Review Open Access
Copyright ©2007 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Feb 21, 2007; 13(7): 985-992
Published online Feb 21, 2007. doi: 10.3748/wjg.v13.i7.985
p53 gene in treatment of hepatic carcinoma: Status quo
Yong-Song Guan, Zi La, Lin Yang, Qing He, Ping Li, Department of Radiology and Oncology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
Author contributions: All authors contributed equally to the work.
Correspondence to: Dr. Yong-Song Guan, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University-Gaopeng Street, Keyuan Road 4, Chengdu 610041, Sichuan Province, China. yongsongguan@yahoo.com
Telephone: +86-28-85422601 Fax: +86-28-85538359
Received: November 30, 2006
Revised: December 12, 2006
Accepted: January 16, 2007
Published online: February 21, 2007

Abstract

Hepatocellular carcinoma (HCC) is one of the 10 most common cancers worldwide. There is no ideal treatment for HCC yet and many researchers are trying to improve the effects of treatment by changing therapeutic strategies. As the majority of human cancers seem to exhibit either abnormal p53 gene or disrupted p53 gene activation pathways, intervention to restore wild-type p53 (wt-p53) activities is an attractive anti-cancer therapy including HCC. Abnormalities of p53 are also considered a predisposition factor for hepatocarcinogenesis. p53 is frequently mutated in HCC. Most HCCs have defects in the p53-mediated apoptotic pathway although they carry wt-p53. High expression of p53 in vivo may exert therapeutic effects on HCC in two aspects: (1) High expression of exogenous p53 protein induces apoptosis of tumor cells by inhibiting proliferation of cells through several biologic pathways and (2) Exogenous p53 renders HCC more sensitive to some chemotherapeutic agents. Several approaches have been designed for the treatment of HCC via the p53 pathway by restoring the tumor suppression function from inactivation, rescuing the mutated p53 gene from instability, or delivering therapeutic exogenous p53. Products with p53 status as the target have been studied extensively in vitro and in vivo. This review elaborates some therapeutic mechanisms and advances in using recombinant human adenovirus p53 and oncolytic virus products for the treatment of HCC.

Key Words: p53 gene, Hepatocellular carcinoma, Therapeutic strategies, Advances, Prospects



INTRODUCTION

The concept of human gene therapy[1,2] derives from fundamental discoveries[3,4] on the nature and working[5] of the gene. Since the essential principles of molecular genetics and gene transfer in bacteria were established in the 1960s, gene transfer into animals and humans using either viral vector and/or genetically modified cultured cells has become inevitable[6]. Since then, this concept has promoted thousands of researchers to attempt to realize their long-cherished dreams of eradicating some of the obstinate human diseases. Broadly defined, the concept of gene therapy involves transfer of genetic materials[6] into cells, tissues, or whole organs, with the goal of eliminating diseases or at least improving the clinical condition of patients[7]. As a milestone of gene therapy, the first approved clinical protocol started trials in September, 1990[8]. Gene therapy provides great opportunities for treating diseases due to genetic disorders, infections and cancer[9]. Hepatocellular carcinoma (HCC) is one of the 10 most common cancers worldwide with its highest prevalence in Southeast Asia and sub-Saharan Africa[10]. Although great efforts have been made to overcome this cancer, no ideal treatment is available at present[11]. In recent years, many researchers are trying to improve the effects of treatment by changing therapeutic strategies[12]. As the majority of human cancers seem to exhibit either abnormal p53 gene[13] or disrupted p53 gene activation pathways[14], intervention to restore wild-type p53 (wt-p53) activities is an attractive anti-cancer therapy[14,15]. This review elaborates some therapeutic mechanisms and advances in using recombinant human adenovirus p53[16] and oncolytic virus[17] products for the treatment of HCC.

CARCINOGENESIS OF LIVER CELLS AND p53 GENE

Cancer predisposition, onset and therapeutic response can be critically determined by the integrity of tumor suppressor p53[15]. Tumor suppressor protein p53 can protect cells from growth and division[18], thus mediating cell-cycle arrest[19], DNA repair[20] and apoptosis[13,14,19,21] after its activation by multiple forms of cellular stresses[19]. p53 mutations[22] in plasma DNA are associated with several cancers, and abnormalities of p53 are also considered a predisposition factor for HCC (Table 1). Mutant p53 (mt-p53) may be a marker of HCC carcinogen exposure[22]. For example, in aflatoxin B1-exposed patients[23], R249S tumor protein p53[24], one of the cancer-associated mutants, encodes p53, and is considered a genetic alteration during hepatocarcinogenesis[23]. Here R249S as a mutant of p53 core domain, is a structural mutation[25], and constitutes one of the “hot spots” associated with cancer.

Table 1 Abnormal p53 status detected in HCC with probable mechanisms.
Endogenous status of p53 or its pathwaysProbable mechanisms
Mutation of p53 genePoint mutation, allele deletion, insertion, etc.
Loss of or decrease in wt-p53 expressionEnhanced p53 degradation, complex formation
Hyperactivities of negative regulators of p53No p53 mutation, over- expressed negative regulators of p53 attenuate its function
Increased diversity of p53 aberrationProgression of HCC
Presence of serum anti-p53 antibodiesOver-expression of wt-p53, presence of mt-p53, or both

There is evidence that the level of p53 alterations is high in HCC, since it was reported that p53 increases the frequency of HCC prediction from 79.5% to 86.3%[26], showing that serum concentration of p53 protein may be a convenient and useful non-invasive screening test for prediction of HCC. A study[27] showed that attenuated p53 function and telomere-induced chromosomal instability play a critical and cooperative role in the progression of chronic liver damage to hepatocellular carcinoma. Loss of p53 expression or presence of abnormal forms of the protein is frequently associated with HCC cell lines. Bressac et al[28] studied the p53 gene at the DNA, RNA, and protein level in seven human HCC-derived cell lines, and found that six of them show p53 abnormalities, suggesting that alterations in p53 may be important events in the transformation of hepatocytes[29] into the malignant phenotype. p53 gene is frequently mutated in high-grade[30] HCC. Inactivation of this multiple tumor suppressor gene plays an important role in the progression of chronic liver damage to hepatocellular carcinoma by directly or indirectly inducing chromosome instability, cell proliferation and neovascularization[31]. Kondo et al[32] performed dual-color fluorescence in situ hybridization to evaluate loss of the p53 gene, and revealed that loss of the p53 gene occurs in HCC, and diversity of the p53 gene aberration increases with the progression of chronic liver damage to HCC.

Other factors for hepatocarcinogenesis are correlated with abnormal functioning of p53, such as hepatitis B virus (HBV)[23,33] and hepatitis C virus (HCV)[34]. HBV gene encodes HBV protein x (HBx)[33], a protein as a transcriptional activator[35] and plays an important role in viral replication in HBV-infected cells. HBx as an oncoprotein[36], can bind to the C terminus of p53 and inhibit several critical p53-mediated cellular processes, including DNA sequence-specific binding, transcriptional transactivation, and apoptosis[37]. HBx integration and inactivation of p53 by mutations and regional allelic deletions are frequently found in tumors associated with HBV infection[38]. HBx up-regulates survivin[39] expression in hepatoma tissues. Survivin is an inhibitor of apoptosis and found in many common human cancers but not in normal tissues. Survivin is suppressed by wt-p53 and over-expressed in 41%-70% of HCCs from Asia, its over-expression is associated with aberrant p53 nuclear positivity[40].

In addition to its aberration, as p53 responds to a variety of genotoxic[34] and cytotoxic[41] agents in the presence of a potent inhibitor[42] of p53, the liver’s ability to handle such toxic agents is influenced, thus inducing hepatocarcinogenesis.

Anti-p53 antibodies (p53-Abs) are products triggered by accumulation of a mutated form of p53 protein and probably a large quantity of wt-p53 protein[43]. As a specific serological marker for p53 gene expression changes in HCC, the presence of p53-Abs is independent of serum alpha fetoprotein and other conventional tumor markers[44]. It was reported that serum p53-Abs have a specificity of 100% for detecting malignancy[44], suggesting that its use in combination with markers may increase the diagnostic sensitivity of cancer.

MECHANISMS OF p53 THERAPY FOR HCC

Restoration of tumor suppression function of p53 has been speculated in several clinical lines for the treatment of HCC (Table 2).

Table 2 Approaches for restoring tumor suppression function of p53 in the treatment of HCC.
StrategiesTherapeutic effects
Chemotherapy or radiotherapyInducing p53-dependent apoptosis in tumor cells with wt-p53
Supplying exogenous wt-p53 by gene deliverySuppressing growth of both mutant and wild-type p53-containing tumor cells
Overexpression of ARFBlocking p53 degradation pathways to induce p53 triggered tumor cell death
Interruption of MDM2-p53 interactionPreventing MDM2-mediated p53 ubiquitinationand degradation to restore transactivation
Molecules stabilizing the active conformation of the proteinRescuing mt-p53 to restore p53 function

p53 is frequently mutated in HCC[31]. Most HCCs have defects in the p53-mediated[37] apoptotic pathway although they carry wt-p53. Sometimes, p53 in HCC is wild-type but has inactive function[45]. Signal to and activation of p53 can lead to wt-p53 expression, thus suppressing the transformed phenotype of hepatocytes and increasing the effects of both chemotherapeutic agents and radiation therapy[46]. Repression of p53 can be partial or complete[45] in HCC, and activation of p53 can be achieved by single small molecules, such as the well known antimalaria drug quinacrine[45]. Supplying exogenous wt-p53 in cancer cells by gene delivery is effective in suppressing tumor growth of both mutant and wild-type p53-containing tumors[14]. The murine double minute 2 gene (MDM2) is an oncogene and contains a p53-DNA binding site and produces a phosphoprotein that forms a tight complex with both mutant and wild-type p53 protein, thus inhibiting p53-mediated transactivation and inducing p53 degradation[47]. There is a MDM2-p53 auto-regulatory feedback loop[48] that regulates the function of p53 protein and expression of the MDM2 gene. Expression of the MDM2 gene can be regulated by the level of wild-type p53 protein, while the MDM2 protein, in turn, can form a complex with p53 and decrease its ability to act as a positive transcription factor at the MDM2 gene-responsive element. Several approaches have now been used to interrupt MDM2-p53 interaction to increase functional p53 levels and p53-mediated therapeutic effectiveness[49]. ADP-ribosylation factor (ARF) proteins are critical regulators of the protein secreting pathway[50]. In a human HCC cell line[51], adenosine diphosphate (ADP) and adenosine triphosphate (ATP) are degraded to adenosine and treatment of HCC cells with adenosine can inhibit growth of HCC cells and activate caspase-3, indicating that adenosine is an apoptotic agent with cytotoxicity. ARF is considered a tumor suppressor[52], and can initiate cellular response to aberrant oncogene activation by binding to and inhibiting the activity of MDM2. The human counterpart[53] of MDM2 (HDM2), like the murine protein, can inactivate the transactivation ability of human p53. ARF and p53 bind to MDM2 on different sites. ARF-p53 complex is formed depending on the mediation by MDM2 and can enhance p53 stability[54]. Blockage of p53 degradation pathways either by over-expression of ARF or interruption of MDM2-p53 interaction can effectively induce p53- triggered tumor cell death[14].

High expression of p53[55] in vivo may exert therapeutic effects on HCC in two aspects: (1) High expression of exogenous p53 protein induces apoptosis of tumor cells by inhibiting proliferation of cells through several biologic pathways and (2) exogenous p53 renders HCC more sensitive to some chemotherapeutic agents.

In addition, p53 down-regulates the expression of genes involved in angiogenesis[56], by the angiogenesis-inhibiting properties of wt-p53 protein[13].

It is very difficult to treat HCC due to drug resistance. It is necessary to reverse multiple drug resistance to human HCC cells and to find out the related mechanisms. The expression and activity of P-glycoprotein as well as the multiple drug resistance gene (MDR) products, are elevated in HCC cells with mt-p53[57]. Expression of mt-p53 enhances drug resistance to HCC cells and reduces their uptake of chemotherapeutic agents. In contrast to the increased expression of MDR by mt-p53, wt-p53 inhibits transcription of MDR[58]. Therefore, restoration of wt-p53 activity in HCC cells leads to the sensitivity of HCC cells to chemotherapeutic agents because of the decreased expression of P-glycoprotein encoded by MDR.

APPROACHES OF p53 THERAPY FOR HCC

Studies indicate that it is necessary to deliver therapeutic genes into cells with high specificity and efficiency in order to increase the effect of gene therapy against cancer[5,7,9,11]. A key factor for the success of gene therapy is the development of delivery systems[5,7,9] that are capable of efficiently transferring genes into a variety of tissues, without any associated pathogenic effects[7]. These techniques permit the isolation and insertion of genes into the recombinant delivery systems[5]. Two kinds of gene transfer vectors, namely viral and non-viral, are available at present.

Viruses are recognized natural gene carriers[5] and provide inspiration of gene therapy. Viruses have been engineered as gene delivery vectors[59]. However, their limits such as selective disadvantage[60], immunogenicity and toxicity[61], inefficient gene transfer and short-lived[62] or inadequate expression in transfected liver cells[63], require us to search other delivery systems.

Non-viral vectors have several advantages over viral vectors[61]. The transferred gene is in the form of a plasmid[64] that is on the surface or in the interior of the vector. Such vectors include liposome[61], molecular conjugates[65], nanoparticles[66] with strong anti-tumor effect on human HCC, naked DNA[67] and complexed DNA[68]. Better transfection efficiency can be achieved with delivery systems such as cationic lipids and cationic polymers[63].

To date, tissue-specific expression[69], self-replicating[70] and integrating plasmid[71] systems have been reported for gene therapy of HCC. Iodized oil emulsion[72] has a particular affinity to hypervascular hepatic tumors and is now commonly used in HCC chemoembolization, suggesting that it can be applied to the liver as a non-viral gene transfer system for intra-arterial gene delivery with selective gene expression in tumor cells.

THERAPEUTIC PRODUCTS WITH p53 STATUS AS TARGET

Recombinant adenovirus p53 (rAd-p53) and oncolytic virus are the promising therapeutic products for the treatment of HCC mostly applied in vitro and in vivo at present. With the introduction of exogenous wt-p53 expressed by the recombinant adenoviral vector, the expression of both p53 and p21 proteins is found to be up-regulated in cells[55]. Inhibition of cell growth and apoptosis can be achieved[55]. Oncolytic viruses[73,74] are a number of defective viruses, which cannot replicate in normal cells but are able to grow in tumor cells, finally leading to their lysis.

The two kinds of viruses and their characteristics were compared (Table 3).

Table 3 Comparison of several characteristics of rAd-p53 with oncolytic virus products.
ProductsCharacteristics
rAd-p53p53 gene is transfected into HCC cells with recombinant adenoviral vector expressing wt-p53
AdvexinLarger host range, low pathogenicity to human, replication-impaired adenoviral vector carrying the p53 gene
GendicineThe first commercial gene therapy product in the world approved by SFDA
SCH58500Replication-deficient type 5 adenovirus vector expressing human wt-p53 under control of cytomegalovirus promoter
Oncolytic virusesIncapable of replicating in normal cells but selectively replicating in p53-defective tumor cells to lyse them
ONYX-015Tumor-selective replicating virus, the prototype for oncolytic adenoviral therapy
CNHK300-mEReplication-competent with advantages of both gene and virus therapies
Recombinant adenovirus p53 (rAd-p53)

Since human adenovirus vector systems have a lager host range and lower pathogenicity to humans[75], and the binding affinity for epithelium[76] is important because most of human tumors are of the epithelial origin, and serotype 4 of species E[77,78] shows a specific binding affinity for HCC cells[76], they are generally used for the expression of proteins in human beings or other species with some advantages. The E1 region of adenovirus has been identified as a subregion of the viral genome present in transformed cells and is responsible for transformation[79]. Recombinant human adenoviruses constructed with the E1 region replaced by exogenous DNA become replication-defective and yield a relatively low degree of acute toxicity[80]. Since recombinant adenoviral vector expresses wt-p53 (Ad-p53), p53 gene can be transfected into HCC cell lines[55]. Experiments showed that tumor cells transduced with the wt-p53 gene can inhibit in vivo tumor growth of adjacent nontransduced cells, suggesting that Ad-p53 is also anti-angiogenic[81], partially by the bystander effect induced by the wt-p53 gene transfer on adjacent tumor cells.

Oncolytic viruses

As HCC cells with p53 defects have lost their cellular surveillance mechanisms, oncolytic viruses interfering with the main surveillance pathways such as those controlled by p53[73], could replicate selectively in them and cause lysis. E1A gene of adenovirus is an apoptosis-inducing gene and E1B gene of adenovirus is an apoptosis-inhibiting gene. The 55-kilodalton (55kDa) protein from the E1B-region of adenovirus binds to and inactivates the p53 gene[82]. Because of a deletion in E1B, the 55-kDa E1B protein is not expressed and the mutant adenovirus, termed ONYX-015[83], is able to replicate only in wt-p53 deficient cells. The E1B55K-defective adenovirus ONYX-015 is a prototype[73,82,83] of oncolytic viruses and can selectively replicate in and kill p53-deficient HCC cells, the success of cancer gene therapy is not promising unless it is carefully designed based on the biology of a specific[84] tumor type. To enhance the efficiency of such oncolytic viruses, another E1B 55kDa-deficient adenovirus armed with a mouse endostatin gene has been constructed for anti-tumor activities against HCC, and termed as CNHK200-mE[85]. With the synergistic effect of carrier virus and therapeutic gene, a novel approach has been established with the vector system termed as gene-viral vector[86] or gene-viral therapeutic system wherein an anti-tumor gene is inserted into the genome of a replicative virus specific for tumor cells to combine the advantages of gene and virus therapies. Using the human telomerase reverse transcriptase (hTERT) promoter to drive the expression of adenovirus E1A gene and clone the therapeutic gene mouse endostatin into the adenovirus genome, CNHK300-murine endostatin (CNHK300-mE)[87] is constructed, showing potential effects in the treatment of HCC.

EMPIRICAL STUDIES OF p53 THERAPY FOR HCC

Activation of p53 by either chemotherapy or radiotherapy induces p53-dependent apoptosis in tumor cells with wt-p53[14].

In the treatment of HCC, p53 products are injected into liver tissue by a variety of routes[55,72,82,88-92]. When p53 products are injected intratumorally[55,88,89], introduction of exogenous wt-p53 can inhibit cell growth, the expression of both p53 and p21 proteins is up-regulated in tumor cells. Intraarterial gene delivery into animal hepatic tumors can lead to selective gene expression in tumor cells[72]. Antegrade intraportal and retrograde intrabiliary routes are compared, induce transgene expression in periportal areas of liver with no significant difference in transduction efficacy, and no apparent complications are observed apart from very mild elevation of serum biochemical parameters[90]. The effect of bile and pancreatic juice on gene delivery has been studied under the guide of endoscopic retrograde cholangiopancreatography (ERCP)[92], showing that neither bile nor pancreatic juice affects transgene expression. Intrasplenic injection of p53 products can transfer gene into the portal venous circulation. When p53 products are injected intravenously, barriers such as the endothelial lining of tumor vasculature impair the efficiency of adenoviral vectors for gene delivery into HCCs[65,82], which can be overcome by direct injection of p53 products into tumor tissues[89]. Other gene delivery routes have also been reported[91].

High-volume hydrodynamic injection of a gene via the hepatic artery with inferior vena cava/portal vein occlusion can achieve a high level of gene expression in HCC rat model[67]. This gene transfer technique may have potential in clinical gene therapy for HCC. Oncolytic adenovirus-mediated gene therapy induces tumor-cell apoptosis and reduces tumor angiogenesis, leading to inhibition of HCC growth in animal model[88]. Because CNHK200-mE is capable of selectively replicating in HCC cells, thus suppressing tumor growth and antiangiogenic activity in nude mice[85], it can be used as a potential agent in the treatment of HCC.

CLINICAL RESEARCHES OF p53 THERAPY FOR HCC

After in vitro and in vivo experiments, different adenovirus-mediated p53 gene therapies for various tumors have been evaluated[46]. It was reported that when recombinant adenovirus p53 (SCH 58500) is administered by hepatic arterial infusion, and it distributes more predominantly in liver tissues than in tumors[93].

Intratumoural injection of Ad-p53 can lead to over-expression of p53 in cancer cells by inducing cell growth arrest and apoptosis, and overcome resistance or increase the effectiveness of radiation therapy and chemotherapy[94].

Hepatic artery embolization for the treatment of HCC was first reported in 1979[95]. Since the 1980s, transcatheter arterial chemo-embolization (TACE) has been applied to various HCCs apart from those with humoral hypercalcemia[96]. TACE as a local ablative treatment is able to control local disease and prolong a similar survival to that of surgical resection[11]. However, the recurrence of HCC after successful control of local tumor spread is high due to the non-curative procedure. In contrast to necrosis resulting from TACE, apoptosis is not commonly accompanied with inflammation that causes collateral cell damage, suggesting that the effects of intra-tumoral or intraarterial injection of p53 products in combination with TACE, on tumor tissue ischemia and necrosis, may be synergic and can improve survival.

In addition, inhibitors of p53-mediated apoptosis might be used to transiently decrease apoptosis in normal tissues when patients receive high doses of radiation or chemotherapy[97]. Emulsion of iodized oil and contrast medium can be used as a nonviral gene transfer system for intraarterial gene delivery[72].

CHALLENGES AND PROSPECTS

p53 is an ideal target for anti-cancer drug design[14]. Blockage of p53 degradation pathways can effectively induce p53 triggered tumor cell death[14]. Since unlike most other tumor suppressor genes, mt-p53 is over-expressed in tumor cells, a promising approach involving reactivation of tumor-suppressing function to mt-p53[21,25]. Further understanding of the mechanisms of how to restore p53 activity, may lead to discovery of more potent analogs and new strategies[11,73,91] for p53-targeting in tumor therapy.

More genes previously unknown have been identified that are involved in the regulation of p53 transcriptional activity and their over-expression inhibits p53 target promoters and p53-mediated apoptosis, suggesting that these genes play a role as p53 inhibitors and may have oncogenic activity[98].

As hepatocarcinogenesis is a multistage process involving a number of genes[99,100], attention should be paid to the target genes whose altered expression actually mediates neoplastic phenotype. There is an urgent need to establish simple and low-cost tests for detecting expression of p53-related genes in HCC[99,100] that are hallmarks of HCC development. Continuous monitoring of serum p53 protein is important in early detection of recurrence of HCC, and immunodetection of serum p53 is valuable[101] for post-operative monitoring during follow-up in preoperatively positive patients.

Nevertheless, about 40% of cancers retain wt-p53, and there may be mutations of other genes in cells with p53 mutations. Therefore, the mechanisms of p53 in HCC therapies should be further studied.

It was reported that hepatic arterial administration of p53 products cannot substantially increase transduction of tumor cells, and ligation of the hepatic artery following infusion of adenovirus or addition of lipiodol infusion has no effect on the transduction of tumor cells[102], suggesting that better administration approaches must be developed for more efficient transduction of tumor cells.

Safety and research ethics must be emphasized. Human gene therapy can lead to serious adverse effects and even death[103]. In a gene therapy experiment in 1999[104], death of Jesse Gelsinger was found to be directly linked to the viral vector used for the treatment. This tragic event has raised new questions about the prospects for human gene therapy, which not only achieves a therapeutic effect but also has potential adverse effects.

Another task at present is to find out the key points of tumor resistance to rAd-p53[105-108]. Since exogenous wt-p53 was introduced, inhibition of tumor growth has become unnecessary for human cancer cells carrying mt-p53[105]. In some cancer cells, wt-p53 is inactivated by different mechanisms. Since the presence of mt-p53 may induce genome instability of human cancer cells and mutator ability, they can escape the effects of exogenous wt-p53 and contribute to the failure of wt-p53 gene therapy. On the contrary, exogenous wt-p53 in other cancers can have effects only on those expressing wt-p53. For example, alterations of p53 gene are uncommon in differentiated thyroid neoplasm but can be frequently detected in anaplastic thyroid carcinoma[107], suggesting that impaired p53 function may contribute to the undifferentiated and highly aggressive phenotype of these tumors. It was reported that exogenous expression of wt-p53 has influence on thyroid tumorigenic properties only in cells bearing altered p53, whereas it has no effect on cells expressing wt-p53 activity[106], indicating that the endogenous p53 status seems to be essential for the effectiveness of p53-based gene therapy for some cancers. Another study[108] evaluated the therapeutic effects of truncated Bid gene (tBid) on p53-resistent HCC and demonstrated that this gene only targets AFP-producing cells but not non-AFP producing ones[109]. The introduction of tBid can not only significantly but also specifically kill HCC cells that produce AFP, indicating that death of HCC cells is induced by an apoptotic pathway independent of p53 status[108].

CONCLUSION

p53 is an ideal target for the design of anti-cancer therapeutic strategies. p53 is frequently mutated in a significant portion of HCCs, and can mediate defective apoptotic pathways in HCC carrying wild type of p53. Several approaches have been designed for the treatment of HCC via the p53 pathway by restoring tumor suppression function from inactivation, rescuing mutated p53 gene from instability, or delivering therapeutic exogenous p53. Products with p53 status as the target have been studied extensively in vitro and in vivo. Although their therapeutic effects are limited, further study is needed to elucidate the mechanisms of p53 in HCC therapies, the role of endogenous p53 status, novel genes involved in the regulation of p53 transcriptional activity, the establishment of simple and low-cost tests for detecting expression of p53-related genes in HCC, better administration approaches of exogenous p53, safety and research ethics, and the key points of resistance to rAd-p53 in HCC.

COMMENTS
Background

Hepatocellular carcinoma (HCC) is one of the 10 most common cancers worldwide. There are no ideal therapies for advanced HCC so far, and many researchers are trying to improve the effects of treatment by changing therapeutic strategies including application of gene therapy.

Research frontiers

About 50% of human cancers are associated with mutations in the core domain of tumor suppressor p53, thus p53 is regarded as the most frequently mutated gene. It is an attractive approach to the treatment of HCC by restoring wild type p53 activity.

Innovations and breakthroughs

Several approaches have been designed for the treatment of HCC via the p53 pathway by restoring tumor suppression function from inactivation, rescuing the mutated p53 gene from instability, or delivering therapeutic exogenous p53. Products with p53 status as the target have been studied extensively in vitro and in vivo.

Applications

p53 pathway can be used as a target in the treatment of HCC.

Terminology

Gene Therapy: The treatment of certain disorders, especially those caused by genetic anomalies or deficiencies, by introducing specifically engineered genes into patient cells. p53 gene: It is a tumor suppressor gene providing instructions for making a protein called tumor protein 53 (TP53). Through the effect of the protein it produces, TP53 is a tumor suppressor gene regulating the cycle of cell division by protecting cells from growing and dividing too fast or in an uncontrolled way. The p53 protein is located in the nuclei of cells throughout the body and can bind directly to DNA. When the DNA in cells becomes damaged, this protein plays a critical role in determining whether the DNA is repaired or the cells undergo programmed cell death (apoptosis). If the DNA can be repaired, p53 activates other genes to repair the damage. If the DNA cannot be repaired, the p53 tumor protein protects cells from dividing and signals it to undergo apoptosis. This process protects cells with mutated or damaged DNA from dividing, which helps prevent the development of tumors. Because the p53 tumor protein is essential for regulating cell division, it has been nicknamed the “guardian of the genome. Vector: Any device of transportation or movement. In this article, it denotes “a virus used to deliver genetic material into cells” or “a piece of DNA carrying DNA fragments into host cells”.

Peer review

This review elaborates some therapeutic mechanisms and advances in using recombinant human adenovirus p53 and oncolytic virus products in the treatment of HCC. This review is helpful for many readers. The prospects should be expanded.

Footnotes

S- Editor Liu Y L- Editor Wang XL E- Editor Lu W

References
1.  Editorial: Playing with genes Br Med J. 1976;1:302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
2.  Stetten D. What men fear. Perspect Biol Med. 1978;21:515-523.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Friedmann T. Progress toward human gene therapy. Science. 1989;244:1275-1281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 400]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
4.  Friedmann T. The evolving concept of gene therapy. Hum Gene Ther. 1990;1:175-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
5.  Gonçalves MA. A concise peer into the background, initial thoughts and practices of human gene therapy. Bioessays. 2005;27:506-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
6.  Wolff JA, Lederberg J. An early history of gene transfer and therapy. Hum Gene Ther. 1994;5:469-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 89]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
7.  Verma IM, Weitzman MD. Gene therapy: twenty-first century medicine. Annu Rev Biochem. 2005;74:711-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 440]  [Cited by in F6Publishing: 455]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
8.  Blaese RM, Culver KW, Miller AD, Carter CS, Fleisher T, Clerici M, Shearer G, Chang L, Chiang Y, Tolstoshev P. T lymphocyte-directed gene therapy for ADA- SCID: initial trial results after 4 years. Science. 1995;270:475-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1074]  [Cited by in F6Publishing: 893]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
9.  Park TG, Jeong JH, Kim SW. Current status of polymeric gene delivery systems. Adv Drug Deliv Rev. 2006;58:467-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 782]  [Cited by in F6Publishing: 745]  [Article Influence: 41.4]  [Reference Citation Analysis (0)]
10.  Seeff LB, Hoofnagle JH. Epidemiology of hepatocellular carcinoma in areas of low hepatitis B and hepatitis C endemicity. Oncogene. 2006;25:3771-3777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 114]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
11.  Müller C. Hepatocellular carcinoma--rising incidence, changing therapeutic strategies. Wien Med Wochenschr. 2006;156:404-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
12.  Llovet JM. Updated treatment approach to hepatocellular carcinoma. J Gastroenterol. 2005;40:225-235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 388]  [Cited by in F6Publishing: 383]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
13.  Cheah PL, Looi LM. p53: an overview of over two decades of study. Malays J Pathol. 2001;23:9-16.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Wang W, Rastinejad F, El-Deiry WS. Restoring p53-dependent tumor suppression. Cancer Biol Ther. 2003;2:S55-S63.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Haupt S, Haupt Y. Importance of p53 for cancer onset and therapy. Anticancer Drugs. 2006;17:725-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
16.  Peng Z. Current status of gendicine in China: recombinant human Ad-p53 agent for treatment of cancers. Hum Gene Ther. 2005;16:1016-1027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 258]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
17.  Ries SJ. Elucidation of the molecular mechanism underlying tumor-selective replication of the oncolytic adenovirus mutant ONYX-015. Future Oncol. 2005;1:763-766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
18.  Levine AJ. p53, the cellular gatekeeper for growth and division. Cell. 1997;88:323-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5482]  [Cited by in F6Publishing: 5379]  [Article Influence: 199.2]  [Reference Citation Analysis (0)]
19.  Bálint E E, Vousden KH. Activation and activities of the p53 tumour suppressor protein. Br J Cancer. 2001;85:1813-1823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 208]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
20.  Selivanova G, Wiman KG. p53: a cell cycle regulator activated by DNA damage. Adv Cancer Res. 1995;66:143-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 82]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
21.  Bykov VJ, Wiman KG. Novel cancer therapy by reactivation of the p53 apoptosis pathway. Ann Med. 2003;35:458-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 33]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
22.  Hagiwara N, Mechanic LE, Trivers GE, Cawley HL, Taga M, Bowman ED, Kumamoto K, He P, Bernard M, Doja S. Quantitative detection of p53 mutations in plasma DNA from tobacco smokers. Cancer Res. 2006;66:8309-8317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
23.  Laurent-Puig P, Zucman-Rossi J. Genetics of hepatocellular tumors. Oncogene. 2006;25:3778-3786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 276]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
24.  Friedler A, DeDecker BS, Freund SM, Blair C, Rüdiger S, Fersht AR. Structural distortion of p53 by the mutation R249S and its rescue by a designed peptide: implications for "mutant conformation". J Mol Biol. 2004;336:187-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 50]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
25.  Bullock AN, Henckel J, DeDecker BS, Johnson CM, Nikolova PV, Proctor MR, Lane DP, Fersht AR. Thermodynamic stability of wild-type and mutant p53 core domain. Proc Natl Acad Sci USA. 1997;94:14338-14342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 330]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
26.  El Far MA, Atwa MA, Yahya RS, El Basuni MA. Evaluation of serum levels of p53 in hepatocellular carcinoma in Egypt. Clin Chem Lab Med. 2006;44:653-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
27.  Farazi PA, Glickman J, Horner J, Depinho RA. Cooperative interactions of p53 mutation, telomere dysfunction, and chronic liver damage in hepatocellular carcinoma progression. Cancer Res. 2006;66:4766-4773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
28.  Bressac B, Galvin KM, Liang TJ, Isselbacher KJ, Wands JR, Ozturk M. Abnormal structure and expression of p53 gene in human hepatocellular carcinoma. Proc Natl Acad Sci USA. 1990;87:1973-1977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 302]  [Cited by in F6Publishing: 321]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
29.  Nagao M, Nakajima Y, Hisanaga M, Kayagaki N, Kanehiro H, Aomatsu Y, Ko S, Yagita H, Yamada T, Okumura K. The alteration of Fas receptor and ligand system in hepatocellular carcinomas: how do hepatoma cells escape from the host immune surveillance in vivo? Hepatology. 1999;30:413-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 82]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
30.  Jeng YM, Peng SY, Lin CY, Hsu HC. Overexpression and amplification of Aurora-A in hepatocellular carcinoma. Clin Cancer Res. 2004;10:2065-2071.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 246]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
31.  Hirohashi S. Pathology and molecular mechanisms of multistage human hepatocarcinogenesis. Princess Takamatsu Symp. 1991;22:87-93.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Kondo M, Marusawa H, Ueda Y, Katsurada A, Kawasome C, Takami S, Kinoshita M, Ikai I, Yamaoka Y, Chiba T. Diverse p53 gene aberration in hepatocellular carcinoma detected by dual-color fluorescence in situ hybridization. J Gastroenterol Hepatol. 2004;19:1066-1073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
33.  Dewantoro O, Gani RA, Akbar N. Hepatocarcinogenesis in viral Hepatitis B infection: the role of HBx and p53. Acta Med Indones. 2006;38:154-159.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Cho JW, Park K, Kweon GR, Park JC, Lee JC, Baek WK, Jang BC, Suh SI, Suh MH. Modulation of cell death sensitivity by mutant p53 in HCV core-expressing cells. Int J Mol Med. 2005;15:475-480.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Wu CG, Salvay DM, Forgues M, Valerie K, Farnsworth J, Markin RS, Wang XW. Distinctive gene expression profiles associated with Hepatitis B virus x protein. Oncogene. 2001;20:3674-3682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 72]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
36.  Mathonnet G, Lachance S, Alaoui-Jamali M, Drobetsky EA. Expression of hepatitis B virus X oncoprotein inhibits transcription-coupled nucleotide excision repair in human cells. Mutat Res. 2004;554:305-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
37.  Elmore LW, Hancock AR, Chang SF, Wang XW, Chang S, Callahan CP, Geller DA, Will H, Harris CC. Hepatitis B virus X protein and p53 tumor suppressor interactions in the modulation of apoptosis. Proc Natl Acad Sci USA. 1997;94:14707-14712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 266]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
38.  Cougot D, Neuveut C, Buendia MA. HBV induced carcinogenesis. J Clin Virol. 2005;34 Suppl 1:S75-S78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 128]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
39.  Zhang X, Dong N, Yin L, Cai N, Ma H, You J, Zhang H, Wang H, He R, Ye L. Hepatitis B virus X protein upregulates survivin expression in hepatoma tissues. J Med Virol. 2005;77:374-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 70]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
40.  Kannangai R, Wang J, Liu QZ, Sahin F, Torbenson M. Survivin overexpression in hepatocellular carcinoma is associated with p53 dysregulation. Int J Gastrointest Cancer. 2005;35:53-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
41.  Lacabanne V, Viguier M, Guillet JG, Choppin J. A wild-type p53 cytotoxic T cell epitope is presented by mouse hepatocarcinoma cells. Eur J Immunol. 1996;26:2635-2639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
42.  Eipel C, Schuett H, Glawe C, Bordel R, Menger MD, Vollmar B. Pifithrin-alpha induced p53 inhibition does not affect liver regeneration after partial hepatectomy in mice. J Hepatol. 2005;43:829-835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
43.  Lutz W, Nowakowska-Swirta E. Gene p53 mutations, protein p53, and anti-p53 antibodies as biomarkers of cancer process. Int J Occup Med Environ Health. 2002;15:209-218.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Müller M, Meyer M, Schilling T, Ulsperger E, Lehnert T, Zentgraf H, Stremmel W, Volkmann M, Galle PR. Testing for anti-p53 antibodies increases the diagnostic sensitivity of conventional tumor markers. Int J Oncol. 2006;29:973-980.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Gurova KV, Hill JE, Guo C, Prokvolit A, Burdelya LG, Samoylova E, Khodyakova AV, Ganapathi R, Ganapathi M, Tararova ND. Small molecules that reactivate p53 in renal cell carcinoma reveal a NF-kappaB-dependent mechanism of p53 suppression in tumors. Proc Natl Acad Sci USA. 2005;102:17448-17453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 219]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
46.  Horowitz J. Adenovirus-mediated p53 gene therapy: overview of preclinical studies and potential clinical applications. Curr Opin Mol Ther. 1999;1:500-509.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Momand J, Zambetti GP, Olson DC, George D, Levine AJ. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992;69:1237-1245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2174]  [Cited by in F6Publishing: 2266]  [Article Influence: 70.8]  [Reference Citation Analysis (0)]
48.  Wu X, Bayle JH, Olson D, Levine AJ. The p53-mdm-2 autoregulatory feedback loop. Genes Dev. 1993;7:1126-1132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1257]  [Cited by in F6Publishing: 1299]  [Article Influence: 41.9]  [Reference Citation Analysis (0)]
49.  Zhang H. MDM2 oncogene as a novel target for human cancer therapy. Curr Pharm Des. 2000;6:393-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 83]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
50.  Stearns T, Willingham MC, Botstein D, Kahn RA. ADP-ribosylation factor is functionally and physically associated with the Golgi complex. Proc Natl Acad Sci USA. 1990;87:1238-1242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 319]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
51.  Wen LT, Knowles AF. Extracellular ATP and adenosine induce cell apoptosis of human hepatoma Li-7A cells via the A3 adenosine receptor. Br J Pharmacol. 2003;140:1009-1018.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 47]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
52.  Rocha S, Perkins ND. ARF the integrator: linking NF-kappaB, p53 and checkpoint kinases. Cell Cycle. 2005;4:756-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
53.  Brown DR, Deb S, Muñoz RM, Subler MA, Deb SP. The tumor suppressor p53 and the oncoprotein simian virus 40 T antigen bind to overlapping domains on the MDM2 protein. Mol Cell Biol. 1993;13:6849-6857.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Lin J, Zhu MH. Interactive pathway of ARF-mdm2-p53. Ai Zheng. 2003;22:328-330.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Guo Y, Zeng Y, Wang K, Zhu X, Luo H, Zheng M, Li M, Chen J. Therapeutic potential of recombinant adenovirus expressing p53 in hepatocellular carcinoma cell lines. Zhonghua GanZangBing ZaZhi. 2001;9 Suppl:43-45.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Oliveira AM, Ross JS, Fletcher JA. Tumor suppressor genes in breast cancer: the gatekeepers and the caretakers. Am J Clin Pathol. 2005;124 Suppl:S16-S28.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Chan KT, Lung ML. Mutant p53 expression enhances drug resistance in a hepatocellular carcinoma cell line. Cancer Chemother Pharmacol. 2004;53:519-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 59]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
58.  Li YX, Lin ZB, Tan HR. Wild type p53 increased chemosensitivity of drug-resistant human hepatocellular carcinoma Bel7402/5-FU cells. Acta Pharmacol Sin. 2004;25:76-82.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Zhang X, Godbey WT. Viral vectors for gene delivery in tissue engineering. Adv Drug Deliv Rev. 2006;58:515-534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 137]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
60.  Büning H, Ried MU, Perabo L, Gerner FM, Huttner NA, Enssle J, Hallek M. Receptor targeting of adeno-associated virus vectors. Gene Ther. 2003;10:1142-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 89]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
61.  Miyazaki M, Obata Y, Abe K, Furusu A, Koji T, Tabata Y, Kohno S. Gene transfer using nonviral delivery systems. Perit Dial Int. 2006;26:633-640.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Palmer DH, Mautner V, Mirza D, Oliff S, Gerritsen W, van der Sijp JR, Hubscher S, Reynolds G, Bonney S, Rajaratnam R. Virus-directed enzyme prodrug therapy: intratumoral administration of a replication-deficient adenovirus encoding nitroreductase to patients with resectable liver cancer. J Clin Oncol. 2004;22:1546-1552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 94]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
63.  Kodama K, Katayama Y, Shoji Y, Nakashima H. The features and shortcomings for gene delivery of current non-viral carriers. Curr Med Chem. 2006;13:2155-2161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 68]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
64.  Noireaux V, Libchaber A. A vesicle bioreactor as a step toward an artificial cell assembly. Proc Natl Acad Sci USA. 2004;101:17669-17674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 889]  [Cited by in F6Publishing: 797]  [Article Influence: 39.9]  [Reference Citation Analysis (0)]
65.  Kim JW, Lee HS. Tumor targeting by doxorubicin-RGD-4C peptide conjugate in an orthotopic mouse hepatoma model. Int J Mol Med. 2004;14:529-535.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Barraud L, Merle P, Soma E, Lefrançois L, Guerret S, Chevallier M, Dubernet C, Couvreur P, Trépo C, Vitvitski L. Increase of doxorubicin sensitivity by doxorubicin-loading into nanoparticles for hepatocellular carcinoma cells in vitro and in vivo. J Hepatol. 2005;42:736-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 165]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
67.  Tada M, Hatano E, Taura K, Nitta T, Koizumi N, Ikai I, Shimahara Y. High volume hydrodynamic injection of plasmid DNA via the hepatic artery results in a high level of gene expression in rat hepatocellular carcinoma induced by diethylnitrosamine. J Gene Med. 2006;8:1018-1026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
68.  Meyer F, Ball V, Schaaf P, Voegel JC, Ogier J. Polyplex-embedding in polyelectrolyte multilayers for gene delivery. Biochim Biophys Acta. 2006;1758:419-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
69.  Uto H, Ido A, Hori T, Hirono S, Hayashi K, Tamaoki T, Tsubouchi H. Hepatoma-specific gene therapy through retrovirus-mediated and targeted gene transfer using an adenovirus carrying the ecotropic receptor gene. Biochem Biophys Res Commun. 1999;265:550-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
70.  Min KA, Lee SK, Kim CK. Improved gene expression pattern using Epstein-Barr virus (EBV)-based plasmid and cationic emulsion. Biomaterials. 2005;26:1063-1070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
71.  Wilson GM, Deeley RG. An episomal expression vector system for monitoring sequence-specific effects on mRNA stability in human cell lines. Plasmid. 1995;33:198-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
72.  Kim YI, Chung JW, Park JH, Han JK, Hong JW, Chung H. Intraarterial gene delivery in rabbit hepatic tumors: transfection with nonviral vector by using iodized oil emulsion. Radiology. 2006;240:771-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
73.  Cherubini G, Petouchoff T, Grossi M, Piersanti S, Cundari E, Saggio I. E1B55K-deleted adenovirus (ONYX-015) overrides G1/S and G2/M checkpoints and causes mitotic catastrophe and endoreduplication in p53-proficient normal cells. Cell Cycle. 2006;5:2244-2252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
74.  O'Shea CC, Johnson L, Bagus B, Choi S, Nicholas C, Shen A, Boyle L, Pandey K, Soria C, Kunich J. Late viral RNA export, rather than p53 inactivation, determines ONYX-015 tumor selectivity. Cancer Cell. 2004;6:611-623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 274]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
75.  Gonçalves MA, de Vries AA. Adenovirus: from foe to friend. Rev Med Virol. 2006;16:167-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 75]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
76.  Zhang LQ, Mei YF, Wadell G. Human adenovirus serotypes 4 and 11 show higher binding affinity and infectivity for endothelial and carcinoma cell lines than serotype 5. J Gen Virol. 2003;84:687-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
77.  Ebner K, Rauch M, Preuner S, Lion T. Typing of human adenoviruses in specimens from immunosuppressed patients by PCR-fragment length analysis and real-time quantitative PCR. J Clin Microbiol. 2006;44:2808-2815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
78.  Jacobs SC, Davison AJ, Carr S, Bennett AM, Phillpotts R, Wilkinson GW. Characterization and manipulation of the human adenovirus 4 genome. J Gen Virol. 2004;85:3361-3366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
79.  Bos JL, Jochemsen AG, Bernards R, Schrier PI, van Ormondt H, van der Eb AJ. Deletion mutants of region E1a of AD12 E1 plasmids: effect on oncogenic transformation. Virology. 1983;129:393-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 28]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
80.  Wills KN, Maneval DC, Menzel P, Harris MP, Sutjipto S, Vaillancourt MT, Huang WM, Johnson DE, Anderson SC, Wen SF. Development and characterization of recombinant adenoviruses encoding human p53 for gene therapy of cancer. Hum Gene Ther. 1994;5:1079-1088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 177]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
81.  Nishizaki M, Fujiwara T, Tanida T, Hizuta A, Nishimori H, Tokino T, Nakamura Y, Bouvet M, Roth JA, Tanaka N. Recombinant adenovirus expressing wild-type p53 is antiangiogenic: a proposed mechanism for bystander effect. Clin Cancer Res. 1999;5:1015-1023.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Heise C, Sampson-Johannes A, Williams A, McCormick F, Von Hoff DD, Kirn DH. ONYX-015, an E1B gene-attenuated adenovirus, causes tumor-specific cytolysis and antitumoral efficacy that can be augmented by standard chemotherapeutic agents. Nat Med. 1997;3:639-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 663]  [Cited by in F6Publishing: 622]  [Article Influence: 23.0]  [Reference Citation Analysis (1)]
83.  Rothmann T, Hengstermann A, Whitaker NJ, Scheffner M, zur Hausen H. Replication of ONYX-015, a potential anticancer adenovirus, is independent of p53 status in tumor cells. J Virol. 1998;72:9470-9478.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Stoff-Khalili MA, Dall P, Curiel DT. Gene therapy for carcinoma of the breast. Cancer Gene Ther. 2006;13:633-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
85.  Li G, Sham J, Yang J, Su C, Xue H, Chua D, Sun L, Zhang Q, Cui Z, Wu M. Potent antitumor efficacy of an E1B 55kDa-deficient adenovirus carrying murine endostatin in hepatocellular carcinoma. Int J Cancer. 2005;113:640-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 27]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
86.  Qian Q, Sham J, Che X, Xu J, Xue H, Cui Z, Zhu B, Wu M. Gene-viral vectors: a promising way to target tumor cells and express anticancer genes simultaneously. Chin Med J (Engl). 2002;115:1213-1217.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Li GC, Yang JM, Nie MM, Su CG, Sun LC, Qian YZ, Fang GE, Sham J, Wu MC, Qian QJ. Potent antitumoral effects of a novel gene-viral therapeutic system CNHK300-mEndostatin in hepatocellular carcinoma. Chin Med J (Engl). 2005;118:179-185.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Ye Z, Wang X, Hao S, Zhong J, Xiang J, Yang J. Oncolytic adenovirus-mediated E1A gene therapy induces tumor-cell apoptosis and reduces tumor angiogenesis leading to inhibition of hepatocellular carcinoma growth in animal model. Cancer Biother Radiopharm. 2006;21:225-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
89.  Yoon SK, Armentano D, Wands JR, Mohr L. Adenovirus-mediated gene transfer to orthotopic hepatocellular carcinomas in athymic nude mice. Cancer Gene Ther. 2001;8:573-579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
90.  Kuriyama S, Yoshiji H, Nakai S, Deguchi A, Uchida N, Kimura Y, Inoue H, Kinekawa F, Ogawa M, Nonomura T. Adenovirus-mediated gene transfer into rat livers: comparative study of retrograde intrabiliary and antegrade intraportal administration. Oncol Rep. 2005;13:69-74.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  White SA, LoBuglio AF, Arani RB, Pike MJ, Moore SE, Barlow DL, Conry RM. Induction of anti-tumor immunity by intrasplenic administration of a carcinoembryonic antigen DNA vaccine. J Gene Med. 2000;2:135-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
92.  Xie X, Forsmark CE, Lau JY. Effect of bile and pancreatic juice on adenoviral-mediated gene delivery: implications on the feasibility of gene delivery through ERCP. Dig Dis Sci. 2000;45:230-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
93.  Atencio IA, Grace M, Bordens R, Fritz M, Horowitz JA, Hutchins B, Indelicato S, Jacobs S, Kolz K, Maneval D. Biological activities of a recombinant adenovirus p53 (SCH 58500) administered by hepatic arterial infusion in a Phase 1 colorectal cancer trial. Cancer Gene Ther. 2006;13:169-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
94.  Roth JA. Adenovirus p53 gene therapy. Expert Opin Biol Ther. 2006;6:55-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 100]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
95.  Roche A, Franco D, Dhumeaux D, Bismuth H, Doyon D. Emergency hepatic arterial embolization for secondary hypercalcemia in hepatocellular carcinoma. Radiology. 1979;133:315-316.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Suzuki K, Kono N, Ono A, Osuga Y, Kiyokawa H, Mineo I, Matsuda Y, Miyoshi S, Kawata S, Minami Y. Transcatheter arterial chemo-embolization for humoral hypercalcemia of hepatocellular carcinoma. Gastroenterol Jpn. 1988;23:29-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
97.  Strom E, Sathe S, Komarov PG, Chernova OB, Pavlovska I, Shyshynova I, Bosykh DA, Burdelya LG, Macklis RM, Skaliter R. Small-molecule inhibitor of p53 binding to mitochondria protects mice from gamma radiation. Nat Chem Biol. 2006;2:474-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 270]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
98.  Llanos S, Efeyan A, Monsech J, Dominguez O, Serrano M. A high-throughput loss-of-function screening identifies novel p53 regulators. Cell Cycle. 2006;5:1880-1885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 41]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
99.  Feitelson MA, Pan J, Lian Z. Early molecular and genetic determinants of primary liver malignancy. Surg Clin North Am. 2004;84:339-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
100.  Zhu MH, Ni CR, Zhu Z, Li FM, Zhang SM. Determination of expression of eight p53-related genes in hepatocellular carcinoma with tissue microarrays. Ai Zheng. 2003;22:680-685.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Attallah AM, Abdel-Aziz MM, El-Sayed AM, Tabll AA. Detection of serum p53 protein in patients with different gastrointestinal cancers. Cancer Detect Prev. 2003;27:127-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 28]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
102.  Maron DJ, Tada H, Moscioni AD, Tazelaar J, Fraker DL, Wilson JM, Spitz FR. Intra-arterial delivery of a recombinant adenovirus does not increase gene transfer to tumor cells in a rat model of metastatic colorectal carcinoma. Mol Ther. 2001;4:29-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
103.  Smith L, Byers JF. Gene therapy in the post-Gelsinger era. JONAS Healthc Law Ethics Regul. 2002;4:104-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
104.  Walters J. Why did Jesse die? Update. 2001;17:E1.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Vinyals A, Peinado MA, Gonzalez-Garrigues M, Monzó M, Bonfil RD, Fabra A. Failure of wild-type p53 gene therapy in human cancer cells expressing a mutant p53 protein. Gene Ther. 1999;6:22-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
106.  Moretti F, Nanni S, Farsetti A, Narducci M, Crescenzi M, Giuliacci S, Sacchi A, Pontecorvi A. Effects of exogenous p53 transduction in thyroid tumor cells with different p53 status. J Clin Endocrinol Metab. 2000;85:302-308.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Moretti F, Farsetti A, Soddu S, Misiti S, Crescenzi M, Filetti S, Andreoli M, Sacchi A, Pontecorvi A. p53 re-expression inhibits proliferation and restores differentiation of human thyroid anaplastic carcinoma cells. Oncogene. 1997;14:729-740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 83]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
108.  Miao J, Chen GG, Chun SY, Yun JP, Chak EC, Ho RL, Lai PB. Adenovirus-mediated tBid overexpression results in therapeutic effects on p53-resistant hepatocellular carcinoma. Int J Cancer. 2006;119:1985-1993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
109.  Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell. 1998;94:481-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2665]  [Cited by in F6Publishing: 2611]  [Article Influence: 100.4]  [Reference Citation Analysis (0)]