Abstract
Nasopharyngeal carcinoma (NPC) is a form of head and neck cancer of multifactorial etiolo-gies that is highly prevalent among men in the population of Southern China and Southeast Asia. NPC has claimed many thousands of lives worldwide; but the low awareness of NPC remains a hindrance in early diagnosis and prevention of the disease. NPC is highly responsive to radiotherapy and chemothera-py, but radiocurable NPC is still dependent on concurrent treatment of megavoltage radiotherapy with chemotherapy. Despite a significant reduction in loco-regional and distant metastases, radiotherapy alone has failed to provide a significant improvement in the overall survival rate of NPC, compared to chemo-therapy. In addition, chemo-resistance persists as the major challenge in the management of metastatic NPC although the survival rate of advanced metastatic NPC has significantly improved with the admin-istration of chemotherapy adjunctive to radiotherapy. In this regard, targeted molecular therapy could be explored for the discovery of alternative NPC therapies. Nutlin-3, a small molecule inhibitor that specifi-cally targets p53-Mdm2 interaction offers new therapeutic opportunities by enhancing cancer cell growth arrest and apoptosis through the restoration of the p53-mediated tumor suppression pathway while pro-ducing minimal cytotoxicity and side effects. This review discusses the potential use of Nutlin-3 as a p53-activating drug and the future directions of its clinical research for NPC treatment.
Keywords: Apoptosis, Epstein-Barr virus (EBV), carcinoma, cisplatin, small molecule inhibitor, targeted molecular therapy
1. INTRODUCTION
Nasopharyngeal carcinoma (NPC) is a common epithelial squamous cell head and neck carcinoma that originates from the nasopharyngeal mucosa layering the upper part of the throat. It is the most common type of nasopharyngeal tumor [1, 2]. NPC has an unusual racial distribution that is strongly associated with multifactorial etiologies, such as gamma herpes Epstein-Barr virus (EBV) infection [3], the intake of salted fish [2], cigarette smoking [4], occupational exposures [5], environmental risk factors and genetic susceptibility [2, 6, 7].
NPC is strongly associated with EBV infection and intake of salted fish. In 1966, Old et al. discovered that NPC is an EBV-associated cancer [8] and almost 95% of NPC tumors were found associated with EBV infection [9]. Salted fish is rich in carcinogenic volatile nitrosamine, an EBV activating agent. The polymorphic nitrosamine metabolizing genes, cytochrome CYP2E1 and CYP2A6 are responsible for NPC susceptibility [5, 10]. Several commonly used herbal plant extracts have the ability to reactivate EBV as well as to increase the titers of anti-EBV antibody in EBV-infected host [5]. A study conducted among Chinese populations revealed that consumption of Chinese medicine is another dietary-related factor for NPC. A significant correlation between traditional herbal medicine consumption and increased NPC risk has been linked with NPC pathogenesis [11]. Alcohol consumption, a common lifestyle in the West and other parts of the world, has been identified as another important risk factor for the development of NPC [12].
2. EPIDEMIOLOGY
NPC is fairly rare in most parts of the world, indicating distinct racial and geographical distribution. Although it is considered as rare, NPC incidence remains significantly high in endemic regions of Southern China [13], Hong Kong, Taiwan, Northern Africa [2] and Southeast Asia. According to the global scale statistics [14], NPC is the 23rd most common cancer with nearly 80,000 new cases diagnosed annually and a mortality rate that exceeds 50,000. NPC is more prevalent in males than females, with the male to female ratio of 2.3:1.0 [15]. High-risk rates of ASR 25-30 (per 100,000 populations) were seen in Southern China notably among the Cantonese in the area of Guangzhou; high incidence rates are also noted in Southeast Asia. In contrast, low incidence rates of ASR < 1 were observed in Europe and North America [14].
NPC is more prevalent among the Chinese populations and this could be attributed to their lifestyles, such as consumption of large amount of carcinogen-contaminated salted fish. Consumption of salted fish is also reported to have a strong correlation with NPC [16]. In addition, early childhood exposure to diet that is high in preserved foods and salted fish is shown to have significant effects on higher NPC risk [13, 17]. Approximately 90% NPC cases from Hong Kong [16], 60% from Malaysian Chinese [18] and 50% from Guangzhou [19] are attributed to frequent consumption of salted fish in childhood. Several case-control studies observed that high-risk NPC populations frequently have high intake of preserved food, pickled vegetables and fermented products, such as beans, bean pastes, eggs and seafood pastes [20, 21].
3. THERAPY COMPLICATIONS
Nasopharyngeal tissue consists of several types of cells, whereby, each type has the ability to transform into different types of tumors. These differences are significant for the classification of the disease subtypes, severity as well as the efficacy of treatment. The selection of treatment option for NPC is dependent on the tumor location and disease extent [22]. Surgery is rarely the main therapeutic option for NPC due to its complex anatomical proximity to critical structures. Traditional surgery leads to transposition of obstructing bone and soft tissue, and has risk of nerves and blood vessels damages, weakness in the arm or lower lip and numbness of the ear [23, 24]. Endoscopic nasopharyngectomy causes significant postoperative complications including surgical pain, headache, nasopharyngeal necrosis, and crusting of the otitis media [25, 26].
NPC is mainly treated with radiotherapy, often in combination with chemotherapy based on the different stages of the disease [27, 28]. The major concerns of radiotherapy are specific organ damage in the head and neck regions due to exposure to harmful radiation. Radiotherapy contributed to radiation-induced sensory neural hearing loss (SNHL) with an incidence rate of 24.2% [29]. Intensity-modulated radiotherapy (IMRT) improves the loco-regional control of NPC, but distant metastasis and recurrence remain the main causes of treatment failure [27, 30, 31].
The most active chemotherapy agents used in NPC are cisplatin and 5-fluorouracil (5-FU) [27, 28]. These drugs are highly toxic yet not cancer cell specific as they are cytotoxic to actively dividing cells during mitotic phase [32, 33]. Cisplatin is extensively used mainly due to its effectiveness in treating difficult to cure cancer, but is associated with nephrotoxicity, neurotoxicity, ototoxicity, alopecia, myelotoxicity, nausea and vomiting [29, 34, 35]. Among the severe adverse effects of 5-FU are chest pain associated with heart disease, fatigue, nausea, mouth sores, photophobia (light sensitivity) and diarrhea.
NPC patients are often adversely affected by post-chemo-radiotherapy effects and tend to experience therapy failure mainly due to local recurrence [36]. In an effort to improve the prognosis and efficacy of NPC therapy, a combination of definitive radiotherapy plus cisplatin-based chemotherapy is recommended [37]. Concurrent advanced combined chemo-radiotherapy (CCRT) technique is the main treatment modality for advanced-stage NPC [28]. However, the use of chemotherapy either before or after radiotherapy for patients with advanced NPC has shown to yield no improvement on the overall survival or relapse-free survival [38, 39]. Severe chemo-radio adverse effects, post-treatment late complications such as osteoradionecrosis and xerostomia, therapy-resistance, loco-regional recurrence and distant metastases remain as major causes of mortality and morbidity in NPC [40-42]. Nearly 50% NPC patients experience cancer recurrence; 80% reported cases of incurring auxiliary late complications of long-term toxicity effect [40] and 43% treated-patients showing distant metastases within four years [37, 43]. The addition of cisplatin to CCRT increased the risk of SNHL, severe hearing impairment or deafness [29]. Furthermore, the emergence of chemo-resistance remains as major obstacle for cisplatin-based chemotherapy which impaired cisplatin-induced apoptosis [44].
The anti-epidermal growth factor receptor (EGFR), cetuximab, is the first monoclonal antibody specifically designed to inhibit EGFR in both normal and cancer cells. Cetuximab enhances the sensitivity of cancer cells to chemo-radiotherapy. It has been approved for use during chemotherapy with cisplatin or along with radiotherapy [45, 46]. Cetuximab therapy rarely has side effects, but patients may develop severe skin allergy, infusion reaction, anemia, cardiac toxicity and lung disease [45].
In addition, early stage NPC may be asymptomatic or can be presented with trivial symptoms, thus likely to escape early diagnosis, which results in undetected progression to stages III or IV of the disease [47, 48]. Late stage NPC is associated with poor prognosis and treatment failures [28, 36]. Although the initial response and local control rates achieved with advance radiotherapy and/or chemotherapy in advanced stage III and IV NPC are high, NPC remains incurable with unacceptable 5- and 10-year survival rates [49]. The 5-year overall survival rate of stage III and IV NPC patients were reported to be 56.2% for radiotherapy and 47.2% for chemotherapy [49]. In fact, recurrence, distant metastases, drug resistance and adverse effects of treatments remain as major challenges in the clinical scenario [40, 42].
Therefore, reducing undesirable complications of chemotherapy drugs is a major goal in pharmaceutical research for NPC treatment. In view of these issues and challenges, the design of modern cancer therapeutic must evolve from non-specific targeting that affects both normal and cancer cells to specifically targeting unique molecular signature of cancer cells and to produce greater antitumor efficacy with less toxicity effects, such as p53-based targeted therapy.
4. p53-Mdm2 INTERACTION: A THERAPEUTIC TARGET
4.1. Tumor Suppressor p53
The p53 gene located on chromosome 17 (17p13.1), contains 11 exons, which encode for a 2.8 kb mRNA, translated into a 53 kDa protein made up of 393 amino acids [50, 51]. The wild type (wt) p53 tumor suppressor gene has been termed as the “guardian of the genome” [52], or “cellular gatekeeper of growth and division” [53] due to its important biological roles in protecting cells from becoming cancerous by regulating the transition of G1-S phase in renewable functional tissue and preventing gene aberration for genome stability by eradicating DNA-damaged cells in a natural way [52].
4.2. Antagonizing Mdm2 to Reactivate p53
The murine double-minute type 2 protein (Mdm2), known in human as Hdm2, is encoded by an oncogene located on the acentromeric extra chromosomal nuclear bodies of chromosome 12 (12q14.3 – q15). Mdm2 acts as an inhibitor of the tumor-suppressive effects of p53. In a normal cell, Mdm2 protein is the main regulator in mastering the stability and activity of p53 protein by E3-ubiquitin ligase activity [54, 55]. The wt p53 is a potent pro-apoptotic protein with a short half-life of 6-20 minutes (Fig. 1). In the absence of stress, p53 which retards inappropriate growth arrest and apoptosis is almost undetectable in proliferating cells. Dissociation of p53-Mdm2 complex occurs in response to cellular stress, DNA damage or activation of oncogene which leads to the activation of p53 [56] and free p53 protein from Mdm2 binding (Fig. 2). The degradation of p53 is inhibited resulting in prolonged p53 half-life to hours and accumulation of p53 in the cell [57]. As a DNA damage-inducible nuclear transcription factor, p53 determines whether the damaged cells are repaired or undergo self-destruction [58]. Reactivation of p53 promotes DNA repair, cell cycle arrest, senescence or cell death by apoptosis [59]; thereby protecting the cell from malignant transformation, suppressing tumor development and eradicating tumor in a natural way. Its tumor suppressing activities are recognized in human cancers [60]; and Mills confirmed that wt p53 contributes tumor-suppressive capabilities compared to mutated p53, which is oncogenic [61].
Fig. (1).
The stabilization of p53 by Mdm2 in normal cell. The regulation of p53 is tightly controlled by its oncogenic E3 ubiquitin ligase negative regulator Mdm2.
Fig. (2).
The wt p53 protects cells from genome mutation, eradicates tumor cells by activating cell cycle arrest, DNA repair and apoptosis.
4.3. Nutlin-3: Inhibitor of the p53-Mdm2 Interaction
Nutlin-3 (C30H30Cl2N4O4, 581.4896 g/mol) is a small molecular weight cis-imidazoline analogue that was designed to compete with Mdm2 for binding to p53 [102]. Nutlin-3 induces the regulation and activation of p53 pathway [102], and is found to be effective and non-genotoxic in stabilizing p53 using experimental models in tumors expressing wt p53 (Fig. 3). Since its discovery, in vitro and in vivo therapeutic-based studies have revealed that Nutlin-3 could
Fig. (3).
p53-dependent effects of Nutlin-3.
be a potential alternative for targeted therapy in the current chemotherapy regime. Nutlin-3 has been reported to selectively enhance apoptosis in wt p53 cancer cells by activating the p53 pathway (Table 1). Nutlin-3 is non-genotoxic and protects normal cells against mitotic toxicity [103, 104] and kidney cells from the cytotoxic effect of cisplatin [105]. Apart from Nutlin-3, other small molecules that reactivate the p53 pathway undergoing clinical trials are shown in (Table 2).
Table 1.
Summary of published experimental Nutlin-3 for human cancer therapy.
Published Research | Cancer Types | p53-dependent Actions of Nutlin-3 |
---|---|---|
Kojima et al., 2005 [62] | AML | Inhibits p53-Mdm2 binding-induced growth arrest and apoptosis in AML cells. |
Stuhmer et al., 2005 [63] | MM | Activates p53 pathway and induces apoptosis in MM cells. |
Tovar et al., 2006 [64] | OS | Induces G1 and G2 phase cell-cycle arrest function of the p53 pathway in OS cells. |
Muller et al., 2007 [65] | LS | Induces apoptosis in LS cells with high wt p53 levels. |
Saddler et al., 2008 [66] | CLL | p53 status is the major determinant of response to Nutlin-3 in CLL cells. |
Miyachi et al., 2009 [67] | RMS | Restores p53 pathway-induced apoptosis and cell cycle arrest in RMS cells. |
Van Maerken et al., 2009 [68] | NB | Activates p53 pathway in chemoresistant NB cells. Induces apoptosis and suppresses distant metastasis in xenograft model NB. |
Hori et al., 2010 [69] | CCa | Sensitizes CCa cells to TRAIL-induced mitochondrial dysfunction, and increases death receptor DR5 promoter. |
Koster et al., 2011 [70] | TC | Upregulates Fas membrane expression and increases Fas death receptor expression in TC cells. |
Sonnemann et al., 2011 [71] | ES | Induces cellular senescence, antineoplastic effects and apoptosis in ES cells. |
Ye et al., 2012 [72] | KS | Activates p53 pathway-induced apoptosis and inhibites “KS-like” tumor growth in nude mice. |
Voon et al., 2015 [73] | NPC | Activates p53 pathway and induces apoptosis in EBV-positive NPC cells. Sensitizes NPC cells to cisplatin-induced apoptosis by modulating pro-apoptotic targets via p53 pathway. |
AML, acute myeloid leukemia; MM, multiple myeloma; OS, osteosarcoma; LS, liposarcoma; CLL, chronic lymphocytic leukemia; RMS, rhabdo-myosarcoma; NB, neuroblastoma; CCa, colon cancer; TC, testicular cancer; ES, Ewing’s sarcoma; KS, Kaposi sarcoma.
Table 2.
Small-molecule p53 activators currently in clinical trials.
Compound | Small-Molecule | Clinical Trial Development | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
Stage of Development | Cancer Types | p53-Dependent Actions | ||||||||
Activate wt p53 by: | ||||||||||
Mdm2 binding: Mimics the p53 amino acid side-chains involved in the binding of the p53 peptide to Mdm2 that inhibits p53-Mdm2 binding |
Nutlins (RG7112, RO5045337) |
cis-imidazoline | Phase I/II [64, 74] |
Advanced solid tumors, sarcoma, liposarcomas, neoplasms, hematological malignancies | Induces cell cycle arrest Induces apoptosis |
|||||
BDA (TDP 665759 TDP 521252) |
Benzodiazepine derivatives | Preclinical [75, 76] |
JAR choriocarcinoma, hepatocellular carcinoma | Inhibits cancer cell proliferation Induces apoptosis |
||||||
Hdm2 binding: Inhibits p53-Hdm2 binding, prevents p53 degradation by its inhibitor Hdm2 and enables p53 activation |
Spiro-oxindoles (MI-219, MI-63, MI-43) |
Computational modeling | Preclinical [77-79] |
Breast cancer, colon carcinoma, prostate cancer, lung cancer, hematological malignancies | Inhibits cancer cell proliferation Induces apoptosis Inhibits tumor growth |
|||||
Serdemetan (JNJ-26854165) |
Tryptamine derivative | Phase I [80, 81] |
B-cell lymphoma, colorectal cancer, lymphoma, melanomas, sarcomas, acute leukemia, lung cancer | Induces apoptosis Increases p53 stability Reduces the rate of DNA synthesis |
||||||
MdmX binding: Mimics the p53 amino acid side-chains involved in the binding of the p53 peptide to MdmX that inhibits p53-MdmX binding |
SJ-172550 | Identified through a peptide-based high throughput screening | Preclinical [82] |
MdmX over-expressing tumor, retinoblastoma | Induces cell cycle arrest Induces apoptosis |
|||||
RO-2443/ RO-5693 |
Indolyl hydantoin | Preclinical [83] |
MdmX over-expressing tumors, breast cancer, osteosarcoma | Induces cell cycle arrest Induces apoptosis |
||||||
XI-011 | Pseudourea derivative | Preclinical [84-86] |
Breast cancer | Induces apoptosis | ||||||
p53 binding: Induces conformational change that prevent p53-Mdm2 binding |
RITA (NSC 652287) |
2,5-bis(5-hydroxymethyl- 2-thienyl)furan |
Preclinical [74, 87, 88] |
Multiple myeloma (mt), AML, CLL, colon carcinoma, lung cancer, breast cancer, Burkitt’s lymphoma | Induces cell cycle arrest Induces apoptosis |
|||||
E3 ubiquitin ligase inhibition: Enhances the autoubiquitination and degradation of Hdm2’s E3 activity that acts as the predominant p53 negative regulator |
HLI98, HLI373 | 7-nitro-10-aryl-5 deazaflavins | Preclinical [89, 90] |
Human retinal pigment epithelial cells, colon carcinoma, melanoma, lung carcinoma, fibrosarcoma, osteosarcoma | Activates p53-dependent transcription Induces apoptosis |
|||||
Protein deacetylators inhibition: Inhibits deacetylase activity of protein deacetylators SirT1 and SirT2; and results in the stabilization of p53 |
Tenovin 1 Tenovin 6 |
Identified by screening of small molecules from Chembridge DIVERSet | Preclinical [91, 92] |
Burkitt’s lymphoma, melanoma, breast cancer, colon carcinoma | Negative regulation of p53 Delays tumor growth in vivo Induces cell cycle arrest Induces apoptosis |
|||||
Restoration of wt function of mt p53 by: | ||||||||||
Conformational stabilization of p53 DBD: Interacts with DNA, reactive oxygen species |
CP-31398 (V173A, R175S, R249S, R273H) |
Styrylquinazoline synthetic | Preclinical [93] |
Osteosarcoma, colon carcinoma, melanoma, lung carcinoma | Stabilizing the active conformation of wt p53 DBD | |||||
Chelation/redox modulation: Restores wt-like DNA binding properties to mt protein p53R273H (DNA contact) or p53R175H (conformational) |
Thiosemicarbazoneg: Zinc |
Metal ion chelators | Preclinical [94, 95] |
p53R175 mt ovarian carcinoma, p53R175 mt breast cancer, p53R273H mt glioblastoma, p53-null lung carcinoma |
Induces apoptosis Induces cell cycle arrest Improves the sensitivity of mt p53-expressing cells to anti-tumor drugs |
|||||
Covalent modifications of cysteine residues: Induces wt-like conformational change and protein folding to mt protein p53R273H or p53R175H |
PRIMA-1MET (APR-246) |
Identified by screening of NCI chemical library that inhibits Saos-2 cell proliferation | Phase I/II [96] |
Hematologic malignancies, prostate cancer, lung cancer, prostate cancer | Induces cell cycle arrest Induces apoptosis |
|||||
Alkylation of cysteine and lysine residues: Reactivates DNA binding and preserves the conformation of mt p53 protein |
MIRA-1 | Maleimide derivative | Preclinical [97, 98] |
Myeloma, osteosarcoma, lung adenocarcinoma, ovarian carcinoma, colon carcinoma | Restores apoptotic activity to mt p53 Induces apoptosis |
|||||
Bindings to the mutation-induced cleft: Raises Tm, slows down the thermal denaturation of mt p53-Y220C |
PhiKan083 | Carbazole derivative | Preclinical [99-101] |
p53-Y220C mt gastric cancer, hepatoblastoma | Regains structural stability and restores wt p53 conformation of mt p53-Y220C Increases p53 half-lives by stabilizing ligands |
AML, acute myeloid leukemia; BDA, benzodiazepinediones; CLL, chronic lymphocytic leukemia; DBD, DNA binding domain; Hdm2, human double minute 2; MIRA-1, mutant p53-dependent induction of rapid apoptosis; mt, mutant; PRIMA-1, p53 reactivation and induction of massive apoptosis; RITA, reactivation of p53 and induction of tumor cell apoptosis; Saos-2, osteosarcoma cells; Tm, melting temperature; wt, wild type.
4.4. Nutlin-3: A Potential Anti-NPC Agent
Mutations in p53 alter its ability for DNA repair, thus contributing to cancer development [106-108]. In a comparative study using several previously sequenced tumors, p53 is the most frequently mutated gene in epithelial malignancies [109]. p53 mutation is common in the early stage of human lung, colon, head and neck cancers [110]. Mutant p53 has a long half-life when compared to wt p53 found in over 60% of head and neck cancer. However, p53 mutations have been reported to be rare in NPC [6, 111, 112]. The findings of a genome-wide association study confirmed that NPC oncogenesis is strongly related to multiple genetic alterations, which involved point mutations of p53 gene detected in hot spots of exons 5, 7 and 8 in <10% of NPC cases [113].
Apart from these reported events, Lin and colleagues [109] recently determined nine significantly mutated genes with the two most commonly found in NPC being PIK3CA and p53. The whole-exome sequencing (WES) and single-nucleotide polymorphism (SNP) array profiles showed detectable G1-S cell cycle transition with most frequent 9p21 and p53 genes deletions detected in 28% of NPCs [109]. However, using the most sensitive combination of WES, targeted deep sequencing and SNP array approaches, it is shown that NPC results in a relatively low level of genomic alteration, as well as rare p53 mutation frequency with <10% cases observed in EBV-associated NPCs [109]. The p53 mutation is rare even in recurrent radiotherapy refractory NPC [114], thus making NPC a potential candidate for treatment with p53 activators, like Nutlin-3. p53 is often overexpressed in NPC cells [115, 116] and overexpression of p53 using an adenoviral vector has been reported effective against NPC [117, 118] indicating that further elevating p53 levels by Nutlin-3 could provide an alternative treatment strategy against NPC.
In our previous study, we have demonstrated that Nutlin-3 could activate the p53 pathway for sensitization of cisplatin-induced apoptosis in EBV-positive NPC cells in a p53-dependent manner [73]. Our findings suggest that restoration of wt p53 with Nutlin-3 maximizes the protection of normal cells while it enhances cytotoxicity of cisplatin. Cisplatin was more cytotoxic to the normal nasopharyngeal cells compared to the NPC cells, while Nutlin-3 was more selective in inhibiting NPC cells. The combination drug treatment of cisplatin and Nutlin-3 showed stronger anti-proliferative effect against NPC cells, demonstrating that combined treatment was more effective than single drug treatment [73]. In addition, combination treatment of NPC cells with cisplatin and Nutlin-3 significantly induced the accumulation of apoptotic cells, concomitant with the upregulation of the protein expressions of apoptosis regulators, BAX and PUMA and detection of an apoptosis marker, cleaved PARP. These indicate that Nutlin-3 sensitizes NPC cells to cisplatin-induced apoptosis by modulating pro-apoptotic targets via the p53 pathway. In this regard, reducing the dose of cisplatin could lead to lesser adverse effects while retaining stronger cytotoxic effect against NPC cells.
Apart from these reported events, we have also determined that the extended treatment of NPC cells with Nutlin-3 did not result in the emergence of p53 mutation, albeit reduced sensitivity to Nutlin-3 was observed [73]. This stresses on the importance of treatment duration and clinical doses optimization to improve the efficacy of Nutlin-3 significantly. Collectively, these findings are important for the development and design of clinical trials of Nutlin-3 for NPC treatment in the near future.
5. FUTURE DIRECTIONS
The margin of success of standard therapy approach for most radiocurable NPC is still dependent on concurrent megavoltage radiotherapy [27, 30, 40]. However, disease relapse, refractory diseases and management of long-term toxicities are critical issues that remain to be addressed. In view of these issues, the National Comprehensive Cancer Network Clinical Practice Guidelines recommend the utilization of multiple alternative strategies as the current standard cancer therapies to improve the prognosis of hardly curable NPC [76]. On the other hand, the need to find new drug targets and establish novel effective therapy for NPC necessitates the focus on specific targeting of unique molecular signature of the cancer cells rather than non-specific targeting that affects both normal and cancer cells.
Despite all the advances in cancer therapies, more drugs that reactivate the p53 pathway are entering clinical trials because of the growing demand for alternative therapy to cure cancers [119]. The preliminary trials on p53-activating therapies showed that appropriate design of a clinical trial remains a crucial issue [119]. Identifying tumors with low incidence p53 mutation and highly responsive to p53-activating drugs, as well as determination of p53 status as a prerequisite for administering the drugs are vital in the design of such clinical trials. Moreover, the contribution of p53 mutation to cancer development is expected to be associated with drug resistance, hence understanding the mechanism of drug resistance induction, particularly the inhibition of p53-induced apoptosis from extended treatments with p53-activating drugs, should be considered as well. Despite the fact that NPC is an aggressive disease characterized by overexpressions of p53 [120] and several p53 gene mutations, p53 mutation is considerably rare compared to other human cancers in general [109, 121]. Hence, antagonizing Mdm2 to reactivate p53 provides an alternative strategy to the current standard cancer therapy [76].
The rationale of using Nutlin-3 to antagonize Mdm2 to reactivate p53 in NPC is formed based on the facts that: (1) Nutlin-3 has been explored as a specific p53 activator in wt-expressing cancer cells and is applicable in NPC which is rarely mutated [73]; (2) Nutlin-3 chemo-protectively conserves normal cells [73, 105]; and (3) Nutlin-3 integrates effectively with radiation, genotoxic or anti-mitotic agents in suppressing various tumors [65, 84-86]. Currently, research for the development of p53-reactivating therapy in NPC is still limited compared to other cancer types where optimized clinical doses, treatment durations and safety profiles remain out of reach for now. Moreover, the effectiveness of Nutlin-3 in preclinical animal models has yet to be established. The synergistic effects via combination of low doses of cisplatin and Nutlin-3, drug interaction and dose-limiting toxicity studies have to be further investigated. Signaling pathways involved in the synergistic effects of the combination could also be delineated for the identification of additional targets. In addition, the mechanism of Nutlin-3-resistant induction and emergence of p53 alteration in NPC should also be investigated to study the pathological significances in NPC to gain deeper understanding to the molecular basis of this disease.
CONCLUSION
Although Nutlin-3 is far from completion for its clinical use for NPC, it is still an attractive alternative drug that should be given attention and explored further, given the poor prognosis and limited treatment choices for NPC. It is anticipated that the mortality and morbidity rate of NPC could be reduced with the combination use of Nutlin-3 and standard chemotherapeutic drugs coupled with the determination of p53 mutation status in each individual.
CONSENT FOR PUBLICATION
Not applicable.
ACKNOWLEDGEMENTS
The authors wish to thank the Director General of Health Malaysia for his permission to publish the paper.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
REFERENCES
- 1.American Cancer society . Cancer Facts & Figures 2013. Atlanta: Ga American Cancer Society; 2013. [Google Scholar]
- 2.Roland N.J., Paleri V.E. Head and Neck Cancer: Multidisciplinary Managment Guidelines. 4th ed. London, England: ENT UK; 2011. pp. 147–197. [Google Scholar]
- 3.Chai S.J., Pua K.C., Amyza S., Yap Y.Y., Lim P.V., Kumar S.S., Lum C.L., Gopala K., Rozita W.M., Teo S.H., Khoo A.S., Yap L.F. Clinical significance of plasma Epstein–Barr Virus DNA loads in a large cohort of Malaysian patients with nasopharyngeal carcinoma. J. Clin. Virol. 2012;55(1):34–39. doi: 10.1016/j.jcv.2012.05.017. [DOI] [PubMed] [Google Scholar]
- 4.Schleper J.R. Prevention, detection, and diagnosis of head and neck cancers. Semin. Oncol. Nurs. 1989;5(3):139–149. doi: 10.1016/0749-2081(89)90086-7. [DOI] [PubMed] [Google Scholar]
- 5.Hildesheim A., Dosemeci M., Chan C.C., Chen C.J., Cheng Y.J., Hsu M.M., Chen I.H., Mittl B.F., Sun B., Levine P.H., Chen J.Y., Brinton L.A., Yang C.S. Occupational exposure to wood, formaldehyde, and solvents and risk of nasopharyngeal carcinoma. Cancer Epidemiol. Biomarkers Prev. 2001;10(11):1145–1153. [PubMed] [Google Scholar]
- 6.Heo D.S., Snyderman C., Gollin S.M., Pan S., Walker E., Deka R., Barnes E.L., Johnson J.T., Herberman R.B., Whiteside T.L. Biology, cytogenetics, and sensitivity to immunological effector cells of new head and neck squamous cell carcinoma lines. Cancer Res. 1989;49(18):5167–5175. [PubMed] [Google Scholar]
- 7.Lo K.W., Chung G.T., To K.F. Deciphering the molecular genetic basis of NPC through molecular, cytogenetic, and epigenetic approaches. Semin. Cancer Biol. 2012;22(2):79–86. doi: 10.1016/j.semcancer.2011.12.011. [DOI] [PubMed] [Google Scholar]
- 8.Old L.J., Boyse E.A., Oettgen H.F., Harven E.D., Geering G., Williamson B., Clifford P. Precipitating antibody in human serum to an antigen present in cultured burkitt’s lymphoma cells. Proc. Natl. Acad. Sci. USA. 1966;56(6):1699–1704. doi: 10.1073/pnas.56.6.1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chou J., Lin Y.C., You L., Xu Z., He B., Jablons D.M. Nasopharyngeal carcinoma review of the molecular mechanisms of tumorigenesis. Head Neck. 2008;30(7):946–963. doi: 10.1002/hed.20833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Tiwawech D., Srivatanakul P., Karalak A., Ishida T. Cytochrome P450 2A6 polymorphism in nasopharyngeal carcinoma. Cancer Lett. 2006;241(1):135–141. doi: 10.1016/j.canlet.2005.10.026. [DOI] [PubMed] [Google Scholar]
- 11.Gallicchio L., Matanoski G., Tao X., Chen L., Lam T.K., Boyd K., Robinson K.A., Balick L., Mickelson S., Caulfield L.E., Herman J.G., Guallar E., Alberg A.J. Adulthood consumption of preserved and nonpreserved vegetables and the risk of nasopharyngeal carcinoma: A systematic review. Int. J. Cancer. 2006;119(5):1125–1135. doi: 10.1002/ijc.21946. [DOI] [PubMed] [Google Scholar]
- 12.Cheng Y.J., Hildesheim A., Hsu M.M., Chen I.H., Brinton L.A., Levine P.H., Chen C.J., Yang C.S. Cigarette smoking, alcohol consumption and risk of nasopharyngeal carcinoma in Taiwan. Cancer Causes Control. 1999;10(3):201–207. doi: 10.1023/a:1008893109257. [DOI] [PubMed] [Google Scholar]
- 13.Yu M.C., Yuan J.M. Epidemiology of nasopharyngeal carcinoma. Semin. Cancer Biol. 2002;12(6):421–429. doi: 10.1016/s1044579x02000858. [DOI] [PubMed] [Google Scholar]
- 14.GLOBOCAN 2012: Estimated cancer incidence, mortality and prevalence worldwide in 2012. International Agency for Research on Cancer 2013. http://globocan.iarc.fr/ 2012
- 15.Parkin D.M., Bray F., Ferlay J., Pisani P. Estimating the world cancer burden: Globocan 2000. Int. J. Cancer. 2001;94(2):153–156. doi: 10.1002/ijc.1440. [DOI] [PubMed] [Google Scholar]
- 16.Yu M.C., Ho J.H., Lai S.H., Henderson B.E. Cantonese-style Salted Fish as a Cause of Nasopharyngeal Carcinoma: Report of a Case-Control Study in Hong Kong. Cancer Res. 1986;46:956–961. [PubMed] [Google Scholar]
- 17.Zheng X., Luo Y., Christensson B., Drettner B. Induction of nasal and nasopharyngeal tumours in Sprague-Dawley rats fed with Chinese salted fish. Acta Otolaryngol. 1994;114(1):98–104. doi: 10.3109/00016489409126024. [DOI] [PubMed] [Google Scholar]
- 18.Armstrong R.W., Chan A.S. Salted fish and nasopharyngeal carcinoma in Malaysia. Soc. Sci. Med. 1983;17(20):1559–1567. doi: 10.1016/0277-9536(83)90100-4. [DOI] [PubMed] [Google Scholar]
- 19.Yu M.C., Huang T.B., Henderson B.E. Diet and nasopharyngeal carcinoma: a case-control study in Guangzhou, China. Int. J. Cancer. 1989;43(6):1077–1082. doi: 10.1002/ijc.2910430621. [DOI] [PubMed] [Google Scholar]
- 20.Lee H.P., Gourley L., Duffy S.W., Esteve J., Lee J., Day N.E. Preserved foods and nasopharyngeal carcinoma: a case-control study among Singapore Chinese. Int. J. Cancer. 1994;59(5):585–590. doi: 10.1002/ijc.2910590502. [DOI] [PubMed] [Google Scholar]
- 21.Yuan J.M., Wang X.L., Xiang Y.B., Gao Y.T., Ross R.K., Yu M.C. Preserved foods in relation to risk of nasopharyngeal carcinoma in Shanghai, China. Int. J. Cancer. 2000;85(3):358–363. doi: 10.1002/(sici)1097-0215(20000201)85:3<358::aid-ijc11>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
- 22.Huang P.Y., Cao K.J., Guo X., Mo H.Y., Guo L., Xiang Y.O., Deng M.Q., Qiu F., Cao S.M., Guo Y. L., Z.; Li, N.W.; Sun, R.; Chen, Q.Y.; Luo, D.H.; Hua, Y.J.; Mai, H.Q.; Hong, M.H. A randomized trial of induction chemotherapy plus concurrent chemoradiotherapy versus induction chemotherapy plus radiotherapy for locoregionally advanced nasopharyngeal carcinoma. Oral Oncol. 2012;48(10):1038–1044. doi: 10.1016/j.oraloncology.2012.04.006. [DOI] [PubMed] [Google Scholar]
- 23.Mould R.F., Tai T.H. Nasopharyngeal carcinoma: treatments and outcomes in the 20th century. Br. J. Radiol. 2002;75(892):307–339. doi: 10.1259/bjr.75.892.750307. [DOI] [PubMed] [Google Scholar]
- 24.Tay H.N., Han H.J., Sudirman S.R. Robotic nasopharyngectomy. Operat. Tech. Otolaryngol. Neck Surgery. 2014;25(3):299–303. [Google Scholar]
- 25.Mai H.Q., Mo H.Y., Deng J.F., Deng M.Q., Mai W.Y., Huang X.M., Guo X., Hong M.H. Endoscopic microwave coagulation therapy for early recurrent T1 nasopharyngeal carcinoma. Eur. J. Cancer. 2009;45(7):1107–1110. doi: 10.1016/j.ejca.2009.02.028. [DOI] [PubMed] [Google Scholar]
- 26.Monteiro E., Witterick I. Endoscopic nasopharyngectomy: Patient selection and surgical execution. Operat. Tech. Otolaryngol. Neck Surgery. 2014;25(3):284–288. [Google Scholar]
- 27.Lee A.W., Lin J.C., Ng W.T. Current management of nasopharyngeal cancer. Semin. Radiat. Oncol. 2012;22(3):233–244. doi: 10.1016/j.semradonc.2012.03.008. [DOI] [PubMed] [Google Scholar]
- 28.Zhang L., Chen Q.Y., Liu H., Tang L.Q., Mai H.Q. Emerging treatment options for nasopharyngeal carcinoma. Drug Des. Devel. Ther. 2013;7:37–52. doi: 10.2147/DDDT.S30753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kwong D.L., Wei W.I., Sham J.S., Ho W.K., Yuen P.W., Chua D.T., Au D.K., Wu P.M., Choy D.T. Sensorineural hearing loss in patients treated for nasopharyngeal carcinoma: a prospective study of the effect of radiation and cisplatin treatment. Int. J. Radiat. Oncol. Biol. Phys. 1996;36(2):281–289. doi: 10.1016/s0360-3016(96)00302-1. [DOI] [PubMed] [Google Scholar]
- 30.Lee A.W., Fee W.E., Jr, Ng W.T., Chan L.K. Nasopharyngeal carcinoma: Salvage of local recurrence. Oral Oncol. 2012;48(9):768–774. doi: 10.1016/j.oraloncology.2012.02.017. [DOI] [PubMed] [Google Scholar]
- 31.Tham I.W., Hee S.W., Yeo R.M., Salleh P.B., Lee J., Tan T.W., Fong K.W., Chua E.T., Wee J.T. Treatment of nasopharyngeal carcinoma using intensity-modulated radiotherapy-the national cancer centre singapore experience. Int. J. Radiat. Oncol. Biol. Phys. 2009;75(5):1481–1486. doi: 10.1016/j.ijrobp.2009.01.018. [DOI] [PubMed] [Google Scholar]
- 32.Chan K.S., Koh C.G., Li H.Y. Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis. 2012;3:e411. doi: 10.1038/cddis.2012.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Choi Y.M., Kim H.K., Shim W., Anwar M.A., Kwon J.W., Kwon H.K., Kim H.J., Jeong H., Kim H.M., Hwang D., Kim H.S., Choi S. Mechanism of Cisplatin-induced cytotoxicity is correlated to impaired metabolism due to mitochondrial ROS generation. PLoS One. 2015;10(8):e0135083. doi: 10.1371/journal.pone.0135083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Anniko M., Sobin A. Cisplatin: Evaluation of its ototoxic potential. Am. J. Otolaryngol. 1986;7(4):276–293. doi: 10.1016/s0196-0709(86)80050-3. [DOI] [PubMed] [Google Scholar]
- 35.Chitapanarux I., Lorvidhaya V., Kamnerdsupaphon P., Sumitsawan Y., Tharavichitkul E., Sukthomya V., Ford J. Chemoradiation comparing cisplatin versus carboplatin in locally advanced nasopharyngeal cancer: randomised, non-inferiority, open trial. Eur. J. Cancer. 2007;43(9):1399–1406. doi: 10.1016/j.ejca.2007.03.022. [DOI] [PubMed] [Google Scholar]
- 36.Chan A.T., Teo P.M., Huang D.P. Pathogenesis and treatment of nasopharyngeal carcinoma. Semin. Oncol. 2004;31(6):794–801. doi: 10.1053/j.seminoncol.2004.09.008. [DOI] [PubMed] [Google Scholar]
- 37.Lee N., Xia P., Quivey J.M., Khalil S., Poon I., Akazawa C., Akazawa P., Weinberg V., Fu K.K. Intensity-modulated radiotherapy in the treatment of nasopharyngeal carcinoma: an update of the UCSF experience. Int. J. Radiat. Oncol. Biol. Phys. 2002;53(1):12–22. doi: 10.1016/s0360-3016(02)02724-4. [DOI] [PubMed] [Google Scholar]
- 38.Chi K.H., Chang Y.C., Guo W.Y., Leung M.J., Shiau C.Y., Chen S.Y., Wang L.W., Lai Y.L., Hsu M.M., Lian S.L., Chang C.H., Liu T.W., Chin Y.H., Yen S.H., Perng C.H., Chen K.Y. A phase III study of adjuvant chemotherapy in advanced nasopharyngeal carcinoma patients. Int. J. Radiat. Oncol. Biol. Phys. 2002;52(5):1238–1244. doi: 10.1016/s0360-3016(01)02781-x. [DOI] [PubMed] [Google Scholar]
- 39.Ma J., Mai H., Hong M.H., Min H.Q., Mao Z.D., Cui N.J., Lu T.X., Mo H.Y. Results of a prospective randomized trial comparing neoadjuvant chemotherapy plus radiotherapy with radiotherapy alone in patients with locoregionally advanced nasopharyngeal carcinoma. J. Clin. Oncol. 2001;19(5):1350–1357. doi: 10.1200/JCO.2001.19.5.1350. [DOI] [PubMed] [Google Scholar]
- 40.Phua V.C., Loo W.H., Md Y.M., Zamaniah W.I., Tho L.M., Ung N.M. Treatment outcome for nasopharyngeal carcinoma in University Malaya Medical Centre from 2004-2008. Asian Pac. J. Cancer Prev. 2013;14(8):4567–4570. doi: 10.7314/apjcp.2013.14.8.4567. [DOI] [PubMed] [Google Scholar]
- 41.Razak A.R., Siu L.L., Liu F.F., Ito E., O’Sullivan B., Chan K. Nasopharyngeal carcinoma: the next challenges. Eur. J. Cancer. 2010;46(11):1967–1978. doi: 10.1016/j.ejca.2010.04.004. [DOI] [PubMed] [Google Scholar]
- 42.Tuan J.K., Ha T., Ong W.S., Siow T.R., Tham I.W., Yap S.P., Tan T.W., Chua E.T., Fong K.W., Wee J.T. Late toxicities after conventional radiation therapy alone for nasopharyngeal carcinoma. Radiother. Oncol. 2012;104(3):305–311. doi: 10.1016/j.radonc.2011.12.028. [DOI] [PubMed] [Google Scholar]
- 43.Sultanem K., Shu H.K., Xia P., Akazawa C., Quivey J.M., Verhey L.J., Fu K.K. Three-dimensional intensity-modulated radiotherapy in the treatment of nasopharyngeal carcinoma: the University of California-San Francisco experience. Int. J. Radiat. Oncol. Biol. Phys. 2000;48(3):711–722. doi: 10.1016/s0360-3016(00)00702-1. [DOI] [PubMed] [Google Scholar]
- 44.Siddik Z.H. Cisplatin: mode of cytotoxic action and molecular basis of resistance. Oncogene. 2003;22(47):7265–7279. doi: 10.1038/sj.onc.1206933. [DOI] [PubMed] [Google Scholar]
- 45.Bou-Assaly W., Mukherji S. Cetuximab (erbitux). AJNR Am. J. Neuroradiol. 2010;31(4):626–627. doi: 10.3174/ajnr.A2054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Martinelli E., De Palma R., Orditura M., De Vita F., Ciardiello F. Anti-epidermal growth factor receptor monoclonal antibodies in cancer therapy. Clin. Exp. Immunol. 2009;158(1):1–9. doi: 10.1111/j.1365-2249.2009.03992.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Khoo A.S., Pua K.C. Diagnosis and clinical evaluation of nasopharyngeal carcinoma. Book title: Nasopharyngeal Carcinoma: Keys for Translational Medicine and Biology. Landes Bioscience; 2013. pp. 1–9. [Google Scholar]
- 48.Pua K.C., Khoo A.S., Yap Y.Y., Subramaniam S.K., Ong C.A., Gopala K.G., Shahid H., Malaysian Nasopharyngeal Carcinoma Study Group Nasopharyngeal Carcinoma Database. Med. J. Malaysia. 2008;63(Suppl. C):59–62. [PubMed] [Google Scholar]
- 49.Leung T.W., Tung S.Y., Sze W.K., Wong F.C., Yuen K.K., Lui C.M., Lo S.H., Ng T.Y. O, S.K. Treatment results of 1070 patients with nasopharyngeal carcinoma: an analysis of survival and failure patterns. Head Neck. 2005;27(7):555–565. doi: 10.1002/hed.20189. [DOI] [PubMed] [Google Scholar]
- 50.Harlow E., Williamson N.M., Ralston R., Helfman D.M., Adams T.E. Molecular cloning and in vitro expression of a cDNA clone for human cellular tumor antigen p53. Mol. Cell. Biol. 1985;5(7):1601–1610. doi: 10.1128/mcb.5.7.1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Matlashewski G., Lamb P., Pim D., Peacock J., Crawford L., Benchimol S. Isolation and characterization of a human p53 cDNA clone: expression of the human p53 gene. EMBO J. 1984;3(13):3257–3262. doi: 10.1002/j.1460-2075.1984.tb02287.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lane D.P. Cancer. p53, guardian of the genome. Nature. 1992;358(6381):15–16. doi: 10.1038/358015a0. [DOI] [PubMed] [Google Scholar]
- 53.Levine A.J. p53, the cellular gatekeeper for growth and division. Cell. 1997;88(3):323–331. doi: 10.1016/s0092-8674(00)81871-1. [DOI] [PubMed] [Google Scholar]
- 54.Schon O., Friedler A., Bycroft M., Freund S.M., Fersht A.R. Molecular mechanism of the interaction between MDM2 and p53. J. Mol. Biol. 2002;323(3):491–501. doi: 10.1016/s0022-2836(02)00852-5. [DOI] [PubMed] [Google Scholar]
- 55.Vassilev L.T. MDM2 inhibitors for cancer therapy. Trends Mol. Med. 2007;13(1):23–31. doi: 10.1016/j.molmed.2006.11.002. [DOI] [PubMed] [Google Scholar]
- 56.Appella E., Anderson C.W. Post-translational modifications and activation of p53. Eur. J. Biochem. 2001;268(10):2764–2772. doi: 10.1046/j.1432-1327.2001.02225.x. [DOI] [PubMed] [Google Scholar]
- 57.Harris S.L., Levine A.J. The p53 pathway: positive and negative feedback loops. Oncogene. 2005;24(17):2899–2908. doi: 10.1038/sj.onc.1208615. [DOI] [PubMed] [Google Scholar]
- 58.Saito S., Goodarzi A.A., Higashimoto Y., Noda Y., Lees-Miller S.P., Appella E., Anderson C.W. ATM mediates phosphorylation at multiple p53 sites, including Ser(46), in response to ionizing radiation. J. Biol. Chem. 2002;277(15):12491–12494. doi: 10.1074/jbc.C200093200. [DOI] [PubMed] [Google Scholar]
- 59.Brown C.J., Lain S., Verma C.S., Fersht A.R., Lane D.P. Awakening guardian angels: drugging the p53 pathway. Nat. Rev. Cancer. 2009;9(12):862–873. doi: 10.1038/nrc2763. [DOI] [PubMed] [Google Scholar]
- 60.Finlay C.A., Hinds P.W., Levine A.J. The p53 proto-oncogene can act as a suppressor of transformation. Cell. 1989;57(7):1083–1093. doi: 10.1016/0092-8674(89)90045-7. [DOI] [PubMed] [Google Scholar]
- 61.Mills A.A. p63: oncogene or tumor suppressor? Curr. Opin. Genet. Dev. 2006;16(1):38–44. doi: 10.1016/j.gde.2005.12.001. [DOI] [PubMed] [Google Scholar]
- 62.Kojima K., Konopleva M., Samudio I.J., Shikami M., Cabreira-Hansen M., McQueen T., Ruvolo V., Tsao T., Zeng Z., Vassilev L.T., Andreeff M. MDM2 antagonists induce p53-dependent apoptosis in AML: implications for leukemia therapy. Blood. 2005;106(9):3150–3159. doi: 10.1182/blood-2005-02-0553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Stuhmer T., Chatterjee M., Hildebrandt M., Herrmann P., Gollasch H., Gerecke C., Theurich S., Cigliano L., Manz R.A., Daniel P.T., Bommert K., Vassilev L.T., Bargou R.C. Nongenotoxic activation of the p53 pathway as a therapeutic strategy for multiple myeloma. Blood. 2005;106(10):3609–3617. doi: 10.1182/blood-2005-04-1489. [DOI] [PubMed] [Google Scholar]
- 64.Tovar C., Rosinski J., Filipovic Z., Higgins B., Kolinsky K., Hilton H., Zhao X., Vu B.T., Qing W., Packman K., Myklebost O., Heimbrook D.C., Vassilev L.T. Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy. Proc. Natl. Acad. Sci. USA. 2006;103(6):1888–1893. doi: 10.1073/pnas.0507493103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Muller C.R., Paulsen E.B., Noordhuis P., Pedeutour F., Saeter G., Myklebost O. Potential for treatment of liposarcomas with the MDM2 antagonist Nutlin-3A. Int. J. Cancer. 2007;121(1):199–205. doi: 10.1002/ijc.22643. [DOI] [PubMed] [Google Scholar]
- 66.Saddler C., Ouillette P., Kujawski L., Shangary S., Talpaz M., Kaminski M., Erba H., Shedden K., Wang S., Malek S.N. Comprehensive biomarker and genomic analysis identifies p53 status as the major determinant of response to MDM2 inhibitors in chronic lymphocytic leukemia. Blood. 2008;111(3):1584–1593. doi: 10.1182/blood-2007-09-112698. [DOI] [PubMed] [Google Scholar]
- 67.Miyachi M., Kakazu N., Yagyu S., Katsumi Y., Tsubai-Shimizu S., Kikuchi K., Tsuchiya K., Iehara T., Hosoi H. Restoration of p53 pathway by nutlin-3 induces cell cycle arrest and apoptosis in human rhabdomyosarcoma cells. Clin. Cancer Res. 2009;15(12):4077–4084. doi: 10.1158/1078-0432.CCR-08-2955. [DOI] [PubMed] [Google Scholar]
- 68.Van Maerken T., Ferdinande L., Taildeman J., Lambertz I., Yigit N., Vercruysse L., Ali R., Michaelis M., Cinatl J.J., Cuvelier C.A., Marine J.C., De Paepe A., Bracke M., Speleman F., Vandesompele J. Antitumor activity of the selective MDM2 antagonist nutlin-3 against chemoresistant neuroblastoma with wild-type p53. J. Natl. Cancer Inst. 2009;101(22):1562–1574. doi: 10.1093/jnci/djp355. [DOI] [PubMed] [Google Scholar]
- 69.Hori T., Kondo T., Masahiko K., Tabuchi Y., Ogawa R., Zhao Q.L., Ahmed K., Yasuda T., Seki S., Suzuki K., Kimura T. Nutlin-3 enhances tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through up-regulation of death receptor 5 (DR5) in human sarcoma HOS cells and human colon cancer HCT116 cells. Cancer Lett. 2010;287(1):98–108. doi: 10.1016/j.canlet.2009.06.002. [DOI] [PubMed] [Google Scholar]
- 70.Koster R., Timmer-Bosscha H., Bischoff R., Gietema J.A., de Jong S. Disruption of the MDM2-p53 interaction strongly potentiates p53-dependent apoptosis in cisplatin-resistant human testicular carcinoma cells via the Fas/FasL pathway. Cell Death Dis. 2011;2:e148. doi: 10.1038/cddis.2011.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sonnemann J., Palani C.D., Wittig S., Becker S., Eichhorn F., Voigt A., Beck J.F. Anticancer effects of the p53 activator nutlin-3 in Ewing’s sarcoma cells. Eur. J. Cancer. 2011;47(9):1432–1441. doi: 10.1016/j.ejca.2011.01.015. [DOI] [PubMed] [Google Scholar]
- 72.Ye F., Abdul L.A., Xie J., Weinberg A., Gao S. Nutlin-3 induces apoptosis, disrupts viral latency and inhibits expression of angiopoietin-2 in Kaposi sarcoma tumor cells. Cell Cycle. 2012;11(7):1393–1399. doi: 10.4161/cc.19756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Voon Y.L., Ahmad M., Wong P.F., Husaini R., Ng W.T., Leong C.O., Lane D.P., Khoo A.S. Nutlin-3 sensitizes nasopharyngeal carcinoma cells to cisplatin-induced cytotoxicity. Oncol. Rep. 2015;34(4):1692–1700. doi: 10.3892/or.2015.4177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Saha M.N., Qiu L., Chang H. Targeting p53 by small molecules in hematological malignancies. J. Hematol. Oncol. 2013;6(23):204–209. doi: 10.1186/1756-8722-6-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Koblish H.K., Zhao S., Franks C.F., Donatelli R.R., Tominovich R.M., LaFrance L.V., Leonard K.A., Gushue J.M., Parks D.J., Calvo R.R., Milkiewicz K.L., Marugan J.J., Raboisson P., Cummings M.D., Grasberger B.L., Johnson D.L., Lu T., Molloy C.J., Maroney A.C. Benzodiazepinedione inhibitors of the Hdm2:p53 complex suppress human tumor cell proliferation in vitro and sensitize tumors to doxorubicin in vivo. Mol. Cancer Ther. 2006;5(1):160–169. doi: 10.1158/1535-7163.MCT-05-0199. [DOI] [PubMed] [Google Scholar]
- 76.Shangary S., Wang S. Small-molecule inhibitors of the MDM2-p53 protein-protein interaction to reactivate p53 function: a novel approach for cancer therapy. Annu. Rev. Pharmacol. Toxicol. 2009;49:223–241. doi: 10.1146/annurev.pharmtox.48.113006.094723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Long J., Parkin B., Ouillette P., Bixby D., Shedden K., Erba H., Wang S., Malek S.N. Multiple distinct molecular mechanisms influence sensitivity and resistance to MDM2 inhibitors in adult acute myelogenous leukemia. Blood. 2010;116(1):71–80. doi: 10.1182/blood-2010-01-261628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Peterson L.F., Mitrikeska E., Giannola D., Lui Y., Sun H., Bixby D., Malek S.N., Donato N.J., Wang S., Talpaz M. p53 stabilization induces apoptosis in chronic myeloid leukemia blast crisis cells. Leukemia. 2011;25(5):761–769. doi: 10.1038/leu.2011.7. [DOI] [PubMed] [Google Scholar]
- 79.Sosin A.M., Burger A.M., Siddiqi A., Abrams J., Mohammad R.M., Al-Katib A.M. HDM2 antagonist MI-219 (spiro-oxindole), but not Nutlin-3 (cis-imidazoline), regulates p53 through enhanced HDM2 autoubiquitination and degradation in human malignant B-cell lymphomas. J. Hematol. Oncol. 2012;5(57):1–18. doi: 10.1186/1756-8722-5-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chargari C., Leteur C., Angevin E., Bashir T., Schoentjes B., Arts J., Janicot M., Bourhis J., Deutsch E. Preclinical assessment of JNJ-26854165 (Serdemetan), a novel tryptamine compound with radiosensitizing activity in vitro and in tumor xenografts. Cancer Lett. 2011;312(2):209–218. doi: 10.1016/j.canlet.2011.08.011. [DOI] [PubMed] [Google Scholar]
- 81.Kojima K., Burks J.K., Arts J., Andreeff M. The novel tryptamine derivative JNJ-26854165 induces wild-type p53- and E2F1-mediated apoptosis in acute myeloid and lymphoid leukemias. Mol. Cancer Ther. 2010;9(9):2545–2557. doi: 10.1158/1535-7163.MCT-10-0337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Reed D., Shen Y., Shelat A.A., Arnold L.A., Ferreira A.M., Zhu F., Mills N., Smithson D.C., Regni C.A., Bashford D., Cicero S.A., Schulman B.A., Jochemsen A.G., Guy R.K., Dyer M.A. Identification and characterization of the first small molecule inhibitor of MDMX. J. Biol. Chem. 2010;285(14):10786–10796. doi: 10.1074/jbc.M109.056747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Graves B., Thompson T., Xia M., Janson C., Lukacs C., Deo D., Di Lello P., Fry D., Garvie C., Huang K.S., Gao L., Tovar C., Lovey A., Wanner J., Vassilev L.T. Activation of the p53 pathway by small-molecule-induced MDM2 and MDMX dimerization. Proc. Natl. Acad. Sci. USA. 2012;109(29):11788–11793. doi: 10.1073/pnas.1203789109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Frei E., III, Luce J.K., Loo T.L. Phase I and phototoxicity studies of pseudourea (NSC-56054). Cancer Chemother. Rep. 1971;55(1):91–97. [PubMed] [Google Scholar]
- 85.Wang H., Ma X., Ren S., Buolamwini J.K., Yan C. A small-molecule inhibitor of MDMX activates p53 and induces apoptosis. Mol. Cancer Ther. 2011;10(1):69–79. doi: 10.1158/1535-7163.MCT-10-0581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wang H., Yan C. A small-molecule p53 activator induces apoptosis through inhibiting MDMX expression in breast cancer cells. Neoplasia. 2011;13(7):611–619. doi: 10.1593/neo.11438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Issaeva N., Bozko P., Enge M., Protopopova M., Verhoef L.G., Masucci M., Pramanik A., Selivanova G. Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors. Nat. Med. 2004;10(12):1321–1328. doi: 10.1038/nm1146. [DOI] [PubMed] [Google Scholar]
- 88.Zhao C.Y., Grinkevich V.V., Nikulenkov F., Bao W., Selivanova G. Rescue of the apoptotic-inducing function of mutant p53 by small molecule RITA. Cell Cycle. 2010;9(9):1847–1855. doi: 10.4161/cc.9.9.11545. [DOI] [PubMed] [Google Scholar]
- 89.Kitagaki J., Agama K.K., Pommier Y., Yang Y., Weissman A.M. Targeting tumor cells expressing p53 with a water-soluble inhibitor of Hdm2. Mol. Cancer Ther. 2008;7(8):2445–2454. doi: 10.1158/1535-7163.MCT-08-0063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yang Y., Ludwig R.L., Jensen J.P., Pierre S.A., Medaglia M.V., Davydov I.V., Safiran Y.J., Oberoi P., Kenten J.H., Phillips A.C., Weissman A.M., Vousden K.H. Small molecule inhibitors of HDM2 ubiquitin ligase activity stabilize and activate p53 in cells. Cancer Cell. 2005;7(6):547–559. doi: 10.1016/j.ccr.2005.04.029. [DOI] [PubMed] [Google Scholar]
- 91.Brooks C.L., Gu W. p53 Activation: a case against Sir. Cancer Cell. 2008;13(5):377–378. doi: 10.1016/j.ccr.2008.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lain S., Hollick J.J., Campbell J., Staples O.D., Higgins M., Aoubala M., McCarthy A., Appleyard V., Murray K.E., Baker L., Thompson A., Mathers J., Holland S.J., Stark M.J., Pass G., Woods J., Lane D.P., Westwood N.J. Discovery, in vivo activity, and mechanism of action of a small-molecule p53 activator. Cancer Cell. 2008;13(5):454–463. doi: 10.1016/j.ccr.2008.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Foster B.A., Coffey H.A., Morin M.J., Rastinejad F. Pharmacological rescue of mutant p53 conformation and function. Science. 1999;286(5449):2507–2510. doi: 10.1126/science.286.5449.2507. [DOI] [PubMed] [Google Scholar]
- 94.Puca R., Nardinocchi L., Porru M., Simon A.J., Rechavi G., Leonetti C., Givol D., D’Orazi G. Restoring p53 active conformation by zinc increases the response of mutant p53 tumor cells to anticancer drugs. Cell Cycle. 2011;10(10):1679–1689. doi: 10.4161/cc.10.10.15642. [DOI] [PubMed] [Google Scholar]
- 95.Yu X., Vazquez A., Levine A.J., Carpizo D.R. Allele-specific p53 mutant reactivation. Cancer Cell. 2012;21(5):614–625. doi: 10.1016/j.ccr.2012.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Lehmann S., Bykov V.J., Ali D., Andren O., Cherif H., Tidefelt U., Uggla B., Yachnin J., Juliusson G., Moshfegh A., Paul C., Wiman K.G., Andersson P.O. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J. Clin. Oncol. 2012;30(29):3633–3639. doi: 10.1200/JCO.2011.40.7783. [DOI] [PubMed] [Google Scholar]
- 97.Bykov V.J., Issaeva N., Zache N., Shilov A., Hultcrantz M., Bergman J., Selivanova G., Wiman K.G. Reactivation of mutant p53 and induction of apoptosis in human tumor cells by maleimide analogs. J. Biol. Chem. 2005;280(34):30384–30391. doi: 10.1074/jbc.M501664200. [DOI] [PubMed] [Google Scholar]
- 98.Selivanova G., Wiman K.G. Reactivation of mutant p53: molecular mechanisms and therapeutic potential. Oncogene. 2007;26(15):2243–2254. doi: 10.1038/sj.onc.1210295. [DOI] [PubMed] [Google Scholar]
- 99.Boeckler F.M., Joerger A.C., Jaggi G., Rutherford T.J., Veprintsev D.B., Fersht A.R. Targeted rescue of a destabilized mutant of p53 by an in silico screened drug. Proc. Natl. Acad. Sci. USA. 2008;105(30):10360–10365. doi: 10.1073/pnas.0805326105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Liu X., Wilcken R., Joerger A.C., Chuckowree I.S., Amin J., Spencer J., Fersht A.R. Small molecule induced reactivation of mutant p53 in cancer cells. Nucleic Acids Res. 2013;41(12):6034–6044. doi: 10.1093/nar/gkt305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Rauf S.M., Endou A., Takaba H., Miyamoto A. Effect of Y220C mutation on p53 and its rescue mechanism: a computer chemistry approach. Protein J. 2013;32(1):68–74. doi: 10.1007/s10930-012-9458-x. [DOI] [PubMed] [Google Scholar]
- 102.Vassilev L.T., Vu B.T., Graves B., Carvajal D., Podlaski F., Filipovic Z., Norman K., Kammlott U., Lukacs C., Klein C., Fotouhi N., Liu E.A. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004;303(5659):844–848. doi: 10.1126/science.1092472. [DOI] [PubMed] [Google Scholar]
- 103.Apontes P., Leontieva O.V., Demidenko Z.N., Li F., Blagosklonny M.V. Exploring long-term protection of normal human fibroblasts and epithelial cells from chemotherapy in cell culture. Oncotarget. 2011;2(3):222–233. doi: 10.18632/oncotarget.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.van Leeuwen I., Lain S. Pharmacological manipulation of the cell cycle and metabolism to protect normal tissues against conventional anticancer drugs. Oncotarget. 2011;2(4):274–276. doi: 10.18632/oncotarget.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Jiang M., Pabla N., Murphy R.F., Yang T., Yin X.M., Degenhardt K., White E., Dong Z. Nutlin-3 protects kidney cells during cisplatin therapy by suppressing Bax/Bak activation. J. Biol. Chem. 2007;282(4):2636–2645. doi: 10.1074/jbc.M606928200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bunz F., Hwang P.M., Torrance C., Waldman T., Zhang Y., Dillehay L., Williams J., Lengauer C., Kinzler K.W., Vogelstein B. Disruption of p53 in human cancer cells alters the responses to therapeutic agents. J. Clin. Invest. 1999;104(3):263–269. doi: 10.1172/JCI6863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.He M., Rennie P.S., Dragowska V., Nelson C.C., Jia W. A mutant P53 can activate apoptosis through a mechanism distinct from those induced by wild type P53. FEBS Lett. 2002;517(1-3):151–154. doi: 10.1016/s0014-5793(02)02609-1. [DOI] [PubMed] [Google Scholar]
- 108.Shiao Y.H., Rugge M., Correa P., Lehmann H.P., Scheer W.D. p53 alteration in gastric precancerous lesions. Am. J. Pathol. 1994;144(3):511–517. [PMC free article] [PubMed] [Google Scholar]
- 109.Lin D.C., Meng X., Hazawa M., Nagata Y., Varela A.M., Xu L., Sato Y., Liu L.Z., Ding L.W., Sharma A., Goh B.C., Lee S.C., Petersson B.F., Yu F.G., Macary P., Oo M.Z., Ha C.S., Yang H., Ogawa S., Loh K.S., Koeffler H.P. The genomic landscape of nasopharyngeal carcinoma. Nat. Genet. 2014;46(8):866–871. doi: 10.1038/ng.3006. [DOI] [PubMed] [Google Scholar]
- 110.Soussi T. The TP53 http://p53.free.fr/p53_info/
- 111.Effert P., McCoy R., Abdel-Hamid M., Flynn K., Zhang Q., Busson P., Tursz T., Liu E., Raab-Traub N. Alterations of the p53 gene in nasopharyngeal carcinoma. J. Virol. 1992;66(6):3768–3775. doi: 10.1128/jvi.66.6.3768-3775.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Huang D.P., Ho J.H., Poon Y.F., Chew E.C., Saw D., Lui M., Li C.L., Mak L.S., Lai S.H., Lau W.H. Establishment of a cell line (NPC/HK1) from a differentiated squamous carcinoma of the nasopharynx. Int. J. Cancer. 1980;26(2):127–132. doi: 10.1002/ijc.2910260202. [DOI] [PubMed] [Google Scholar]
- 113.Sun Y., Hegamyer G., Cheng Y.J., Hildesheim A., Chen J.Y., Chen I.H., Cao Y., Yao K.T., Colburn N.H. An infrequent point mutation of the p53 gene in human nasopharyngeal carcinoma. Proc. Natl. Acad. Sci. USA. 1992;89(14):6516–6520. doi: 10.1073/pnas.89.14.6516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Chang K.P., Hao S.P., Lin S.Y., Tsao K.C., Kuo T.T., Tsai M.H., Tseng C.K., Tsang N.M. A lack of association between p53 mutations and recurrent nasopharyngeal carcinomas refractory to radiotherapy. Laryngoscope. 2002;112(11):2015–2019. doi: 10.1097/00005537-200211000-00019. [DOI] [PubMed] [Google Scholar]
- 115.Chen R., Zhu D., Yin W. A hot-spot mutation of p53 gene in nasopharyngeal carcinoma. Zhonghua Zhong Liu Za Zhi. 1995;17(6):401–404. [PubMed] [Google Scholar]
- 116.Hoe S.L., Lee E.S., Khoo A.S., Peh S.C. p53 and nasopharyngeal carcinoma: A Malaysian study. Pathology. 2009;41(6):561–565. doi: 10.1080/00313020903071504. [DOI] [PubMed] [Google Scholar]
- 117.Pan J.J., Zhang S.W., Chen C.B., Xiao S.W., Sun Y., Liu C.Q., Su X., Li D.M., Xu G., Xu B., Lu Y.Y. Effect of recombinant adenovirus-p53 combined with radiotherapy on long-term prognosis of advanced nasopharyngeal carcinoma. J. Clin. Oncol. 2009;27(5):799–804. doi: 10.1200/JCO.2008.18.9670. [DOI] [PubMed] [Google Scholar]
- 118.Weinrib L., Li J.H., Donovan J., Huang D., Liu F.F. Cisplatin chemotherapy plus adenoviral p53 gene therapy in EBV-positive and -negative nasopharyngeal carcinoma. Cancer Gene Ther. 2001;8(5):352–360. doi: 10.1038/sj.cgt.7700319. [DOI] [PubMed] [Google Scholar]
- 119.Khoo K.H., Verma C.S., Lane D.P. Drugging the p53 pathway: understanding the route to clinical efficacy. Nat. Rev. Drug Discov. 2014;13(3):217–236. doi: 10.1038/nrd4236. [DOI] [PubMed] [Google Scholar]
- 120.Sheu L.F., Chen A., Lee H.S., Hsu H.Y., Yu D.S. Cooperative interactions among p53, bcl-2 and Epstein-Barr virus latent membrane protein 1 in nasopharyngeal carcinoma cells. Pathol. Int. 2004;54(7):475–485. doi: 10.1111/j.1440-1827.2004.01654.x. [DOI] [PubMed] [Google Scholar]
- 121.Momand J., Jung D., Wilczynski S., Niland J. The MDM2 gene amplification database. Nucleic Acids Res. 1998;26(15):3453–3459. doi: 10.1093/nar/26.15.3453. [DOI] [PMC free article] [PubMed] [Google Scholar]