Biomarkers for enhancing the radiosensitivity of nasopharyngeal carcinoma ========================================================================= * Wei Chen * Guo-Hua Hu ## Abstract Nasopharyngeal carcinoma (NPC) is a common head and neck malignancy. The incidence of NPC is higher in Southern China and Southeast Asia compared with Western countries. Given its high radiosensitivity, the standard treatment for NPC is radiotherapy. However, radioresistance remains a serious obstacle to successful treatment. Radioresistance can cause local recurrence and distant metastases in some patients after treatment by radiation. Thus, special emphasis has been given to the discovery of effective radiosensitizers. This review aims to discuss the biomarkers, classified according to the main mechanisms of radiosensitization, which can enhance the sensitivity of NPC cells to ionizing radiation. * Nasopharyngeal carcinoma (NPC) * radiotherapy * radiosensitization * biomarkers ## Introduction Nasopharyngeal carcinoma (NPC) is a highly metastatic cancer that originates from the epithelial lining of the nasopharynx. NPC occurs mostly in the pharyngeal recess (fossa of Rosenmüller) and the eustachian tube opening in the nasopharynx1. Compared with other head and neck malignancies, NPC is unique in the aspects of epidemiology, pathology, and clinical manifestation. The etiology involves various factors, including genetic susceptibility, Epstein-Barr virus (EBV) infection, and exposure to chemical carcinogens1–3. According to the histological classification of World Health Organization, NPC is divided into three types: keratinizing squamous cell carcinoma (type I), non-keratinizing squamous cell carcinoma (type II), and undifferentiated carcinomas (type III)1. In comparison with the differentiated NPC, undifferentiated NPC is more aggressive, with a higher incidence of distant metastasis4. NPC exhibits a remarkable geographic distribution that the incidence in southern parts of China and Southeast Asia, usually belonging to types II and III, is higher than that in Western countries. In the West, NPC occurs sporadically and usually belongs to type I1. Distinguished by its unique clinical and pathologic characteristics, NPC is highly radiosensitive. Compared with type I, types II and III have a higher response rate to ionizing radiation4. Thus, radiotherapy serves as a standard treatment that is effective to control the early tumor with good prognosis, achieving a 5-year overall survival of 90% and 84% for early stage I and IIA disease, respectively5. However, along with radiotherapy, radioresistance also exists in many cases. Moreover, radioresistance can cause locoregional recurrence and distant metastasis after radiotherapy in some patients, especially when the tumor is in the advanced stage (stage III or IV)6. Therefore, for the relatively advanced stage, combined radio-chemotherapy may increase survival rate, which is 50%-70% as reported6. However, radioresistance still exists in these cases. Along with the damage of radiation to normal tissues, radioresistance remains a serious obstacle to successful treatment. Hence, methods of decreasing NPC radioresistance and enhancing radiosensitivity are urgently needed. In this context, special emphasis is given to the exploitation of effective radiosensitizers according to the three main categories of radiosensitization mechanisms7. ## Biomarkers for enhancing radiosensitivity ### Repair inhibition Cellular radiation repair is a type of ability that is required and vital for cells. Equipped with this ability, cells can correct radiation damage in various ways. First, cellular systems may prevent damage before it occurs. Second, cells may contain molecules that can restore damaged sites chemically. Third, different types of molecules and enzymatic systems can identify and repair damaged substrates through a set of complex and ordered reactions7. As follows, some biomarkers, participating in the repair of radiation damage, are related to radiosensitivity of NPC. ### NFBD1/MDC1 Nuclear factor with BRCA1 C-terminal (BRCT) domain 1/mediator of the DNA damage checkpoint protein 1 (NFBD1/MDC1) is one of the BRCT superfamily members that contains a forkhead-associated domain at the amino-terminus, two BRCT domains at carboxy-terminus, and central Pro-Ser-Thr repeat8. Previous studies showed that in response to DNA damage, the ataxia telangiectasia mutated (ATM) kinase catalyzes the phosphorylation of histone variant H2AX to recognize and marks the sites of damaged DNA9. NFBD1 then recruits repair factors including MRE11, RAD50, and NBS1 complex to the marked sites to form a nuclear foci, thus contributing to an increase in the fidelity of genomic integrity8. Furthermore, Lou *et al*.10 found that the depletion of endogenous NFBD1 can cause a complete loss of nuclear foci formation and induce genomic instability and DNA repair defects. This finding demonstrates that NFBD1 has a close relationship with the initiation and promotion of DNA repair process. *In vivo*, the generation of *MDC1* knockout mice had the characteristics of growth retardation, male infertility, defective class switching, and increased radiation sensitivity10. The expression level of *NFBD1* in NPC cells was absolutely high, and the suppression of *NFBD1* could increase the rate of apoptosis of NPC cells and the sensitivity to chemotherapy11,12. Recently, Wang *et al*.13 demonstrates that Lentivirus-mediated shRNA targeting *NFBD1* can steadily silence the expression of *NFBD1* gene and enhance the radiosensitivity of CNE-1 cells. These results may eventually contribute to the clinical investigation of prediction markers and prognostic factors, as well to the development of radiosensitizing molecules for therapeutic use. ### Cell cycle redistribution Radiosensitivity of the cells varies according to their positions in the cell cycle. Cells in G2/M are approximately 3-fold more sensitive to radiation than cells in late S-phase/early G1, but the exact cause of variation is currently not completely known14. Thus, agents that can block the progression of a cell cycle in a radiosensitive phase may induce significant radiosensitization. #### COX-2 Cyclooxygenase or prostaglandin endoperoxidase synthase (COX) is an enzyme that participates in the formation of prostaglandins (PGs) and is also recognized as an important chemical mediator for inflammation. COX has two isoforms: COX-1 and COX-2. COX-2, not usually expressed in most normal tissues, has a close relationship with the synthesis of PGs in inflamed and neoplastic tissues15. COX-2 overexpression has been reported in most cancers, such as esophageal, gastric, pancreatic, colorectal, lung, and breast cancers16–18, suggesting that COX-2 overexpression has a close relationship with cancer progression15. Some preclinical studies have investigated the role of COX-2 in carcinogenesis19. In these studies, upregulation of COX-2 can regulate angiogenesis, resist apoptosis, and influence cell proliferation successfully through induction of vascular endothelial growth factor (VEGF) and translocation of HIF-1α protein, increase of the expression of proapoptotic Bcl-2 proteins, inhibition of cytochrome c released from mitochondria, and control of G1 and S phase cyclins20. Thus, the selective COX-2 inhibitors can be regarded as anticancer agents. Recently, a preclinical study showed that high *COX-2* expression was found in more than 70% of NPC, and these tumors had earlier regional lymph node metastasis, distant metastasis, and poor responses to chemotherapy, as well as a lower 5-year survival rate compared with low *COX-2* expression or negative tumors21. Meanwhile, an *in vitro* study demonstrated that adding the selective COX-2 inhibitor NS-398 in NPC cells could increase the radiation-induced cell death22. Furthermore, Sun *et al*.23 showed that *COX-2* overexpressing proteins in NPC cells have radioprotective effects, and silence of the expression of *COX-2* can reduce radiation-induced damage repair, thereby enhancing cellular radiosensitivity; this finding lays a theoretical foundation on new gene-radiation combined therapy for NPC. The underlying mechanism may be attributable to the G2-M cell phase arrest and enhancement of cell apoptosis24. #### *GP96* or *GDF-15* The 96 kDa glycoprotein (GP96, also known as GPR94) is an endoplasmic reticulum resident protein that belongs to the heat shock protein 90 family. This glycoprotein is involved in the package and transport of membrane-bound oligomeric proteins, such as immunoglobulins, epidermal growth factor receptor (EGFR), and integrins. It can also induce a variety of immune responses, including antitumor immune responses and the productions of antigen-processing peptides, to collect MHC class I molecules25. Growth differentiation factor 15 (GDF-15) is a member of the transforming growth factor β superfamily. In response to DNA damage, GDF-15 is also a significant downstream mediator. Moreover, high expression of GDF-15 can induce apoptosis and G1 cell cycle arrest26. Chang *et al*.27 used siRNA to suppress the expression of *GP96* and *GDF-15* in NPC-radioresistant cells, which demonstrated that silence of these genes could cause cell growth delay, G2-M cell cycle arrest, and a reduction of clonogenic survival. They also verified that knockdown of the genes could increase the radiosensitivity of NPC cells27. Although further clinical studies are needed, these results may eventually contribute to the development of novel radiosensitizing therapeutics. #### GnT-V Protein glycosylation, a type of post-translational modification, is vital for the glycoprotein, which can affect cell growth, differentiation, and tumor metastasis28. The glycosyltransferase, located in the Golgi apparatus, plays an important role in protein glycosylation. The glycosyltransferase contains at least six N-acetylglucosaminyltransferases (GnTs), defined as GnT-I-VI. N-glycosyltransferase-V (GnT-V) is recognized as a primary member of the glycosyltransferase family, catalyzing the formation of β1, 6 GlcNAc branching structures. Many studies have shown that upregulation of the β1, 6 GlcNAc branched N-glycans structure is related to malignant transformation through various ways, such as inhibiting cell apoptosis and enhancing cell proliferation29. Previous studies have shown that GnT-V plays an important role in malignant tumors. Recently, Wei *et al*.30 demonstrated that downregulation of GnT-V inhibited NPC cell line CNE-2 clonogenic survival after radiation *in vitro*. Another study investigated the role of GnT-V on the radiosensitivity of NPC cells and its underlying mechanisms. These results showed that downregulation of GnT-V in NPC enhanced radiosensitivity *in vitro* and *in vivo*. The underlying mechanisms may be linked to the cell cycle G2-M arrest and the reduction of Bcl-2/Bax ratio. GnT-V may be a potential target for predicting NPC response to radiotherapy31. #### Jab1/CSN5 C-Jun activation domain-binding protein-1 (Jab1) is a modulator of intracellular signaling. Through its existence as the fifth component of the constitutive photomorphogenic-9 signalosome (CSN5), Jab1/CSN5 can regulate cellular proliferation and apoptosis by functionally deactivating some key negative regulatory proteins and tumor suppressors, such as p53 and p2732. In previous studies, overexpression of *Jab1* was found to be correlated with poor prognosis in some tumor types, such as pancreatic adenocarcinomas33, breast cancers34, and NPC35. Other previous studies had found that Jab1 played an important role in the pathologenesis and radioresistance in NPC36. Curcumin is a common chemopreventive agent. Although it plays an important role in anticancer activities in tumor cells37, many previous studies showed that curcumin was limited in its application in anticancer therapy because of its poor bioavailability, rapid metabolism, and instability under several physiologic conditions38. Thus, analogues of curcumin, which have increased anticancer activity and similar safety profiles as curcumin, have been developed in recent years39,40. Pan *et al.*41 used T83 (a new 4-arylidene curcumin analogue) to inhibit the expression of *Jab1* in NPC cells, demonstrating that inhibition of Jab1 could reduce tumor cell growth, induce G2/M arrest, and increase tumor cell apoptosis, thus enhancing the sensitivities of NPC cells to radiotherapy. This study verified T83 as a potential targeted therapy, which may increase the radiosensitivities of NPC cells, providing a promising and effective treatment for NPC patients. ### Increased initial damage Through production of free radical species and direct ionization of target molecules, ionizing radiation can cause cellular damage. DNA is the standard target of ionizing radiation damage, and radiation induced double-strand DNA breaks are fatal. Although we do not completely know the exact type of radiation-induced damage, we can use the agents that could cause more initial damage to cellular targets to achieve an increasing cytotoxic effect of radiation. #### DNA topoisomerase I DNA topoisomerases, first discovered in 1971, are important enzymes that can regulate the transformation of DNA topological isomer. DNA topoisomerases have two types: DNA topoisomerase I and DNA topoisomerase II42. Topoisomerase I (Top I), which is ATP-independent, catalyzes the relaxation of superhelical DNA by cutting off a single chain, allowing the intact chain through the broken chain (Top IA) or leaving the broken chain free to rotate around the intact chain (Top IB). Topoisomerase II (Top II), which is ATP-dependent, mediates the generation of breaks in both chains of the DNA duplex43. Both topoisomerase I and II play important roles in many aspects of DNA metabolism, such as DNA replication and DNA transcription, especially the topoisomerase I, which is very essential for the maintenance of the genomic stability42. Furthermore, drugs that interfere with topoisomerase I-mediated cleavage rejoining of DNA chains can be widely used in the treatment of cancer44. These drugs have also been regarded as effective radiosensitizers when used before radiotherapy, including derivatives of camptothecin45. Topotecan (TPT) is a derivative of camptothecin, which can inhibit topoisomerase I in S phase cells. TPT has been suggested to be effective in the radiotherapy of NPC. Zhang *et al.*46 established a xenografted human NPC model in nude mice and investigated the synergistic effect of TPT and chronoradiotherapy. The results showed that TPT has a radiosensitizing effect on xenografted human NPC. More importantly, TPT has a circadian dependence, thus contributing to further optimization of therapeutic schedules for NPC. Moreover, the potential mechanism of chronomodulated radiosensitization of TPT may be related to the circadian rhythm of tumor hypoxia and the radiation reluctance of cells in S phase, causing G2/M phase arrest. #### Nitric oxide synthases NO is a gaseous molecule that has two types: enzyme-dependent NO and enzyme-independent NO. In human cells, NO is enzyme-dependent and comes from L-arginine catalyzed by NO synthases (NOS). NO plays a very important role in cell signaling pathway through cell-mediated immune responses, cytotoxicity, formation of guanylate cyclase, and cyclic guanosine monophosphate47. Previous studies showed that NO is a hypoxic cell radiosensitizer when combined with radiotherapy48. Recent evidence showed that NO is an initiator of apoptotic signals due to its ability to form peroxynitrite, which can activate oxidative stress-induced apoptosis49. Stimulated by cytokines, inducible NOS (iNOS) can produce much NO. Thus, increased iNOS is associated with increased apoptosis. To explore the function of iNOS in NPC, Jayasurya *et al*.50 demonstrated that low *iNOS* expression comes with high incidence of tumor local recurrence and metastasis, probably because of decreased apoptosis. In addition, NPC cells with high *iNOS* expressions may be more sensitive to radiotherapy. ### Others The mechanisms of the following biomarkers do not belong to the three main categories or are not completely known right now. #### *EGFR* and *IGF-1R* EGFR and insulin-like growth factor-1 receptor (IGF-1R) are key members of the tyrosine kinase receptor family, whose ligands are respectively epidermal growth factor and insulin-like growth factor-1 (IGF-1). The combinations of both receptor and ligand are involved in the development of malignant tumor through modulation of tumor cell cycles, apoptosis, and interactions with the surrounding environment51. Moreover, IGF-1R plays a crucial role in the generation of drug resistance during anticancer therapy and also in the proliferation and transformation of tumor cells. Meanwhile, EGFR significantly contributes to promotion of cell proliferation52. Early experiments showed that combining short hairpin RNA segments to *IGF-1* and *EGFRs* via plasmid or virus transfection could significantly inhibit NPC cell growth, induce apoptosis, and increase chemotherapy sensitivity *in vitro*53. Meanwhile, another study found that overexpression of *EGFR* and *IGF-1R* in advanced and recurrent NPC tissues affected the radiosensitivity of NPC cells54. Recently, Liu *et al*.55 verified that the dual-silencing of *IGF-1R* and *EGFR*, which are capable to decrease the expression of NPC cyclins and to block cell cycles, may promote the radiosensitivities and apotosis of NPC cells. #### VEGF VEGF, earlier known as vascular permeability factor, is a specific heparin-binding growth factor of vascular endothelial cells that can induce angiogenesis to support the growth of both normal and tumor tissues *in vivo*. As it is very essential for the progressive enlargement of malignant tumor, the function of VEGF in cancer treatment has been widely studied. Some studies found that VEGF made a great contribution to radioresistance in cancer radiotherapy both *in vitro* and *in vivo*. Those results suggested that targeting VEGF may inhibit cancer cell growth, decrease radioresistance, and prevent metastasis56. Angiotensin II (Ang II) is an important part of the renin–angiotensin-aldosterone system, which can regulate the expression of vasoconstriction, vasopressin, and aldosterone; induce angiogenesis by upregulating VEGF; and promote cell proliferation. In the circulatory system, almost all the physiological functions of Ang II are generated by stimulating the angiotensin II type 1 receptors (AT1R)57. Valsartan is a type of AT1R antagonist that can cut off the binding of Ang II to AT1R to reduce the production of VEGF. Previous studies reported that NPC cells could produce VEGF58, and its expression had a close relationship with the clinical stage59 and the prognosis of NPC60. In exploring whether valsartan plays a role in the treatment of NPC, Wang *et al*.61 revealed that the expression of AT1R was very high in NPC cells, and AT1R blocker valsartan could suppress the secretion of VEGF in NPC cells, thus inhibiting cancer cell growth. All these results implied that AT1R blocker valsartan could increase the radiosensitivity of NPC cells. Meanwhile, Zhou *et al*.62 used RNA interference (RNAi) technology to block *VEGF* expression of NPC cells, thus suppressing the proliferation and migration of NPC cells, inducing tumor apoptosis, and increasing the therapeutic efficacy of ionizing radiation. Therefore, methods to decrease VEGF in NPC cells may have great potential as novel therapeutic strategies against NPC. #### LMP-1 EBV, a prototype gamma herpes virus that infects the great majority of the population worldwide, has been implicated in the pathogenesis of several human malignancies including lymphoproliferative malignancies, such as Burkitt’s and Hodgkin’s lymphomas, as well as epithelial tumors, such as NPC and gastric carcinoma. EBV infection in NPC is classified as latency type II, which is characterized by the expression of a limited series of latent genes, including EBV nuclear antigen-1, latent membrane protein-1 (LMP1), LMP2, and EBV early RNA63. Considering its oncogenic potential, LMP1 is speculated to be an essential factor in the tumorigenesis of NPC64. The LMP1 protein is an integral membrane protein and can be subdivided into three domains: a short intracellular N terminus, which orientates the LMP1 protein to the plasma membrane; six hydrophobic transmembrane domains, which are involved in self-aggregation and oligomerization65; and an intracellular carboxyl-terminal activating region, which activates most of LMP1’s signaling activities66 and induces a variety of downstream pathological changes in cell proliferation, anti-apoptosis, and metastasis67,68. Therefore, Yang *et al*.69 successfully obtained a phosphorothioate-modified “10-23” DNAzyme, named DZ1, which could downregulate the expression of *LMP1* in NPC cells, to explore the potential use of DNAzymes for the new treatment of NPC. All results demonstrated that low expression of *LMP1* in NPC cells could inhibit cell proliferation and metastasis, promote apoptosis, and enhance radiosensitivity of NPC. In addition, the potential mechanism is through the interferences of signal pathways, which are launched by LMP1 including NF-κB, AP-1, and STAT3 signal pathways69. Recently, Ma *et al*.70 confirmed that LMP1 could regulate the expression of *ATM* via the NF-κB pathways, thus resulting in the change of radiosensitivity in NPC cells. Moreover, Xiao *et al*.71 used a metabolomics approach to investigate EBV-modulated metabolic changes, indicating that there is a close relationship between EBV and glycolysis in NPC. Significantly, LMP1 plays a key role in the reprogramming of EBV-mediated glycolysis in NPC cells, and anti-glycolytic therapy can effectively enhance the sensitivity of *LMP1*-overexpressing NPC cells to radiation. Thus, anti-glycolytic therapy may be a novel therapeutic strategy for NPC patients71. #### Protein kinase CK2 Protein kinase CK2 (formerly known as casein kinase 2 or II) is a common serine/threonine protein kinase that is localized in the cytoplasmic and nuclear compartments of the cell. Protein kinase CK2 contains two regulatory (28-kDaβ) subunits and two catalytic (42-kDaα and 38-kDaα') subunits, forming the heterotetrameric configurations α2β2, αα'β2, and α'2β2. The role of CK2, which is multifunctional, in normal and abnormal cell growth, proliferation, and apoptosis has been recognized for a long time. CK2 exerts its anti-apoptotic effects through the phosphorylation of a series factors, and inhibition of CK2 can induce apoptosis and promote tumor cell proliferation72. Furthermore, Liu *et al*.73 used RNAi technique to downregulate the *protein kinase CK2α* expression in NPC cells and demonstrated that *CK2α* knockdown significantly decreased the clonogenic activity and increased the radiosensitivity of the NPC cells. Hence, suppression of the expression of *CK2* may be a potential therapeutic approach to NPC. #### Bcl-2 *Bcl-2* gene (also known as B-cell lymphoma/leukemia gene-2) is one of the strongest genes that have an anti-apoptotic effect and is recognized as a human longevity gene74. *Bcl-2* gene can inhibit many apoptotic signal-induced apoptosis, promote cell survival, and disorder regulatory mechanism of apoptosis *in vivo*; this gene is found expressed in a variety of malignancies, such as NPC, lung cancer, and cervical cancer75. Previously, a study found that *Bcl-2* gene played a significant role in the promotion of tumor invasion and metastasis, and its evolvement had a close relationship with the progression of NPC76. Other studies demonstrated that Bcl-2 and Bcl-2/bax could be used as important indicators of radiation sensitivity in treating NPC cells77. Furthermore, Tian *et al*.78 successfully suppressed the expression of Bcl-2 protein by using RNAi technology, thus contributing to an increase of the radiosensitivity of NPC cells. All results suggested that *Bcl-2* may be a potential indicator for predicting the responses of NPC cells to radiotherapy. #### Notch signaling pathway Notch gene, first found in 1917 by Morgan79, is involved in the Notch signaling pathway, which is essential to determine cell fate, maintain the stability of stem cell, and regulate the pattern formation. In addition, its dysfunction is related to various defects of developments and human pathologies, including tumorigenesis80. Notch signaling pathway is found highly expressed in many malignant tumors, such as breast cancer81, glioma82, and NPC83. This pathway can be activated by EBV-coded latent membrane protein 2A, which is also an important factor to NPC oncogenesis84. Thus, Notch signaling pathway plays a vital role in NPC tumorigenesis. γ-secretase, a type of proteolytic enzyme, is very essential for the Notch signaling pathway, which can release the Notch intracellular domain. Given that Notch signaling pathway contributes to the progression of NPC, γ-secretase inhibitors (GSIs) that block Notch signaling pathway may achieve better effects when combined with radiotherapy85. Yu *et al*.86 investigated the effect of one GSI (N-[(3,5-difluorophenyl)acetyl]-L-alanyl-2-phenyl]glycine-1,1-dimethylethylester, DAPT) combined with radiotherapy. This study demonstrated that downregulation of Notch signaling pathway could increase the radiosensitivity of NPC cells, thus indicating GSIs as radiosensitizers for NPC treatment86. ## Conclusion NPC is a common head and neck cancer. The incidence rate is higher in southern China and Southeast Asia than in Western countries. Given its high radiosensitivity, radiotherapy is considered the primary treatment for NPC. However, radiotherapy is challenging for NPC because of the radiosensitive structures that surround the nasopharynx, including the brain stem, spinal cord, pituitary-hypothalamic axis, temporal lobes, eyes, ears, and parotid glands87. In addition, radiotherapy often brings adverse effects, such as xerostomia and temporal lobe necrosis. Many NPCs are resistant to radiation, which brings difficulties to treatment87. To improve the efficacy of radiotherapy, the use of radiosensitizers combined with ionizing radiation is beneficial. The concept of radiosensitizers was first proposed in 1958. All the chemical and biological methods that can affect the sensitivity to radiation are named radiosensitizers, including chemotherapeutic drugs and molecular targeted agents88,89. Chemotherapeutics, such as 5-fluorouracil, platinum analogs, and DNA topoisomerase I-targeting drugs, are commonly used when combined with radiotherapy and can achieve better local-regional therapeutic effects90. Molecular targeted agents that target DNA or not are also helpful to radiation therapy, such as *EGFR* blockers and *COX-2* inhibitors89. In this article, we have summarized some of the biomarkers, classified according to the main mechanisms of radiosensitization, to enhance the responsiveness of NPC cells to radiation treatment (Table 1). View this table: [Table 1](http://www.cancerbiomed.org/content/12/1/23/T1) Table 1 Biomarkers for enhancing the radiosensitivity of nasopharyngeal carcinoma (NPC) As described above, certain useful biomarkers can be silenced in NPC radiotherapy to enhance radiosensitivity. Meanwhile, to modify the expression of unwanted molecular biomarkers, short nucleic acid-based cancer therapeutics containing short interfering RNAs, antisense oligonucleotides, DNAzymes, and ribozymes were employed91. In addition, some agents can be used to silence or inhibit the expression of unwanted biomarkers, including topotecan, valsartan, and analogues of curcumin. Targeting the biomarkers to enhance the radiosensitivity of NPC is very promising and practicable. However, some problems need to be resolved. First, most of the candidate biomarkers are examined only in the molecular level studies and are not yet evaluated in large population clinical trials. Second, the mechanisms of some biomarkers leading to radiosensitization of NPC are not yet completely known. Third, whether some of the biomarkers are specific only to NPC or are common in other cancers is unclear. Hence, large cohort clinical trials are needed to verify the feasibility, availability, and safety of biomarkers to enhance the radiosensitivity of NPC cells. ## Footnotes * **Conflict of interest statement** No potential conflicts of interest are disclosed. * Received October 12, 2014. * Accepted January 7, 2015. * Copyright: © 2015, Cancer Biology & Medicine [https://creativecommons.org/licenses/by/4.0/](https://creativecommons.org/licenses/by/4.0/) This is an open access article distributed under the terms of the [Creative Commons Attribution License (CC BY) 4.0](https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited. ## References 1. 1. Wei WI, Sham JS. Nasopharyngeal carcinoma. Lancet 2005;365:2041–2054. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0140-6736(05)66698-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15950718&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000229701400026&link_type=ISI) 2. 2. Tao Q, Chan AT. Nasopharyngeal carcinoma: molecular pathogenesis and therapeutic developments. Expert Rev Mol Med 2007;9:1–24. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1017/S146239940700035X&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17565757&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 3. 3. Lo KW, Chung GT, To KF. Deciphering the molecular genetic basis of NPC through molecular, cytogenetic, and epigenetic approaches. Semin Cancer Biol 2012;22:79–86. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.semcancer.2011.12.011&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22245473&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000302450100002&link_type=ISI) 4. 4. Barnett GC, West CM, Dunning AM, Elliott RM, Coles CE, Pharoah PD, et al. Normal tissue reactions to radiotherapy: towards tailoring treatment dose by genotype. Nat Rev Cancer 2009;9:134–142. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nrc2587&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19148183&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000263180900015&link_type=ISI) 5. 5. Lee AW, Sze WM, Au JS, Leung SF, Leung TW, Chua DT, et al. Treatment results for nasopharyngeal carcinoma in the modern era: the Hong Kong experience. Int J Radiat Oncol Biol Phys 2005;61:1107–1116. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ijrobp.2004.07.702&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15752890&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000227604200017&link_type=ISI) 6. 6. Rottey S, Madani I, Deron P, Van Belle S. Modern treatment for nasopharyngeal carcinoma: current status and prospects. Curr Opin Oncol 2011;23:254–258. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1097/CCO.0b013e328344f527&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21330921&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 7. 7. Poggi MM, Coleman CN, Mitchell JB. Sensitizers and protectors of radiation and chemotherapy. Curr Probl Cancer 2001;25:334–411. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1067/mcn.2001.120122&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11740469&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000172822200001&link_type=ISI) 8. 8. Stewart GS, Wang B, Bignell CR, Taylor AM, Elledge SJ. MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature 2003;421:961–966. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature01446&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12607005&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000181186900053&link_type=ISI) 9. 9. Fernandez-Capetillo O, Chen HT, Celeste A, Ward I, Romanienko PJ, Morales JC, et al. DNA damage-induced G2-M checkpoint activation by histone H2AX and 53BP1. Nat Cell Biol 2002;4:993–997. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ncb884&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12447390&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000179571800021&link_type=ISI) 10. 10. Lou Z, Minter-Dykhouse K, Franco S, Gostissa M, Rivera MA, Celeste A, et al. MDC1 maintains genomic stability by participating in the amplification of ATM-dependent DNA damage signals. Mol Cell 2006;21:187–200. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.molcel.2005.11.025&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16427009&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000234943000008&link_type=ISI) 11. 11. Liu DD, Hu GH, Liu XL, Bu YQ. Effect of siRNA-mediated NFBD1 depletion on chemosensitivity in nasopharyngeal carcinoma cells. J Chongqing Med Univ 2012;37:393–396. 12. 12. Qiu ZL, Zeng Q, Liu XL, Hu GH. Expressions of NFBD1 in nasopharyngeal carcinoma and their clinical significances. J Chongqing Med Univ 2013;38:126–129. 13. 13. Wang ZH, Zeng Q, Chen T, Liao K, Bu YQ, Hong SL, et al. Effect of NFBD1 gene silencing by RNA interference on radiosensitivity of nasopharyngeal carcinoma CNE-1 cells. Tumor 2014;34:101–107. 14. 14. Norbury CJ, Hickson ID. Cellular responses to DNA damage. Annu Rev Pharmacol Toxicol 2001;41:367–401. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1146/annurev.pharmtox.41.1.367&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11264462&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000168439400015&link_type=ISI) 15. 15. Ulrich CM, Bigler J, Potter JD. Non-steroidal anti-inflammatory drugs for cancer prevention: promise, perils and pharmacogenetics. Nat Rev Cancer 2006;6:130–140. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nrc1801&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16491072&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000234844000011&link_type=ISI) 16. 16. Wang D, Dubois RN. Cyclooxygenase-2: a potential target in breast cancer. Semin Oncol 2004;31:64–73. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1053/j.seminoncol.2004.01.008&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15052544&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000220589900008&link_type=ISI) 17. 17. Hill R, Li Y, Tran LM, Dry S, Calvopina JH, Garcia A, et al. Cell intrinsic role of COX-2 in pancreatic cancer development. Mol Cancer Ther 2012;11:2127–2137. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6Im1vbGNhbnRoZXIiO3M6NToicmVzaWQiO3M6MTA6IjExLzEwLzIxMjciO3M6NDoiYXRvbSI7czoxNzoiL2NibS8xMi8xLzIzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 18. 18. Zhao Y, Hao Y, Ji H, Fang Y, Guo Y, Sha W, et al. Combination effects of salvianolic acid B with low-dose celecoxib on inhibition of head and neck squamous cell carcinoma growth in vitro and in vivo. Cancer Prev Res (Phila) 2010;3:787–796. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNhbnByZXZyZXMiO3M6NToicmVzaWQiO3M6NzoiMy82Lzc4NyI7czo0OiJhdG9tIjtzOjE3OiIvY2JtLzEyLzEvMjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 19. 19. Mutter R, Lu B, Carbone DP, Csiki I, Moretti L, Johnson DH, et al. A phase II study of celecoxib in combination with paclitaxel, carboplatin, and radiotherapy for patients with inoperable stage IIIA/B non-small cell lung cancer. Clin Cancer Res 2009;15:2158–2165. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6OToiMTUvNi8yMTU4IjtzOjQ6ImF0b20iO3M6MTc6Ii9jYm0vMTIvMS8yMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 20. 20. Shin YK, Park JS, Kim HS, Jun HJ, Kim GE, Suh CO, et al. Radiosensitivity enhancement by celecoxib, a cyclooxygenase (COX)-2 selective inhibitor, via COX-2-dependent cell cycle regulation on human cancer cells expressing differential COX-2 levels. Cancer Res 2005;65:9501–9509. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI2NS8yMC85NTAxIjtzOjQ6ImF0b20iO3M6MTc6Ii9jYm0vMTIvMS8yMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 21. 21. Tan KB, Putti TC. Cyclooxygenase 2 expression in nasopharyngeal carcinoma: immunohistochemical findings and potential implications. J Clin Pathol 2005;58:535–538. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToiamNsaW5wYXRoIjtzOjU6InJlc2lkIjtzOjg6IjU4LzUvNTM1IjtzOjQ6ImF0b20iO3M6MTc6Ii9jYm0vMTIvMS8yMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 22. 22. Chen WC, McBride WH, Chen SM, Lee KF, Hwang TZ, Jung SM, et al. Prediction of poor survival by cyclooxygenase-2 in patients with T4 nasopharyngeal cancer treated by radiation therapy: clinical and in vitro studies. Head Neck 2005;27:503–512. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15772955&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 23. 23. Sun QQ, Liu X, Liu Y, Wang L, Peng XY, Zeng FF, et al. Effect of shRNAmir lentiviral vector-mediated COX-2 silencing on the radiosensitivity of nasopharyngeal carcinoma cells. Nan Fang Yi Ke Da Xue Xue Bao 2011;31:599–603. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21515451&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 24. 24. Zhang SX, Qiu QH, Chen WB, Liang CH, Huang B. Celecoxib enhances radiosensitivity via induction of G2-M phase arrest and apoptosis in nasopharyngeal carcinoma. Cell Physiol Biochem 2014;33:1484–1497. 25. 25. Srivastava P. Interaction of heat shock proteins with peptides and antigen presenting cells: chaperoning of the innate and adaptive immune responses. Annu Rev Immunol 2002;20:395–425. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1146/annurev.immunol.20.100301.064801&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11861608&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000175012600015&link_type=ISI) 26. 26. Li PX, Wong J, Ayed A, Ngo D, Brade AM, Arrowsmith C, et al. Placental transforming growth factor-beta is a downstream mediator of the growth arrest and apoptotic response of tumor cells to DNA damage and p53 overexpression. J Biol Chem 2000;275:20127–20135. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEyOiIyNzUvMjYvMjAxMjciO3M6NDoiYXRvbSI7czoxNzoiL2NibS8xMi8xLzIzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 27. 27. Chang JT, Chan SH, Lin CY, Lin TY, Wang HM, Liao CT, et al. Differentially expressed genes in radioresistant nasopharyngeal cancer cells: gp96 and GDF15. Mol Cancer Ther 2007;6:2271–2279. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6Im1vbGNhbnRoZXIiO3M6NToicmVzaWQiO3M6ODoiNi84LzIyNzEiO3M6NDoiYXRvbSI7czoxNzoiL2NibS8xMi8xLzIzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 28. 28. Ohtsubo K, Marth JD. Glycosylation in cellular mechanisms of health and disease. Cell 2006;126:855–867. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2006.08.019&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16959566&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000240675000014&link_type=ISI) 29. 29. Przybyło M, Martuszewska D, Pocheć E, Hoja-Łukowicz D, Lityńska A. Identification of proteins bearing beta1-6 branched N-glycans in human melanoma cell lines from different progression stages by tandem mass spectrometry analysis. Biochim Biophys Acta 2007;1770:1427–1435. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17600626&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 30. 30. Wei T, Li Y, Zhuo E, Zhu W, Meng H, Zhang J. Down-regulation of GnT-V inhibits nasopharyngeal carcinoma cell CNE-2 malignancy in vitro and in vivo. Cancer Lett 2011;309:151–161. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21676538&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 31. 31. Zhuo E, He J, Wei T, Zhu W, Meng H, Li Y, et al. Down-regulation of GnT-V enhances nasopharyngeal carcinoma cell CNE-2 radiosensitivity in vitro and in vivo. Biochem Biophys Res Commun 2012;424:554–562. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22780953&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 32. 32. Shackleford TJ, Claret FX. JAB1/CSN5: a new player in cell cycle control and cancer. Cell Div 2010;5:26. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1186/1747-1028-5-26&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20955608&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 33. 33. Kouvaraki MA, Korapati AL, Rassidakis GZ, Tian L, Zhang Q, Chiao P, et al. Potential role of Jun activation domain-binding protein 1 as a negative regulator of p27kip1 in pancreatic adenocarcinoma. Cancer Res 2006;66:8581–8589. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI2Ni8xNy84NTgxIjtzOjQ6ImF0b20iO3M6MTc6Ii9jYm0vMTIvMS8yMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 34. 34. Kouvaraki MA, Rassidakis GZ, Tian L, Kumar R, Kittas C, Claret FX. Jun activation domain-binding protein 1 expression in breast cancer inversely correlates with the cell cycle inhibitor p27(Kip1). Cancer Res 2003;63:2977–2981. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI2My8xMS8yOTc3IjtzOjQ6ImF0b20iO3M6MTc6Ii9jYm0vMTIvMS8yMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 35. 35. Pan Y, Zhang Q, Tian L, Wang X, Fan X, Zhang H, et al. Jab1/CSN5 negatively regulates p27 and plays a role in the pathogenesis of nasopharyngeal carcinoma. Cancer Res 2012;72:1890–1900. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjk6IjcyLzcvMTg5MCI7czo0OiJhdG9tIjtzOjE3OiIvY2JtLzEyLzEvMjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 36. 36. Pan Y, Zhang Q, Atsaves V, Yang H, Claret FX. Suppression of Jab1/CSN5 induces radio- and chemo-sensitivity in nasopharyngeal carcinoma through changes to the DNA damage and repair pathways. Oncogene 2013;32:2756–2766. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/onc.2012.294&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22797071&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 37. 37. Ji JL, Huang XF, Zhu HL. Curcumin and its formulations: potential anti-cancer agents. Anticancer Agents Med Chem 2012;12:210–218. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22044005&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 38. 38. Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB. Bioavailability of curcumin: problems and promises. Mol Pharm 2007;4:807–818. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1021/mp700113r&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17999464&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 39. 39. Debata PR, Castellanos MR, Fata JE, Baggett S, Rajupet S, Szerszen A, et al. A novel curcumin-based vaginal cream Vacurin selectively eliminates apposed human cervical cancer cells. Gynecol Oncol 2013;129:145–153. 40. 40. Manohar S, Khan SI, Kandi SK, Raj K, Sun G, Yang X, et al. Synthesis, antimalarial activity and cytotoxic potential of new monocarbonyl analogues of curcumin. Bioorg Med Chem Lett 2013;23:112–116. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.bmcl.2012.11.004&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23218718&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 41. 41. Pan Y, Wang M, Bu X, Zuo Y, Wang S, Wang D, et al. Curcumin analogue T83 exhibits potent antitumor activity and induces radiosensitivity through inactivation of Jab1 in nasopharyngeal carcinoma. BMC Cancer 2013;13:323. 42. 42. Chen SH, Chan NL, Hsieh TS. New mechanistic and functional insights into DNA topoisomerases. Annu Rev Biochem 2013;82:139–170. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1146/annurev-biochem-061809-100002&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23495937&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000321698100006&link_type=ISI) 43. 43. Koster DA, Croquette V, Dekker C, Shuman S, Dekker NH. Friction and torque govern the relaxation of DNA supercoils by eukaryotic topoisomerase IB. Nature 2005;434:671–674. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature03395&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15800630&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000228011000051&link_type=ISI) 44. 44. Staker BL, Feese MD, Cushman M, Pommier Y, Zembower D, Stewart L, et al. Structures of three classes of anticancer agents bound to the human topoisomerase I-DNA covalent complex. J Med Chem 2005;48:2336–2345. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1021/jm049146p&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15801827&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000228111500013&link_type=ISI) 45. 45. Chen AY, Chou R, Shih SJ, Lau D, Gandara D. Enhancement of radiotherapy with DNA topoisomerase I-targeted drugs. Crit Rev Oncol Hematol 2004;50:111–119. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15157660&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 46. 46. Zhang Y, Chen X, Ren P, Su Z, Cao H, Zhou J, et al. Synergistic effect of combination topotecan and chronomodulated radiation therapy on xenografted human nasopharyngeal carcinoma. Int J Radiat Oncol Biol Phys 2013;87:356–362. 47. 47. Carreras MC, Poderoso JJ. Mitochondrial nitric oxide in the signaling of cell integrated responses. Am J Physiol Cell Physiol 2007;292:C1569–C1580. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1152/ajpcell.00248.2006&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17496232&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000247936500001&link_type=ISI) 48. 48. De Ridder M, Van Esch G, Engels B, Verovski V, Storme G. Hypoxic tumor cell radiosensitization: role of the iNOS/NO pathway. Bull Cancer 2008;95:282–291. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18390408&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 49. 49. Shigyo H, Nonaka S, Katada A, Bandoh N, Ogino T, Katayama A, et al. Inducible nitric oxide synthase expression in various laryngeal lesions in relation to carcinogenesis, angiogenesis, and patients’ prognosis. Acta Otolaryngol 2007;127:970–979. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17712678&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 50. 50. Jayasurya A, Dheen ST, Yap WM, Tan NG, Ng YK, Bay BH. Inducible nitric oxide synthase and bcl-2 expression in nasopharyngeal cancer: correlation with outcome of patients after radiotherapy. Int J Radiat Oncol Biol Phys 2003;56:837–845. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0360-3016(03)00122-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12788193&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000183361800031&link_type=ISI) 51. 51. Lorch JH, Posner MR, Wirth LJ, Haddad RI. Seeking alternative biological therapies: the future of targeted molecular treatment. Oral Oncol 2009;45:447–453. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19027348&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 52. 52. Dziadziuszko R, Merrick DT, Witta SE, Mendoza AD, Szostakiewicz B, Szymanowska A, et al. Insulin-like growth factor receptor 1 (IGF1R) gene copy number is associated with survival in operable non-small-cell lung cancer: a comparison between IGF1R fluorescent in situ hybridization, protein expression, and mRNA expression. J Clin Oncol 2010;28:2174–2180. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyOC8xMy8yMTc0IjtzOjQ6ImF0b20iO3M6MTc6Ii9jYm0vMTIvMS8yMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 53. 53. Yuan YL, Zhou XH, Song J, Qiu XP, Li J, Ye LF. Dual silencing of type 1 insulin-like growth factor and epidermal growth factor receptors to induce apoptosis of nasopharyngeal cancer cells. J Laryngol Otol 2008;122:952–960. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1017/S0022215107000606&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17908353&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 54. 54. Yuan Y, Zhou X, Song J, Qiu X, Li J, Ye L, et al. Expression and clinical significance of epidermal growth factor receptor and type 1 insulin-like growth factor receptor in nasopharyngeal carcinoma. Ann Otol Rhinol Laryngol 2008;117:192–200. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18444479&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 55. 55. Liu GL, Yuan YL, Liu YJ, Zhou XH. Dual Silencing of Insulin-Like Growth Factor-1 and Epidermal Growth Factor Receptors to Induce the Radioactivity of Nasopharyngeal Cancer Cells. Med J Wuhan Univ 2012;33:307–311. 56. 56. Ye WJ, Min HQ, Cao XP, Chen KT, Hou JH, Li Y. Correlations of biomolecular markers, such as P53 protein and vascular endothelial growth factor, to radiosensitivity of nasopharyngeal carcinoma. Ai Zheng 2006;25:1168–1172. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16965664&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 57. 57. Mehta PK, Griendling KK. Angiotensin II cell signaling: physiological and pathological effects in the cardiovascular system. Am J Physiol Cell Physiol 2007;292:C82–C97. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1152/ajpcell.00287.2006&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16870827&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000243425900010&link_type=ISI) 58. 58. Yang HF, Tang WP. Vascular endothelial growth factor gene expression in nasopharyngeal carcinoma cell line induced by hypoxia in vitro. Ai Zheng 2003;22:160–163. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12600291&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 59. 59. Sha D, He YJ. Expression and clinical significance of VEGF and its receptors Flt-1 and KDR in nasopharyngeal carcinoma. Ai Zheng 2006;25:229–234. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16480593&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 60. 60. Krishna SM, James S, Balaram P. Expression of VEGF as prognosticator in primary nasopharyngeal cancer and its relation to EBV status. Virus Res 2006;115:85–90. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.virusres.2005.07.010&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16139912&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000234574400010&link_type=ISI) 61. 61. Wang Q, Zhao W, Wu G. Valsartan inhibits NPC cell line CNE-2 proliferation and invasion and promotes its sensitivity to radiation. Eur J Cancer Prev 2009;18:510–517. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19687742&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 62. 62. Zhou HB, Yin YF, Hu Y, Li X, Zou LY, Li YJ, et al. Suppression of vascular endothelial growth factor via siRNA interference modulates the biological behavior of human nasopharyngeal carcinoma cells. Jpn J Radiol 2011;29:615–622. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21956366&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 63. 63. Chou J, Lin YC, Kim J, You L, Xu Z, He B, et al. Nasopharyngeal carcinoma--review of the molecular mechanisms of tumorigenesis. Head Neck 2008;30:946–963. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/hed.20833&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18446839&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000257480700018&link_type=ISI) 64. 64. Morris MA, Dawson CW, Young LS. Role of the Epstein-Barr virus-encoded latent membrane protein-1, LMP1, in the pathogenesis of nasopharyngeal carcinoma. Future Oncol 2009;5:811–825. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.2217/fon.09.53&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19663731&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000269434400014&link_type=ISI) 65. 65. Zheng H, Li LL, Hu DS, Deng XY, Cao Y. Role of Epstein-Barr virus encoded latent membrane protein 1 in the carcinogenesis of nasopharyngeal carcinoma. Cell Mol Immunol 2007;4:185–196. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17601372&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000249759000003&link_type=ISI) 66. 66. Hatzivassiliou E, Mosialos G. Cellular signaling pathways engaged by the Epstein-Barr virus transforming protein LMP1. Front Biosci 2002;7:d319–d329. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.2741/hatziva&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=11779697&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 67. 67. Horikawa T, Yang J, Kondo S, Yoshizaki T, Joab I, Furukawa M, et al. Twist and epithelial-mesenchymal transition are induced by the EBV oncoprotein latent membrane protein 1 and are associated with metastatic nasopharyngeal carcinoma. Cancer Res 2007;67:1970–1978. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjk6IjY3LzUvMTk3MCI7czo0OiJhdG9tIjtzOjE3OiIvY2JtLzEyLzEvMjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 68. 68. Dawson CW, Laverick L, Morris MA, Tramoutanis G, Young LS. Epstein-Barr virus-encoded LMP1 regulates epithelial cell motility and invasion via the ERK-MAPK pathway. J Virol 2008;82:3654–3664. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjk6IjgyLzcvMzY1NCI7czo0OiJhdG9tIjtzOjE3OiIvY2JtLzEyLzEvMjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 69. 69. Yang L, Lu Z, Ma X, Cao Y, Sun LQ. A therapeutic approach to nasopharyngeal carcinomas by DNAzymes targeting EBV LMP-1 gene. Molecules 2010;15:6127–6139. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.3390/molecules15096127&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20877211&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 70. 70. Ma X, Yang L, Xiao L, Tang M, Liu L, Li Z, et al. Down-regulation of EBV-LMP1 radio-sensitizes nasal pharyngeal carcinoma cells via NF-κB regulated ATM expression. PLoS One 2011;6:e24647. 71. 71. Xiao L, Hu ZY, Dong X, Tan Z, Li W, Tang M, et al. Targeting Epstein-Barr virus oncoprotein LMP1-mediated glycolysis sensitizes nasopharyngeal carcinoma to radiation therapy. Oncogene 2014;33:4568–4578. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/onc.2014.32&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24662831&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 72. 72. Litchfield DW. Protein kinase CK2: structure, regulation and role in cellular decisions of life and death. Biochem J 2003;369:1–15. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBwYmlvY2hlbWoiO3M6NToicmVzaWQiO3M6NzoiMzY5LzEvMSI7czo0OiJhdG9tIjtzOjE3OiIvY2JtLzEyLzEvMjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 73. 73. Liu L, Zou JJ, Luo HS, Wu DH. Effect of protein kinase CK2 gene silencing on radiosensitization in human nasopharyngeal carcinoma cells. Nan Fang Yi Ke Da Xue Xue Bao 2009;29:1551–1553. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19726289&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 74. 74. Wang X, Wang Y, Kim HP, Nakahira K, Ryter SW, Choi AM. Carbon monoxide protects against hyperoxia-induced endothelial cell apoptosis by inhibiting reactive oxygen species formation. J Biol Chem 2007;282:1718–1726. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEwOiIyODIvMy8xNzE4IjtzOjQ6ImF0b20iO3M6MTc6Ii9jYm0vMTIvMS8yMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 75. 75. Deng WT, Wang SL. Research of the relationship between p53, p16, bcl-2, COX-2 and nasopharyngeal carcinoma. J Practi Med 2010;26:1849–1851. 76. 76. Li J, Pan YD, Li ZQ, Guo XT, Li LZ. Relations between the expression of survivin, Bcl-2, nm23-H1 and the standardized uptake value (SUV) of 18F-fluorodeoxyglucose in NPC. Guangdong Med J 2010;31:1106–1108. 77. 77. Huang T, Chen MH, Wu MY, Wu XY, Huang WJ, Ou HQ. The Relationship between the Expression of bcl-2 and bax Protein and the Radiosensitivity of Nasopharyngeal Carcinoma. CJCO 2004;31:1051–1053. 78. 78. Tian XF, Liu DM, Zhang SH, Li GW. Effect of siRNA-mediated Bcl-2 silencing on the radiosensitivity of nasopharyngeal carcinoma cells. J Practi Med 2012;28:1411–1413. 79. 79. Lai EC. Notch signaling: control of cell communication and cell fate. Development 2004;131:965–973. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGV2ZWxvcCI7czo1OiJyZXNpZCI7czo5OiIxMzEvNS85NjUiO3M6NDoiYXRvbSI7czoxNzoiL2NibS8xMi8xLzIzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 80. 80. Purow B. Notch inhibitors as a new tool in the war on cancer: a pathway to watch. Curr Pharm Biotechnol 2009;10:154–160. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.2174/138920109787315060&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19199947&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 81. 81. Guo S, Liu M, Gonzalez-Perez RR. Role of Notch and its oncogenic signaling crosstalk in breast cancer. Biochim Biophys Acta 2011;1815:197–213. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.bbcan.2010.12.002&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21193018&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 82. 82. Jiang L, Wu J, Chen Q, Hu X, Li W, Hu G. Notch1 expression is upregulated in glioma and is associated with tumor progression. J Clin Neurosci 2011;18:387–390. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.jocn.2010.07.131&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21251836&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 83. 83. Zhang Y, Peng J, Zhang H, Zhu Y, Wan L, Chen J, et al. Notch1 signaling is activated in cells expressing embryonic stem cell proteins in human primary nasopharyngeal carcinoma. J Otolaryngol Head Neck Surg 2010;39:157–166. 84. 84. Anderson LJ, Longnecker R. An auto-regulatory loop for EBV LMP2A involves activation of Notch. Virology 2008;371:257–266. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.virol.2007.10.009&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17980397&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000253060200006&link_type=ISI) 85. 85. Lin J, Zhang XM, Yang JC, Ye YB, Luo SQ. γ-secretase inhibitor-I enhances radiosensitivity of glioblastoma cell lines by depleting CD133+ tumor cells. Arch Med Res 2010;41:519–529. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.arcmed.2010.10.006&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21167391&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 86. 86. Yu S, Zhang R, Liu F, Hu H, Yu S, Wang H. Down-regulation of Notch signaling by a γ-secretase inhibitor enhances the radiosensitivity of nasopharyngeal carcinoma cells. Oncol Rep 2011;26:1323–1328. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21805038&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 87. 87. Lee CC, Ho CY. Post-treatment late complications of nasopharyngeal carcinoma. Eur Arch Otorhinolaryngol 2012;269:2401–2409. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s00405-011-1922-2&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22249834&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 88. 88. Spalding AC, Lawrence TS. New and emerging radiosensitizers and radioprotectors. Cancer Invest 2006;24:444–456. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1080/07357900600705706&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16777698&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000238414500015&link_type=ISI) 89. 89. Kvols LK. Radiation sensitizers: a selective review of molecules targeting DNA and non-DNA targets. J Nucl Med 2005;46 Suppl 1:187S–190S. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam51bWVkIjtzOjU6InJlc2lkIjtzOjE1OiI0Ni8xX3N1cHBsLzE4N1MiO3M6NDoiYXRvbSI7czoxNzoiL2NibS8xMi8xLzIzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 90. 90. Cooper JS, Ang KK. Concomitant chemotherapy and radiation therapy certainly improves local control. Int J Radiat Oncol Biol Phys 2005;61:7–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ijrobp.2004.09.034&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15629587&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom) 91. 91. Zhang L, Yang L, Li JJ, Sun L. Potential use of nucleic acid-based agents in the sensitization of nasopharyngeal carcinoma to radiotherapy. Cancer Lett 2012;323:1–10. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.canlet.2012.03.030&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22484469&link_type=MED&atom=%2Fcbm%2F12%2F1%2F23.atom)