TAp73 and the complex TAp73-PML-YAP1 are essential for the completion of this process (red oval; refs. 51, 67). The crucial steps of phospho-acetylation and protein-protein interactions are subtly regulated by oncotoxic drugs and DNA-damaging agents (29, 35, 4345, 5053, 67). p300, p300 acetyltransferase; P, phosphorylation; ac, acetylation.

Fig. 3.

Mechanisms of TAp73 activation through phospho-acetylation and protein-protein interactions. The tyrosine kinase c-Abl acts as a tumor suppressor by phosphorylating TAp73 in response to chemotherapeutic drugs and DNA damage (29, 4346). The support of p38 mitogen-activated protein kinase is essential to the process of TAp73 regulation by c-Abl (47). At the same time, c-Abl phosphorylates YAP1, thus increasing its affinity to TAp73 (52); PML physically interacts with YAP1, causing PML-mediated sumoylation and stabilization of the molecule (67). The potent transactivation of proapoptotic target genes by TAp73 is only possible after acetylation by the acetyltransferase p300 in a c-Abl–dependent manner (50). The binding of prolyl isomerase Pin1 to TAp73 and the complex TAp73-PML-YAP1 are essential for the completion of this process (red oval; refs. 51, 67). The crucial steps of phospho-acetylation and protein-protein interactions are subtly regulated by oncotoxic drugs and DNA-damaging agents (29, 35, 4345, 5053, 67). p300, p300 acetyltransferase; P, phosphorylation; ac, acetylation.

Close modal

New light could be thrown on the mechanisms of p73-dependent drug sensitivity through an analysis of the relationship with microRNAs (miRNA). A crucial relationship for inducing apoptosis has recently been shown to exist between specific miRNAs and p53 (68). Recently, Sampath et al. (69) found that drug-induced deacetylase inhibition induces miRNA 106b, which regulates p73-dependent apoptotic signaling and death in quiescent B chronic lymphocytic leukemia cells. Induction of miRNA 106b is associated with a downregulation in the levels of the E3 ubiquitin ligase Itch. Decreases in Itch protein levels lead to an accumulation of its proapoptotic substrate TAp73. The discovery of this interesting mechanism could lead to the development of further studies on miRNA and p73 relations.

After its discovery in 1997, more than 1,200 articles have been published over the last decade on the study of p73 expression in tumors and its involvement in carcinogenesis, but the real value of this molecule as a potential antitumor target was not apparent. For this reason, the recent recognition of molecules specifically designed toward p73 proteins or their close and direct regulators that inhibit tumor cell growth by activating p73 has generated great interest. Although confirmatory clinical studies are needed, many tumors can be potential targets because the p53-related protein p73 functions similarly, but unlike p53, it is rarely lost or mutated in cancer (70). Figure 4 depicts some strategies that have recently been tested in preclinical settings for targeting p73 to induce the transcription of proapoptotic genes.

Fig. 4.

Targeting p53 family proteins to treat tumors by transactivation of proapoptotic genes. A, the strategy to use small molecules to restore p53 and TAp73 tumor control is an innovative approach (71, 7577). 37AA is a small peptide that contains 37 amino acids from p53 capable of alleviating p53 and TAp73 from inhibitor of ASPP (iASPP)–mediated repression, allowing them to activate downstream transcription. When p53 is mutated or not functional, 37AA activation of endogenous proapoptotic genes via p73 gives a substantial contribution to tumor death (71). B, new siRNA-based therapeutic strategies will specifically target ΔN isoforms and down-modulate their prosurvival activities. Inhibition by siRNA of ΔNp73 can increase the balance between TAp73 and ΔNp73 that is reduced in several tumors (25, 27, 31, 36) and block the heterocomplex formation between ΔNp73 and wild-type p53 or TAp73 (25). Inhibition by siRNA of ΔNp63 hampers the suppression of TAp73-dependent apoptosis in squamous cell carcinomas and in breast cancers (28, 29). ΔN isoform inhibition allows both p53 and TAp73 to activate downstream transcription of proapoptotic target genes. The approaches reported in A and B have been tested in preclinical settings but provide strong grounds for clinical trials in the near future.

Fig. 4.

Targeting p53 family proteins to treat tumors by transactivation of proapoptotic genes. A, the strategy to use small molecules to restore p53 and TAp73 tumor control is an innovative approach (71, 7577). 37AA is a small peptide that contains 37 amino acids from p53 capable of alleviating p53 and TAp73 from inhibitor of ASPP (iASPP)–mediated repression, allowing them to activate downstream transcription. When p53 is mutated or not functional, 37AA activation of endogenous proapoptotic genes via p73 gives a substantial contribution to tumor death (71). B, new siRNA-based therapeutic strategies will specifically target ΔN isoforms and down-modulate their prosurvival activities. Inhibition by siRNA of ΔNp73 can increase the balance between TAp73 and ΔNp73 that is reduced in several tumors (25, 27, 31, 36) and block the heterocomplex formation between ΔNp73 and wild-type p53 or TAp73 (25). Inhibition by siRNA of ΔNp63 hampers the suppression of TAp73-dependent apoptosis in squamous cell carcinomas and in breast cancers (28, 29). ΔN isoform inhibition allows both p53 and TAp73 to activate downstream transcription of proapoptotic target genes. The approaches reported in A and B have been tested in preclinical settings but provide strong grounds for clinical trials in the near future.

Close modal

Indeed, a p53-derived small peptide, called 37AA, as it contains 37 amino acids from p53, has been described as inducing cell death in multiple tumor cell types irrespective of p53 status, but not in normal cells (71). This peptide has the capacity to bind inhibitor of apoptosis stimulating protein of p53 (iASPP), a common negative regulator of p53 family members that antagonizes positive regulators such as ASPP1 and ASPP2 (7274). Remarkably, in p53-null cells, this peptide derepresses TAp73, causing p73-mediated gene activation and cell death (71). Moreover, systemic nanoparticle delivery of a transgene expressing this peptide is able to induce p73-dependent tumor regression in vivo. This study supports future strategies to directly and selectively activate the therapeutic potential of TAp73.

The screening by different laboratories of small molecules structurally related to or dissimilar from p53 that activate p53 response by increasing TAp73 or releasing this molecule from the blocking complex with mutated p53 has developed reagents with strong antitumor activity that could become successful drugs (7577). In some cases, the efficacy of these compounds has been tested in vivo in p53-deficient human tumor xenografts (77).

In tumors that express high levels of ΔNp73, strategies would be aimed at interfering by means of small molecules with the expression and function of these proteins (78, 79). The development of siRNA-based therapeutic agents with improved viability and capability to target specific tissues with poor toxicity (80) would make it possible to specifically target the expression and activity of p53 family Δ isoforms that exert an antiapoptotic, pro-proliferative function (Fig. 4B). This strategy would be used in combination treatment with compounds that positively modulate TAp73 after testing the p53 family status of individual tumors during diagnosis and relapses.

Recently, interest has grown in the small molecule Nutlin-3, which can induce p53-dependent apoptosis in AML cells and B chronic lymphocytic leukemia cells that express a p53 inhibited by murine double minute-2 overexpression (about 50% of AML patients; refs. 81, 82). The demonstration of the property of Nutlin-3 to disrupt murine double minute-2-TAp73 binding in tumor cells with null or mutated p53 deserves attention for cancer treatment (83).

Besides developing small molecules active toward p73 and related molecules, we believe that all new classes of compounds that are candidate anticancer drugs (e.g., aurora kinase inhibitors) should be investigated to assess the effect on p53 family status. Dar et al. (84) investigated the role of Aurora kinase A in regulating p73-dependent apoptosis using p53-deficient cancer cell lines. Interestingly, the authors showed that Aurora kinase A hampers endogenous TAp73 protein expression and its transcriptional activity. The Aurora kinase A inhibitor MLN8054 suppresses cell viability and induces TAp73 and its downstream proapoptotic targets PUMA, BH3-only protein NOXA, and p53AIP1, promoting apoptosis. Because interest in targeting Aurora kinases for the purpose of treating cancer is constantly growing, this article could pave the way for further studies.

It might be worth mentioning that an important class of drugs, cyclooxygenase inhibitors, has recently been shown to induce apoptosis independent of p53 and differentially modulate endogenous TAp73 and ΔNp73 isoforms in fresh neuroblastoma cells (85). Cyclooxygenase downregulation of ΔNp73 is associated with decreased levels of transcription factor E2F-1. These results encourage preclinical combination studies to enhance chemosensitivity in tumors with deregulated E2F-1 and in those with wild-type or mutant p53.

The mammalian target of rapamycin (mTOR) kinase pathway, which has a potentially great drug effect (86), has recently been focused by a gene signature–based approach to be a regulator of p73 (87). Indeed, treatment of both primary cells and cancer cell lines with rapamycin resulted in an increase in p73 levels, as did RNA interference knockdown of mTOR (87). Moreover, the intriguing observation that mTOR regulates autophagy-associated genes downstream of p73 through an unknown mechanism (8789) could draw attention to alternative, still unexplored, ways of killing cancer.

This review primarily stresses the important acquisition of the role of p73 in human tumor chemosensitivity—a great success of translational and clinical oncology. This scenario leads to investigating the mechanisms and relations with p73 of new classes of compounds (e.g., Aurora kinases, cyclooxygenase, and mTOR inhibitors) that are candidates for cancer therapy. These studies could help identify dysregulated molecules that are potential targets for synergistic combination therapies.

A further great challenge concerns the design of specific small molecules directly targeted to dysregulated p73 proteins or their close regulators. Recently, to overcome laborious screens of genetic or chemical libraries, innovative methods have been proposed, such as the Connectivity Map project (i.e., a means of connecting a disease with a disease-modifying gene product and a chemical modulator of that protein by a systematized and centralized process using gene expression signatures; refs. 90, 91). In this light, the need for reliable and rapid tests to detect all p53 family proteins in the clinical setting is a major objective for research and development efforts in the near future. The integration of expression profiling tools, such as dedicated microarrays or automated multiple real-time PCR reagents, reliable immunohistochemistry reagents and methods, and protein microarray techniques, will help achieve the aim of a more personalized therapy for cancer patients.

The authors declare that they do not have conflicts of interest.

The authors apologize for not being able to cite several interesting articles due to the space limitations created by the need to target primarily translational topics, in keeping with the purpose of this review.

1
Lane
DP
,
Crawford
LV
. 
T antigen is bound to a host protein in SV40-transformed cells
.
Nature
1978
;
278
:
261
3
.
2
Linzer
DI
,
Levine
AJ
. 
Characterization of a 54K dalton SV40 tumor antigen present in SV40-transformed cells and uninfected embryonal carcinoma cells
.
Cell
1979
;
17
:
43
52
.
3
Kaghad
M
,
Bonnet
H
,
Yang
A
, et al
. 
Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers
.
Cell
1997
;
90
:
809
19
.
4
Yang
A
,
Kaghad
M
,
Wang
Y
, et al
. 
p63, a p53 homolog at 3q27-29, encodes multiple products with transactivating, death-inducing, and dominant-negative activities
.
Mol Cell
1998
;
2
:
305
16
.
5
Augustin
M
,
Bamberger
C
,
Paul
D
,
Schmale
H
. 
Cloning and chromosomal mapping of the human p53-related KET gene to chromosome 3q27 and its murine homolog Ket to mouse chromosome 16
.
Mamm Genome
1998
;
9
:
899
902
.
6
Osada
M
,
Ohba
M
,
Kawahara
C
, et al
. 
Cloning and functional analysis of human p51, which structurally and functionally resembles p53
.
Nat Med
1998
;
4
:
839
43
.
7
Senoo
M
,
Seki
N
,
Ohira
M
, et al
. 
A second p53-related protein, p73L, with high homology to p73
.
Biochem Biophys Res Commun
1998
;
248
:
603
7
.
8
Yang
A
,
Kaghad
M
,
Caput
D
,
McKeon
F
. 
On the shoulders of giants: p63, p73 and the rise of p53
.
Trends Genet
2002
;
18
:
90
5
.
9
Yang
A
,
Walker
N
,
Bronson
R
, et al
. 
p73-deficient mouse have neurological pheromonal and inflammatory defects but lack spontaneous tumors
.
Nature
2000
;
404
:
99
103
.
10
Pozniak
CD
,
Radinovic
S
,
Yang
A
,
McKeon
F
,
Kaplan
DR
,
Miller
FD
. 
An anti-apoptotic role for the p53 family member, p73, during developmental neuron death
.
Science
2000
;
289
:
304
6
.
11
Bourdon
JC
,
Fernandes
K
,
Murray-Zmijewski
F
, et al
. 
p53 isoforms can regulate p53 transcriptional activity
.
Genes Dev
2005
;
19
:
2122
37
.
12
Grob
TJ
,
Novak
U
,
Maisse
C
, et al
. 
Human ΔNp73 regulates a dominant negative feedback loop for TAp73 and p53
.
Cell Death Differ
2001
;
8
:
1213
23
.
13
Ishimoto
O
,
Kawahara
C
,
Enjo
K
,
Obinata
M
,
Nukiwa
T
,
Ikawa
S
. 
Possible oncogenic potential of ΔNp73: a newly identified isoform of human p73
.
Cancer Res
2002
;
62
:
636
41
.
14
Fillippocich
I
,
Sorokina
N
,
Gatei
M
, et al
. 
Transactivation-deficient p73 α (p73Δexon2) inhibits apoptosis and competes with p53
.
Oncogene
2001
;
20
:
514
22
.
15
Stiewe
T
. 
The p53 family in differentiation and tumorigenesis
.
Nat Rev Cancer
2007
;
7
:
165
8
.
16
De Laurenzi
V
,
Costanzo
A
,
Barcaroli
D
, et al
. 
Two new p73 splice variants, γ and δ, with different transcriptional activity
.
J Exp Med
1998
;
88
:
1763
8
.
17
De Laurenzi
V
,
Catani
MV
,
Terrinoni
A
, et al
. 
Additional complexity in p73: induction by mitogens in lymphoid cells and identification of two new splicing variants ϵ and ζ
.
Cell Death Differ
1999
;
6
:
389
90
.
18
Flores
ER
,
Sengupta
S
,
Miller
JB
, et al
. 
Tumor predisposition in mice mutant for p63 and p73: evidence for broader tumor suppression functions for the p53 family
.
Cancer Cell
2005
;
7
:
363
73
.
19
Carroll
DK
,
Carroll
JS
,
Leong
CO
, et al
. 
p63 regulates an adhesion programme and cell survival in epithelial cells
.
Nat Cell Biol
2006
;
8
:
551
61
.
20
Beitzinger
M
,
Hofmann
L
,
Oswald
C
, et al
. 
p73 poses a barrier to malignant transformation by limiting anchorage-independent growth
.
EMBO J
2008
;
27
:
792
803
.
21
Tomasini
R
,
Tsuchihara
K
,
Wilhelm
M
, et al
. 
TAp73 knockout shows genomic instability with infertility and tumor suppressor functions
.
Genes Dev
2008
;
22
:
2677
91
.
22
Irwin
MS
,
Kondo
K
,
Marin
MC
,
Cheng
LS
,
Hahn
WC
,
Kaelin
WG,
 Jr
. 
Chemosensitivity linked to p73 function
.
Cancer Cell
2003
;
3
:
403
10
.
23
Bergamaschi
D
,
Gasco
M
,
Hiller
L
, et al
. 
p53 polymorphism influences response in cancer chemotherapy via modulation of p73-dependent apoptosis
.
Cancer Cell
2003
;
3
:
387
402
.
24
Concin
N
,
Hofstetter
G
,
Berger
A
, et al
. 
Clinical relevance of dominant-negative p73 isoforms for responsiveness to chemotherapy and survival in ovarian cancer: evidence for a crucial p53–73 cross-talk in vivo
.
Clin Cancer Res
2005
;
11
:
8372
83
.
25
Zaika
A
,
Slade
N
,
Erster
SH
, et al
. 
ΔNp73, a dominant negative inhibitor of wild type p53 and TAp73, is upregulated in human tumors
.
J Exp Med
2002
;
196
:
765
80
.
26
Concin
N
,
Becker
K
,
Slade
N
, et al
. 
Transdominant ΔTAp73 isoforms are frequently up-regulated in ovarian cancer, evidence for their role as epigenetic p53 inhibitors in vivo
.
Cancer Res
2004
;
64
:
2449
60
.
27
Cam
H
,
Griesmann
H
,
Beitzinger
M
, et al
. 
p53 family members in myogenic differentiation and rhabdomyosarcoma development
.
Cancer Cell
2006
;
10
:
281
93
.
28
Rocco
JW
,
Leong
CO
,
Kuperwasser
N
,
DeYoung
MP
,
Ellisen
LW
. 
p63 mediates survival in squamous cell carcinoma by suppression of p73-dependent apoptosis
.
Cancer Cell
2006
;
9
:
45
56
.
29
Leong
CO
,
Vidnovic
N
,
DeYoung
MP
,
Sgroi
D
,
Ellisen
LW
. 
The p63/p73 network mediates chemosensitivity to cisplatin in a biologically defined subset of primary breast cancers
.
J Clin Invest
2007
;
117
:
1370
80
.
30
Frasca
F
,
Vella
V
,
Aloisi
A
, et al
. 
p73 tumor-suppressor activity is impaired in human thyroid cancer
.
Cancer Res
2003
;
63
:
5829
37
.
31
Muller
M
,
Schilling
T
,
Sayan
AE
, et al
. 
TAp73/ΔNp73 influences apoptotic response, chemosensitivity and prognosis in hepatocellular carcinoma
.
Cell Death Differ
2005
;
12
:
1564
77
.
32
Casciano
I
,
Mazzocco
K
,
Boni
L
, et al
. 
Expression of ΔNp73 is a molecular marker for adverse outcome in neuroblastoma patients
.
Cell Death Differ
2002
;
9
:
246
51
.
33
Wager
M
,
Guilhot
J
,
Blanc
JL
, et al
. 
Prognostic value of increase in transcript levels of Tp73 ΔEx2-3 isoforms in low-grade glioma patients
.
Br J Cancer
2006
;
95
:
1062
9
.
34
Becker
K
,
Pancoska
P
,
Concin
N
, et al
. 
Patterns of p73 N-terminal isoform expression and p53 status have prognostic value in gynecological cancers
.
Int J Oncol
2006
;
29
:
889
902
.
35
Lunghi
P
,
Costanzo
A
,
Levrero
M
,
Bonati
A
. 
Treatment with arsenic trioxide (ATO) and MEK1 inhibition activates the p73-53AIP1 apoptotic pathway in leukemia cells
.
Blood
2004
;
104
:
519
25
.
36
Lunghi
P
,
Costanzo
A
,
Salvatore
L
, et al
. 
MEK1 inhibition sensitizes primary acute myelogenous leukemia to arsenic trioxide-induced apoptosis
.
Blood
2006
;
107
:
4549
53
.
37
Rizzo
MG
,
Giombini
E
,
Diverio
D
, et al
. 
Analysis of p73 expression pattern in acute myeloid leukemias: lack of ΔN-p73 expression is a frequent feature of acute promyelocytic leukemia
.
Leukemia
2004
;
18
:
1804
9
.
38
Mainardi
S
,
Pelosi
A
,
Palescandolo
E
, et al
. 
ΔNp73 is a transcriptional target of the PML/RARα oncogene in myeloid differentiation
.
Cell Death Differ
2007
;
14
:
1968
71
.
39
Dominguez
G
,
Garcia
JM
,
Pena
C
, et al
. 
ΔTAp73 upregulation correlates with poor prognosis in human tumors: putative in vivo network involving p73 isoforms, p53, and E2F-1
.
J Clin Oncol
2006
;
24
:
1
11
.
40
Bozzetti
C
,
Nizzoli
R
,
Musolino
M
, et al
. 
p73 and p53 pathway in human breast cancers
.
J Clin Oncol
2007
;
25
:
1541
3
.
41
Di Vinci
A
,
Sessa
F
,
Casciano
I
, et al
. 
Different intracellular compartmentalization of TA and ΔNp73 in non-small cell lung cancer
.
Int J Oncol
2009
;
34
:
449
56
.
42
Uramoto
H
,
Sugio
K
,
Oyama
T
, et al
. 
Expression of δNp73 predicts poor prognosis in lung cancer
.
Clin Cancer Res
2004
;
10
:
6905
11
.
43
Gong
JG
,
Costanzo
A
,
Yang
HQ
, et al
. 
The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin-induced DNA damage
.
Nature
1999
;
399
:
806
9
.
44
Agami
R
,
Blandino
G
,
Oren
M
,
Shaul
Y
. 
Interaction of c-Abl and p73α and their collaboration to induce apoptosis
.
Nature
1999
;
399
:
809
13
.
45
Yuan
Z-M
,
Shioya
H
,
Ishiko
T
, et al
. 
p73 is regulated by tyrosine kinase c-Abl in the apoptotic response to DNA damage
.
Nature
1999
;
399
:
814
7
.
46
Tsai
KK
,
Yuan
ZM
. 
c-Abl stabilizes p73 by a phosphorylation-augmented interaction
.
Cancer Res
2003
;
63
:
3418
24
.
47
Sanchez-Prieto
R
,
Sanchez-Arevalo
VJ
,
Servitja
JM
,
Gutkind
JS
. 
Regulation of p73 by c-Abl through the p38 MAP kinase pathway
.
Oncogene
2002
;
21
:
974
9
.
48
Vella
V
,
Zhu
J
,
Frasca
F
, et al
. 
Exclusion of c-Abl from the nucleus restrains the p73 tumor suppression function
.
J Biol Chem
2003
;
278
:
25151
7
.
49
Vigneri
P
,
Wang
JY
. 
Induction of apoptosis in chronic myelogenous leukemia cells through nuclear entrapment of BCR-ABL tyrosine kinase
.
Nat Med
2001
;
7
:
228
34
.
50
Costanzo
A
,
Merlo
P
,
Pediconi
N
, et al
. 
DNA damage-dependent acetylation of p73 dictates the selective activation of apoptotic target genes
.
Mol Cell
2002
;
9
:
175
86
.
51
Mantovani
F
,
Piazza
S
,
Gostissa
M
, et al
. 
Pin1 links the activities of c-Abl and p300 in regulating p73 function
.
Mol Cell
2004
;
14
:
625
36
.
52
Strano
S
,
Monti
O
,
Pediconi
N
, et al
. 
The transcriptional coactivator Yes-associated protein drives p73 gene-target specificity in response to DNA damage
.
Mol Cell
2005
;
18
:
447
59
.
53
Levy
D
,
Adamovich
Y
,
Reuven
N
,
Shaul
Y
. 
Yap1 phosphorylation by c-Abl is a critical step in selective activation of pro-apoptotic genes in response to DNA damage
.
Mol Cell
2008
;
29
:
350
61
.
54
Jiménez-Velasco
A
,
Roman-Gomez
J
,
Agirre
X
, et al
. 
Down-regulation of the large tumor suppressor 2 (LATS2/KPM) gene is associated with poor prognosis in acute lymphoblastic leukemia
.
Leukemia
2005
;
19
:
2347
50
.
55
Takahashi
Y
,
Miyoshi
Y
,
Takahata
C
, et al
. 
Downregulation of LATS1 and LATS2mRNA expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers
.
Clin Cancer Res
2005
;
11
:
1380
5
.
56
Kawahara
M
,
Hori
T
,
Chonabayashi
K
,
Oka
T
,
Sudol
M
,
Uchiyama
T
. 
Kpm/Lats2 is linked to chemosensitivity of leukemic cells through the stabilization of p73
.
Blood
2008
;
112
:
3856
66
.
57
Milella
M
,
Kornblau
SM
,
Estrov
Z
, et al
. 
Therapeutic targeting of the MEK/MAPK signal transduction module in acute myeloid leukemia
.
J Clin Invest
2001
;
108
:
851
9
.
58
Lee
JT
,
McCubrey
JA
. 
The Raf/MEK/ERK (MAPK) signal transduction cascade as a target for chemotherapeutic intervention in leukemia
.
Leukemia
2002
;
16
:
486
507
.
59
Lunghi
P
,
Tabilio
A
,
Dall'Aglio
PP
, et al
. 
Down-modulation of ERK activity inhibits the proliferation and induces the apoptosis of primary acute myelogenous leukemia blasts
.
Leukemia
2003
;
17
:
1783
93
.
60
Oda
K
,
Arakawa
H
,
Tanaka
T
, et al
. 
p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46-phosphorylated p53
.
Cell
2000
;
102
:
849
62
.
61
Matsuda
K
,
Yoshida
K
,
Taya
Y
,
Nakamura
Y
,
Arakawa
H
. 
p53AIP1 regulates the mitochondrial apoptotic pathway
.
Cancer Res
2002
;
62
:
2883
9
.
62
Lunghi
P
,
Giuliani
N
,
Mazzera
L
, et al
. 
Targeting MEK/MAPK signal transduction module potentiates ATO-induced apoptosis in multiple myeloma cells through multiple signaling pathways
.
Blood
2008
;
112
:
2450
62
.
63
Shammas
MA
,
Neri
P
,
Koley
H
, et al
. 
Specific killing of multiple myeloma cells by (-)-epigallocatechin-3-gallate extracted from green tea: biologic activity and therapeutic implications
.
Blood
2006
;
108
:
2804
10
.
64
Raab
MS
,
Breitkreutz
I
,
Tonon
G
, et al
. 
Targeting PKC: a novel role for β catenin in ER stress and apoptosis signalling
.
Blood
2009
;
12
:
1513
21
.
65
Bernassola
F
,
Salomoni
P
,
Oberst
A
, et al
. 
Ubiquitin-dependent degradation of p73 is inhibited by PML
.
J Exp Med
2004
;
199
:
1545
7
.
66
Ito
K
,
Bernardi
R
,
Morotti
A
, et al
. 
PML targeting eradicates quiescent leukaemia-initiating cells
.
Nature
2008
;
453
:
1072
8
.
67
Lapi
E
,
Di Agostino
S
,
Donzelli
S
, et al
. 
PML, YAP, and p73 are components of a proapoptotic autoregulatory feedback loop
.
Mol Cell
2008
;
32
:
803
14
.
68
Hermeking
H
. 
p53 enters the microRNA world
.
Cancer Cell
2007
;
12
:
414
8
.
69
Sampath
D
,
Calin
GA
,
Puduvalli
VK
, et al
. 
Specific activation of microRNA106b enables the p73 apoptotic response in chronic lymphocytic leukemia by targeting the ubiquitin ligase Itch for degradation
.
Blood
2009
;
113
:
3744
53
.
70
Benard
J
,
Douc-Rasy
S
,
Ahomadegbe
JC
. 
TP53 family members and human cancers
.
Hum Mutat
2003
;
21
:
182
91
.
71
Bell
HS
,
Dufes
C
,
O'Prey
J
, et al
. 
A p53-derived apoptotic peptide derepresses p73 to cause tumor regression in vivo
.
J Clin Invest
2007
;
117
:
1008
18
.
72
Bergamaschi
D
,
Samuels
Y
,
Jin
B
,
Duraisingham
S
,
Crook
T
,
Lu
X
. 
ASPP1 and ASPP2: common activators of p53 family members
.
Mol Cell Biol
2004
;
24
:
1341
50
.
73
Bergamaschi
D
,
Samuels
Y
,
Sullivan
A
, et al
. 
iASPP preferentially binds p53 proline-rich region and modulates apoptotic function of codon 72-polymorphic p53
.
Nat Genet
2006
;
38
:
1133
41
.
74
Mantovani
F
,
Tocco
F
,
Girardini
J
, et al
. 
The prolyl isomerase Pin1 orchestrates p53 acetylation and dissociation from the apoptosis inhibitor iASSP
.
Nat Struct Mol Biol
2007
;
14
:
912
20
.
75
Wang
W
,
Kim
S-H
,
El-Deiry
WS
. 
Small-molecule modulators of p53 family signaling and antitumor effects in p53-deficient human colon tumor xenografts
.
Proc Natl Acad Sci U S A
2006
;
103
:
1103
8
.
76
Kravchenko
JE
,
Ilyinskaya
GV
,
Komarov
PG
, et al
. 
Small-molecule RETRA suppresses mutant p53-bearing cancer cells through a p73-dependent salvage pathway
.
Proc Natl Acad Sci U S A
2008
;
105
:
6302
7
.
77
Di Agostino
S
,
Cortese
G
,
Monti
O
, et al
. 
The disruption of the protein complex mutantp53/p73 increases selectively the response of tumor cells to anticancer drugs
.
Cell Cycle
2008
;
7
:
3440
7
.
78
Irwin
MS
. 
Family feud in chemosensitivity. P73 and mutant p53
.
Cell Cycle
2004
;
3
:
319
23
.
79
Bell
HS
,
Ryan
KM
. 
iASPP inhibition: increased options in targeting the p53 family for cancer therapy
.
Cancer Res
2008
;
68
:
4959
62
.
80
Whitehead
KA
,
Langer
R
,
Anderson
DG
. 
Knocking down barriers: advances in siRNA delivery
.
Nat Rev Drug Discov
2009
;
8
:
129
38
.
81
Kojima
K
,
Konopleva
M
,
McQueen
T
,
O'Brien
S
,
Plunkett
W
,
Andreeff
M
. 
Mdm2 inhibitor Nutlin-3a induces p53-mediated apoptosis by transcription-dependent and transcription independent mechanisms and may overcome Atm-mediated resistance to fludarabine in chronic lymphocytic leukemia
.
Blood
2006
;
108
:
993
1000
.
82
Kojima
K
,
Konopleva
M
,
Samudio
IJ
, et al
. 
MDM2 antagonists induce p53-dependent apoptosis in AML: implications for leukemia therapy
.
Blood
2005
;
106
:
3150
9
.
83
Lau
LMS
,
Nugent
JK
,
Zhao
X
,
Irwin
MS
. 
HDM2 antagonist Nutlin-3 disrupts p73-2 binding and enhances p73 function
.
Oncogene
2008
;
27
:
997
1003
.
84
Dar
AA
,
Belkhiri
A
,
Ecsedy
J
,
Zaika
A
,
El-Rifai
W
. 
Aurora kinase A inhibition leads to p73-dependent apoptosis in p53-deficient cancer cells
.
Cancer Res
2008
;
68
:
8998
9004
.
85
Lau
LMS
,
Wolter
JK
,
Lau
JTML
, et al
. 
Cyclooxygenase inhibitors differentially modulate p73 isoforms in neuroblastoma
.
Oncogene
2009
;
28
:
2024
33
.
86
Meric-Berstein
F
,
Gonzalez-Angulo
AM
. 
Targeting the mTOR signaling network for cancer therapy
.
J Clin Oncol
2009
;
27
:
2278
87
.
87
Rosenbluth
JM
,
Mays
DJ
,
Pino
MF
,
Tang
LJ
,
Pietenpol
JA
. 
A gene signature-based approach identifies mTOR as a regulator of p73
.
Mol Cell Biol
2008
;
28
:
5951
64
.
88
Crighton
D
,
O'Prey
J
,
Bell
HS
,
Ryan
KM
. 
p73 regulates DRAM-independent autophagy that does not contribute to programmed cell death
.
Cell Death Differ
2007
;
14
:
1071
9
.
89
Rosenbluth
JM
,
Pietenpol
JA
. 
mTOR regulates autophagy-associated genes downstream of p73
.
Autophagy
2009
;
5
:
114
6
.
90
Lamb
J
,
Crawford
ED
,
Peck
D
, et al
. 
The Connectivity Map: using gene expression signatures to connect small molecules, genes, and disease
.
Science
2006
;
313
:
1929
35
.
91
Lamb
J
. 
The Connectivity Map: a new tool for biomedical research
.
Nat Rev Cancer
2007
;
7
:
54
60
.