Next Article in Journal
Preclinical Models in Prostate Cancer: Resistance to AR Targeting Therapies in Prostate Cancer
Next Article in Special Issue
Breast Cancer Patient Prognosis Is Determined by the Interplay between TP53 Mutation and Alternative Transcript Expression: Insights from TP53 Long Amplicon Digital PCR Assays
Previous Article in Journal
A MYC-Driven Plasma Polyamine Signature for Early Detection of Ovarian Cancer
Previous Article in Special Issue
TAp73β Can Promote Hepatocellular Carcinoma Dedifferentiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Isoforms of the p53 Family and Gastric Cancer: A Ménage à Trois for an Unfinished Affair

1
Laboratory “Streinth” (STress REsponse and INnovative Therapies in Cancer), Inserm UMR_S 1113, IRFAC, ITI InnoVec, Université de Strasbourg, 67200 Strasbourg, France
2
ICANS (Institut du Cancer Strasbourg Europe), 67200 Strasbourg, France
*
Author to whom correspondence should be addressed.
Submission received: 31 December 2020 / Revised: 6 February 2021 / Accepted: 17 February 2021 / Published: 22 February 2021

Abstract

:

Simple Summary

The p53 family is a complex family of transcription factors with different cellular functions that are involved in several physiological processes. A massive amount of data has been accumulated on their critical role in the tumorigenesis and the aggressiveness of cancers of different origins. If common features are observed, there are numerous specificities that may reflect particularities of the tissues from which the cancers originated. In this regard, gastric cancer tumorigenesis is rather remarkable, as it is induced by bacterial and viral infections, various chemical carcinogens, and familial genetic alterations, which provide an example of the variety of molecular mechanisms responsible for cell transformation and how they impact the p53 family. This review summarizes the knowledge gathered from over 40 years of research on the role of the p53 family in gastric cancer, which still displays one of the most elevated mortality rates amongst all types of cancers.

Abstract

Gastric cancer is one of the most aggressive cancers, with a median survival of 12 months. This illustrates its complexity and the lack of therapeutic options, such as personalized therapy, because predictive markers do not exist. Thus, gastric cancer remains mostly treated with cytotoxic chemotherapies. In addition, less than 20% of patients respond to immunotherapy. TP53 mutations are particularly frequent in gastric cancer (±50% and up to 70% in metastatic) and are considered an early event in the tumorigenic process. Alterations in the expression of other members of the p53 family, i.e., p63 and p73, have also been described. In this context, the role of the members of the p53 family and their isoforms have been investigated over the years, resulting in conflicting data. For instance, whether mutations of TP53 or the dysregulation of its homologs may represent biomarkers for aggressivity or response to therapy still remains a matter of debate. This uncertainty illustrates the lack of information on the molecular pathways involving the p53 family in gastric cancer. In this review, we summarize and discuss the most relevant molecular and clinical data on the role of the p53 family in gastric cancer and enumerate potential therapeutic innovative strategies.

1. p53 and the Original Sin

The p53 protein was originally discovered in 1979 as a cellular binding partner of the Large T antigen of the Simian Virus 40 [1,2]. It was soon shown to be a phosphorylated protein [3,4] more highly expressed in cancer cells than in normal cells [5], characterized by a low stability that was controlled by posttranslational processes [6], and able to bind DNA [7]. At the cellular level, the p53 protein was found to be capable of inhibiting cell growth [8]. Later on, the TP53 gene that encodes the p53 protein was established as a tumor suppressor gene and was found to be the most commonly mutated gene in cancers, with approximately 50% penetrance [9,10]. About 20 years later, two paralogs of TP53, TP63, and TP73 were discovered. The successive investigations performed on p53 mutations in gastric cancer (GC) over the years is an archetype of the research done to combat cancer that have been paved with successes, defeats, and confrontative theories.

2. Gastric Cancer, a Disregarded Health Issue in Most of the Western Countries

Gastric cancer (GC) is the fifth most common cancer in the world, mostly affecting men (sex ratio of 2.5). Its mortality remains strikingly high compared to other types of cancers (third in the world and 800,000 deaths in 2018), with a significant geographic disparity between Asia and Europe [11,12]. For instance, in Europe, the five-year survival rate is less than 25% in all stages combined, and the median survival is about 11 months. The GC incidence is higher in Asia (32 per 100,000 among males) than in Western countries (5.6 per 100,000), probably due to food habits and genetic differences. Hence, research in GC is more developed in Asia in contrast to most Western countries where less effort and resources are invested in research and treatment, especially by the pharmaceutical companies. For instance, in Japan, 70% of localized gastric cancers are cured in the setting of early diagnosis owing to a systematic screening policy. Risk factors include bacterial (Helicobacter pylori) and viral (Epstein-Barr virus, EBV) agents, as well as rare hereditary mutations in the CDH1 gene or in Mismatch Repair genes involved in the hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndrome [13]. In addition, lifestyle factors like food preferences (e.g., salty or smoked foods), tobacco, and alcohol are important risk factors accounting for the presence of synchronous cancers in the stomach and oral cavity [14]. Inversely, the consumption of vegetables (e.g., carrots, lettuce) reduces the risk of GC [15,16].
There are two major histological GC subtypes representing 90% of cases: intestinal and diffuse (e.g., signet ring cell type) cancers. The intestinal subtype presents a cellular organization of the cancer cells similar to intestinal epithelial glands (Figure 1). The diffuse subtype is characterized by the dispersion of cancer cells within the stroma [17]. More recently, a molecular classification has been established identifying four molecular groups. [18]. The most important subgroup, matching mostly the intestinal subtype, is “Chromosomal Instability” (CIN, 49%), characterized by TP53 mutations and activation of the RTK-RAS pathway. The “Epstein-Barr virus” subgroup (EBV, 9%) is associated with DNA hypermethylation, frequent PIK3CA mutation, CDKN2A silencing, and overexpression of PD-L1/2. The “Microsatellite Instable” (MSI, 22%) subgroup has MLH1 silencing and a hypermutation phenotype, making these last two subgroups candidates for immunotherapy with immune checkpoints inhibitors [19]. Finally, the “Genomic Stable” subgroup (GS, 20%) corresponds to CDH1 and RHOA mutations and matches the diffuse GC subgroup. TP53 mutations are also found in the MSI and GS subgroups.
Surgery with perioperative platinum-based chemotherapy (e.g., oxaliplatin) is the gold standard treatment for localized GC, but surgical morbidity is reported to be as high as 39% [20]. The low five-year survival rate (25%) in Europe highlights an unmet medical need, which is due to inherent or acquired resistance mechanisms, like EGFR overexpression [21] or the aberrant activation of various circulating microRNAs (miRNAs) [22]. So far, the molecular classification has not led to the introduction of new routine treatments, and even immunotherapy strategies have led to only limited advances with less than 20% of responders. One of the challenges is to identify the patients that have a better chance of responding to immunotherapy.
The unmet need for truly efficient treatments in advanced GC illustrates the unique complexity of this cancer. Indeed, despite the classifications and molecular data, we are still missing some crucial information required to develop new treatments that would improve the patients’ management for survival benefit. This requires going beyond the existing classifications and refinement of our understanding of the molecular mechanisms that are often oversimplified and/or overlooked despite their extreme complexity. For instance, pan-genomic analyses do not include data on the relative expression of isoforms generated by alternative splicing or promoters or on protein expression levels that can be controlled through complex post-transcriptional modifications impacting protein stability, subcellular localization, and activity.
A good example of this is the complex p53 family of transcription factors that is present at the center of a dense interdigitated network of signaling pathways. The p53 family is composed of three genes: TP53 (p53), TP63 (p63), and TP73 (p73), and each generates multiple isoforms via alternative promoters and splicing (Figure 2). Importantly, the function of these isoforms can be opposing, as they generate isoforms with a N-terminus transactivation domain (TA isoforms) or without it (∆N isoforms). The function of these genes in cancers have been investigated over the years and were found to be critical for tumorigenesis, response to treatment, and metastasis [23]. In this comprehensive review that compiles about 400 articles out of 2600 published on the p53 family in GC, we are focusing on their expression and function in GC to highlight what has been clearly established but, also, what remains to be understood for a successful transfer to patient care.

3. TP53, a Guardian Against Gastric Cancer?

3.1. Incidence of TP53 Mutations in Gastric Cancer

Drs. Sano and Tahara were the first to report a loss of heterozygosity (LOH) in the TP53 locus, 17p13.1, in 68% of gastric tumors (n = 48) [24]. The TP53 LOH frequency was higher in well-differentiated cancers and in advanced cancers (up to 70% in metastatic cancers). Simultaneously, Drs. Tamura and Hirohashi detected TP53 mutations in exons 4–8 (codons deletions 137–139, V173M, I251S, insertion CTCA 252–253) in 64% (n = 24) of aneuploid tumors but none in diploid tumors [25]. They suggested that TP53 mutations were occurring at a late stage of the carcinogenesis, as subsequently supported by other studies (e.g., codons V216L and R248W) [26,27]. As seen in other types of cancers, the mutations were mostly centered in the most conserved region of the p53 protein, the DNA-binding domain [27]. The accumulation of mutated p53 proteins in malignant GC cells was confirmed by Dr. Lane using monoclonal antibodies [28]. These mutations were also observed in GC cell lines that are still widely used nowadays, such as KATOIII (deleted for TP53), NUGC-3 (p53 Y220C), MKN28 (p53 I251L), or MKN1 (p53 V143A) [29]. However, a year later, Drs. Yokosaki and Tahara showed that, even in primary tumors, TP53 was mutated (codons G117A, C141T, W158C, M160K, G226A, D228T, G245S, I254T, and V272E) in 56% of the samples, including early-stage cancers I and II [30] (Figure 3). It turned out that TP53 mutations were homogeneously present within the tumors, even at the early stages [31,32,33], including stomach precancer states such as incomplete intestinal metaplasia [34] and gastritis induced by H. pylori infection [35]. Altogether, these observations indicate that TP53 mutations occur before clonal expansion and, therefore, are likely a founding event in GC carcinogenesis and critical for the transformation of stomach epithelial cells.
In addition, familial mutations in TP53, like at aa 175 for Li-Fraumeni or the atypical truncation E287X [36], lead to GC (±5% incidence) but less frequently to colon cancer [37,38,39]. Hence, the carcinogenesis processes seem to be different between GC and colon cancer, where alterations of APC are more prominent than of TP53 [40]. However, the low penetrance of GC in Li-Fraumeni patients, which is higher in families of Asian origin than in the Western population, indicates the existence of genetic/environmental modulators and raises interesting questions about complementary/compensatory mechanisms [41].
The prominent role of p53 in GC raises the questions of both the origin of these events and their influence on gastric epithelial cell transformations towards the development of particular GC subtypes.

3.2. Association of p53 Mutations with Specific Types of Gastric Cancers

The pattern of TP53 mutations in GC shows complexity, as they are frequently present at an early stage of the intestinal subtype (41% n = 130) and rarely in the diffuse subtype (4%), but the mutation frequency increases significantly up to 33% in the late stage of the diffuse subtype [42]. This suggested a differential role of p53 in both types of cancers, which was further supported by several studies [43,44]. TP53 mutations were found more likely to be associated with H. pylori infection, a major cause for the development of GC of the intestinal subtype [35,45]. In addition, the involvement of TP53 mutations in the intestinal type is well-demonstrated by mouse genetic experiments in which precancerous lesions (polyps) and carcinomas following the exogenous overexpression of the intestinal differentiation transcription factor CDX2 are favored by the inactivation of tp53 (50 weeks, 50% penetrance vs. 80–100 weeks) [46]. The inactivation of TP53 might still be needed for the development of the diffuse type, since the amplification of the major p53 inhibitor, MDM2, was described to be more frequently found in this subtype [47]. However, a detailed analysis of the TCGA (The Cancer Genome Atlas) data do not support this early observation (Figure 1, personal data). The differential role of TP53 mutations is also observed with regards to the location of the tumor. For instance, tumors of the cardia seem to have more frequent mutations (54%) than tumors of the antrum (25%) [48]. In addition, TP53 mutations are also present in the rare gastric carcinomas “small cell carcinoma” [49].
Pan-genomic analyses of GC defined the molecular subgroups and indicated that TP53 mutations are mostly associated with the CIN molecular subgroup that represents about 49% of GC [18] (Figure 1). This observation is in agreement with previous observation, as the CIN molecular subgroup mostly corresponds to the intestinal subtype often induced by H. pylori infection. However, a careful analysis of the TCGA shows that TP53 mutations are also present in the MSI (24% of TP53 missense mutations) and the GS subgroups (11% of missense mutations). In addition, truncating mutations are present in 28%, 9%, and 2% of the CIN, MSI, and GS subgroups, respectively (personal data).
In addition to pan-genomic analyses, TP53 mutations have been correlated with precise molecular alterations of genes that may or may not be related to p53. For instance, TP53 mutations are more frequently (81%) found in tumors overexpressing EGFR/TGFα (26%) [50] or CHK1/2 [51]. In contrast, the epigenetic silencing of XAF1 by promoter methylation, which interacts with the DIABLO proapoptotic protein, is exclusive of TP53 mutations, suggesting that they participate in the same pathway [52] (Figure 4). Indeed, XAF1 interferes in a complex way with p53 function [53], and p53 represses the expression of XAF1 at the transcriptional level in GC cells [54,55]. Similar observations were made with HRK/DP5 [56]. More generally, pan-genomic analyses established that TP53 mutations are exclusive with the methylation of several tumor suppressor genes, such as PGP9.5, NMDAR2B, or CCNA1 [57].

3.3. Causes for TP53 Mutations in Gastric Cancer

It is proposed that TP53 mutations are caused by dietary carcinogens that induce DNA deamination [32]. This is supported by mouse experiments showing that the food carcinogen MelQ (2-Amino-3,4-dimethylimidazo [4,5-f]quinoline) and the alkylating agent N-methyl-N-nitrosourea (MNU) can induce GC with p53 mutations (aa 171 or 113) [58,59,60]. In addition, the transformation ability of MNU is higher (incidence and aggressivity with invading mucosa) in mice with inactivated tp53 [61,62]. Moreover, it was observed that patients drinking alcohol with polymorphisms in detoxifying enzymes (ex., CYP1A1 and CYP2E1) have more mutations in TP53 [63] (Figure 3), which further supports the correlation between carcinogens and TP53 mutations.
Another source of TP53 mutations is H. pylori infection, which highly correlates with the infected gastric mucosa of humans and Japanese monkeys [64]. the infection by H. pylori causes genomic DNA oxidation in various genes due to inflammation and the production of radical oxidized species (ROS), leading to mutations in the TP53 gene even at the gastritis stage [35,65,66] (Figure 3). Additional mechanisms have been proposed, such as the cytidine deamination of the TP53 DNA sequence by the induction of AID (Activation-Induced cytidine Deaminase) following NF-kB (Nuclear Factor kappa B) activation by the H. pylori protein CagA [67,68]. Indeed, accumulated mutations were predominantly C:G > T:A transitions in GpCpX motifs—a marker of cytidine deamination by AID. In addition, the expression of AID in mice induces TP53 mutation [66]. Finally, TP53 mutations following H. pylori infection also involved CHAC1 (Cation transport regulator 1) that has γ-glutamylcyclotransferase activity to degrade glutathione, an antioxidant [69].
The role of food carcinogens, as well as the specificity of the H. pylori strain, may explain some observations made towards ethnical differences. For instance, the Europeans show more G:C to A:T transitions (90% vs. 48%) and the Oriental population more A:T to G:C transitions (22% vs. 2%) [70]. Similarly, more frequent TP53 mutations are found in the GC of African Americans compared to other ethnic groups [71].

3.4. TP53 Polymorphism and Gastric Cancer

TP53 presents polymorphisms in exon 4 at codon 36 and 72 that impact the prognosis of different cancers [72]. The most common polymorphism is at codon 72 (pro vs. arg) and differs between populations. The impact of this polymorphism in GC is still debated in the literature. Several studies or meta-analyses suggest that there is no difference in polymorphism representation in GC patients compared to the general population [73]. However, other analyses suggest that the impact of the polymorphism may be dependent on specific subtypes (lower frequency of Pro/Pro in poorly differentiated cancer) and the ethnicity (in Asians: less cancer with Arg/Arg compared to the general population, Pro/Pro more present in the cardia subtype, and Arg/Arg more present in advanced cancers) [74]. The more common Arg/Arg codon 72 polymorphism in Caucasians correlates with a tendency for cardia GC, whereas the more common Pro allele in African Americans correlates with a tendency for antral GC [72,75]. In addition, the Arg/Arg polymorphism favors the survival of patients with non-cardia cancer [75], although it increases slightly the risk (adjusted odds ratio (OR) = 1.67, 95% confidence interval (CI) = 1.00–2.77) of non-cardia GC, especially in alcoholics [76]. Furthermore, the Pro polymorphism could also favor the development of GC from gastritis [77]. In addition, the Arg/Pro and Pro/Pro genotypes significantly correlate with a lower response rate to chemotherapy when compared to the Arg/Arg genotype (35.7 vs. 66.7%, p-value 0.019) [78,79], but the Arg/Arg polymorphism is of negative predictive value for 5-fluorouracil (FU) -based therapy [80]. Altogether, it seems that aa 72 polymorphisms may have a predictive significance in GC, but their exact roles are complex.
Based on the fact that TP53 mutations occur early during the gastric epithelial cell transformation and that p53 is an important factor in the response to DNA damages, it seems relevant to consider that the p53 status (mutation or expression level) might be used as a biomarker for personalized therapy.

3.5. Are TP53 Mutations Biomarker for Personalized Therapy in Gastric Cancer?

Two studies initially showed that the mutational status of TP53 was an independent biomarker for a reduced five-year survival (9% vs. 42%, n = 116 patients) [81] and for a poor response to cisplatin associated with a low P21 expression and reduced apoptosis (heterozygote for R162P, R175H, P177S, A178D, R213P, Y220C, R273C, and R282L, n = 11 patients) [82]. However, soon after, the value of p53 mutations as a prognosis marker was debated between studies supporting the early results (ex., [83]), including by using circulating DNA (ctDNA) to detect TP53 mutations [84], and others with the opposite conclusion (ex., [85]). These discrepancies may reflect the small size and/or the heterogeneity (grades, subtypes, ages, etc.) of the investigated cohorts. However, a further meta-analysis clearly indicated then that TP53 mutations should be considered as a predictive marker for the response to chemotherapy [86]. This mutational status of TP53 as a biomarker for a poor prognosis is in agreement with additional observations that GC patients with TP53 mutations tend to develop more aggressive tumors characterized by frequent liver metastasis (86% vs. 40%) [87] and the presence of a hypoxic response [88] (Figure 3).
In order to increase the specificity and perform personalized therapy using the TP53 status as a biomarker, combined analyses of different genes associated with TP53 mutations have been tested. For instance, the combined mutation of TP53 with BAX [89] or NRXN1 expression [90] drastically worsened the prognosis for GC patients. Inversely, TP53 mutations associated with an elevated expression of the oncogenic YAP protein tend, surprisingly, to favor GC patient survival [91]. Although the molecular mechanisms explaining this unexpected observation remain unclear, one may speculate that the pro-proliferative activity of YAP and mutant p53 facilitates the activity of cytotoxic drugs.
More recently, pan-genomic analyses on large cohorts have shown that TP53 mutations have an impact on the prognosis in specific molecular subgroups. For instance, TP53 mutations in tumors with a MSS (microsatellite stable) status negatively impact the patients’ prognosis [92], and the association of p53 mutants with the CIMP (CpG island methylated phenotype) characteristic shows a higher prognosis quality [93]. Similarly, TP53 mutations are significantly associated with a decrease in the overall survival in patients with diffuse or stage III or IV tumors [94].
A better understanding of the molecular impact of each type of mutation on p53 activity has led to studies querying whether these mutations impact patients’ prognosis differently. Oncogenic TP53 missense mutations are mostly found in the core domain. Many of these mutations result in protein misfolding and accelerated aggregation. TP53 mutations also frequently result in the alteration or loss of zinc at the DNA-binding site, which increases the aggregation via nucleation with zinc-bound p53. The top 10 most frequent mutations (hotspots) affect codons of the core residues R175H, Y220C, G245S/D/C, R248Q/W, R249S/C/D, R273C/H/S, and R282H and are classified into structural mutants, which alter the p53 core structure, and contact mutants that affect residues at the DNA interface [95]. Both structural and contact mutations result in a loss of function for binding to p53-specific DNA elements. In the last decade, several studies have provided compelling evidence that certain hotspot mutations also confer gain of function (GOF) properties to p53, which support a number of oncogenic activities from cell survival to proliferation [96]. In general, such GOF activities result from newly acquired interactions (“neo-interactions”) of p53 mutants with transcription factors (TFs) or coactivators such as YAP, HSF1, p73, ETS1 and 2, NF-kB subunits, and NRF2, which can be defined as “neo-interactants” [97]. These novel interactions create “neo-TFs”, leading to alterations in the gene expression patterns that inhibit cell death and favor cell growth. In gastric cancers, mutations in the L3 loop; the LSH motif; and mutations considered as hotspots (R175, G245, R248, R273, and R282) correlate more strongly with a poor prognosis [98,99]. Typically, these mutations correlated with a venous invasion in advanced-stage diseases, significantly worse overall survival, and the recurrence of free survival (p = 0.001). The negative impact of some of these mutations (e.g., R175H, R248G, and R273H) on cancer cells has been attributed to gain of function properties. Inversely, p53 mutants with residual activity favor survival in males [100]. The impact of these mutations at the molecular level specifically in GC cells has not been well-characterized, except for p53 V143A, which interacts with S100A4 in MKN1 GC cells to stabilize p53 [101]. The silencing of S100A4 reduced the expression of two target genes of p53 V143A: MYC and ID2.
In summary, there are strong lines of evidence that TP53 mutations have a negative impact on GC, either by impacting the response to chemotherapy or by favoring the development of metastasis. However, the extent of this impact appears to depend on additional molecular features of the tumors, such as the microsatellite status or mutations in other genes. So far, these potential biomarkers have not been used in the clinic, as no therapeutic alternatives have been or are proposed to treat patients besides chemotherapy. However, in the era of immunotherapy, the recent findings on p53 mutants might point to new therapeutic directions.
Indeed, studies using immunogenics (characterization of the immune landscape using pan-genomics) suggest that TP53 mutations reduce GC immunogenicity [102]. Strikingly, pan-genomic analyses show that almost all immune gene sets are significantly downregulated in mutated TP53 vs. wild-type (WT) TP53 GC. These downregulated immune pathways and cell types in TP53-mutated tumors include 15 immune cell types and functions, tumor-infiltrating lymphocytes, regulatory T cells, immune checkpoints, cytokines and cytokine receptors, human leukocyte antigens, and proinflammatory and para-inflammations. Moreover, a number of p53-mediated pathways are significantly associated with immune activities. These observations are confirmed in another cohort in which a low expression of PD-L1 and reduced number of infiltrating lymphocytes correlates with the TP53 mutation [103]. The level of PD-L1 seems to be particularly lower in tumors with a p53 mutant and elevated HER2 expression [104]. As an immunotherapy, anti-PD-1 immunotherapy shows a limited efficacy in GC; these observations may open the way for a therapeutic strategy based on a combined use of a p53 reactivator and trastuzumab (anti-HER2). In addition, p53 mutants promote a hypoxic response in GC tumors, which likely impacts the immune response [88].

3.6. Is the TP53 Expression Level a Biomarker for Personalized Therapy in Gastric Cancer?

Another approach often used in pathology for establishing a prognosis uses the protein expression level, independently of the mutation status. Drs Martin and Sylvester were the first to take advantage of Dr. Lane’s p53 monoclonal antibodies and investigate by immunohistochemistry the correlation between the p53 protein level and a GC patient’s prognosis [105]. This initial study found that 57% of the tumors expressed a high level of p53 protein and that these patients had a lower five-year prognosis (24% vs. 56% with low level of p53 proteins; p = 0.02, Mantel-Cox Test; odds ratio of death, 2.09; 95% confidence interval I 0.02 to 4.25; n = 126). A high expression of p53 was present in the intestinal (62%) and diffuse (48%) types of GC, including the signet cell type (71%), and lymph node metastases (64%). Similar results were later confirmed on larger cohorts (n > 200), showing that the elevated p53 expression could be an independent factor for a poor prognosis in patients that undergo curative resection [106] or in general, with a significant difference in the five-year survival rate (62% vs. 43%) [107], and that a positive nuclear p53 immunoreaction can be significantly associated with a shorter overall survival [108,109]. It was also shown that GC displaying the combination of a high p53 expression and aneuploidy carries the worst prognosis [110].
However, these results on the prognostic value of p53 protein expression were contradicted and revealed a more complex picture [111,112,113,114,115,116]. For instance, some studies identified p53 expression as a poor prognosis marker only in the diffuse subgroup [117] or only in a univariable analysis (p < 0.0005) but not in the multivariable analysis, although relatively close to be statistically significant (ex., p = 0.08) [81,115,118].
These discrepancies illustrate the complexity of GC and the difficulties encountered in studies differing by the patients’ profiles, the antibodies used, and the cut-off scores chosen for positivity, as well as the statistical methods. In particular, these studies did not analyze the mutational status of TP53; thus, the relative representation of the TP53 mutations between the cohorts might explain some differences in the outcomes. Hence, several meta-analyses have been performed and concluded that a high expression of p53 proteins should be considered a biomarker for a poor prognosis (ex., [119]). Therefore, an elevated p53 protein level represents an indicator of a poor prognosis for GC patients.
In addition to the patient prognosis, multiple studies have shown that a high p53 expression was a marker for a weak response to chemotherapy [120,121,122,123,124]. For example, a study combining three large cohorts (1150 patients) indicated that p53 expression (DO7 antibody) was a predictive marker for survival (p < 0.001) and a response to therapy (5-FU+folinate+oxaliplatin, p < 0.01) [125], but such a correlation was not always observed [126]. However, a meta-analysis of more than 800 articles concluded that the p53 expression should be considered an interesting biomarker for the therapy response [86,127].
To increase the selectivity and sensitivity of the prognosis and therapeutic prediction, the co-expression of p53 with known positive or negative regulators of the cell cycle and apoptosis have been investigated in GC. For instance, the elevated co-expression of p53 with BCL2 [128], TS GSH [129], or VEGF [130] correlate with a poor response to 5-FU and cisplatin. Similar results were obtained for an association between p53 and MCL1 [131], Cyclin E [132], HIF1A [133], or MMP7 [127]. In addition, an inverse correlation was found between the p53 and Survivin expression [134], which is repressed directly by p53 [135].
Besides establishing a correlation between the p53 protein expression and survival, investigators have tried to define the characteristics of the tumors expressing high levels of p53. For instance, the nuclear accumulation of p53 protein was frequently seen in papillary adenocarcinoma, well-to-moderately differentiated tubular adenocarcinoma and poorly differentiated adenocarcinoma with solid nests or focal tubular structures but was rarely seen in signet-ring cell carcinoma, mucinous adenocarcinoma, or poorly differentiated adenocarcinoma growing in a scattered manner [136,137]. In a French cohort, similar results were observed, showing that 67% of the intestinal type of GC but only 24% of the diffuse type express high levels of nuclear p53 [138]. There were also differences in the p53 expression between proximal and distal CG, respectively, 38% vs. 20% [139]. A similar observation was made for MDM2 and p14-ARF [140]. In addition, H. pylori-infected GC show higher p53 staining [141], whilst the eradication of H. pylori reduces the p53 and MDM2 protein levels [142]. High levels of p53 expression occur in primary and metastatic GC, supporting a correlation with elevated aneuploidy and the proliferation rate [137,143,144]. In contrast, the absence or low expression of p53 was observed in the very early stage of gastric carcinogenesis, such as chronic gastritis or metaplasia [145,146]. The expression of p53 appears to increase with the carcinogenesis [147]. However, some studies did not see the same correlation between a high p53 expression and cancer stage [114].
One of the difficulties is to make a precise assessment of the p53 status, mutation and expression-wise. The studies described in this section did not assess the mutational status of p53 in parallel to the expression level. A plausible hypothesis is that the elevated p53 protein level could be explained by the presence of a mutation in the p53 that would inhibit its transcriptional activity, hence reducing the expression of the main p53 repressor and target gene the MDM2 responsible for p53 degradation. Thus, the p53 expression and TP53 mutations might be considered equally concerning the p53 status. However, several studies have shown discrepancies between the expression level of p53 and the TP53 mutation status in GC [32,148,149,150]. For instance, a third of the tumors with high p53 expression have no TP53 mutation and vice versa. Hence, studies considering only the expression level have to be taken cautiously. Overall, assessing both, the protein expression level, as well as the mutational status, seem to have a clinical interest, since accumulation of the wild-type p53 protein in tumors correlates with a lower risk of metastasis [151].
To simultaneously assess the expression level and the mutation status of TP53, an interesting approach is to test for the presence of autoantibodies directed against p53 in patients’ blood. Several studies, including meta-analyses, showed that the presence of p53 autoantibodies correlated with the stage and prognosis [152,153,154,155,156,157] but were not always confirmed [158]. More recently, the detection of p53 autoantibodies combined with other autoantibodies has been evaluated with success to diagnose GC [159].
Hence, to summarize, an elevated expression level and/or mutation in p53 should be considered as prognosis and predictive markers in GC, despite some contradictory results. These contradictory results may also reflect the complexity of GC and may also justify why, in the particular case of GC, the status of p53 (mutation or expression level) is not used in current clinical practice to stratify the patients and make a reasoned therapeutic decision. Another explanation is that there is a lack of alternative therapies to treat GC if chemotherapy is not used.

3.7. Alteration of p53 Regulators in Gastric Cancer

When no TP53 mutation is present, inhibition of the p53 function can also occur by alternative means that impact negatively on the p53 protein level and/or activity, favoring tumorigenesis. Multiple mechanisms have been described in GC—in particular, alterations that target the expression or activity of MDM2.

3.7.1. TP53 and MDM2 Alterations in Gastric Cancer

The E3 ubiquitin ligase MDM2 is the major negative regulator of p53 through a direct interaction that drives p53 ubiquitination and degradation by the proteasome. MDM2 does not seem to be overexpressed at precancerous stages [160], but MDM2 amplification was found in 42% of GC and in a majority of the diffuse type [47]. A detailed analysis of the TCGA data shows that 20% of GC display high MDM2 mRNA levels or gene amplification in both the intestinal and diffuse types (personal data). Surprisingly, the gain or overexpression of MDM2 is similarly found in tumors with WT or mutated p53 (24%, personal data). Interestingly, tumors infected by EBV display significantly more MDM2 mRNA. In addition, a polymorphism (SNP309 G/G) in the promoter of MDM2, which induces MDM2 transcription, correlates with an increased risk of GC by hazard ratio, 3.16; 95% CI, 1.22 to 8.20; p = 0.018 [161,162]. The elevated expression of MDM2 in GC is also caused by the loss of miR-518 that targets MDM2 [163].
Besides the alterations of the MDM2 gene, other molecular mechanisms that affect the MDM2 protein activity have been described in GC. The elevated expression of positive MDM2 regulators—namely MDM4 (MDMX) [164], ENIGMA [165], AURKA [166], and CDT2 [167]—correlates with the MDM2 protein level and has an impact on the prognosis. For instance, MDM4 has an elevated expression that correlates with lymph node metastasis [164]. Inversely, the downregulation of MDM4 increases slightly the activity of cytotoxic drugs (5-FU) in GC cells [168]. Another example is the LIM domain protein ENIGMA, whose overexpression is caused by SRF [165]. At the molecular level, ENIGMA interacts with MDM2 to inhibit its self-ubiquitination, thereby stabilizing it and inducing p53 ubiquitination and degradation. Similarly, the Aurora A kinase (AURKA) stabilizes MDM2 by phosphorylation at S166 to reduce the p53 protein levels by ubiquitination [166].
Conversely, several negative regulators of MDM2 are lost in GC. For example, a mutation of SMAD1 at S239 found in GC favors the interaction of MDM2 with p53 by avoiding the phosphorylation of SMAD1 by ATM, a DNA damage response kinase, thereby reducing the cell death caused by doxorubicin or BMP2, a member of the TGF-β family [169]. However, the frequency of this SMAD1 mutation seems to be very low, as a TCGA analysis on a cohort with 478 patients did not show any mutation and, rather, showed a selective increase in its expression in the singlet cell GC subtype (personal data). Another example is the reduced expression of PICT1 in GC that correlates with a better prognosis (n = 70 patients; p = 0.046) [170]. At the molecular level, PICT1 appears to sequester the ribosomal protein RPL11 in the nucleus to destabilize MDM2. UBTB1 also has a reduced expression in GC that correlates with a poor patient survival (p = 0.012, stage I/II). This ubiquitin ligase induces the degradation of MDM2 to stabilize p53 and induces senescence [171]. The expression of FHIT is mainly lost in diffuse GC that rarely display TP53 mutations [172]. FHIT (fragile histidine triad), a P1-P3-bis(5′-adenosyl) triphosphate hydrolase, is a tumor suppressor gene frequently lost in various cancers that inhibits the MDM2-dependent degradation of p53 [173].

3.7.2. Other p53 Regulators Altered in Gastric Cancer

Besides molecular alterations that affect MDM2, other silenced or overexpressed genes in GC alter the p53 activity and impact the prognosis. A few of these genes have their expression diminished by promoter methylations. For instance, the promoter methylation of PAX5 [174], OSR [175], PKNOX2 [176], and BCL6B [177] occurs in GC, and the restoration of their expression, by overexpression or treatment with 5-Aza, increases the p53 activity on P21, BAX, FAS, or IGFBP5 expression. Importantly, the methylation of OSR correlated with a poor prognosis in a multivariate analysis (p = 0.04). Another example is the loss of a positive regulator of p53, TIP30, that correlates with a reduced survival in GC patients [178]. Its re-expression in GC cells restores the p53 protein levels and induces the expression of the proapoptotic target gene of p53 BAX.
Conversely, the elevated expression of p53-negative regulators is also observed in GC, such as ubiquitin ligase TRIM59, which is upregulated in the T2 and T3 stages and correlates with a poor prognosis (n = 111, p = 0.447) [179]. TRIM59 interacts with and drives p53 ubiquitination, whilst TRIM59 silencing reduces the cell growth and migration that correlates with upregulation of the p53 protein and NOXA mRNA levels. Other examples are the HSP70-type protein CREP that interacts and negatively regulates p53 [180] and ZBTB2, whose expression is increased in GC due to the loss of miR-149 expression [181].
The deregulation of additional genes that correlate with p53 activity have been described, such as the low expression of CARP (caspase-associated recruitment domain-containing protein) [182], Ku70, and CHK2, which leads to impairment of the DNA damage response (DDR) mechanisms [183], Ubiquilin 4 (Ubqln4), encoding a ubiquitin ligase interacting with RNF114 [184], and CHD5 (chromodomain helicase DNA-binding) [185]. Inversely, the elevated expression of AP-4 [186], the bHLH-LZ interactant of the large T antigen of SV40 Mortalin [187], DITT4 [188], Mucin 17 (MCU17) [189], PDRG1 (P53 and DNA Damage-Regulated Gene 1) [190], MPP8 (M-phase phosphoprotein 8) [191], CCAT2 [192], HBXIP (Hepatitis B X-interacting protein) [193], IRTKS [194], and the Cdc42 interacting kinase gene ACK1 [195] correlates in GC with a reduced p53 activity, and their silencing restores p53 functions characterized by cell growth arrest or cell death. However, the precise mechanisms connecting the change in p53 activity to the deregulation of these genes and their impact on patient prognosis has not yet been established.
Another mechanism that impacts p53 expression involves noncoding RNAs. The downregulation of miR-3174 [196] or miR-370 [197] increases the expression of their targets, ARHGAP10 and PTEN, respectively, which, in turn, inhibits p53 [196]. Inversely, the elevated expression of miR-27b [198] or miR-100 [199] downregulates the expression of p53 activators CCNG1 and the ubiquitin ligase RNF144B (an inhibitor of the p53-ubiquitin ligase PIRH2), respectively. Several long noncoding RNA (lncRNA) also induce the expression of p53 in GC. For instance, MEG3 lncRNA increases the p53 protein levels to mediate its antitumor activity [200]. Its elevated expression results in a good prognosis for GC (n = 72, p < 0.001), but its expression can be downregulated by methylation. Similarly, GAS5 lncRNA stabilizes p53 in GC cells by interacting with TP53 exon 12, reducing the proliferation and migration [201]. Inversely, several oncogenic lncRNA (OnclncRNA) can inactivate the p53 pathway to promote gastric tumorigenesis, such as OnclncRNA-626 through an interaction with SRSF1 [202], VCAN-AS1 lncRNA through a competitive interaction with eiF4A3 that correlates with a poor prognosis [203], and lncRNA ZFPM2-AS via the stabilization of the MIF (macrophage inhibitory factor) [204]. Finally, the circular RNA Hsa_circ_0001546 acts as a miRNA-421 sponge to inhibit the chemoresistance of GC cells via an ATM/Chk2/p53-dependent pathway, correlating with a poor prognosis [205].

3.8. Alteration of p53 Target Genes in Gastric Cancer

Alterations found in GC also impact the downstream components of the p53 pathway. More precisely, the known p53 target genes involved in cell growth or death are altered. In several cases, the change in expression is most likely due to the presence of p53 mutations, such as for the elevated expression of HDGF, which is normally repressed by the binding of WT p53 to its promoter [206]. Inversely, the expression of RHOE [207], XEDAR (X-linked ectodermal dysplasia receptor) [208], and FBXW7 [209] is low in GC, while the overexpression of p53 restores their expression. Surprisingly, the p53 target gene TIGAR, involved in the cell metabolism, has an elevated expression in GC, where it protects cells from RAS and favors cell proliferation [210]. Hence, the expression of TIGAR in GC appears independent from the p53 status. Noncoding RNAs are also impacted. For instance, the lncRNA TUSC7 is a direct target of p53, whose expression is inhibited in GC and correlates with a poor prognosis (n = 78, disease-free p = 0.0019) [211]. Similarly, p53 can directly induce the expression of miR-508-5p in GC cells, which targets ABCB1 mRNA to favor the response to therapy [198]. However, there are also more interesting examples, such as the p53 target gene BGT4 that is repressed through promoter methylation (66%), and the restoration of its expression using 5-Aza-DCT induces cell death and reduces tumor growth [212,213]. Another example is the mutations in the p53 target and pro-cell death receptor DR5 that are exclusive of TP53 mutations [214].
Overall, it is interesting to note that there are only two examples of alterations of the p53 target genes BTG4 and DR5 that are independent from the p53 status. This highlights how p53 seems to represent a key node involved in gastric epithelial cell homeostasis, which is supported by a study identifying a signature of p53 target genes that correlates the most with patients’ survival [215].

3.9. Functions and Regulations of p53 in Gastric Cancer

3.9.1. Cellular and Molecular Activities of p53 in Gastric Cancer Cells

In 1991, Baker and Vogelstein showed that wild-type p53 could inhibit cell growth [8]. It was soon after confirmed that wild-type p53 inhibits growth of GC cells but not mutated p53 V143A [216]. The evidence of a dominant negative activity of the p53 mutant over WT p53 was also pointed out in GC, since half of the TP53 mutations (54%) occurred in samples without allelic loss [217]. However, the role of p53 in response to DNA damage [218] and chemotherapy [219] was, at first, unclear, as an initial report suggested that WT p53 was not induced by these stresses in MKN74 cells and did not participate in their response. However, it is of note that a lower band around 44 kDa detected by the p53 Pab1801 antibody was significantly induced and correlated inversely with the protein level of BCL2, an anti-apoptotic gene known to be repressed by p53. At that time, the nature of this band was unknown, but now, we can speculate that it could correspond to the p53 splice variant, p53β, which was described in GC cells treated with cisplatin [220]. The function of p53β in the GC cells remains to be established.
Since then, multiple studies have indicated that p53 is involved in the response to chemotherapy in GC cells. Typically, cell lines with mutated p53 (SK-GT1-5 and MKN28) are less sensitive towards 5-FU, cisplatin, or mitomycin C compared to cells that express WT p53 (MKN74, NUGC-4, and MKN45) [221,222]. Furthermore, the reintroduction of WT p53 in mutated p53 cells favors apoptosis induced by cisplatin or 5-FU [223] via the downregulation of AKT phosphorylation [224]. As observed in other biological systems, these drugs (ex., 5-FU) induced a stabilization of WT p53 and a change in expression of the p53 target genes, such as BCL2 and BAX [225,226], but not mutated p53 [221].
These observations confirmed the initial findings made by Kastan et al. that p53 plays a role in the DNA damage response [227] and that the introduction of WT p53 in p53-null cancer cells favors a response to cisplatin in vivo by inducing apoptosis [228]. However, it is of note that most of the studies performed on GC cells based their conclusion on a correlation rather than on functional experiments. In 2000, Drs. Jiang and Lam were the first to use the p53 loss of function (antisense) in AGS cells to show that p53 was required to induce apoptosis caused by arsenic trioxide [229]. More recently, the function of p53 in DNA damage and chemotherapy responses in GC cells was confirmed by showing that the silencing of p53 and ATM synergize to favor the cytotoxicity induced by an inhibitor (Olaparib) of the DNA damage response enzyme PARP-1 [230]. However, the demonstration that the mutational status of p53 dictates the response to chemotherapy in mouse models of GC remains to be established [231].
The activity of p53 upon treatments has been assessed by the regulation of well-known p53 target genes, such as P21 and BAX. However, other target genes have been also investigated. For instance, chemotherapies (cisplatin, 5-FU, etc.) can induce the promoter activity of the receptor for pro-death ligand FAS CD95 via p53 in GC cells [232,233]. In contrast, p53 can repress the TFF2 gene, involved in wound healing in the digestive tract, by competing with the AP1 transcription factor, inhibiting cell migration and inducing apoptosis [234]. In contrast, mutant p53 R175H favors TFF2 expression. Similarly, P53 inhibits the E2F1-dependent induction of CHK1 upon radiation treatment in AGS cells, explaining why high level of nuclear CHK1 correlated with a poor prognosis in GC [235].
Numerous other mechanisms positively (ex., CHK1/2 kinases) and negatively regulating p53 (ex., MDM2) have been identified in cancers. Only a handful of them have been characterized in GC. More precisely, hSRBC, a putative tumor-suppressor gene seems to be an inducer of p53 in response to chemotherapy (5-FU, etoposide) in GC [236]. The topoisomerase TOP3a is required for p53 activity through a complex mechanism involving its binding to the p53 promoter and a physical interaction with the p53 protein [237]. Hence, growth inhibition induced by TOP3a requires p53, and therefore, TOP3a expression is logically found downregulated in GC. Inversely, XIAP seems to be a p53 inhibitor reducing apoptosis caused by chemotherapy (ex., cisplatin) in p53 WT GC cells [238]. Other examples are a diminished expression of p53 by miR375 by interacting with the p53 3′UTR [239] or the inhibition of PI3Kcat leading to cell death that correlates with an increased expression of p53 and PUMA [240]. However, the significance of these mechanisms for the patient outcome has not been established yet.
Beside chemotherapies, other drugs and stresses inducing growth arrest or cell death have been described to modulate the p53 activity in GC cells. TNFα induces apoptosis via an increased p53 expression, which, in turn, induces BAX [241] and IGFBP3 [242] expression. TRAIL (TNF-related apoptosis-inducing ligand) is more active in MKN45 cells expressing WT p53 [243]. Besides these proapoptotic ligands, the activation of PPARg by troglitazone induces apoptosis via p53 and the downregulation of BCL2 in GC cells [244] [245]. Inversely, p53 is inhibited in GC by persisting pollutants (ex., PFDA) [246].
P53 has also been a marker to explore the potential of novel anticancer therapies. For instance, the p53 and BAX protein levels are induced by S-propargyl-cysteine (SPRC), which causes H2S production by the CSE enzyme of the GSH metabolism pathway (trans-sulphuration pathway) and cell death [247]. Copper ternary complexes with amino acid chains [248] or iron chelators (ex., DpdtC (2,2′-di-pyridylketone dithiocarbamate) [249] induce p53 activity in GC cells. Finally, several molecules derived from Chinese medicine, such as Pteris semipinnata L, induce cell death more efficiently in GC cells with WT p53, [250,251], involving the inhibition of MDM2 in the case of triptolide [252].
However, p53 does not seem to be involved in the response to all pro-cell death signals in GC cells. For instance, TGF-β causes apoptosis in GC cells by a p53-independent mechanism [253,254]. However, TGF-β alterations (20% mutations in the TGF-β RII receptor) are exclusive to TP53 mutations in GC and correlate with the microsatellite instability [33]. In addition, the genetic inactivation of the TGF-β signaling component, SMAD4, does not favor TP53 mutations in mice while inducing gastric polyps [255]. Hence, this would suggest that p53 and TGF-β either participate in the same molecular pathway in gastric tumorigenesis or that p53 inactivation is not necessary for the tumorigenesis of the MSI subtype. Other drugs or stresses were shown to act independently of p53, such as cadmium [256], paclitaxel [257], cetuximab [258], arsenide tetroxide [259], and ruthenium complexes, representing interesting molecules to treat p53-mutated GC [260,261]. This is the case for nonsteroid anti-inflammatory molecules (NSAID) (ex., indomethacin) that target the COX2 enzyme and display anticancer activities. Several p53-target genes, such as PUMA, are activated by NSAID [262], but NSAID anticancer activities seem to be independent of p53 in GC [263,264] and might involve ER stress pathway effectors (ATF4 and CHOP) instead [265]. Still, there is a functional interaction between p53 and COX2, since the expression of COX2 is repressed by p53, which competes with TATAA-binding proteins on the COX2 promoter [266]. This may explain why the expression of COX2 is more elevated in GC with TP53 mutations [267]. Hence, targeting COX2 might represent an interesting strategy in tumors with TP53 mutations that are often more resistant to chemotherapies.

3.9.2. p53 Mouse Genetics and Gastric Cancer

P53−/− null mice are viable but were shown to be highly susceptible to spontaneous tumorigenesis, with a predominance of malignant lymphomas, osteosarcomas, and soft tissue sarcomas [268,269]. To our knowledge, no spontaneous tumor formation in the stomach has been reported in p53−/−. However, when p53−/− null mice are treated with the carcinogen N-nitrosomethylbenzylamine (NMBA), about 50% of p53−/− null mice develop tumors in the stomach. Interestingly, zinc depletion makes these mice more susceptible to NMBA, and 100% of p53−/− null mice develop tumors within 30 days [270]. This suggests a role for p53 in regulating zinc metabolism in the stomach, as seen in HEPG2 cells [271]. Hence, the loss of tp53 may be a facilitator for the early step of gastric tumorigenesis, which is supported by the observation that gastric organoids are deleted for tp53 to evolve toward dysplasia and then adenocarcinomas when implanted in vivo [272]. In the following years, many other groups combined p53−/− null mice with other mouse models to induce and study tumorigenesis in the stomach. Importantly, these studies revealed that, depending on which mouse model(s) are combined with p53−/− null mice, tumorigenesis can be directed towards the development of intestinal carcinoma, diffuse with the presence of ring cell carcinomas, or a mixed type of GC.
More precisely, when p53−/− null mice are combined with
(1)
the loss of cdh1, mice develop poorly differentiated GC with the presence of ring cell carcinomas [273].
(2)
In addition to the loss of tp53 and cdh1, the loss of smad4 drives the development of the diffuse type of GC with the presence of signet ring cell carcinomas [274]. In this model, the osteopontin cytokine, Zeb2, and thymosin-β4 (Tβ4) were later identified as relays for tumor formation [275].
(3)
Replacing, in the latter case, the smad4 loss by the gain of expression of the oncogene krasG12D, generating a compound mouse with the loss of tp53 and cdh1 and gain of the krasG12D oncogene expression results in the development of diffuse and intestinal type of tumors [276].
These different models clearly show that, by combining the loss of tp53 with the loss of cdh1 and smad4 or gain of krasG12D, one can drive gastric tumorigenesis either towards a diffuse type of gastric cancer or mixed type or diffuse and intestinal. However, the major drawback of these models is that they are not time-controllable, as the Cre driver lines used are already active at the embryonic stages. In addition, they seem to be not active in all cell types of the gastric epithelium, and/or as in the case of pdx1-Cre, which are not restricted to the gastric epithelium. Instead, they induce tumors in other organs, like the small intestines or the pancreas. Ideally, one would need a mouse model where GC can be induced at adult stages specifically only in the stomach and where the tumor development is not only directed towards diffuse or mixed types of tumors but, more precisely, towards the CIN, GS, EBV, and MSI types of gastric adenocarcinomas.
In this respect, Seidlitz and coworkers set out to fulfill these needs [277] by taking advantage of various mouse tissue gene expression databases (Gene Expression Omnibus, MGI-Mouse Gene Expression Database (GXD), and BioGPS) to identify a stomach-specific gene to drive a tamoxifen-inducible Cre recombinase (CreERT2) and improve our knowledge concerning the genetic characteristics of the different GC subtypes. Concerning the first aspect, they identified the anxa10 gene locus to be specifically expressed in all cell types of the gastric epithelium only and generated the tamoxifen-inducible anxa10-CreERT2 driver mouse line. Combining anxa10-CreERT2 mice with
(1)
Tp53R172H + KrasG12D + Smad4fl/fl drove gastric tumorigenesis towards the CIN type with intestinal morphology, glandular structures, and the development of lung and liver metastasis.
(2)
Cdh1fl/fl + KrasG12D + Smad4fl/fl gave rise to a diffuse GS type of gastric adenocarcinomas with diffuse morphology, poorly differentiated, signet ring cells, and peritoneal carcinomatosis and lung metastases.
(3)
Cdh1fl/fl + KrasG12D + Apcfl/fl gave rise to the newly identified GS subtype “Serrated adenomatous type cancer” with serrated tooth like morphology and adenomatous morphology [278].
Importantly, by using CreERT2, they could induce GC at an adult stage, permitting them to follow the tumorigenesis over time. Taken together, their mouse models hold many promises, but one has to wait to see if they will also succeed in generating EBV and MSI GC subtype mouse models.
In addition to these sophisticated mouse models, other published models have also shown the role of p53 in the tumorigenesis of GC. For example, heterozygotes mice for tp53+/− and lkb1+/− develop more polyps in the pylorus region than lkb1+/− mice only, 92% vs. 69%, respectively, indicating their cooperation in GC development [279]. The deletion of tp53 and rb in LGR5/CD44 cells strongly induced the development of gastroesophageal carcinomas in the gastric epithelial junction and a low frequency (10%) of benign tumors, adenomas, in the antral region [280]. Transgenic mice expressing mutant p53 V143 develop a higher percentage of GC (20% vs. 4%) when treated with 4-nitroquinoline-1-oxide [281]. As expected, this increased tumorigenesis correlates with a decreased expression of genes involved in cell cycle inhibition and apoptosis. Interestingly, it also correlates with a decrease in genes involved in inflammation (ex., tnfa and cxcl9) and the immune response (ex., irf1, 3, and 7). Finally, the deletion of tp53 in a wnt1-activated background induces the EMT (Epithelial Mesenchymal Transition) and gastric epithelial cell transformations with the cooperation of the microenvironment via COX2 activation [282]. Although not directly addressing the role of p53 in GC, Guo and coworkers demonstrated that the loss of pten (SP-A-Cre; ptenflox/flox) induces differentiated GC within 80 days depending, in part, on the expression of p53 [283]. More precisely, they demonstrated that the loss of pten leads to an upregulation of Akt activity, which represses the p53 expression, resulting in a decreased expression of miR-365, as it is a target gene of p53. Consequently, there is an increased expression of two of its target mRNAs: cyclin D1 and cdc25 and enhanced proliferation. Relevantly, the miR-365 expression is reduced in GC in patients, which inversely correlates with the pten expression (r = 0.68; p < 0.001).
Mouse genetics has brought interesting observations about the relationship between gastric tumorigenesis and mutations of TP53, pointing to their importance for the development of intestinal/CIN carcinogenesis. However, no information exists about p53 isoforms or mouse inactivated for p63 or p73 isoforms regarding gastric tumorigenesis, despite the fact that several studies pointed to their alterations in GC.

3.9.3. Deregulation of the p53 Pathway by H. pylori

An infection with H. pylori is a major factor causing GC with intestinal and chromosomal instability characteristics (CIN) that predominantly display TP53 mutations. In mice, p53 is a protector against H. pylori-induced proliferation during gastritis but does not seem to provide protection against GC development, possibly due to a reduced Th1 immune response [284,285]. In Mongolian gerbils, an infection with H. pylori induced a dynamic change of the p53 protein levels in the gastric tissues, peaking four hours after infection but decreasing to normal level after 12 weeks [286]. In vitro, an infection with H. pylori or expression of the H. pylori protein VacA toxin in GC cells inhibits cell growth, induces apoptosis, and causes a transient increased p53 expression at two hours with a subsequently progressive decrease up to 72h conjointly with the p53 target genes (P21) [287,288]. Inversely, the p73 expression increases to reach a peak at 72h, but the identity of the isoform, TA or ∆N, remains to be characterized.
The delayed reduction of the p53 protein level is caused by degradation involving multiple mechanisms. For instance, the CagA protein secreted by H. pylori induces mechanisms that destabilize p53—namely, the phosphorylation of MDM2 by AKT [286], the induction of an ARF-BP1 ubiquitin ligase complex [289], and the subversion of ASPP2 to target p53 for degradation [290]. Inversely, H. pylori represses the expression of the mechanisms that stabilized p53, such as E-cadherin that destabilizes the p53/MDM2 interactions [291], USP7 that deubiquitinates and stabilizes p53 [292], and USF1 (promoter methylation) that reduces the USF1 ability to interact with and stabilize p53 in the nucleus by competing with MDM2 [293]. Typically, the silencing of E-cadherin by siRNA restores the p53 expression that correlates with the induction of NOXA and P21. The importance of the p53/MDM2 interaction in CagA activity is illustrated by the fact that the inhibitor of the p53/MDM2 interaction, Nutlin-3, restores the p53 protein level after CagA exposure.
Besides affecting the p53 expression, a H. pylori infection increases the expression of the p53 isoform ∆133p53 at the transcriptional level via an AP1-binding site targeted by c-Jun [294]. The expression of ∆133p53 inhibits the activity of p53 and p73 and induces the NF-kB pathway (p53 expression and target genes IL-6, IL-8, and BCL2) by inhibiting the IKK expression to reduce apoptosis.

3.9.4. Deregulation of the p53 pathway by the Epstein–Barr Virus

The Epstein–Barr virus (EBV) is associated with several human malignancies, including GC [295,296,297]. Infection by the EBV defines a specific molecular subclass that represents about 9% of GC (Figure 1). However, it has been suggested that the EBV subgroup might be heterogeneous. For instance, EBV infection (detected by EBER staining) is of poor prognosis (Hazard ration, HR 2.9) in GC with the intestinal characteristics and of good prognosis (HR 0.4) in GC with diffuse characteristics [298]. However, EBV tumors have no TP53 mutation, and the EBV infection (viral EBER1 mRNA) is homogeneously present in the tumor, suggesting that the infection is an early event in the carcinogenesis process [299,300,301,302]. If EBV infection is exclusive of TP53 mutations, in contrast to H. pylori, few of the 70 open reading frames (ORFs) contained by the 184-kb linear double-stranded DNA EBV genome, which code for latent and lytic viral proteins, can have an impact on p53 activity [296]. For instance, the EBV viral protein EBNA1 downregulates p53 activity via USP7 (a p53 regulator) and PML bodies, reducing the response to DNA damage in GC cells [303,304]. In other types of EBV-induced cancers, additional viral proteins were shown to be involved in p53 inhibition. For instance, the EBV immediate early transcription factor BZLF1, which is the main regulator of the viral life cycle by controlling the switch between its latent and lytic stages, interacts with p53. This interaction allows the binding of p53 to the elongin BC–cullin 5–SOCS box ubiquitin ligase complex, causing p53 degradation independently of MDM2 [305]. In addition, the latent phase viral protein EBNA3C interacts directly with the p53 at its C-terminus, reducing the binding of p53 to its target genes and some of its regulators, such as MDM2 [306]. In addition to proteins, EBV encodes about 44 miRNAs and circular RNAs. Some target the p53 mRNA, such as miR-BART5-3p [307] and miR-BART3-3p [308]. The EBV-encoded circular RNA ebv-circLMP2A plays a crucial role in maintaining stemness phenotypes through targeting the miR-3908/TRIM59/p53 axis, significantly increasing the metastasis and correlating with a poor prognosis [309].
However, the relationship between EBV and p53 is more complex. In EBV latently infected cells, the LMP1 viral protein can promote p53 accumulation by reducing its ubiquitination mediated by MDM2 at two sites, K48 and K63, and the interaction with tumor necrosis factor receptor-associated factor 2 (TRAF2) [310]. These mechanisms may explain an increase in the p53 protein level despite a reduction of TP53 mRNA in some EBV-infected GC [311]. In addition, the functional interaction between the EBV and p53 is reciprocal, as p53/ATM [312] and the hypoxia inducible factor HIF1A [313] participate in the reactivation of the viral lytic phase through their binding to a hypoxia response element (HRE) located within the promoter of the EBV latent-lytic switch BZLF1 gene Zp.
Finally, it is of note that EBV is not the only virus associated with GC. Indeed, the lesser-known JC virus is also suspected to cause GC. In this case, the transformation process seems to involve the multifunctional viral protein T-Ag that inhibits p53 [314,315].

3.10. Interactions of p53 with Oncogenic and Antitumoral Signaling Pathways in GC

3.10.1. Interplay between p53 and HIF1A/Angiogenesis Mechanisms

As p53 is a key player in the cellular stress response, its role in the response to hypoxia has been extensively investigated—in particular, its interaction with the hypoxia inducible factor HIF1A. HIF1A is a key transcription factor that allows the cancer cells to adapt to the hypoxia environment present within the tumor in order to stimulate neo-angiogenesis, via the induction of VEGF, and to redirect the oxidative metabolism towards a glycolytic metabolism—via the induction of GLUT1, for example. The relationship existing between HIF1A and p53 is complex and has been detailed in previous reviews (ex., [316]). In this review, we are focusing specifically on what has been observed in GC. In GC, several studies showed that a high p53 expression, possibly caused by TP53 mutations, correlated with elevated angiogenesis (anti-CD34) and necrosis [317,318,319] and is indicative of a poor prognosis [133]. At the molecular level, hypoxia stabilizes HIF1A while reducing the p53 and VHL protein levels in AGS cells expressing WT p53 but not in MKN28 cells expressing mutated p53 [320]. This favors a resistance to 5-FU chemotherapy [321]. In addition, HIF1A favors NF-kB binding on the promoter of the antiapoptotic genes cIPA1 and A20. However, p53 expression also affects HIF1A, as p53 mutants in GC cells cause a hypoxia response and favor cell growth [88].
In addition to hypoxia, other metabolic stresses impact p53. For instance, deoxycholic acid (DCA), an inhibitor of the PK2 enzyme, induces p53 protein levels in SGC-7901 cells, which correlate with a change in the BAX/BCL2 ratio and cell death [322,323].

3.10.2. Interplay between p53 and NF-kB

There is a complex physical and functional interaction between p53 and the NF-kB pathway, a pathway important to control the immune response, notably via interleukins and interferons. However, little is known of its interaction with p53 in GC. Initially, it was shown that an inhibitor of NF-kB nuclear translocation, SN50, can induce p53 and PUMA protein levels in GC cells [324]. More precisely, SN50 induces p53 at the mRNA level, which contributes to the induction of autophagy via BECLIN 1 [325]. It was also shown that inhibition of the NF-kB inhibitor, IKK2, leads to cell death and reduced migration in GC cells in vitro and in vivo, which correlates with induction of p53 and caspase 3 cleavage [326]. Inversely, STAT3, an activator of the NF-kB pathway, is a resistance factor toward chemotherapy in GC cells [327]. These studies indicate that NF-kB can repress p53 expression and activity in GC, at least partly, but NF-kB can also induce p53. In the MKN45 cells, 5-FU can induce NF-kB, and the silencing of a NF-kB family member RELA reduces the p53 protein levels and expression of p53 target genes (ex., PUMA) [328]. Similarly, NF-kB activation by Trastuzumab via AKT and GSK3β upregulates p53 and PUMA to induce NCI-N87 cell apoptosis [329]. Supporting the possibility that NF-kB may upregulate p53 is the finding that an elevated expression of miR18a in GC cells represses IRF2 (interferon response factor 2), an activator of NF-kB [330], decreasing the p53 protein levels, leading to higher cell motility and correlating with a poor prognosis [331].
These data highlight the complexity of the interactions between p53 and the NF-kB pathway, likely reflecting the importance of the physiological context. Understanding these interactions is particularly crucial, as they may provide key information to explain how the immune landscape of GC expressing p53 mutants is deprived of immune cells compared to tumors with WT p53.

3.10.3. Interplay between p53, the Hippo Pathway, and the Extracellular Matrix

The Hippo pathway has been widely described in different types of cancer to be altered and involved in tumor aggressiveness [332]. It is a mechano-transduction pathway that relays mechanical stress signals from the membrane towards the nucleus to drive adaptative gene expression. Hence, it has been connected to tight junction and extracellular matrix signaling. Several studies on GC have shown that the silencing of activators (e.g., MST1/2) of the Hippo pathway leads to nuclear accumulation of the oncogenic transcriptional cofactor YAP, favoring tumor aggressiveness and metastasis [333]. However, there are only two studies describing a functional interaction between p53, or a p53 family member, and the Hippo pathway in GC. The most interesting, however surprising, shows that mutations in p53, when associated with an elevated expression of the oncogenic YAP protein, favor GC patient survival [91]. The molecular mechanism mediating this counterintuitive association remains unclear. One explanation proposed by the authors is that the pro-proliferative activity of YAP and mutant p53 facilitates the activity of cytotoxic drugs. The second study suggested that a chemical, DIM, extracted from a Chinese vegetable has anticancer activity that correlates with an increased p53 protein level and activation of the Hippo pathway in GC cells [334]. Unfortunately, this limited information does not allow a good understanding of the functional interactions between the p53 family and the Hippo pathway in GC, highlighting the need for additional studies.
However, several components of the extracellular matrix or the tight junctions, which interact with the Hippo pathway, can impact p53. For instance, Cadherin 17 was found to be overexpressed in GC, and its silencing induces p53 protein levels to inhibit cell growth [335]. Similarly, the inhibition of E-cadherin expression by H. pylori induces p53 protein levels and the expression of its target genes (NOXA and P21) [291]. This inhibition is mediated by the MDM2-ERK pathway and the H. pylori CagA protein. In contrast, the extracellular matrix protein Periostin increases the p53 protein levels in GC cells, which is correlated with a decreased proliferation [336].
In summary, the activity of p53 seems to be regulated by extracellular signals caused by extracellular matrix components, tight junctions, and cell adhesion elements. However, whether this is mediated by the Hippo pathway remains an open question.

3.10.4. Interplay between p53 and Epigenetic Effectors (HDAC, HAT, and MET)

The p53 protein has been shown to be the subject of multiple and complex post-translational modifications by several epigenetic effectors, such as histone acetylases (HAT) or methylases, which participate in the tight control of its activity [337]. Numerous studies have demonstrated that several of these epigenetic effectors are deregulated in cancers and can be targeted by various chemicals as potential therapeutic approaches. Surprisingly, only a small part of these findings has been investigated and confirmed in GC. For instance, the expression of p53 target genes (i.e., P21, BAX, BAK, PIG3, NOXA, and PUMA) is induced by two histone deacetylase (HDAC) inhibitors (HDACi): TSA and sodium butyrate, in different GC cells [338,339,340]. This activation is mediated by acetylation at K320 and K373 on p53 [339], or the K382 following inhibition of HDAC1, induced by the silencing of RNT overexpressed in GC [341]. The induction of PUMA [340] and DTWD1 [342] by TSA is p53- and HDAC3-dependent. A low expression of PUMA and DTWD1 correlates with a poor prognosis, while TSA may represent an interesting therapeutic approach. However, TSA, as well as sodium butyrate, are pan-HDACi and, therefore, are not selective towards a specific HDAC, increasing the risk of side effects and/or complex antagonistic effects between different HDACs. Hence, more selective inhibitors have been developed. For instance, HDAC4 has an elevated expression in GC, and its alteration (mutation or deletion) correlates with a better prognosis [343]. The inhibition of HDAC4 by small chemicals enhances cisplatin-induced apoptosis in vitro and in vivo. This is partly mediated by p53, p73, and their target gene BIK [343]. Similarly, the elevated expression of the HDAC SIRT1 in GC correlates with a poor prognosis (p = 0.008) and with a low expression of p53 [344]. The inhibition of SIRT1 with Tenovin-6 induces p53 acetylation and DR5 in GC cells [345].
The exact impact of HDACi on p53 protein levels is controversial, as an early study showed that, although it induces p53 target gene expression, TSA decreases the p53 protein level in GC cells [338]. Similarly, the silencing of HDAC4 reduces the p53 protein levels [343]. However, another study contradicted these results by showing the synergistic effects of cotreatments between the HDACi TSA and chemotherapies (5-FU, oxaliplatin, and paclitaxel) to increase p53 expression in the MKN74 cells [346]. Another HDACi, FK228, also acts in synergy with the overexpression of p53, requiring BAX and NOXA [347].
The chemical inhibition or silencing of methylases leads to growth arrest or apoptosis in GC cells, and p53 seems to be involved, but the exact molecular mechanisms are still debated. For instance, the pan-methylases inhibitor 5-AZA-cdr induces cytotoxicity and p53 mRNA and the protein levels in MKN45 cells [348,349]. However, the reverse was also observed [350], as well as the involvement of a post-translational modification of p53 at S15 caused by ATM in AGS cells [351]. The selective inhibition (using siRNA or a selective inhibitor 3-Deazaneplanocin A, DZNep) of the histone methyltransferase EZH2 of the polycomb PRC2 complex also induces cytotoxicity in GC cells via p53 [352,353]. At the molecular level, EZH2 inhibition increases the p53 and MDM2 protein levels via the reduction of ubiquitination, inducing p53 target gene expression (DR5, P21, FAS, and GADD45α). In addition, the expression of EZH2 in GC inversely correlates with the activity of the p53 pathway (gene signature) [352,353]. Interestingly, the histone demethylases JMJD2B [354] and KDM5C [355] are overexpressed in GC, and their silencing induces the p53 and P21 proteins in AGS cells, causing cell death in a p53-dependent manner. However, how the p53 protein level is regulated by these two histone methylases is not known. Inversely, another histone demethylase, PHF2, interacts directly with p53 to induce its activity on its target genes (P21 and MDM2) [356]. This also seems to have clinical relevance, since the expression of PHF2 is downregulated or mutated in GC [357].
In summary, it appears that different inhibitors of HDACs, histone methylases, or histone demethylases can represent interesting strategies to treat gastric cancers. However, the molecular mechanisms involved are still largely elusive—in particular, regarding p53—handicapping their use for personalized therapy.

4. Developing the Magic Bullet to Target p53 Family Alteration in Gastric Cancer

Based on its critical role in GC, p53 has been investigated as a potential therapeutic target. The first approach used in vitro and in vivo was the adenovirus transduction of WT p53 (AdCAp53) into MKN1 cells [358] (Figure 5). AdCAp53 induces growth inhibition and apoptosis in MKN1 p53-mutated GC cells but not in cells expressing WT p53. The activity of the transduced p53 can be increased by treating the cells with an HDAC inhibitor (Sodium butyrate) [359]. Importantly, AdCAp53 acts in synergy with chemotherapies (e.g., oxaliplatin) in vitro and in vivo [360,361]. Preclinical proof of concept has been published in GC, but this strategy (GendicineTM and OncorineTM) is only used in China to treat head and neck cancers. Clinical trials for other indications are ongoing. As alternative, the transduction of an adenovirus expressing the ribosomal protein RPL23, which inhibits the interaction between p53 and MDM2, was performed in GC cells to induce cell death in vitro and reduce tumor growth in vivo [362]. Later, a bicistronic virus co-expressing TP53 and RPL23 was developed and showed a higher efficacy in vitro and in vivo than Ad-p53 alone, including in a GC orthotopic model [363]. An alternative to directly targeting p53 is to use downstream targets of p53. For instance, an adenovirus expressing BAX shows a high efficiency in killing GC cells, even if they are resistant to p53 overexpression [364]. Similarly, an adenovirus expressing PUMA under the control of the β-catenin TCF-controlled promoter significantly increases chemotherapy cytotoxicity in AGS cells in vitro [365]. The p53 family members have also been targeted. An adenovirus expressing TAp63γ (p51A) was shown to reduce the viability in GC cells (MKN45 expressing p63γ and MKN28 non-expressing p63γ) in vitro [366]. These p53 gene therapy approaches have been validated for some cancers in China, but they are not used in Western countries.
Alternative strategies have been chosen in Europe and the US by focusing on small chemicals targeting p53 regulators or p53 mutants. For instance, the first inhibitors developed for clinical use were inhibitors of the interaction between p53 and MDM2, Nutlins, that bind to the p53-binding pocket of MDM2, resulting in p53 accumulation. The most common, Nutlin-3, induces a strong toxicity (IC50 ± 2–3 µM) in GC cells in vitro and in vivo [367]. In contrast, Nutlin-3 showed no cytotoxicity in mutated p53 cells (NUGC-3) up to 20 µM. Nutlin-3 stabilizes p53 and MDM2 after four to eight hours of treatment (10 µM) in p53 WT GC cells but not NUGC-3 (p53 Y220C), leading to apoptosis. Nutlin-3 seems to have synergistic effect with cisplatin or 5-FU. In vivo, Nutlin-3 reduced MKN45 xenografts growth (−40%; 40 mg/Kg) slightly less than 5-FU (−60%; 40 mg/Kg), but combining Nutlin-3 and 5-FU blocks tumor growth (−80%). Nutlin-3 has been the leading compound from the Nutlin family and the base for the development of the derived compounds, such as RG7112, which has been tested in phase I for leukemias. Another MDM2-p53 interaction inhibitor, APG-115, induces cell cycle arrest and apoptosis in GC cells and acts synergistically with radiotherapy [368]. An alternative strategy is to inhibit p53 nuclear export by using KPT330, which targets XOP1 (exportin1) often overexpressed in tumors and associated with a poor prognosis for patients [369]. KPT330 favors apoptosis of GC cells and acts synergistically with irinotecan to inhibit tumor growth in vivo.
The forced activation of p53 by inhibiting the p53/MDM2 interaction is an interesting approach, but it raises questions on the possible side effects on healthy tissues, since p53 is ubiquitously expressed. Hence, a promising approach consists of targeting p53 mutants selectively, as this may solely impact cancer cells, preserving healthy cells. However, the limit of this approach is that each mutation has a different impact on p53 structure and function [95]. Thus, a library of molecules able to target the various mutations is needed. For instance, p53 Y220C harbors a mutation in the cysteine that participates in an SH-stabilization of the Zinc, which forms the Zinc finger of the p53 DNA-binding domain. PK7088, developed by Drs. J. Spencer and A. Fersht, partially restored p53 Y220C activity in NUGC-3 GC cells when applied at high concentration (200 µM) [370]. To reduce the quantity of drugs required, we developed a bifunctional molecule able to interact with the mutant p53-Y220C and to restore zinc binding to p53 [371]. This molecule, L5, induces death in GC cells partly via reactivation of p53 Y220C function. However, it suffers from off-target effects, as cytotoxicity is also observed in cells expressing WT p53. This led us to optimize the structure and design novel compounds based on L5 by targeting zinc loss and protein aggregation in p53 mutants. One of these compounds was found to be cytotoxic in GC cells expressing p53 Y220C while having no toxicity on healthy organoids [372]. Another type of molecules targeting p53 mutants, variants of CP-31398, have been tested in GC [373]. For instance, the variant 10ah showed an interesting cytotoxicity in vitro and in vivo, reactivating the expression of BIM, BAK, and BAX. However, the selectivity of these complexes for the p53 mutant is relative, since additional mechanisms, such as reactive oxygen species, seem to be involved independently of p53 [374]. Another small molecule, NA20, a naphthalimide derivative, exhibits a selective growth inhibition of p53 mutant GC cell lines [375]. After coimmunoprecipitation and mass spectrometry (Co-IP/MS), it was found that NA20 binds directly to p53 and is able to target DNA to trigger cytotoxicity. More precisely, NA20 induces cell cycle arrest and apoptosis, which correlates with the activation of p21 and caspase 3/9 and inhibits tumor growth.
Altogether, besides the use of adenoviruses to induce p53, relatively little preclinical proofs of concepts have been provided for targeting the p53 pathway in GC models. In addition, some of the molecules tested have significant off-target effects. This highlights the necessity of further investigations using in vivo models and detailed verification of the mode of action of these strategies and the development of more selective compounds.

5. TAp73 and ∆Np73, the Yin and the Yang of Gastric Carcinogenesis

As a member of the p53 family, p73 presents two major types of isoforms (Figure 2). The TAp73 isoforms have a transactivation domain in the N-terminal and are generally considered to be proapoptotic. The ∆Np73 isoforms lacking the N-terminus transactivation domain are considered to be anti-apoptotic with a dominant negative function over TAp73 and p53. These two types of isoforms are generated by alternative promoters [23]. Each of these two types of isoforms have additional variants in the C-terminus that are generated by alternative splicing. It is generally accepted that ∆Np73 has oncogenic properties and TAp73 has tumor suppressor function, such as induction of apoptosis in response to DNA damage [23,376]. However, these properties remain debated. Interestingly, the pro-apoptotic function of TAp73 can be inhibited by a direct interaction with p53 mutants [377]. Based on its role in the response to therapy and tumorigenesis, the alterations of p73 in GC have been investigated.
As observed in other types of cancers, p73 is very rarely mutated in GC [378,379]. For instance, our analysis of the TCGA indicates that 5 tumors out of 440 show mutations in TP73, located at A79T and A79V in the transactivation domain, P483L in the DNA binding domain, D611G just before the SAM domain and a non-sense mutation at position 205 within the DNA binding domain (personal data). These mutations are present in all molecular subgroups of GC, suggesting that their occurrence is not a consequence of the high mutational context of the MSI subgroup. However, the impact of these mutations on p73 function is unknown, but their presence in the DNA binding domain may drastically diminish p73 function, as seen for p53 or p63 [23]. Similarly, mutations in the transactivation domain might reduce p73 transcriptional activity. Beside somatic mutations, a polymorphism C4C14-to-A4T14 at exon 2 of TP73 has been described in several cancers. This polymorphism does not seem to show any significant correlation with the risk of GC [380,381,382,383,384]. However, one study points out the possibility that the homozygous genotype favors GC of the intestinal subtype among the Caucasian population [385].
If p73 is rarely mutated, changes in its expression pattern have been observed in GC. A first detailed investigation indicated that p73 expression is detected in 50% of GC [386]. In addition, p73 has an elevated expression in adjacent mucosa of the tumors, suggesting a role in inflammatory tissues [387]. An elevated expression of p73 correlates with the loss of Reprimo due to methylation and lymph node metastasis [388]. Unfortunately, these studies did not clarify whether it was TA or ∆Np73 isoforms that were involved. A more detailed analysis showed that ∆Np73 can be found overexpressed in gastric tumors (62%, 15 out of 24) [389], which was confirmed by two additional studies [390,391]. This overexpression of ∆Np73 decreases the transcriptional activity of TAp73 on reporter genes [392] and “typical” p73 target genes, but induces the expression of the drug resistance gene MDR1, via a direct binding of ∆Np73 on the MDR1 promoter [390]. In contrast, ∆Np73 increases the β-catenin protein level and the activity of TCF-driven transcription. Importantly, ∆Np73 overexpression correlates with a poor prognosis in GC patients (20 months vs. 47 months) [391]. Hence, ∆Np73 seems to present oncogenic properties in GC and might impact on drug activity.
At the molecular level, one mechanism explaining the elevated expression of ∆Np73 in GC is the loss of HIC1 that negatively regulates the ∆Np73 promoter [391]. However, the correlation between HIC1 and ∆Np73 expression in GC is relatively weak (r = 0.22, p = 0.2), in contrast to what is observed in esophagus cancers (r = 0.68, p = 0.03). Similarly, our own analysis of the TCGA shows no correlation (r = −0.07) between the mRNA levels of TP73 and HIC1 in GC (personal data), suggesting that other mechanisms should be involved. For instance, expression of ∆Np73 can be induced by gastric acid via pro-inflammatory cytokines (TNFα, IL-1β) [393]. In fact, only a few studies have focused on the regulation of p73 expression in GC. For instance, without identifying which isoform, it was shown that p73 expression is increased by serum starvation, inhibition of methylases (5-Aza-DC), and the extracellular matrix protein Periostin in GC cell lines [336,394].
The effect of methylase inhibitors might be explained by the methylation of p73 promoter in EBV infected GC [395,396]. EBV impacts also on TP73 at the protein level. For instance, the viral protein EBNA3C directly interferes with p73 in the nucleus to stabilize ΔNp73 [295]. In addition, elevated expression of ∆Np73 is caused by an increased binding of p73 on ∆Np73 promoter consecutive to the displacement by the EBV protein LMP-1 of the repressive polycomb 2 complex component EZH2 and the induction of epigenetic changes by JNK-1. As these protein-protein interactions have been described in other types of cancer, they remain to be confirmed in GC cells.
The expression and activity of p73 is also affected by H. pylori, contributing to the response of the gastric epithelium to the infection. For instance, H. pylori infected tissues show a strong p73 staining [397]. In vitro, the infection leads to induction of TAp73 protein stability (but decreases the mRNA levels) and activation of various target genes such as PUMA, NOXA and FASR in GC cells and primary mouse gastric epithelial cells. In parallel, the infection decreases the binding of TAp73 to ∆Np63 due to a decrease of ∆Np63 protein and mRNA levels. The elevated expression of TAp73 is necessary for the infection to induce cell death. The increase in p73 protein levels is progressive and peaks at 72h post-infection in AGS cells [286]. Hence, TAp73 displays anti-tumoral properties to protect the gastric epithelium from H. pylori-induced transformation. However, the relationship between H. pylori and p73 seems to be more complex, as H. pylori can also decrease TAp73 transcriptional activity through the induction of the isoform ∆133p53 [294].
P73 interacts with another oncogenic pathway, the TGF-β pathway [398]. The transforming growth factor-β (TGF-β) is a multifunctional cytokine involved in several biological processes such as cell differentiation, proliferation, and apoptosis. In GC, mRNA level of the ligands TGF-β1/2 are significantly higher [399] and TAp73α participates in a TGF-β-induced apoptosis [400]. At the molecular level, TGF-β increases the TAp73 protein level, the TP73 overall mRNA level, and TP63 to a lesser extent. Induction of TAp73 induces BAX and PUMA, which is required for TGF-β-induced apoptosis. Hence, these studies suggest that TAp73 has antitumor activity by transducing the effect of TGF-β.
It has been accepted that TAp73 is a mediator of the response to chemotherapy in various biological systems. DNA damage induces TAp73 activity via phosphorylation by the tyrosine kinase cAbl [401] and increases TAp73 mRNA levels through an upregulation of E2F1 after its activation by the DNA damage inducible kinases CHK1/2 [402,403]. This was also observed in non-cancerous cells [376]. Similarly, induction of TAp73 expression upon cisplatin treatment was observed in GC cells [343]. In this case, the expression of TAp73 is partly positively controlled by HDAC4. Interestingly, HDAC4 expression is elevated in GC, especially in the diffuse subtype, correlates with poor prognosis, and its expression is downregulated upon treatment with cytotoxic drugs. Hence, HDAC4 participates in a complex regulatory control of p73 expression in GC in response to chemotherapy. However, this precept has been questioned recently in GC, as Dr. Qiang showed that overexpression of TAp73 seems to inhibit BAX and NOXA expression, to increase Cyclin D1 expression, and confers resistance to 5-FU and doxorubicin [404,405]. In addition, the protein expression of TAp73 appears to be downregulated in AGS cells upon 24h of treatment with these drugs. This hypothesis of a pro-survival activity of TAp73 has also been brought forward by two additional studies suggesting that TAp73 may play a role in GC cell metabolism. For instance, the casein kinase 2 inhibitor CX-4945 elicits an anti-Warburg effect and inhibits gastric tumorigenesis through the downregulation of TAp73 [406]. However, this study has to be taken with precaution as some results are difficult to interpret. Nonetheless, this pro-survival function of TAp73 fits with the observation that TAp73-induced phospho-fructokinase-1 transcription promotes the Warburg effect and cell proliferation [407]. Hence, the exact role of TAp73 in GC tumorigenesis and aggressiveness remains debated, highlighting the need for further and more precise investigations.
In summary, ∆Np73 seems to have an elevated expression in GC, which correlated with a poor prognosis and inhibition of p53/TAp73 activity. This is in agreement with the hypothesis that ∆Np73 has oncogenic properties in GC. However, the causes of the ∆Np73 overexpression during tumorigenesis are unclear as well as the transcriptome that is under ∆Np73 control. Therefore, there remains a lack of understanding of the exact role of TAp73 due to conflicting data. Overall, additional studies are required to fully understand the molecular events that linked TA and ∆Np73 to GC tumorigenesis and aggressiveness.

6. TP63, a Third Wheel Yet to Be Explored

TP63 is the ancestral member from which gene duplication occurred to produce TP73 and TP53 [23,408]. Hence, TP63 produces two types of N-terminus variants, based on two promoters generating TAp63 and ∆Np63 isoforms, and C-terminus variants due to alternative splicing (Figure 2). ΔNp63α has been described as the predominant isoform. The function of TP63 is mostly described as to be critical for the balance between proliferation and differentiation in epithelial tissues. Besides this physiological function, mutations, or deregulated expression have been associated with different cancers.
Tannapfel et al. were the first to investigate p63 expression using in situ hybridization and immunohistochemistry in GC [386]. They showed that p63 is expressed in about 40% of gastric tumors (n = 32) and is preferentially expressed in diffuse and high-grade tumors. In addition, expression was also seen in metaplasia and gastritis. These results were later confirmed showing that 48.5% of the tumors (n = 101) are expressing p63 [409]. As the expression of p63 in healthy stomach epithelium seems to be low [410], these observations suggested that p63 protein levels are increased upon the transformation process and tumor grade, and that it might be preferentially involved in the development of the diffuse subtype. However, it remains to identify which isoforms (TA or ∆N) are concerned by this increased expression in GC. It is of note that a partial analysis of GC cell lines with various degrees of differentiation suggested that ∆Np63 expression is higher in the poorly differentiated cell lines and overexpression of ∆Np63 induces GATA6 expression and proliferation in MKN28 cells [408].
∆Np63 mRNA and protein levels are strongly decreased in H. pylori-infected GC cells, while TAp63 remains unchanged [397]. The decrease in ∆Np63 expression reduces its interaction with TAp73, favoring an increase in the transcriptional activity of TAp73. Concomitantly, the TAp73β protein level is increased by stability, contributing to apoptosis by the induction of NOXA and PUMA. Hence, the silencing of ∆Np63 in GC cells stimulates apoptosis [408]. The inhibition of ∆Np63 expression seems to be mediated by an increased expression of MDM2 caused by the H. pylori lipopolysaccharide (LPS) via the downregulation of DICER and miR-375 that targets MDM2 [411].
TP63 may also be involved in EBV-mediated transformation. Indeed, elevated expression of ∆Np63 allows a prolonged latency phase of the virus cycle by directly inhibiting the expression of BARF1 [412]. BARF1 is considered a viral oncogene in epithelial cells, has immune-modulating properties and is expressed as an early gene regulated by the immediate early EBV protein R during viral lytic replication. The repression of BARF1 by ∆Np63 could explain why the latency phase is restricted to esophageal and GC since ∆Np63 expression is selectively elevated in those cancers, but not in lymphomas.
In summary, p63 expression is induced in GC and absent in normal gastric epithelium. This expression is specifically increased in diffuse and advanced cancers and is affected by H. pylori and EBV infection. Most of the existing information about the expression and role of p63 concerns ∆Np63 showing elevated expression and that it could inhibit the activity of TAp73 in vitro. However, no information exists on TAp63 or any type of physical or functional interaction with p53 or p53 mutants. In addition, there are no animal studies indicating any function of p63 in GC. Hence, additional studies are required to understand the exact function of the different isoforms of p63 in GC aggressiveness.

7. Discussion and Perspective: p53 and Its Galaxy in Gastric Cancer

Since its discovery in the 90s, the p53 family has been intensively investigated and a vast quantity of information regarding fundamental molecular mechanisms and translational/applied observations have been published. It is clear that in various cancers TP53 and its paralogues are playing an important role in tumorigenesis and the response to chemotherapies, and likely to other therapies as well.
In gastric cancer, the presence of TP53 mutations at an early stage and its presence homogeneously within the tumor indicates a key role in gastric epithelium tumorigenesis and should be considered as “driver” mutations. These mutations, in particular 5 hot spot mutations (R175H, Y220C, R248Q/W, R273C/H/S, and R282W), impact on multiple levels on the patient by favoring a hypoxic response, impairing the immune tumoral landscape, increasing metastasis, reducing the response to chemotherapy, thus impacting negatively on patient prognosis. However, the importance of these mutations varies depending on the histological and molecular subtype of GC (Table 1). For instance, it is likely that TP53 mutations are more important in the intestinal/CIN subgroup, as supported by mouse genetics and patients’ data. However, we should not overlook that p53 mutants are also present in other subtypes and that p53 can be inactivated by multiple different means, including viral oncogenes, amplification of inhibitors and methylation of positive regulators. The exact importance of p53 isoforms, as well as p63 and p73 isoforms, remains an open question since no mouse genetic experiments have yet supported any in vitro or patient-based observations. In particular, the exact role of TAp73 remains debated. It is clear, however, that p53 family isoforms are at the center of multiple pathways in GC, including HIF1A, Hippo/YAP, TGF-β, TNF-α and NF-kB, which can account for the profound and diverse impact of p53 mutants on the tumor characteristics.
The data gathered over the years clearly allows a better understanding of the molecular mechanisms causing stomach tumorigenesis and gastric cancer elevated aggressiveness. However, these data have not been used for daily clinical routine in treating gastric cancer. The explanation of this relative failure to transfer to the clinic for the treatment of gastric cancer may reside in two facts. The first fact is the inherent complexity of the p53 family that contains more than 30 different isoforms, with sometimes opposite functions, and that participate in a multitude of signaling pathways. The second fact is the imperfections of the tools and techniques we are using to approach and investigate this complex family. Similarly, the access to tumor samples in sufficient number and diversity, their quality and their analysis using immunohistochemistry techniques that are antibody- and experimenter-dependent may also explain some contradictory results that impact translation to the clinic. In addition, the therapeutic strategies developed have been limited by the complexity of developing highly selective small chemicals targeting non-enzymatic proteins, the diversity of the mutations/alterations, the lack of pertinent animal models, and the length and cost of clinical trials. Finally, the use of the p53 family as biomarkers or in therapeutic approaches is also confronted by the lack of alternative therapies to chemotherapy in GC, as in several other types of cancer.

8. Conclusions

Hence, a long road is still ahead of us in further understanding this complex family. This might require the development of innovative tools, such as highly selective nanobodies for p53 family isoforms, functional assays that can be applied to tissue samples or liquid biopsies, and automated-immunohistochemistry scanning coupled to artificial intelligence. It is also likely that the increasing use of pan-genomics and -proteomics approaches at the individual cellular level will contribute significantly in providing crucial information in the understanding of the functions of isoforms of the p53 family in the complex ecosystem developed by the cancer cells. In particular, dissecting how isoforms of the p53 family impact their environment via the expression of diffusible factors and membrane ligand/receptors, defining an “exteriome”, might provide clues to explain their impact on the immune landscape in the tumor ecosystem and provide strategies to improve the response to immunotherapy.

Author Contributions

Conceptualization, C.G.; writing—original draft preparation, A.B. (Anais Blanchet), A.B. (Agathe Bourgmayer), G.M., and C.G.; writing—review and editing, A.B. (Anais Blanchet), A.B. (Agathe Bourgmayer), G.M., J.-E.K., and C.G.; and funding acquisition, C.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Inserm, CNRS, Ligue Contre le Cancer, Itmo Cancer, Idex Unistra, Alsace Contre le Cancer. This work of the Interdisciplinary Thematic Institute InnoVec, as part of the ITI program of the University of Strasbourg, CNRS and Inserm, was supported by IdEx Unistra (ANR-10-IDEX-0002), and by SFRI-STRAT’US project (ANR-20-SFRI-0012) under the framework of the French Investments for the Future Program.

Data Availability Statement

Publicly available datasets were analyzed in this study. This data can be found here: https://www.cbioportal.org/study/summary?id=stad_tcga_pub (accessed on 31 November 2020).

Acknowledgments

The authors thanks E. Mathern for her administrative work that supported our research and Nicole Clark for the English proofreading of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lane, D.P.; Crawford, L.V. T antigen is bound to a host protein in SV40-transformed cells. Nature 1979, 278, 261–263. [Google Scholar] [CrossRef]
  2. Linzer, D.I.; Levine, A.J. Characterization of a 54K dalton cellular SV40 tumor antigen present in SV40-transformed cells and uninfected embryonal carcinoma cells. Cell 1979, 17, 43–52. [Google Scholar] [CrossRef]
  3. Rotter, V.; Boss, M.A.; Baltimore, D. Increased concentration of an apparently identical cellular protein in cells transformed by either Abelson murine leukemia virus or other transforming agents. J. Virol. 1981, 38, 336–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Jay, G.; Khoury, G.; DeLeo, A.B.; Dippold, W.G.; Old, L.J. p53 transformation-related protein: Detection of an associated phosphotransferase activity. Proc. Natl. Acad. Sci. USA 1981, 78, 2932–2936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Benchimol, S.; Pim, D.; Crawford, L. Radioimmunoassay of the cellular protein p53 in mouse and human cell lines. EMBO J. 1982, 1, 1055–1062. [Google Scholar] [CrossRef]
  6. Oren, M.; Maltzman, W.; Levine, A.J. Post-translational regulation of the 54K cellular tumor antigen in normal and transformed cells. Mol. Cell. Biol. 1981, 1, 101–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Bargonetti, J.; Friedman, P.N.; Kern, S.E.; Vogelstein, B.; Prives, C. Wild-type but not mutant p53 immunopurified proteins bind to sequences adjacent to the SV40 origin of replication. Cell 1991, 65, 1083–1091. [Google Scholar] [CrossRef]
  8. Baker, S.J.; Markowitz, S.; Fearon, E.R.; Willson, J.K.; Vogelstein, B. Suppression of human colorectal carcinoma cell growth by wild-type p53. Science 1990, 249, 912–915. [Google Scholar] [CrossRef] [PubMed]
  9. Soussi, T.; Wiman, K.G. Shaping genetic alterations in human cancer: The p53 mutation paradigm. Cancer Cell 2007, 12, 303–312. [Google Scholar] [CrossRef] [Green Version]
  10. Lane, D.; Levine, A. p53 Research: The past thirty years and the next thirty years. Cold Spring Harb. Perspect. Biol. 2010, 2, a000893. [Google Scholar] [CrossRef] [Green Version]
  11. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bickenbach, K.; Strong, V.E. Comparisons of Gastric Cancer Treatments: East vs. West. J. Gastric Cancer 2012, 12, 55–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef] [Green Version]
  14. Fukuzawa, K.; Noguchi, Y.; Yoshikawa, T.; Saito, A.; Doi, C.; Makino, T.; Takanashi, Y.; Ito, T.; Tsuburaya, A. High incidence of synchronous cancer of the oral cavity and the upper gastrointestinal tract. Cancer Lett. 1999, 144, 145–151. [Google Scholar] [CrossRef]
  15. Palli, D.; Caporaso, N.E.; Shiao, Y.H.; Saieva, C.; Amorosi, A.; Masala, G.; Rice, J.M.; Fraumeni, J.F., Jr. Diet, Helicobacter pylori, and p53 mutations in gastric cancer: A molecular epidemiology study in Italy. Cancer Epidemiol Biomarkers Prev. 1997, 6, 1065–1069. [Google Scholar]
  16. Fedriga, R.; Calistri, D.; Nanni, O.; Cortesi, L.; Saragoni, L.; Amadori, D. Relation between food habits and p53 mutational spectrum in gastric cancer patients. Int. J. Oncol. 2000, 17, 127–133. [Google Scholar] [CrossRef] [PubMed]
  17. Liu, X.; Cai, H.; Sheng, W.; Yu, L.; Long, Z.; Shi, Y.; Wang, Y. Clinicopathological Characteristics and Survival Outcomes of Primary Signet Ring Cell Carcinoma in the Stomach: Retrospective Analysis of Single Center Database. PLoS ONE 2015, 10, e0144420. [Google Scholar] [CrossRef]
  18. Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Yang, L.; Wang, Y.; Wang, H. Use of immunotherapy in the treatment of gastric cancer. Oncol. Lett. 2019, 18, 5681–5690. [Google Scholar] [CrossRef]
  20. Bosing, N.M.; Goretzki, P.E.; Roher, H.D. Gastric cancer: Which patients benefit from systematic lymphadenectomy? Eur. J. Surg. Oncol. 2000, 26, 498–505. [Google Scholar] [CrossRef]
  21. Song, H.; Zhu, J.; Lu, D. Molecular-targeted first-line therapy for advanced gastric cancer. Cochrane Database Syst. Rev. 2016, 7, CD011461. [Google Scholar] [CrossRef] [PubMed]
  22. Mirzaei, H.; Khataminfar, S.; Mohammadparast, S.; Sales, S.S.; Maftouh, M.; Mohammadi, M.; Simonian, M.; Parizadeh, S.M.; Hassanian, S.M.; Avan, A. Circulating microRNAs as Potential Diagnostic Biomarkers and Therapeutic Targets in Gastric Cancer: Current Status and Future Perspectives. Curr. Med. Chem. 2016, 23, 4135–4150. [Google Scholar] [CrossRef] [PubMed]
  23. Bourdon, J.C. p53 Family isoforms. Curr. Pharm. Biotechnol. 2007, 8, 332–336. [Google Scholar] [CrossRef] [PubMed]
  24. Sano, T.; Tsujino, T.; Yoshida, K.; Nakayama, H.; Haruma, K.; Ito, H.; Nakamura, Y.; Kajiyama, G.; Tahara, E. Frequent loss of heterozygosity on chromosomes 1q, 5q, and 17p in human gastric carcinomas. Cancer Res. 1991, 51, 2926–2931. [Google Scholar] [PubMed]
  25. Tamura, G.; Kihana, T.; Nomura, K.; Terada, M.; Sugimura, T.; Hirohashi, S. Detection of frequent p53 gene mutations in primary gastric cancer by cell sorting and polymerase chain reaction single-strand conformation polymorphism analysis. Cancer Res. 1991, 51, 3056–3058. [Google Scholar]
  26. Yamada, Y.; Yoshida, T.; Hayashi, K.; Sekiya, T.; Yokota, J.; Hirohashi, S.; Nakatani, K.; Nakano, H.; Sugimura, T.; Terada, M. p53 gene mutations in gastric cancer metastases and in gastric cancer cell lines derived from metastases. Cancer Res. 1991, 51, 5800–5805. [Google Scholar]
  27. Kim, J.H.; Takahashi, T.; Chiba, I.; Park, J.G.; Birrer, M.J.; Roh, J.K.; De Lee, H.; Kim, J.P.; Minna, J.D.; Gazdar, A.F. Occurrence of p53 gene abnormalities in gastric carcinoma tumors and cell lines. J. Natl. Cancer Inst. 1991, 83, 938–943. [Google Scholar] [CrossRef]
  28. Bartek, J.; Bartkova, J.; Vojtesek, B.; Staskova, Z.; Lukas, J.; Rejthar, A.; Kovarik, J.; Midgley, C.A.; Gannon, J.V.; Lane, D.P. Aberrant expression of the p53 oncoprotein is a common feature of a wide spectrum of human malignancies. Oncogene 1991, 6, 1699–1703. [Google Scholar]
  29. Matozaki, T.; Sakamoto, C.; Matsuda, K.; Suzuki, T.; Konda, Y.; Nakano, O.; Wada, K.; Uchida, T.; Nishisaki, H.; Nagao, M.; et al. Missense mutations and a deletion of the p53 gene in human gastric cancer. Biochem. Biophys. Res. Commun. 1992, 182, 215–223. [Google Scholar] [CrossRef]
  30. Yokozaki, H.; Kuniyasu, H.; Kitadai, Y.; Nishimura, K.; Todo, H.; Ayhan, A.; Yasui, W.; Ito, H.; Tahara, E. p53 point mutations in primary human gastric carcinomas. J. Cancer Res. Clin. Oncol. 1992, 119, 67–70. [Google Scholar] [CrossRef]
  31. Strickler, J.G.; Zheng, J.; Shu, Q.; Burgart, L.J.; Alberts, S.R.; Shibata, D. p53 mutations and microsatellite instability in sporadic gastric cancer: When guardians fail. Cancer Res. 1994, 54, 4750–4755. [Google Scholar] [PubMed]
  32. Poremba, C.; Yandell, D.W.; Huang, Q.; Little, J.B.; Mellin, W.; Schmid, K.W.; Bocker, W.; Dockhorn-Dworniczak, B. Frequency and spectrum of p53 mutations in gastric cancer—A molecular genetic and immunohistochemical study. Virchows Arch. 1995, 426, 447–455. [Google Scholar] [CrossRef]
  33. Renault, B.; Calistri, D.; Buonsanti, G.; Nanni, O.; Amadori, D.; Ranzani, G.N. Microsatellite instability and mutations of p53 and TGF-beta RII genes in gastric cancer. Hum. Genet. 1996, 98, 601–607. [Google Scholar] [CrossRef] [PubMed]
  34. Ochiai, A.; Yamauchi, Y.; Hirohashi, S. p53 mutations in the non-neoplastic mucosa of the human stomach showing intestinal metaplasia. Int. J. Cancer 1996, 69, 28–33. [Google Scholar] [CrossRef]
  35. Morgan, C.; Jenkins, G.J.; Ashton, T.; Griffiths, A.P.; Baxter, J.N.; Parry, E.M.; Parry, J.M. Detection of p53 mutations in precancerous gastric tissue. Br. J. Cancer 2003, 89, 1314–1319. [Google Scholar] [CrossRef] [Green Version]
  36. Kim, I.J.; Kang, H.C.; Shin, Y.; Park, H.W.; Jang, S.G.; Han, S.Y.; Lim, S.K.; Lee, M.R.; Chang, H.J.; Ku, J.L.; et al. A TP53-truncating germline mutation (E287X) in a family with characteristics of both hereditary diffuse gastric cancer and Li-Fraumeni syndrome. J. Hum. Genet. 2004, 49, 591–595. [Google Scholar] [CrossRef]
  37. Varley, J.M.; McGown, G.; Thorncroft, M.; Tricker, K.J.; Teare, M.D.; Santibanez-Koref, M.F.; Martin, J.; Birch, J.M.; Evans, D.G. An extended Li-Fraumeni kindred with gastric carcinoma and a codon 175 mutation in TP53. J. Med. Genet. 1995, 32, 942–945. [Google Scholar] [CrossRef] [Green Version]
  38. Oliveira, C.; Ferreira, P.; Nabais, S.; Campos, L.; Ferreira, A.; Cirnes, L.; Alves, C.C.; Veiga, I.; Fragoso, M.; Regateiro, F.; et al. E-Cadherin (CDH1) and p53 rather than SMAD4 and Caspase-10 germline mutations contribute to genetic predisposition in Portuguese gastric cancer patients. Eur. J. Cancer 2004, 40, 1897–1903. [Google Scholar] [CrossRef] [Green Version]
  39. Masciari, S.; Dewanwala, A.; Stoffel, E.M.; Lauwers, G.Y.; Zheng, H.; Achatz, M.I.; Riegert-Johnson, D.; Foretova, L.; Silva, E.M.; Digianni, L.; et al. Gastric cancer in individuals with Li-Fraumeni syndrome. Genet. Med. 2011, 13, 651–657. [Google Scholar] [CrossRef] [Green Version]
  40. Maesawa, C.; Tamura, G.; Suzuki, Y.; Ogasawara, S.; Sakata, K.; Kashiwaba, M.; Satodate, R. The sequential accumulation of genetic alterations characteristic of the colorectal adenoma-carcinoma sequence does not occur between gastric adenoma and adenocarcinoma. J. Pathol. 1995, 176, 249–258. [Google Scholar] [CrossRef]
  41. Ariffin, H.; Chan, A.S.; Oh, L.; Abd-Ghafar, S.; Ong, G.B.; Mohamed, M.; Razali, H.; Juraida, E.; Teo, S.H.; Karsa, M.; et al. Frequent occurrence of gastric cancer in Asian kindreds with Li-Fraumeni syndrome. Clin. Genet. 2015, 88, 450–455. [Google Scholar] [CrossRef]
  42. Ranzani, G.N.; Luinetti, O.; Padovan, L.S.; Calistri, D.; Renault, B.; Burrel, M.; Amadori, D.; Fiocca, R.; Solcia, E. p53 gene mutations and protein nuclear accumulation are early events in intestinal type gastric cancer but late events in diffuse type. Cancer Epidemiol. Biomark. Prev. 1995, 4, 223–231. [Google Scholar]
  43. Solcia, E.; Fiocca, R.; Luinetti, O.; Villani, L.; Padovan, L.; Calistri, D.; Ranzani, G.N.; Chiaravalli, A.; Capella, C. Intestinal and diffuse gastric cancers arise in a different background of Helicobacter pylori gastritis through different gene involvement. Am. J. Surg. Pathol. 1996, 20 (Suppl. S1). [Google Scholar] [CrossRef]
  44. Uchino, S.; Noguchi, M.; Ochiai, A.; Saito, T.; Kobayashi, M.; Hirohashi, S. p53 mutation in gastric cancer: A genetic model for carcinogenesis is common to gastric and colorectal cancer. Int. J. Cancer 1993, 54, 759–764. [Google Scholar] [CrossRef]
  45. Deguchi, R.; Takagi, A.; Kawata, H.; Inoko, H.; Miwa, T. Association between CagA+ Helicobacter pylori infection and p53, bax and transforming growth factor-beta-RII gene mutations in gastric cancer patients. Int. J. Cancer 2001, 91, 481–485. [Google Scholar] [CrossRef]
  46. Mutoh, H.; Sakurai, S.; Satoh, K.; Tamada, K.; Kita, H.; Osawa, H.; Tomiyama, T.; Sato, Y.; Yamamoto, H.; Isoda, N.; et al. Development of gastric carcinoma from intestinal metaplasia in Cdx2-transgenic mice. Cancer Res. 2004, 64, 7740–7747. [Google Scholar] [CrossRef] [Green Version]
  47. Gunther, T.; Schneider-Stock, R.; Hackel, C.; Kasper, H.U.; Pross, M.; Hackelsberger, A.; Lippert, H.; Roessner, A. Mdm2 gene amplification in gastric cancer correlation with expression of Mdm2 protein and p53 alterations. Mod. Pathol. 2000, 13, 621–626. [Google Scholar] [CrossRef] [PubMed]
  48. Tolbert, D.; Fenoglio-Preiser, C.; Noffsinger, A.; De Voe, G.; MacDonald, J.; Benedetti, J.; Stemmermann, G.N. The relation of p53 gene mutations to gastric cancer subsite and phenotype. Cancer Causes Control 1999, 10, 227–231. [Google Scholar] [CrossRef]
  49. Han, B.; Mori, I.; Wang, X.; Nakamura, M.; Nakamura, Y.; Kakudo, K. Combined small-cell carcinoma of the stomach: p53 and K-ras gene mutational analysis supports a monoclonal origin of three histological components. Int. J. Exp. Pathol. 2005, 86, 213–218. [Google Scholar] [CrossRef] [PubMed]
  50. Espinoza, L.A.; Tone, L.G.; Neto, J.B.; Costa, R.S.; Wang, Q.J.; Ballejo, G. Enhanced TGFalpha-EGFR expression and P53 gene alterations contributes to gastric tumors aggressiveness. Cancer Lett. 2004, 212, 33–41. [Google Scholar] [CrossRef]
  51. Shigeishi, H.; Yokozaki, H.; Oue, N.; Kuniyasu, H.; Kondo, T.; Ishikawa, T.; Yasui, W. Increased expression of CHK2 in human gastric carcinomas harboring p53 mutations. Int. J. Cancer 2002, 99, 58–62. [Google Scholar] [CrossRef] [PubMed]
  52. Byun, D.S.; Cho, K.; Ryu, B.K.; Lee, M.G.; Kang, M.J.; Kim, H.R.; Chi, S.G. Hypermethylation of XIAP-associated factor 1, a putative tumor suppressor gene from the 17p13.2 locus, in human gastric adenocarcinomas. Cancer Res. 2003, 63, 7068–7075. [Google Scholar] [PubMed]
  53. Lee, M.G.; Han, J.; Jeong, S.I.; Her, N.G.; Lee, J.H.; Ha, T.K.; Kang, M.J.; Ryu, B.K.; Chi, S.G. XAF1 directs apoptotic switch of p53 signaling through activation of HIPK2 and ZNF313. Proc. Natl. Acad. Sci. USA 2014, 111, 15532–15537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zou, B.; Chim, C.S.; Pang, R.; Zeng, H.; Dai, Y.; Zhang, R.; Lam, C.S.; Tan, V.P.; Hung, I.F.; Lan, H.Y.; et al. XIAP-associated factor 1 (XAF1), a novel target of p53, enhances p53-mediated apoptosis via post-translational modification. Mol. Carcinog. 2012, 51, 422–432. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, W.; Guo, Z.; Jiang, B.; Niu, L.; Xia, G.; Wang, X.; Cheng, T.; Zhang, Y.; Wang, J. Identification of a functional p53 responsive element within the promoter of XAF1 gene in gastrointestinal cancer cells. Int. J. Oncol. 2010, 36, 1031–1037. [Google Scholar] [CrossRef]
  56. Obata, T.; Toyota, M.; Satoh, A.; Sasaki, Y.; Ogi, K.; Akino, K.; Suzuki, H.; Murai, M.; Kikuchi, T.; Mita, H.; et al. Identification of HRK as a target of epigenetic inactivation in colorectal and gastric cancer. Clin. Cancer Res. 2003, 9, 6410–6418. [Google Scholar] [PubMed]
  57. Waraya, M.; Yamashita, K.; Ema, A.; Katada, N.; Kikuchi, S.; Watanabe, M. Exclusive Association of p53 Mutation with Super-High Methylation of Tumor Suppressor Genes in the p53 Pathway in a Unique Gastric Cancer Phenotype. PLoS ONE 2015, 10, e0139902. [Google Scholar] [CrossRef] [Green Version]
  58. Ushijima, T.; Makino, H.; Okonogi, H.; Hosoya, Y.; Sugimura, T.; Nagao, M. Mutation, loss of heterozygosity, and recombination of the p53 gene in mouse forestomach tumors induced by 2-amino-3,4-dimethylimidazo[4,5-f]quinoline. Mol. Carcinog. 1995, 12, 23–30. [Google Scholar] [CrossRef] [PubMed]
  59. Masui, T.; Tezuka, N.; Nakanishi, H.; Inada, K.; Miyashita, N.; Tatematsu, M. Induction of invasive squamous cell carcinomas in the forestomach of (C3H × MSM)F1, MSM, and C3H mice by N-methyl-N-nitrosourea and mutational analysis of the H-ras and p53 genes. Cancer Lett. 1997, 111, 97–104. [Google Scholar] [CrossRef]
  60. Furihata, C.; Tatematsu, M.; Saito, M.; Ishida, S.; Nakanishi, H.; Inada, K.; Tei, H.; Hattori, M.; Ito, T.; Sakaki, Y. Rare occurrence of ras and p53 gene mutations in mouse stomach tumors induced by N-methyl-N-nitrosourea. Jpn. J. Cancer Res. 1997, 88, 363–368. [Google Scholar] [CrossRef]
  61. Ohgaki, H.; Fukuda, M.; Tohma, Y.; Huang, H.; Stoica, G.; Tatematsu, M.; Donehower, L.A. Effect of intragastric application of N-methylnitrosourea in p53 knockout mice. Mol. Carcinog. 2000, 28, 97–101. [Google Scholar] [CrossRef]
  62. Yamamoto, M.; Tsukamoto, T.; Sakai, H.; Shirai, N.; Ohgaki, H.; Furihata, C.; Donehower, L.A.; Yoshida, K.; Tatematsu, M. p53 knockout mice (−/−) are more susceptible than (+/−) or (+/+) mice to N-methyl-N-nitrosourea stomach carcinogenesis. Carcinogenesis 2000, 21, 1891–1897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Suzuki, S.; Muroishi, Y.; Nakanishi, I.; Oda, Y. Relationship between genetic polymorphisms of drug-metabolizing enzymes (CYP1A1, CYP2E1, GSTM1, and NAT2), drinking habits, histological subtypes, and p53 gene point mutations in Japanese patients with gastric cancer. J. Gastroenterol. 2004, 39, 220–230. [Google Scholar] [CrossRef]
  64. Murakami, K.; Fujioka, T.; Kodama, M.; Honda, S.; Okimoto, T.; Oda, T.; Nishizono, A.; Sato, R.; Kubota, T.; Kagawa, J.; et al. Analysis of p53 mutations and Helicobacter pylori infection in human and animal models. J. Gastroenterol. 2002, 37 (Suppl. S13), 1–5. [Google Scholar] [CrossRef] [PubMed]
  65. Choi, J.; Yoon, S.H.; Kim, J.E.; Rhee, K.H.; Youn, H.S.; Chung, M.H. Gene-specific oxidative DNA damage in Helicobacter pylori-infected human gastric mucosa. Int. J. Cancer 2002, 99, 485–490. [Google Scholar] [CrossRef] [PubMed]
  66. Shimizu, T.; Marusawa, H.; Matsumoto, Y.; Inuzuka, T.; Ikeda, A.; Fujii, Y.; Minamiguchi, S.; Miyamoto, S.; Kou, T.; Sakai, Y.; et al. Accumulation of somatic mutations in TP53 in gastric epithelium with Helicobacter pylori infection. Gastroenterology 2014, 147, 407–417. [Google Scholar] [CrossRef] [PubMed]
  67. Takaishi, S.; Wang, T.C. Providing AID to p53 mutagenesis. Nat. Med. 2007, 13, 404–406. [Google Scholar] [CrossRef] [PubMed]
  68. Matsumoto, Y.; Marusawa, H.; Kinoshita, K.; Endo, Y.; Kou, T.; Morisawa, T.; Azuma, T.; Okazaki, I.M.; Honjo, T.; Chiba, T. Helicobacter pylori infection triggers aberrant expression of activation-induced cytidine deaminase in gastric epithelium. Nat. Med. 2007, 13, 470–476. [Google Scholar] [CrossRef] [Green Version]
  69. Wada, Y.; Takemura, K.; Tummala, P.; Uchida, K.; Kitagaki, K.; Furukawa, A.; Ishige, Y.; Ito, T.; Hara, Y.; Suzuki, T.; et al. Helicobacter pylori induces somatic mutations in TP53 via overexpression of CHAC1 in infected gastric epithelial cells. FEBS Open Bio 2018, 8, 671–679. [Google Scholar] [CrossRef] [Green Version]
  70. Hongyo, T.; Buzard, G.S.; Palli, D.; Weghorst, C.M.; Amorosi, A.; Galli, M.; Caporaso, N.E.; Fraumeni, J.F., Jr.; Rice, J.M. Mutations of the K-ras and p53 genes in gastric adenocarcinomas from a high-incidence region around Florence, Italy. Cancer Res. 1995, 55, 2665–2672. [Google Scholar]
  71. Van Beek, E.; Hernandez, J.M.; Goldman, D.A.; Davis, J.L.; McLaughlin, K.; Ripley, R.T.; Kim, T.S.; Tang, L.H.; Hechtman, J.F.; Zheng, J.; et al. Rates of TP53 Mutation are Significantly Elevated in African American Patients with Gastric Cancer. Ann. Surg. Oncol. 2018, 25, 2027–2033. [Google Scholar] [CrossRef] [PubMed]
  72. Shepherd, T.; Tolbert, D.; Benedetti, J.; Macdonald, J.; Stemmermann, G.; Wiest, J.; DeVoe, G.; Miller, M.A.; Wang, J.; Noffsinger, A.; et al. Alterations in exon 4 of the p53 gene in gastric carcinoma. Gastroenterology 2000, 118, 1039–1044. [Google Scholar] [CrossRef]
  73. Zhang, Z.W.; Laurence, N.J.; Hollowood, A.; Newcomb, P.; Moorghen, M.; Gupta, J.; Feakins, R.; Farthing, M.J.; Alderson, D.; Holly, J. Prognostic value of TP53 codon 72 polymorphism in advanced gastric adenocarcinoma. Clin. Cancer Res. 2004, 10, 131–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhou, Y.; Li, N.; Zhuang, W.; Liu, G.J.; Wu, T.X.; Yao, X.; Du, L.; Wei, M.L.; Wu, X.T. P53 codon 72 polymorphism and gastric cancer: A meta-analysis of the literature. Int. J. Cancer 2007, 121, 1481–1486. [Google Scholar] [CrossRef]
  75. Zhang, Z.W.; Newcomb, P.; Hollowood, A.; Feakins, R.; Moorghen, M.; Storey, A.; Farthing, M.J.; Alderson, D.; Holly, J. Age-associated increase of codon 72 Arginine p53 frequency in gastric cardia and non-cardia adenocarcinoma. Clin. Cancer Res. 2003, 9, 2151–2156. [Google Scholar] [PubMed]
  76. Shen, H.; Solari, A.; Wang, X.; Zhang, Z.; Xu, Y.; Wang, L.; Hu, X.; Guo, J.; Wei, Q. P53 codon 72 polymorphism and risk of gastric cancer in a Chinese population. Oncol. Rep. 2004, 11, 1115–1120. [Google Scholar] [CrossRef]
  77. Xi, Y.G.; Ding, K.Y.; Su, X.L.; Chen, D.F.; You, W.C.; Shen, Y.; Ke, Y. p53 polymorphism and p21WAF1/CIP1 haplotype in the intestinal gastric cancer and the precancerous lesions. Carcinogenesis 2004, 25, 2201–2206. [Google Scholar] [CrossRef] [PubMed]
  78. Kim, J.G.; Sohn, S.K.; Chae, Y.S.; Song, H.S.; Kwon, K.Y.; Do, Y.R.; Kim, M.K.; Lee, K.H.; Hyun, M.S.; Lee, W.S.; et al. TP53 codon 72 polymorphism associated with prognosis in patients with advanced gastric cancer treated with paclitaxel and cisplatin. Cancer Chemother. Pharmacol. 2009, 64, 355–360. [Google Scholar] [CrossRef] [PubMed]
  79. Huang, Z.H.; Hua, D.; Du, X. Polymorphisms in p53, GSTP1 and XRCC1 predict relapse and survival of gastric cancer patients treated with oxaliplatin-based adjuvant chemotherapy. Cancer Chemother. Pharmacol. 2009, 64, 1001–1007. [Google Scholar] [CrossRef]
  80. Wang, S.; Chen, L.; Zhao, Q.; Rong, H.; Wang, M.; Gong, W.; Zhou, J.; Wu, D.; Zhang, Z. Effect of TP53 codon 72 and MDM2 SNP309 polymorphisms on survival of gastric cancer among patients who receiving 5-fluorouracil-based postoperative adjuvant chemotherapy. Cancer Chemother. Pharmacol. 2013, 71, 1073–1082. [Google Scholar] [CrossRef]
  81. Lim, B.H.; Soong, R.; Grieu, F.; Robbins, P.D.; House, A.K.; Iacopetta, B.J. p53 accumulation and mutation are prognostic indicators of poor survival in human gastric carcinoma. Int. J. Cancer 1996, 69, 200–204. [Google Scholar] [CrossRef]
  82. Hamada, M.; Fujiwara, T.; Hizuta, A.; Gochi, A.; Naomoto, Y.; Takakura, N.; Takahashi, K.; Roth, J.A.; Tanaka, N.; Orita, K. The p53 gene is a potent determinant of chemosensitivity and radiosensitivity in gastric and colorectal cancers. J. Cancer Res. Clin. Oncol. 1996, 122, 360–365. [Google Scholar] [CrossRef] [PubMed]
  83. Bataille, F.; Rummele, P.; Dietmaier, W.; Gaag, D.; Klebl, F.; Reichle, A.; Wild, P.; Hofstadter, F.; Hartmann, A. Alterations in p53 predict response to preoperative high dose chemotherapy in patients with gastric cancer. Mol. Pathol. 2003, 56, 286–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hamakawa, T.; Kukita, Y.; Kurokawa, Y.; Miyazaki, Y.; Takahashi, T.; Yamasaki, M.; Miyata, H.; Nakajima, K.; Taniguchi, K.; Takiguchi, S.; et al. Monitoring gastric cancer progression with circulating tumour DNA. Br. J. Cancer 2015, 112, 352–356. [Google Scholar] [CrossRef] [PubMed]
  85. Ott, K.; Vogelsang, H.; Mueller, J.; Becker, K.; Muller, M.; Fink, U.; Siewert, J.R.; Hofler, H.; Keller, G. Chromosomal instability rather than p53 mutation is associated with response to neoadjuvant cisplatin-based chemotherapy in gastric carcinoma. Clin. Cancer Res. 2003, 9, 2307–2315. [Google Scholar] [PubMed]
  86. Xu, H.Y.; Xu, W.L.; Wang, L.Q.; Chen, M.B.; Shen, H.L. Relationship between p53 status and response to chemotherapy in patients with gastric cancer: A meta-analysis. PLoS ONE 2014, 9, e95371. [Google Scholar] [CrossRef]
  87. Ikari, N.; Serizawa, A.; Mitani, S.; Yamamoto, M.; Furukawa, T. Near-Comprehensive Resequencing of Cancer-Associated Genes in Surgically Resected Metastatic Liver Tumors of Gastric Cancer. Am. J. Pathol. 2019, 189, 784–796. [Google Scholar] [CrossRef]
  88. Sethi, N.; Kikuchi, O.; McFarland, J.; Zhang, Y.; Chung, M.; Kafker, N.; Islam, M.; Lampson, B.; Chakraborty, A.; Kaelin, W.G., Jr.; et al. Mutant p53 induces a hypoxia transcriptional program in gastric and esophageal adenocarcinoma. JCI Insight 2019, 4. [Google Scholar] [CrossRef]
  89. Mrozek, A.; Petrowsky, H.; Sturm, I.; Kraus, J.; Hermann, S.; Hauptmann, S.; Lorenz, M.; Dorken, B.; Daniel, P.T. Combined p53/Bax mutation results in extremely poor prognosis in gastric carcinoma with low microsatellite instability. Cell Death Differ. 2003, 10, 461–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Park, S.; Lee, J.; Kim, Y.H.; Park, J.; Shin, J.W.; Nam, S. Clinical Relevance and Molecular Phenotypes in Gastric Cancer, of TP53 Mutations and Gene Expressions, in Combination With Other Gene Mutations. Sci. Rep. 2016, 6, 34822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Pallocca, M.; Goeman, F.; De Nicola, F.; Melucci, E.; Sperati, F.; Terrenato, I.; Pizzuti, L.; Casini, B.; Gallo, E.; Amoreo, C.A.; et al. Coexisting YAP expression and TP53 missense mutations delineates a molecular scenario unexpectedly associated with better survival outcomes in advanced gastric cancer. J. Transl. Med. 2018, 16, 247. [Google Scholar] [CrossRef] [PubMed]
  92. Cristescu, R.; Lee, J.; Nebozhyn, M.; Kim, K.M.; Ting, J.C.; Wong, S.S.; Liu, J.; Yue, Y.G.; Wang, J.; Yu, K.; et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 2015, 21, 449–456. [Google Scholar] [CrossRef] [PubMed]
  93. Tahara, T.; Tahara, S.; Horiguchi, N.; Okubo, M.; Terada, T.; Yamada, H.; Yoshida, D.; Omori, T.; Osaki, H.; Maeda, K.; et al. Molecular subtyping of gastric cancer combining genetic and epigenetic anomalies provides distinct clinicopathological features and prognostic impacts. Hum. Mutat. 2019, 40, 347–354. [Google Scholar] [CrossRef] [PubMed]
  94. Nemtsova, M.V.; Kalinkin, A.I.; Kuznetsova, E.B.; Bure, I.V.; Alekseeva, E.A.; Bykov, I.I.; Khorobrykh, T.V.; Mikhaylenko, D.S.; Tanas, A.S.; Kutsev, S.I.; et al. Clinical relevance of somatic mutations in main driver genes detected in gastric cancer patients by next-generation DNA sequencing. Sci. Rep. 2020, 10, 504. [Google Scholar] [CrossRef] [Green Version]
  95. Baugh, E.H.; Ke, H.; Levine, A.J.; Bonneau, R.A.; Chan, C.S. Why are there hotspot mutations in the TP53 gene in human cancers? Cell Death Differ. 2018, 25, 154–160. [Google Scholar] [CrossRef]
  96. Mantovani, F.; Collavin, L.; Del Sal, G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ. 2019, 26, 199–212. [Google Scholar] [CrossRef] [PubMed]
  97. Kim, M.P.; Lozano, G. Mutant p53 partners in crime. Cell Death Differ. 2018, 25, 161–168. [Google Scholar] [CrossRef]
  98. Migliavacca, M.; Ottini, L.; Bazan, V.; Agnese, V.; Corsale, S.; Macaluso, M.; Lupi, R.; Dardanoni, G.; Valerio, M.R.; Pantuso, G.; et al. TP53 in gastric cancer: Mutations in the l3 loop and LSH motif DNA-binding domains of TP53 predict poor outcome. J. Cell. Physiol. 2004, 200, 476–485. [Google Scholar] [CrossRef]
  99. Tahara, T.; Shibata, T.; Okamoto, Y.; Yamazaki, J.; Kawamura, T.; Horiguchi, N.; Okubo, M.; Nakano, N.; Ishizuka, T.; Nagasaka, M.; et al. Mutation spectrum of TP53 gene predicts clinicopathological features and survival of gastric cancer. Oncotarget 2016, 7, 42252–42260. [Google Scholar] [CrossRef] [Green Version]
  100. Fischer, N.W.; Prodeus, A.; Gariepy, J. Survival in males with glioma and gastric adenocarcinoma correlates with mutant p53 residual transcriptional activity. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  101. Shen, W.; Chen, D.; Liu, S.; Chen, L.; Yu, A.; Fu, H.; Sun, X. S100A4 interacts with mutant p53 and affects gastric cancer MKN1 cell autophagy and differentiation. Int. J. Oncol. 2015, 47, 2123–2130. [Google Scholar] [CrossRef] [PubMed]
  102. Jiang, Z.; Liu, Z.; Li, M.; Chen, C.; Wang, X. Immunogenomics Analysis Reveals that TP53 Mutations Inhibit Tumor Immunity in Gastric Cancer. Transl. Oncol. 2018, 11, 1171–1187. [Google Scholar] [CrossRef]
  103. Kim, J.Y.; Kim, W.G.; Kwon, C.H.; Park, D.Y. Differences in immune contextures among different molecular subtypes of gastric cancer and their prognostic impact. Gastric Cancer 2019, 22, 1164–1175. [Google Scholar] [CrossRef] [PubMed]
  104. Sun, Y.; Yu, W.; Guan, W.; Cai, L.; Qiao, M.; Zheng, L.; Jiang, R.; Wang, R.; Wang, L. Integrated assessment of PD-L1 expression and molecular classification facilitates therapy selection and prognosis prediction in gastric cancer. Cancer Manag. Res. 2019, 11, 6397–6410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Martin, H.M.; Filipe, M.I.; Morris, R.W.; Lane, D.P.; Silvestre, F. p53 expression and prognosis in gastric carcinoma. Int. J. Cancer 1992, 50, 859–862. [Google Scholar] [CrossRef]
  106. Starzynska, T.; Markiewski, M.; Domagala, W.; Marlicz, K.; Mietkiewski, J.; Roberts, S.A.; Stern, P.L. The clinical significance of p53 accumulation in gastric carcinoma. Cancer 1996, 77, 2005–2012. [Google Scholar] [CrossRef]
  107. Maehara, Y.; Tomoda, M.; Hasuda, S.; Kabashima, A.; Tokunaga, E.; Kakeji, Y.; Sugimachi, K. Prognostic value of p53 protein expression for patients with gastric cancer—A multivariate analysis. Br. J. Cancer 1999, 79, 1255–1261. [Google Scholar] [CrossRef] [Green Version]
  108. Yonemura, Y.; Fushida, S.; Tsugawa, K.; Ninomiya, I.; Fonseca, L.; Yamaguchi, A.; Miyazaki, I.; Urano, T.; Shiku, H. Correlation of p53 expression and proliferative activity in gastric cancer. Anal. Cell Pathol. 1993, 5, 277–288. [Google Scholar]
  109. Joypaul, B.V.; Hopwood, D.; Newman, E.L.; Qureshi, S.; Grant, A.; Ogston, S.A.; Lane, D.P.; Cuschieri, A. The prognostic significance of the accumulation of p53 tumour-suppressor gene protein in gastric adenocarcinoma. Br. J. Cancer 1994, 69, 943–946. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, Z.; Ikeguchi, M.; Maeta, M.; Kaibara, N. The correlation between the expression of p53 protein and DNA ploidy in patients with gastric cancer that has invaded the serosa. Anticancer Res. 1997, 17, 3701–3705. [Google Scholar]
  111. Hurlimann, J.; Saraga, E.P. Expression of p53 protein in gastric carcinomas. Association with histologic type and prognosis. Am J. Surg. Pathol. 1994, 18, 1247–1253. [Google Scholar] [CrossRef] [PubMed]
  112. Gomyo, Y.; Ikeda, M.; Osaki, M.; Tatebe, S.; Tsujitani, S.; Ikeguchi, M.; Kaibara, N.; Ito, H. Expression of p21 (waf1/cip1/sdi1), but not p53 protein, is a factor in the survival of patients with advanced gastric carcinoma. Cancer 1997, 79, 2067–2072. [Google Scholar] [CrossRef]
  113. Kim, J.P.; Oh, S.T.; Hwang, T.S.; Chi, J.G. The prognostic significance of c-erbB-2 and p53 protein expressions in gastric carcinoma—A multivariate analysis of prognostic factors. J. Korean Med. Sci. 1994, 9, 248–253. [Google Scholar] [CrossRef] [Green Version]
  114. Gabbert, H.E.; Muller, W.; Schneiders, A.; Meier, S.; Hommel, G. The relationship of p53 expression to the prognosis of 418 patients with gastric carcinoma. Cancer 1995, 76, 720–726. [Google Scholar] [CrossRef]
  115. Victorzon, M.; Nordling, S.; Haglund, C.; Lundin, J.; Roberts, P.J. Expression of p53 protein as a prognostic factor in patients with gastric cancer. Eur. J. Cancer 1996, 32, 215–220. [Google Scholar] [CrossRef]
  116. Liu, X.P.; Tsushimi, K.; Tsushimi, M.; Kawauchi, S.; Oga, A.; Furuya, T.; Sasaki, K. Expression of p21(WAF1/CIP1) and p53 proteins in gastric carcinoma: Its relationships with cell proliferation activity and prognosis. Cancer Lett. 2001, 170, 183–189. [Google Scholar] [CrossRef]
  117. Liu, X.P.; Tsushimi, K.; Tsushimi, M.; Oga, A.; Kawauchi, S.; Furuya, T.; Sasaki, K. Expression of p53 protein as a prognostic indicator of reduced survival time in diffuse-type gastric carcinoma. Pathol. Int. 2001, 51, 440–444. [Google Scholar] [CrossRef]
  118. Muller, W.; Borchard, F. Prognostic influence of p53 expression in gastric cancer. J. Pathol. 1996, 178, 255–258. [Google Scholar] [CrossRef]
  119. Wei, K.; Jiang, L.; Wei, Y.; Wang, Y.; Qian, X.; Dai, Q.; Guan, Q. The prognostic significance of p53 expression in gastric cancer: A meta-analysis. J. Cancer Res. Clin. Oncol. 2015, 141, 735–748. [Google Scholar] [CrossRef] [PubMed]
  120. Nakata, B.; Chung, K.H.; Ogawa, M.; Ogawa, Y.; Yanagawa, K.; Muguruma, K.; Inoue, T.; Yamashita, Y.; Onoda, N.; Maeda, K.; et al. p53 protein overexpression as a predictor of the response to chemotherapy in gastric cancer. Surg. Today 1998, 28, 595–598. [Google Scholar] [CrossRef]
  121. Cascinu, S.; Graziano, F.; Del Ferro, E.; Staccioli, M.P.; Ligi, M.; Carnevali, A.; Muretto, P.; Catalano, G. Expression of p53 protein and resistance to preoperative chemotherapy in locally advanced gastric carcinoma. Cancer 1998, 83, 1917–1922. [Google Scholar] [CrossRef]
  122. Diez, M.; Medrano, M.J.; Gutierrez, A.; Lopez, A.; Muguerza, J.M.; Hernandez, P.; Lozano, O.; Noguerales, F.; Ruiz, A.; Granell, J. P53 protein expression in gastric adenocarcinoma. Negative predictor of survival after postoperative adjuvant chemotherapy. Anticancer Res. 2000, 20, 3929–3933. [Google Scholar]
  123. Fondevila, C.; Metges, J.P.; Fuster, J.; Grau, J.J.; Palacin, A.; Castells, A.; Volant, A.; Pera, M. p53 and VEGF expression are independent predictors of tumour recurrence and survival following curative resection of gastric cancer. Br. J. Cancer 2004, 90, 206–215. [Google Scholar] [CrossRef]
  124. Kamoshida, S.; Suzuki, M.; Shimomura, R.; Sakurai, Y.; Komori, Y.; Uyama, I.; Tsutsumi, Y. Immunostaining of thymidylate synthase and p53 for predicting chemoresistance to S-1/cisplatin in gastric cancer. Br. J. Cancer 2007, 96, 277–283. [Google Scholar] [CrossRef] [Green Version]
  125. Liu, X.; Wang, S.; Xia, X.; Chen, Y.; Zhou, Y.; Wu, X.; Zhang, J.; He, S.; Tan, Y.; Qiang, F.; et al. Synergistic role between p53 and JWA: Prognostic and predictive biomarkers in gastric cancer. PLoS ONE 2012, 7, e52348. [Google Scholar] [CrossRef] [Green Version]
  126. Yeh, K.H.; Shun, C.T.; Chen, C.L.; Lin, J.T.; Lee, W.J.; Lee, P.H.; Chen, Y.C.; Cheng, A.L. Overexpression of p53 is not associated with drug resistance of gastric cancers to 5-fluorouracil-based systemic chemotherapy. Hepatogastroenterology 1999, 46, 610–615. [Google Scholar]
  127. Sawada, T.; Yashiro, M.; Sentani, K.; Oue, N.; Yasui, W.; Miyazaki, K.; Kai, K.; Fushida, S.; Fujimura, T.; Ohira, M.; et al. New molecular staging with G-factor supplements TNM classification in gastric cancer: A multicenter collaborative research by the Japan Society for Gastroenterological Carcinogenesis G-Project committee. Gastric Cancer 2015, 18, 119–128. [Google Scholar] [CrossRef]
  128. Boku, N.; Chin, K.; Hosokawa, K.; Ohtsu, A.; Tajiri, H.; Yoshida, S.; Yamao, T.; Kondo, H.; Shirao, K.; Shimada, Y.; et al. Biological markers as a predictor for response and prognosis of unresectable gastric cancer patients treated with 5-fluorouracil and cis-platinum. Clin. Cancer Res. 1998, 4, 1469–1474. [Google Scholar] [PubMed]
  129. Maeda, K.; Kang, S.M.; Onoda, N.; Ogawa, M.; Sawada, T.; Nakata, B.; Kato, Y.; Chung, Y.S.; Sowa, M. Expression of p53 and vascular endothelial growth factor associated with tumor angiogenesis and prognosis in gastric cancer. Oncology 1998, 55, 594–599. [Google Scholar] [CrossRef] [PubMed]
  130. Xiangming, C.; Hokita, S.; Natsugoe, S.; Tanabe, G.; Baba, M.; Takao, S.; Kuroshima, K.; Aikou, T. p21 expression is a prognostic factor in patients with p53-negative gastric cancer. Cancer Lett. 2000, 148, 181–188. [Google Scholar] [CrossRef]
  131. Maeta, Y.; Tsujitani, S.; Matsumoto, S.; Yamaguchi, K.; Tatebe, S.; Kondo, A.; Ikeguchi, M.; Kaibara, N. Expression of Mcl-1 and p53 proteins predicts the survival of patients with T3 gastric carcinoma. Gastric Cancer. 2004, 7, 78–84. [Google Scholar] [CrossRef] [Green Version]
  132. Bani-Hani, K.E.; Almasri, N.M.; Khader, Y.S.; Sheyab, F.M.; Karam, H.N. Combined evaluation of expressions of cyclin E and p53 proteins as prognostic factors for patients with gastric cancer. Clin. Cancer Res. 2005, 11, 1447–1453. [Google Scholar] [CrossRef] [Green Version]
  133. Sumiyoshi, Y.; Kakeji, Y.; Egashira, A.; Mizokami, K.; Orita, H.; Maehara, Y. Overexpression of hypoxia-inducible factor 1alpha and p53 is a marker for an unfavorable prognosis in gastric cancer. Clin. Cancer Res. 2006, 12, 5112–5117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lu, C.D.; Altieri, D.C.; Tanigawa, N. Expression of a novel antiapoptosis gene, survivin, correlated with tumor cell apoptosis and p53 accumulation in gastric carcinomas. Cancer Res. 1998, 58, 1808–1812. [Google Scholar]
  135. Hoffman, W.H.; Biade, S.; Zilfou, J.T.; Chen, J.; Murphy, M. Transcriptional repression of the anti-apoptotic survivin gene by wild type p53. J. Biol. Chem. 2002, 277, 3247–3257. [Google Scholar] [CrossRef] [Green Version]
  136. Uchino, S.; Noguchi, M.; Hirota, T.; Itabashi, M.; Saito, T.; Kobayashi, M.; Hirohashi, S. High incidence of nuclear accumulation of p53 protein in gastric cancer. Jpn. J. Clin. Oncol. 1992, 22, 225–231. [Google Scholar]
  137. Brito, M.J.; Williams, G.T.; Thompson, H.; Filipe, M.I. Expression of p53 in early (T1) gastric carcinoma and precancerous adjacent mucosa. Gut 1994, 35, 1697–1700. [Google Scholar] [CrossRef] [Green Version]
  138. Flejou, J.F.; Muzeau, F.; Potet, F.; Lepelletier, F.; Fekete, F.; Henin, D. Overexpression of the p53 tumor suppressor gene product in esophageal and gastric carcinomas. Pathol. Res. Pract. 1994, 190, 1141–1148. [Google Scholar] [CrossRef]
  139. Tang, H.; Hokita, S.; Che, X.; Baba, M.; Aridome, K.; Kijima, F.; Tanabe, G.; Takao, S.; Aikou, T. Comparison of p53 expression in proximal and distal gastric cancer: Histopathologic correlation and prognostic significance. Ann. Surg. Oncol. 1997, 4, 470–474. [Google Scholar] [CrossRef] [PubMed]
  140. Yao, D.; Wang, Y.; Xue, L.; Wang, H.; Zhang, J.; Zhang, X. Different expression pattern and significance of p14ARF-Mdm2-p53 pathway and Bmi-1 exist between gastric cardia and distal gastric adenocarcinoma. Hum. Pathol. 2013, 44, 844–851. [Google Scholar] [CrossRef]
  141. Shibata, A.; Parsonnet, J.; Longacre, T.A.; Garcia, M.I.; Puligandla, B.; Davis, R.E.; Vogelman, J.H.; Orentreich, N.; Habel, L.A. CagA status of Helicobacter pylori infection and p53 gene mutations in gastric adenocarcinoma. Carcinogenesis 2002, 23, 419–424. [Google Scholar] [CrossRef] [Green Version]
  142. Kodama, M.; Fujioka, T.; Murakami, K.; Okimoto, T.; Sato, R.; Watanabe, K.; Nasu, M. Eradication of Helicobacter pylori reduced the immunohistochemical detection of p53 and MDM2 in gastric mucosa. J. Gastroenterol. Hepatol. 2005, 20, 941–946. [Google Scholar] [CrossRef]
  143. Kakeji, Y.; Korenaga, D.; Tsujitani, S.; Baba, H.; Anai, H.; Maehara, Y.; Sugimachi, K. Gastric cancer with p53 overexpression has high potential for metastasising to lymph nodes. Br. J. Cancer 1993, 67, 589–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Oiwa, H.; Maehara, Y.; Ohno, S.; Sakaguchi, Y.; Ichiyoshi, Y.; Sugimachi, K. Growth pattern and p53 overexpression in patients with early gastric cancer. Cancer 1995, 75, 1454–1459. [Google Scholar] [CrossRef]
  145. Lauwers, G.Y.; Wahl, S.J.; Melamed, J.; Rojas-Corona, R.R. p53 expression in precancerous gastric lesions: An immunohistochemical study of PAb 1801 monoclonal antibody on adenomatous and hyperplastic gastric polyps. Am. J. Gastroenterol. 1993, 88, 1916–1919. [Google Scholar]
  146. Starzynska, T.; Marsh, P.J.; Stern, P.L. p53 overexpression as a marker of malignancy in gastric biopsies. Surg. Oncol. 1993, 2, 321–324. [Google Scholar] [CrossRef]
  147. Wang, L.D.; Shi, S.T.; Zhou, Q.; Goldstein, S.; Hong, J.Y.; Shao, P.; Qiu, S.L.; Yang, C.S. Changes in p53 and cyclin D1 protein levels and cell proliferation in different stages of human esophageal and gastric-cardia carcinogenesis. Int. J. Cancer 1994, 59, 514–519. [Google Scholar] [CrossRef]
  148. Seruca, R.; David, L.; Holm, R.; Nesland, J.M.; Fangan, B.M.; Castedo, S.; Sobrinho-Simoes, M.; Borresen, A.L. P53 mutations in gastric carcinomas. Br. J. Cancer 1992, 65, 708–710. [Google Scholar] [CrossRef] [Green Version]
  149. Fushida, S.; Urano, T.; Tsuruta, H.; Yonemura, Y.; Miyazaki, I.; Shiku, H. Frequent p53 mutations on exons-5 and exon-8 in human primary gastric-cancer. Int. J. Oncol. 1993, 2, 563–568. [Google Scholar] [CrossRef]
  150. Hong, S.I.; Hong, W.S.; Jang, J.J.; Lee, D.S.; Cho, N.S.; Jung, M.E.; Kim, H.B.; Ha, G.W.; Park, I.C.; Cho, D.S.; et al. Alterations of p53 gene in primary gastric cancer tissues. Anticancer Res. 1994, 14, 1251–1255. [Google Scholar]
  151. Shiao, Y.H.; Palli, D.; Caporaso, N.E.; Alvord, W.G.; Amorosi, A.; Nesi, G.; Saieva, C.; Masala, G.; Fraumeni, J.F., Jr.; Rice, J.M. Genetic and immunohistochemical analyses of p53 independently predict regional metastasis of gastric cancers. Cancer Epidemiol. Biomark. Prev. 2000, 9, 631–633. [Google Scholar]
  152. Wurl, P.; Weigmann, F.; Meye, A.; Fittkau, M.; Rose, U.; Berger, D.; Rath, F.W.; Dralle, H.; Taubert, H. Detection of p53 autoantibodies in sera of gastric cancer patients and their prognostic relevance. Scand. J. Gastroenterol. 1997, 32, 1147–1151. [Google Scholar] [CrossRef]
  153. Shiota, G.; Ishida, M.; Noguchi, N.; Takano, Y.; Oyama, K.; Okubo, M.; Katayama, S.; Harada, K.; Hori, K.; Ashida, K.; et al. Clinical significance of serum P53 antibody in patients with gastric cancer. Res. Commun. Mol. Pathol. Pharmacol. 1998, 99, 41–51. [Google Scholar] [PubMed]
  154. Maehara, Y.; Kakeji, Y.; Watanabe, A.; Baba, H.; Kusumoto, H.; Kohnoe, S.; Sugimachi, K. Clinical implications of serum anti-p53 antibodies for patients with gastric carcinoma. Cancer 1999, 85, 302–308. [Google Scholar] [CrossRef]
  155. Soussi, T. p53 Antibodies in the sera of patients with various types of cancer: A review. Cancer Res. 2000, 60, 1777–1788. [Google Scholar]
  156. Wu, C.W.; Lin, Y.Y.; Chen, G.D.; Chi, C.W.; Carbone, D.P.; Chen, J.Y. Serum anti-p53 antibodies in gastric adenocarcinoma patients are associated with poor prognosis, lymph node metastasis and poorly differentiated nuclear grade. Br. J. Cancer 1999, 80, 483–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Zhang, J.; Xu, Z.; Yu, L.; Chen, M.; Li, K. Assessment of the potential diagnostic value of serum p53 antibody for cancer: A meta-analysis. PLoS ONE 2014, 9, e99255. [Google Scholar] [CrossRef] [PubMed]
  158. Nakajima, K.; Suzuki, T.; Shimada, H.; Hayashi, H.; Takeda, A.; Ochiai, T. Detection of preoperative serum anti-p53 antibodies in gastric cancer. Tumour Biol. 1999, 20, 147–152. [Google Scholar] [CrossRef]
  159. Werner, S.; Chen, H.; Butt, J.; Michel, A.; Knebel, P.; Holleczek, B.; Zornig, I.; Eichmuller, S.B.; Jager, D.; Pawlita, M.; et al. Evaluation of the diagnostic value of 64 simultaneously measured autoantibodies for early detection of gastric cancer. Sci. Rep. 2016, 6, 25467. [Google Scholar] [CrossRef] [PubMed]
  160. Blok, P.; Craanen, M.E.; Dekker, W.; Offerhaus, G.J.; Tytgat, G.N. No evidence for functional inactivation of wild-type p53 protein by MDM2 overexpression in gastric carcinogenesis. J. Pathol. 1998, 186, 36–40. [Google Scholar] [CrossRef]
  161. Ohmiya, N.; Taguchi, A.; Mabuchi, N.; Itoh, A.; Hirooka, Y.; Niwa, Y.; Goto, H. MDM2 promoter polymorphism is associated with both an increased susceptibility to gastric carcinoma and poor prognosis. J. Clin. Oncol. 2006, 24, 4434–4440. [Google Scholar] [CrossRef] [PubMed]
  162. Ma, Y.; Bian, J.; Cao, H. MDM2 SNP309 rs2279744 polymorphism and gastric cancer risk: A meta-analysis. PLoS ONE 2013, 8, e56918. [Google Scholar] [CrossRef] [Green Version]
  163. Feng, C.; Xian, Q.; Liu, S. Micro RNA-518 inhibits gastric cancer cell growth by inducing apoptosis via targeting MDM2. Biomed. Pharmacother. 2018, 97, 1595–1602. [Google Scholar] [CrossRef] [PubMed]
  164. Bao, J.; Nanding, A.; Song, H.; Xu, R.; Qu, G.; Xue, Y. The overexpression of MDM4: An effective and novel predictor of gastric adenocarcinoma lymph node metastasis. Oncotarget 2016, 7, 67212–67222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Jung, C.R.; Lim, J.H.; Choi, Y.; Kim, D.G.; Kang, K.J.; Noh, S.M.; Im, D.S. Enigma negatively regulates p53 through MDM2 and promotes tumor cell survival in mice. J. Clin. Investig. 2010, 120, 4493–4506. [Google Scholar] [CrossRef] [Green Version]
  166. Sehdev, V.; Katsha, A.; Arras, J.; Peng, D.; Soutto, M.; Ecsedy, J.; Zaika, A.; Belkhiri, A.; El-Rifai, W. HDM2 regulation by AURKA promotes cell survival in gastric cancer. Clin. Cancer Res. 2014, 20, 76–86. [Google Scholar] [CrossRef] [Green Version]
  167. Li, J.; Ng, E.K.; Ng, Y.P.; Wong, C.Y.; Yu, J.; Jin, H.; Cheng, V.Y.; Go, M.Y.; Cheung, P.K.; Ebert, M.P.; et al. Identification of retinoic acid-regulated nuclear matrix-associated protein as a novel regulator of gastric cancer. Br. J. Cancer 2009, 101, 691–698. [Google Scholar] [CrossRef] [Green Version]
  168. Imanishi, M.; Yamamoto, Y.; Wang, X.; Sugaya, A.; Hirose, M.; Endo, S.; Natori, Y.; Yamato, K.; Hyodo, I. Augmented antitumor activity of 5-fluorouracil by double knockdown of MDM4 and MDM2 in colon and gastric cancer cells. Cancer Sci. 2019, 110, 639–649. [Google Scholar] [CrossRef] [Green Version]
  169. Chau, J.F.; Jia, D.; Wang, Z.; Liu, Z.; Hu, Y.; Zhang, X.; Jia, H.; Lai, K.P.; Leong, W.F.; Au, B.J.; et al. A crucial role for bone morphogenetic protein-Smad1 signalling in the DNA damage response. Nat. Commun. 2012, 3, 836. [Google Scholar] [CrossRef]
  170. Uchi, R.; Kogo, R.; Kawahara, K.; Sudo, T.; Yokobori, T.; Eguchi, H.; Sugimachi, K.; Maehama, T.; Mori, M.; Suzuki, A.; et al. PICT1 regulates TP53 via RPL11 and is involved in gastric cancer progression. Br. J. Cancer 2013, 109, 2199–2206. [Google Scholar] [CrossRef] [Green Version]
  171. Zhang, X.W.; Wang, X.F.; Ni, S.J.; Qin, W.; Zhao, L.Q.; Hua, R.X.; Lu, Y.W.; Li, J.; Dimri, G.P.; Guo, W.J. UBTD1 induces cellular senescence through an UBTD1-Mdm2/p53 positive feedback loop. J. Pathol. 2015, 235, 656–667. [Google Scholar] [CrossRef]
  172. Chang, Y.T.; Wu, M.S.; Chang, C.J.; Huang, P.H.; Hsu, S.M.; Lin, J.T. Preferential loss of Fhit expression in signet-ring cell and Krukenberg subtypes of gastric cancer. Lab. Investig. 2002, 82, 1201–1208. [Google Scholar] [CrossRef] [Green Version]
  173. Nishizaki, M.; Sasaki, J.; Fang, B.; Atkinson, E.N.; Minna, J.D.; Roth, J.A.; Ji, L. Synergistic tumor suppression by coexpression of FHIT and p53 coincides with FHIT-mediated MDM2 inactivation and p53 stabilization in human non-small cell lung cancer cells. Cancer Res. 2004, 64, 5745–5752. [Google Scholar] [CrossRef] [Green Version]
  174. Li, X.; Cheung, K.F.; Ma, X.; Tian, L.; Zhao, J.; Go, M.Y.; Shen, B.; Cheng, A.S.; Ying, J.; Tao, Q.; et al. Epigenetic inactivation of paired box gene 5, a novel tumor suppressor gene, through direct upregulation of p53 is associated with prognosis in gastric cancer patients. Oncogene 2012, 31, 3419–3430. [Google Scholar] [CrossRef]
  175. Otani, K.; Dong, Y.; Li, X.; Lu, J.; Zhang, N.; Xu, L.; Go, M.Y.; Ng, E.K.; Arakawa, T.; Chan, F.K.; et al. Odd-skipped related 1 is a novel tumour suppressor gene and a potential prognostic biomarker in gastric cancer. J. Pathol. 2014, 234, 302–315. [Google Scholar] [CrossRef] [Green Version]
  176. Zhang, L.; Li, W.; Cao, L.; Xu, J.; Qian, Y.; Chen, H.; Zhang, Y.; Kang, W.; Gou, H.; Wong, C.C.; et al. PKNOX2 suppresses gastric cancer through the transcriptional activation of IGFBP5 and p53. Oncogene 2019, 38, 4590–4604. [Google Scholar] [CrossRef] [Green Version]
  177. Xu, L.; Li, X.; Chu, E.S.; Zhao, G.; Go, M.Y.; Tao, Q.; Jin, H.; Zeng, Z.; Sung, J.J.; Yu, J. Epigenetic inactivation of BCL6B, a novel functional tumour suppressor for gastric cancer, is associated with poor survival. Gut 2012, 61, 977–985. [Google Scholar] [CrossRef]
  178. Li, X.; Zhang, Y.; Cao, S.; Chen, X.; Lu, Y.; Jin, H.; Sun, S.; Chen, B.; Liu, J.; Ding, J.; et al. Reduction of TIP30 correlates with poor prognosis of gastric cancer patients and its restoration drastically inhibits tumor growth and metastasis. Int. J. Cancer 2009, 124, 713–721. [Google Scholar] [CrossRef]
  179. Zhou, Z.; Ji, Z.; Wang, Y.; Li, J.; Cao, H.; Zhu, H.H.; Gao, W.Q. TRIM59 is up-regulated in gastric tumors, promoting ubiquitination and degradation of p53. Gastroenterology 2014, 147, 1043–1054. [Google Scholar] [CrossRef]
  180. Sun, M.; Si, G.; Sun, H.S.; Si, F.C. Inhibition of CREPT restrains gastric cancer growth by regulation of cycle arrest, migration and apoptosis via ROS-regulated p53 pathway. Biochem. Biophys. Res. Commun. 2018, 496, 1183–1190. [Google Scholar] [CrossRef] [PubMed]
  181. Wang, Y.; Zheng, X.; Zhang, Z.; Zhou, J.; Zhao, G.; Yang, J.; Xia, L.; Wang, R.; Cai, X.; Hu, H.; et al. MicroRNA-149 inhibits proliferation and cell cycle progression through the targeting of ZBTB2 in human gastric cancer. PLoS ONE 2012, 7, e41693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Lu, F.; Xue, J.X.; Hu, Y.C.; Gan, L.; Shi, Y.; Yang, H.S.; Wei, Y.Q. CARP is a potential tumor suppressor in gastric carcinoma and a single-nucleotide polymorphism in CARP gene might increase the risk of gastric carcinoma. PLoS ONE 2014, 9, e97743. [Google Scholar] [CrossRef] [PubMed]
  183. Arai, H.; Wada, R.; Ishino, K.; Kudo, M.; Uchida, E.; Naito, Z. Expression of DNA damage response proteins in gastric cancer: Comprehensive protein profiling and histological analysis. Int. J. Oncol. 2018, 52, 978–988. [Google Scholar] [CrossRef] [Green Version]
  184. Huang, S.; Li, Y.; Yuan, X.; Zhao, M.; Wang, J.; Li, Y.; Li, Y.; Lin, H.; Zhang, Q.; Wang, W.; et al. The UbL-UBA Ubiquilin4 protein functions as a tumor suppressor in gastric cancer by p53-dependent and p53-independent regulation of p21. Cell Death Differ. 2019, 26, 516–530. [Google Scholar] [CrossRef] [Green Version]
  185. Hashimoto, T.; Kurokawa, Y.; Wada, N.; Takahashi, T.; Miyazaki, Y.; Tanaka, K.; Makino, T.; Yamasaki, M.; Nakajima, K.; Mori, M.; et al. Clinical significance of chromatin remodeling factor CHD5 expression in gastric cancer. Oncol. Lett. 2020, 19, 1066–1073. [Google Scholar] [CrossRef] [Green Version]
  186. Liu, X.; Zhang, B.; Guo, Y.; Liang, Q.; Wu, C.; Wu, L.; Tao, K.; Wang, G.; Chen, J. Down-regulation of AP-4 inhibits proliferation, induces cell cycle arrest and promotes apoptosis in human gastric cancer cells. PLoS ONE 2012, 7, e37096. [Google Scholar] [CrossRef] [Green Version]
  187. Ando, K.; Oki, E.; Zhao, Y.; Ikawa-Yoshida, A.; Kitao, H.; Saeki, H.; Kimura, Y.; Ida, S.; Morita, M.; Kusumoto, T.; et al. Mortalin is a prognostic factor of gastric cancer with normal p53 function. Gastric Cancer. 2014, 17, 255–262. [Google Scholar] [CrossRef] [Green Version]
  188. Du, F.; Sun, L.; Chu, Y.; Li, T.; Lei, C.; Wang, X.; Jiang, M.; Min, Y.; Lu, Y.; Zhao, X.; et al. DDIT4 promotes gastric cancer proliferation and tumorigenesis through the p53 and MAPK pathways. Cancer Commun. 2018, 38, 45. [Google Scholar] [CrossRef] [Green Version]
  189. Yang, B.; Wu, A.; Hu, Y.; Tao, C.; Wang, J.M.; Lu, Y.; Xing, R. Mucin 17 inhibits the progression of human gastric cancer by limiting inflammatory responses through a MYH9-p53-RhoA regulatory feedback loop. J. Exp. Clin. Cancer Res. 2019, 38, 283. [Google Scholar] [CrossRef] [PubMed]
  190. Zhang, Y.J.; Li, J.Q.; Li, H.Z.; Song, H.; Wei, C.S.; Zhang, S.Q. PDRG1 gene silencing contributes to inhibit the growth and induce apoptosis of gastric cancer cells. Pathol. Res. Pract. 2019, 215, 152567. [Google Scholar] [CrossRef]
  191. Wang, Y.; Xiao, H.; Wang, C.; Wu, H.; He, H.; Yao, C.; Cui, J.; Li, W. M-phase phosphoprotein 8 promotes gastric cancer growth and metastasis via p53/Bcl-2 and EMT-related signaling pathways. J. Cell. Biochem. 2020, 121, 2330–2342. [Google Scholar] [CrossRef] [PubMed]
  192. Lin, S.; Wang, H.; Yang, W.; Wang, A.; Geng, C. Silencing of Long Non-Coding RNA Colon Cancer-Associated Transcript 2 Inhibits the Growth and Metastasis of Gastric Cancer Through Blocking mTOR Signaling. Onco Targets Ther. 2020, 13, 337–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Qiu, L.; Lu, F.; Zhang, L.; Wang, G.; Geng, R.; Miao, Y. HBXIP Regulates Gastric Cancer Glucose Metabolism and Malignancy Through PI3K/AKT and p53 Signaling. Onco Targets Ther. 2020, 13, 3359–3374. [Google Scholar] [CrossRef] [Green Version]
  194. Huang, L.Y.; Wang, X.; Cui, X.F.; Li, H.; Zhao, J.; Wu, C.C.; Min, L.; Zhou, Z.; Wan, L.; Wang, Y.P.; et al. IRTKS is correlated with progression and survival time of patients with gastric cancer. Gut 2018, 67, 1400–1409. [Google Scholar] [CrossRef] [PubMed]
  195. Xu, S.H.; Huang, J.Z.; Chen, M.; Zeng, M.; Zou, F.Y.; Chen; Yan, G.R. Amplification of ACK1 promotes gastric tumorigenesis via ECD-dependent p53 ubiquitination degradation. Oncotarget 2017, 8, 12705–12716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Li, B.; Wang, L.; Li, Z.; Wang, W.; Zhi, X.; Huang, X.; Zhang, Q.; Chen, Z.; Zhang, X.; He, Z.; et al. miR-3174 Contributes to Apoptosis and Autophagic Cell Death Defects in Gastric Cancer Cells by Targeting ARHGAP10. Mol. Ther. Nucleic Acids 2017, 9, 294–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Zeng, Y.; Fu, M.; Wu, G.W.; Zhang, A.Z.; Chen, J.P.; Lin, H.Y.; Fu, Y.A.; Jia, J.; Cai, Z.D.; Wu, X.J.; et al. Upregulation of microRNA-370 promotes cell apoptosis and inhibits proliferation by targeting PTEN in human gastric cancer. Int. J. Oncol. 2016, 49, 1589–1599. [Google Scholar] [CrossRef] [Green Version]
  198. Shang, Y.; Feng, B.; Zhou, L.; Ren, G.; Zhang, Z.; Fan, X.; Sun, Y.; Luo, G.; Liang, J.; Wu, K.; et al. The miR27b-CCNG1-P53-miR-508-5p axis regulates multidrug resistance of gastric cancer. Oncotarget 2016, 7, 538–549. [Google Scholar] [CrossRef] [Green Version]
  199. Yang, G.; Gong, Y.; Wang, Q.; Wang, L.; Zhang, X. miR-100 antagonism triggers apoptosis by inhibiting ubiquitination-mediated p53 degradation. Oncogene 2017, 36, 1023–1037. [Google Scholar] [CrossRef]
  200. Sun, M.; Xia, R.; Jin, F.; Xu, T.; Liu, Z.; De, W.; Liu, X. Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumour Biol. 2014, 35, 1065–1073. [Google Scholar] [CrossRef]
  201. Liu, Y.; Yin, L.; Chen, C.; Zhang, X.; Wang, S. Long non-coding RNA GAS5 inhibits migration and invasion in gastric cancer via interacting with p53 protein. Dig. Liver Dis. 2020, 52, 331–338. [Google Scholar] [CrossRef]
  202. Wu, Z.H.; Liu, C.C.; Zhou, Y.Q.; Hu, L.N.; Guo, W.J. OnclncRNA-626 promotes malignancy of gastric cancer via inactivated the p53 pathway through interacting with SRSF1. Am. J. Cancer Res. 2019, 9, 2249–2263. [Google Scholar]
  203. Feng, L.; Li, J.; Li, F.; Li, H.; Bei, S.; Zhang, X.; Yang, Z. Long noncoding RNA VCAN-AS1 contributes to the progression of gastric cancer via regulating p53 expression. J. Cell. Physiol. 2020, 235, 4388–4398. [Google Scholar] [CrossRef] [PubMed]
  204. Kong, F.; Deng, X.; Kong, X.; Du, Y.; Li, L.; Zhu, H.; Wang, Y.; Xie, D.; Guha, S.; Li, Z.; et al. ZFPM2-AS1, a novel lncRNA, attenuates the p53 pathway and promotes gastric carcinogenesis by stabilizing MIF. Oncogene 2018, 37, 5982–5996. [Google Scholar] [CrossRef]
  205. Wu, Q.; Wang, H.; Liu, L.; Zhu, K.; Yu, W.; Guo, J. Hsa_circ_0001546 acts as a miRNA-421 sponge to inhibit the chemoresistance of gastric cancer cells via ATM/Chk2/p53-dependent pathway. Biochem. Biophys. Res. Commun. 2020, 521, 303–309. [Google Scholar] [CrossRef] [PubMed]
  206. Sasaki, Y.; Negishi, H.; Idogawa, M.; Yokota, I.; Koyama, R.; Kusano, M.; Suzuki, H.; Fujita, M.; Maruyama, R.; Toyota, M.; et al. p53 negatively regulates the hepatoma growth factor HDGF. Cancer Res. 2011, 71, 7038–7047. [Google Scholar] [CrossRef] [Green Version]
  207. Zhu, Y.; Zhou, J.; Xia, H.; Chen, X.; Qiu, M.; Huang, J.; Liu, S.; Tang, Q.; Lang, N.; Liu, Z.; et al. The Rho GTPase RhoE is a p53-regulated candidate tumor suppressor in cancer cells. Int. J. Oncol. 2014, 44, 896–904. [Google Scholar] [CrossRef] [Green Version]
  208. Yang, L.; Huang, X.; Wang, W.; Jiang, T.; Ding, F. XEDAR inhibits the proliferation and induces apoptosis of gastric cancer cells by regulating JNK signaling pathway. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef]
  209. Yokobori, T.; Mimori, K.; Iwatsuki, M.; Ishii, H.; Onoyama, I.; Fukagawa, T.; Kuwano, H.; Nakayama, K.I.; Mori, M. p53-Altered FBXW7 expression determines poor prognosis in gastric cancer cases. Cancer Res. 2009, 69, 3788–3794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Liu, Z.; Wu, Y.; Zhang, Y.; Yuan, M.; Li, X.; Gao, J.; Zhang, S.; Xing, C.; Qin, H.; Zhao, H.; et al. TIGAR Promotes Tumorigenesis and Protects Tumor Cells From Oxidative and Metabolic Stresses in Gastric Cancer. Front. Oncol. 2019, 9, 1258. [Google Scholar] [CrossRef] [Green Version]
  211. Qi, P.; Xu, M.D.; Shen, X.H.; Ni, S.J.; Huang, D.; Tan, C.; Weng, W.W.; Sheng, W.Q.; Zhou, X.Y.; Du, X. Reciprocal repression between TUSC7 and miR-23b in gastric cancer. Int. J. Cancer 2015, 137, 1269–1278. [Google Scholar] [CrossRef] [PubMed]
  212. Zhang, N.; Jiang, T.; Wang, Y.; Wang, S.; Hu, L.; Bu, Y. BTG4 is A Novel p53 Target Gene That Inhibits Cell Growth and Induces Apoptosis. Genes 2020, 11, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Dong, W.; Tu, S.; Xie, J.; Sun, P.; Wu, Y.; Wang, L. Frequent promoter hypermethylation and transcriptional downregulation of BTG4 gene in gastric cancer. Biochem. Biophys. Res. Commun. 2009, 387, 132–138. [Google Scholar] [CrossRef] [PubMed]
  214. Park, W.S.; Lee, J.H.; Shin, M.S.; Park, J.Y.; Kim, H.S.; Kim, Y.S.; Park, C.H.; Lee, S.K.; Lee, S.H.; Lee, S.N.; et al. Inactivating mutations of KILLER/DR5 gene in gastric cancers. Gastroenterology 2001, 121, 1219–1225. [Google Scholar] [CrossRef]
  215. Wang, P.; Wang, Y.; Hang, B.; Zou, X.; Mao, J.H. A novel gene expression-based prognostic scoring system to predict survival in gastric cancer. Oncotarget 2016, 7, 55343–55351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Matozaki, T.; Sakamoto, C.; Suzuki, T.; Matsuda, K.; Uchida, T.; Nakano, O.; Wada, K.; Nishisaki, H.; Konda, Y.; Nagao, M.; et al. p53 gene mutations in human gastric cancer: Wild-type p53 but not mutant p53 suppresses growth of human gastric cancer cells. Cancer Res. 1992, 52, 4335–4341. [Google Scholar] [PubMed]
  217. Renault, B.; van den Broek, M.; Fodde, R.; Wijnen, J.; Pellegata, N.S.; Amadori, D.; Khan, P.M.; Ranzani, G.N. Base transitions are the most frequent genetic changes at P53 in gastric cancer. Cancer Res. 1993, 53, 2614–2617. [Google Scholar] [PubMed]
  218. Yanagihara, K.; Numoto, M.; Tauchi, H.; Akama, Y.; Yokozaki, H.; Tahara, E.; Kamiya, K.; Seito, T. Genetic status of p53 and induction of apoptosis by radiation or isoflavones in human gastric carcinoma cell lines. Int. J. Oncol. 1996, 9, 95–102. [Google Scholar] [CrossRef]
  219. Ikeguchi, M.; Tatebe, S.; Kaibara, N.; Ito, H. Changes in levels of expression of p53 and the product of the bcl-2 in lines of gastric cancer cells during cisplatin-induced apoptosis. Eur. Surg. Res. 1997, 29, 396–402. [Google Scholar] [CrossRef]
  220. Ji, W.; Ma, J.; Zhang, H.; Zhong, H.; Li, L.; Ding, N.; Jiao, J.; Gao, Z. Role of p53beta in the inhibition of proliferation of gastric cancer cells expressing wild-type or mutated p53. Mol. Med. Rep. 2015, 12, 691–695. [Google Scholar] [CrossRef]
  221. Nabeya, Y.; Loganzo, F., Jr.; Maslak, P.; Lai, L.; de Oliveira, A.R.; Schwartz, G.K.; Blundell, M.L.; Altorki, N.K.; Kelsen, D.P.; Albino, A.P. The mutational status of p53 protein in gastric and esophageal adenocarcinoma cell lines predicts sensitivity to chemotherapeutic agents. Int. J. Cancer 1995, 64, 37–46. [Google Scholar] [CrossRef]
  222. Matsuhashi, N.; Saio, M.; Matsuo, A.; Sugiyama, Y.; Saji, S. The evaluation of gastric cancer sensitivity to 5-FU/CDDP in terms of induction of apoptosis: Time- and p53 expression-dependency of anti-cancer drugs. Oncol. Rep. 2005, 14, 609–615. [Google Scholar] [CrossRef]
  223. Matsuhashi, N.; Saio, M.; Matsuo, A.; Sugiyama, Y.; Saji, S. Expression of p53 protein as a predictor of the response to 5-fluorouracil and cisplatin chemotherapy in human gastrointestinal cancer cell lines evaluated with apoptosis by use of thin layer collagen gel. Int. J. Oncol. 2004, 24, 807–813. [Google Scholar] [CrossRef] [PubMed]
  224. Kim, C.W.; Lu, J.N.; Go, S.I.; Jung, J.H.; Yi, S.M.; Jeong, J.H.; Hah, Y.S.; Han, M.S.; Park, J.W.; Lee, W.S.; et al. p53 restoration can overcome cisplatin resistance through inhibition of Akt as well as induction of Bax. Int. J. Oncol. 2013, 43, 1495–1502. [Google Scholar] [CrossRef] [PubMed]
  225. Osaki, M.; Tatebe, S.; Goto, A.; Hayashi, H.; Oshimura, M.; Ito, H. 5-Fluorouracil (5-FU) induced apoptosis in gastric cancer cell lines: Role of the p53 gene. Apoptosis Int. J. Program. Cell Death 1997, 2, 221–226. [Google Scholar] [CrossRef] [PubMed]
  226. Litvak, D.A.; Papaconstantinou, H.T.; Hwang, K.O.; Kim, M.; Evers, B.M.; Townsend, C.M., Jr. Inhibition of gastric cancer by camptothecin involves apoptosis and multiple cellular pathways. Surgery 1999, 126, 223–230. [Google Scholar] [CrossRef]
  227. Kastan, M.B.; Onyekwere, O.; Sidransky, D.; Vogelstein, B.; Craig, R.W. Participation of p53 protein in the cellular response to DNA damage. Cancer Res. 1991, 51, 6304–6311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Fujiwara, T.; Grimm, E.A.; Mukhopadhyay, T.; Zhang, W.W.; Owen-Schaub, L.B.; Roth, J.A. Induction of chemosensitivity in human lung cancer cells in vivo by adenovirus-mediated transfer of the wild-type p53 gene. Cancer Res. 1994, 54, 2287–2291. [Google Scholar] [PubMed]
  229. Jiang, X.H.; Wong, B.C.; Yuen, S.T.; Jiang, S.H.; Cho, C.H.; Lai, K.C.; Lin, M.C.; Kung, H.F.; Lam, S.K. Arsenic trioxide induces apoptosis in human gastric cancer cells through up-regulation of p53 and activation of caspase-3. Int. J. Cancer 2001, 91, 173–179. [Google Scholar] [CrossRef] [Green Version]
  230. Kubota, E.; Williamson, C.T.; Ye, R.; Elegbede, A.; Peterson, L.; Lees-Miller, S.P.; Bebb, D.G. Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines. Cell Cycle. 2014, 13, 2129–2137. [Google Scholar] [CrossRef] [Green Version]
  231. Koike, M.; Fujita, F.; Komori, K.; Katoh, F.; Sugimoto, T.; Sakamoto, Y.; Matsuda, M.; Fujita, M. Dependence of chemotherapy response on p53 mutation status in a panel of human cancer lines maintained in nude mice. Cancer Sci. 2004, 95, 541–546. [Google Scholar] [CrossRef] [Green Version]
  232. Muller, M.; Wilder, S.; Bannasch, D.; Israeli, D.; Lehlbach, K.; Li-Weber, M.; Friedman, S.L.; Galle, P.R.; Stremmel, W.; Oren, M.; et al. p53 activates the CD95 (APO-1/Fas) gene in response to DNA damage by anticancer drugs. J. Exp. Med. 1998, 188, 2033–2045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Wu, G.S.; Burns, T.F.; McDonald, E.R., 3rd; Jiang, W.; Meng, R.; Krantz, I.D.; Kao, G.; Gan, D.D.; Zhou, J.Y.; Muschel, R.; et al. KILLER/DR5 is a DNA damage-inducible p53-regulated death receptor gene. Nat. Genet. 1997, 17, 141–143. [Google Scholar] [CrossRef]
  234. Tu, S.P.; Chi, A.L.; Ai, W.; Takaishi, S.; Dubeykovskaya, Z.; Quante, M.; Fox, J.G.; Wang, T.C. p53 inhibition of AP1-dependent TFF2 expression induces apoptosis and inhibits cell migration in gastric cancer cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 297, G385–G396. [Google Scholar] [CrossRef] [PubMed]
  235. Bargiela-Iparraguirre, J.; Prado-Marchal, L.; Fernandez-Fuente, M.; Gutierrez-Gonzalez, A.; Moreno-Rubio, J.; Munoz-Fernandez, M.; Sereno, M.; Sanchez-Prieto, R.; Perona, R.; Sanchez-Perez, I. CHK1 expression in Gastric Cancer is modulated by p53 and RB1/E2F1: Implications in chemo/radiotherapy response. Sci. Rep. 2016, 6, 21519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Lee, J.H.; Byun, D.S.; Lee, M.G.; Ryu, B.K.; Kang, M.J.; Chae, K.S.; Lee, K.Y.; Kim, H.J.; Park, H.; Chi, S.G. Frequent epigenetic inactivation of hSRBC in gastric cancer and its implication in attenuated p53 response to stresses. Int. J. Cancer 2008, 122, 1573–1584. [Google Scholar] [CrossRef] [PubMed]
  237. Hsieh, M.Y.; Fan, J.R.; Chang, H.W.; Chen, H.C.; Shen, T.L.; Teng, S.C.; Yeh, Y.H.; Li, T.K. DNA topoisomerase III alpha regulates p53-mediated tumor suppression. Clin. Cancer Res. 2014, 20, 1489–1501. [Google Scholar] [CrossRef] [Green Version]
  238. Tong, Q.S.; Zheng, L.D.; Wang, L.; Zeng, F.Q.; Chen, F.M.; Dong, J.H.; Lu, G.C. Downregulation of XIAP expression induces apoptosis and enhances chemotherapeutic sensitivity in human gastric cancer cells. Cancer Gene Ther. 2005, 12, 509–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Liu, Y.; Xing, R.; Zhang, X.; Dong, W.; Zhang, J.; Yan, Z.; Li, W.; Cui, J.; Lu, Y. miR-375 targets the p53 gene to regulate cellular response to ionizing radiation and etoposide in gastric cancer cells. DNA Repair 2013, 12, 741–750. [Google Scholar] [CrossRef]
  240. Xing, C.; Zhu, B.; Liu, H.; Yao, H.; Zhang, L. Class I phosphatidylinositol 3-kinase inhibitor LY294002 activates autophagy and induces apoptosis through p53 pathway in gastric cancer cell line SGC7901. Acta Biochim. Biophys. Sin. 2008, 40, 194–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Park, I.C.; Park, M.J.; Lee, S.H.; Choe, T.B.; Jang, J.J.; Hong, S.I. Increased susceptibility of the c-Myc overexpressing cell line, SNU-16, to TNF-alpha. Cancer Lett. 1998, 125, 17–23. [Google Scholar] [CrossRef]
  242. Fukui, T.; Matsui, K.; Kato, H.; Takao, H.; Sugiyama, Y.; Kunieda, K.; Saji, S. Significance of apoptosis induced by tumor necrosis factor-alpha and/or interferon-gamma against human gastric cancer cell lines and the role of the p53 gene. Surg. Today 2003, 33, 847–853. [Google Scholar] [CrossRef]
  243. Shimoyama, S.; Mochizuki, Y.; Kusada, O.; Kaminishi, M. Supra-additive antitumor activity of 5FU with tumor necrosis factor-related apoptosis-inducing ligand on gastric and colon cancers in vitro. Int. J. Oncol. 2002, 21, 643–648. [Google Scholar] [CrossRef] [PubMed]
  244. Leung, W.K.; Bai, A.H.; Chan, V.Y.; Yu, J.; Chan, M.W.; To, K.F.; Wu, J.R.; Chan, K.K.; Fu, Y.G.; Chan, F.K.; et al. Effect of peroxisome proliferator activated receptor gamma ligands on growth and gene expression profiles of gastric cancer cells. Gut 2004, 53, 331–338. [Google Scholar] [CrossRef] [Green Version]
  245. Nagamine, M.; Okumura, T.; Tanno, S.; Sawamukai, M.; Motomura, W.; Takahashi, N.; Kohgo, Y. PPAR gamma ligand-induced apoptosis through a p53-dependent mechanism in human gastric cancer cells. Cancer Sci. 2003, 94, 338–343. [Google Scholar] [CrossRef]
  246. Liu, F.; Fan, Z.; Song, N.; Han, M.; Yan, M.; Guo, L.H.; Jihui, J.; Liu, S. XRCC4, which is inhibited by PFDA, regulates DNA damage repair and cell chemosensitivity. J. Cell. Biochem. 2019, 120, 12665–12676. [Google Scholar] [CrossRef]
  247. Ma, K.; Liu, Y.; Zhu, Q.; Liu, C.H.; Duan, J.L.; Tan, B.K.; Zhu, Y.Z. H2S donor, S-propargyl-cysteine, increases CSE in SGC-7901 and cancer-induced mice: Evidence for a novel anti-cancer effect of endogenous H2S? PLoS ONE 2011, 6, e20525. [Google Scholar] [CrossRef] [PubMed]
  248. Ma, T.; Xu, J.; Wang, Y.; Yu, H.; Yang, Y.; Liu, Y.; Ding, W.; Zhu, W.; Chen, R.; Ge, Z.; et al. Ternary copper(II) complexes with amino acid chains and heterocyclic bases: DNA binding, cytotoxic and cell apoptosis induction properties. J. Inorg. Biochem. 2015, 144, 38–46. [Google Scholar] [CrossRef]
  249. Feng, J.; Li, C.; Xu, R.; Li, Y.; Hou, Q.; Feng, R.; Wang, S.; Zhang, L.; Li, C. DpdtC-Induced EMT Inhibition in MGC-803 Cells Was Partly through Ferritinophagy-Mediated ROS/p53 Pathway. Oxid. Med. Cell Longev. 2020, 2020, 9762390. [Google Scholar] [CrossRef]
  250. Liu, Z.; Ng, E.K.; Liang, N.C.; Deng, Y.F.; Leung, B.C.; Chen, G.G. Cell death induced by Pteris semipinnata L. is associated with p53 and oxidant stress in gastric cancer cells. FEBS Lett. 2005, 579, 1477–1487. [Google Scholar] [CrossRef] [Green Version]
  251. Sun, B.; Geng, S.; Huang, X.; Zhu, J.; Liu, S.; Zhang, Y.; Ye, J.; Li, Y.; Wang, J. Coleusin factor exerts cytotoxic activity by inducing G0/G1 cell cycle arrest and apoptosis in human gastric cancer BGC-823 cells. Cancer Lett. 2011, 301, 95–105. [Google Scholar] [CrossRef] [PubMed]
  252. Wang, B.Y.; Cao, J.; Chen, J.W.; Liu, Q.Y. Triptolide induces apoptosis of gastric cancer cells via inhibiting the overexpression of MDM2. Med. Oncol. 2014, 31, 270. [Google Scholar] [CrossRef] [PubMed]
  253. Yamamoto, M.; Maehara, Y.; Sakaguchi, Y.; Kusumoto, T.; Ichiyoshi, Y.; Sugimachi, K. Transforming growth factor-beta 1 induces apoptosis in gastric cancer cells through a p53-independent pathway. Cancer 1996, 77, 1628–1633. [Google Scholar] [CrossRef]
  254. Akagi, M.; Yasui, W.; Akama, Y.; Yokozaki, H.; Tahara, H.; Haruma, K.; Kajiyama, G.; Tahara, E. Inhibition of cell growth by transforming growth factor beta 1 is associated with p53-independent induction of p21 in gastric carcinoma cells. Jpn. J. Cancer Res. 1996, 87, 377–384. [Google Scholar] [CrossRef]
  255. Takaku, K.; Miyoshi, H.; Matsunaga, A.; Oshima, M.; Sasaki, N.; Taketo, M.M. Gastric and duodenal polyps in Smad4 (Dpc4) knockout mice. Cancer Res. 1999, 59, 6113–6117. [Google Scholar]
  256. Liu, Z.M.; Chen, G.G.; Ng, E.K.; Leung, W.K.; Sung, J.J.; Chung, S.C. Upregulation of heme oxygenase-1 and p21 confers resistance to apoptosis in human gastric cancer cells. Oncogene 2004, 23, 503–513. [Google Scholar] [CrossRef] [Green Version]
  257. Matsuhashi, N.; Saio, M.; Matsuo, A.; Sugiyama, Y.; Saji, S. Apoptosis induced by 5-fluorouracil, cisplatin and paclitaxel are associated with p53 gene status in gastric cancer cell lines. Int. J. Oncol. 2005, 26, 1563–1567. [Google Scholar] [CrossRef]
  258. Liu, X.; Guo, W.J.; Zhang, X.W.; Cai, X.; Tian, S.; Li, J. Cetuximab enhances the activities of irinotecan on gastric cancer cell lines through downregulating the EGFR pathway upregulated by irinotecan. Cancer Chemother. Pharmacol. 2011, 68, 871–878. [Google Scholar] [CrossRef]
  259. Li, Y.; Qu, X.; Qu, J.; Zhang, Y.; Liu, J.; Teng, Y.; Hu, X.; Hou, K.; Liu, Y. Arsenic trioxide induces apoptosis and G2/M phase arrest by inducing Cbl to inhibit PI3K/Akt signaling and thereby regulate p53 activation. Cancer Lett. 2009, 284, 208–215. [Google Scholar] [CrossRef]
  260. Licona, C.; Delhorme, J.B.; Riegel, G.; Vidimar, V.; Ceron-Camacho, R.; Boff, B.; Venkatasamy, A.; Tomasetto, C.; Gomes, P.; Rognan, D.; et al. Anticancer activity of ruthenium and osmium cyclometalated compounds: Identification of ABCB1 and EGFR as resistance mechanisms. Inorg. Chem. Front. 2020, 7, 678–688. [Google Scholar] [CrossRef]
  261. Licona, C.; Spaety, M.E.; Capuozzo, A.; Ali, M.; Santamaria, R.; Armant, O.; Delalande, F.; Van Dorsselaer, A.; Cianferani, S.; Spencer, J.; et al. A ruthenium anticancer compound interacts with histones and impacts differently on epigenetic and death pathways compared to cisplatin. Oncotarget 2017. [Google Scholar] [CrossRef] [Green Version]
  262. Bonelli, P.; Tuccillo, F.M.; Calemma, R.; Pezzetti, F.; Borrelli, A.; Martinelli, R.; De Rosa, A.; Esposito, D.; Palaia, R.; Castello, G. Changes in the gene expression profile of gastric cancer cells in response to ibuprofen: A gene pathway analysis. Pharmacogenom. J. 2011, 11, 412–428. [Google Scholar] [CrossRef] [Green Version]
  263. Zhu, G.H.; Wong, B.C.; Eggo, M.C.; Ching, C.K.; Yuen, S.T.; Chan, E.Y.; Lai, K.C.; Lam, S.K. Non-steroidal anti-inflammatory drug-induced apoptosis in gastric cancer cells is blocked by protein kinase C activation through inhibition of c-myc. Br. J. Cancer 1999, 79, 393–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Zhu, G.H.; Wong, B.C.; Ching, C.K.; Lai, K.C.; Lam, S.K. Differential apoptosis by indomethacin in gastric epithelial cells through the constitutive expression of wild-type p53 and/or up-regulation of c-myc. Biochem. Pharmacol. 1999, 58, 193–200. [Google Scholar] [CrossRef]
  265. Ishihara, T.; Hoshino, T.; Namba, T.; Tanaka, K.; Mizushima, T. Involvement of up-regulation of PUMA in non-steroidal anti-inflammatory drug-induced apoptosis. Biochem. Biophys. Res. Commun. 2007, 356, 711–717. [Google Scholar] [CrossRef]
  266. Subbaramaiah, K.; Altorki, N.; Chung, W.J.; Mestre, J.R.; Sampat, A.; Dannenberg, A.J. Inhibition of cyclooxygenase-2 gene expression by p53. J. Biol. Chem. 1999, 274, 10911–10915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Leung, W.K.; To, K.F.; Ng, Y.P.; Lee, T.L.; Lau, J.Y.; Chan, F.K.; Ng, E.K.; Chung, S.C.; Sung, J.J. Association between cyclo-oxygenase-2 overexpression and missense p53 mutations in gastric cancer. Br. J. Cancer 2001, 84, 335–339. [Google Scholar] [CrossRef] [Green Version]
  268. Harvey, M.; McArthur, M.J.; Montgomery, C.A., Jr.; Butel, J.S.; Bradley, A.; Donehower, L.A. Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice. Nat. Genet. 1993, 5, 225–229. [Google Scholar] [CrossRef] [PubMed]
  269. Donehower, L.A.; Harvey, M.; Slagle, B.L.; McArthur, M.J.; Montgomery, C.A., Jr.; Butel, J.S.; Bradley, A. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 1992, 356, 215–221. [Google Scholar] [CrossRef]
  270. Fong, L.Y.; Ishii, H.; Nguyen, V.T.; Vecchione, A.; Farber, J.L.; Croce, C.M.; Huebner, K. p53 deficiency accelerates induction and progression of esophageal and forestomach tumors in zinc-deficient mice. Cancer Res. 2003, 63, 186–195. [Google Scholar]
  271. Reaves, S.K.; Fanzo, J.C.; Arima, K.; Wu, J.Y.; Wang, Y.R.; Lei, K.Y. Expression of the p53 tumor suppressor gene is up-regulated by depletion of intracellular zinc in HepG2 cells. J. Nutr. 2000, 130, 1688–1694. [Google Scholar] [CrossRef] [Green Version]
  272. Li, X.; Nadauld, L.; Ootani, A.; Corney, D.C.; Pai, R.K.; Gevaert, O.; Cantrell, M.A.; Rack, P.G.; Neal, J.T.; Chan, C.W.; et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat. Med. 2014, 20, 769–777. [Google Scholar] [CrossRef] [Green Version]
  273. Shimada, S.; Mimata, A.; Sekine, M.; Mogushi, K.; Akiyama, Y.; Fukamachi, H.; Jonkers, J.; Tanaka, H.; Eishi, Y.; Yuasa, Y. Synergistic tumour suppressor activity of E-cadherin and p53 in a conditional mouse model for metastatic diffuse-type gastric cancer. Gut 2012, 61, 344–353. [Google Scholar] [CrossRef] [PubMed]
  274. Park, J.W.; Jang, S.H.; Park, D.M.; Lim, N.J.; Deng, C.; Kim, D.Y.; Green, J.E.; Kim, H.K. Cooperativity of E-cadherin and Smad4 loss to promote diffuse-type gastric adenocarcinoma and metastasis. Mol. Cancer Res. 2014, 12, 1088–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Park, J.W.; Kim, M.S.; Voon, D.C.; Kim, S.J.; Bae, J.; Mun, D.G.; Ko, S.I.; Kim, H.K.; Lee, S.W.; Kim, D.Y. Multi-omics analysis identifies pathways and genes involved in diffuse-type gastric carcinogenesis induced by E-cadherin, p53, and Smad4 loss in mice. Mol. Carcinog. 2018, 57, 947–954. [Google Scholar] [CrossRef] [PubMed]
  276. Till, J.E.; Yoon, C.; Kim, B.J.; Roby, K.; Addai, P.; Jonokuchi, E.; Tang, L.H.; Yoon, S.S.; Ryeom, S. Oncogenic KRAS and p53 Loss Drive Gastric Tumorigenesis in Mice That Can Be Attenuated by E-Cadherin Expression. Cancer Res. 2017, 77, 5349–5359. [Google Scholar] [CrossRef] [Green Version]
  277. Seidlitz, T.; Chen, Y.T.; Uhlemann, H.; Scholch, S.; Kochall, S.; Merker, S.R.; Klimova, A.; Hennig, A.; Schweitzer, C.; Pape, K.; et al. Mouse Models of Human Gastric Cancer Subtypes With Stomach-Specific CreERT2-Mediated Pathway Alterations. Gastroenterology 2019, 157, 1599–1614. [Google Scholar] [CrossRef] [Green Version]
  278. Rubio, C.A. Serrated neoplasia of the stomach: A new entity. J. Clin. Pathol. 2001, 54, 849–853. [Google Scholar] [CrossRef]
  279. Wei, C.; Amos, C.I.; Stephens, L.C.; Campos, I.; Deng, J.M.; Behringer, R.R.; Rashid, A.; Frazier, M.L. Mutation of Lkb1 and p53 genes exert a cooperative effect on tumorigenesis. Cancer Res. 2005, 65, 11297–11303. [Google Scholar] [CrossRef] [Green Version]
  280. Fu, D.J.; Wang, L.; Chouairi, F.K.; Rose, I.M.; Abetov, D.A.; Miller, A.D.; Yamulla, R.J.; Schimenti, J.C.; Flesken-Nikitin, A.; Nikitin, A.Y. Gastric squamous-columnar junction contains a large pool of cancer-prone immature osteopontin responsive Lgr5(−)CD44(+) cells. Nat. Commun. 2020, 11, 84. [Google Scholar] [CrossRef] [Green Version]
  281. Zhang, Z.; Wang, Y.; Yao, R.; Li, J.; Lubet, R.A.; You, M. p53 Transgenic mice are highly susceptible to 4-nitroquinoline-1-oxide-induced oral cancer. Mol. Cancer Res. 2006, 4, 401–410. [Google Scholar] [CrossRef] [Green Version]
  282. Ohtsuka, J.; Oshima, H.; Ezawa, I.; Abe, R.; Oshima, M.; Ohki, R. Functional loss of p53 cooperates with the in vivo microenvironment to promote malignant progression of gastric cancers. Sci. Rep. 2018, 8, 2291. [Google Scholar] [CrossRef] [PubMed]
  283. Guo, L.; Chen, C.; Shi, M.; Wang, F.; Chen, X.; Diao, D.; Hu, M.; Yu, M.; Qian, L.; Guo, N. Stat3-coordinated Lin-28-let-7-HMGA2 and miR-200-ZEB1 circuits initiate and maintain oncostatin M-driven epithelial-mesenchymal transition. Oncogene 2013, 32, 5272–5282. [Google Scholar] [CrossRef] [PubMed]
  284. Fox, J.G.; Li, X.; Cahill, R.J.; Andrutis, K.; Rustgi, A.K.; Odze, R.; Wang, T.C. Hypertrophic gastropathy in Helicobacter felis-infected wild-type C57BL/6 mice and p53 hemizygous transgenic mice. Gastroenterology 1996, 110, 155–166. [Google Scholar] [CrossRef] [PubMed]
  285. Fox, J.G.; Sheppard, B.J.; Dangler, C.A.; Whary, M.T.; Ihrig, M.; Wang, T.C. Germ-line p53-targeted disruption inhibits helicobacter-induced premalignant lesions and invasive gastric carcinoma through down-regulation of Th1 proinflammatory responses. Cancer Res. 2002, 62, 696–702. [Google Scholar]
  286. Wei, J.; Nagy, T.A.; Vilgelm, A.; Zaika, E.; Ogden, S.R.; Romero-Gallo, J.; Piazuelo, M.B.; Correa, P.; Washington, M.K.; El-Rifai, W.; et al. Regulation of p53 tumor suppressor by Helicobacter pylori in gastric epithelial cells. Gastroenterology 2010, 139, 1333–1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Shirin, H.; Sordillo, E.M.; Oh, S.H.; Yamamoto, H.; Delohery, T.; Weinstein, I.B.; Moss, S.F. Helicobacter pylori inhibits the G1 to S transition in AGS gastric epithelial cells. Cancer Res. 1999, 59, 2277–2281. [Google Scholar] [PubMed]
  288. Cho, S.J.; Kang, N.S.; Park, S.Y.; Kim, B.O.; Rhee, D.K.; Pyo, S. Induction of apoptosis and expression of apoptosis related genes in human epithelial carcinoma cells by Helicobacter pylori VacA toxin. Toxicon 2003, 42, 601–611. [Google Scholar] [CrossRef]
  289. Wei, J.; Noto, J.M.; Zaika, E.; Romero-Gallo, J.; Piazuelo, M.B.; Schneider, B.; El-Rifai, W.; Correa, P.; Peek, R.M.; Zaika, A.I. Bacterial CagA protein induces degradation of p53 protein in a p14ARF-dependent manner. Gut 2015, 64, 1040–1048. [Google Scholar] [CrossRef]
  290. Buti, L.; Spooner, E.; Van der Veen, A.G.; Rappuoli, R.; Covacci, A.; Ploegh, H.L. Helicobacter pylori cytotoxin-associated gene A (CagA) subverts the apoptosis-stimulating protein of p53 (ASPP2) tumor suppressor pathway of the host. Proc. Natl. Acad. Sci. USA 2011, 108, 9238–9243. [Google Scholar] [CrossRef] [Green Version]
  291. Bhardwaj, V.; Noto, J.M.; Wei, J.; Andl, C.; El-Rifai, W.; Peek, R.M.; Zaika, A.I. Helicobacter pylori bacteria alter the p53 stress response via ERK-HDM2 pathway. Oncotarget 2015, 6, 1531–1543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Coombs, N.; Sompallae, R.; Olbermann, P.; Gastaldello, S.; Goppel, D.; Masucci, M.G.; Josenhans, C. Helicobacter pylori affects the cellular deubiquitinase USP7 and ubiquitin-regulated components TRAF6 and the tumour suppressor p53. Int. J. Med. Microbiol. 2011, 301, 213–224. [Google Scholar] [CrossRef] [PubMed]
  293. Costa, L.; Corre, S.; Michel, V.; Le Luel, K.; Fernandes, J.; Ziveri, J.; Jouvion, G.; Danckaert, A.; Mouchet, N.; Da Silva Barreira, D.; et al. USF1 defect drives p53 degradation during Helicobacter pylori infection and accelerates gastric carcinogenesis. Gut 2020, 69, 1582–1591. [Google Scholar] [CrossRef] [PubMed]
  294. Wei, J.; Noto, J.; Zaika, E.; Romero-Gallo, J.; Correa, P.; El-Rifai, W.; Peek, R.M.; Zaika, A. Pathogenic bacterium Helicobacter pylori alters the expression profile of p53 protein isoforms and p53 response to cellular stresses. Proc. Natl. Acad. Sci. USA 2012, 109, E2543–E2550. [Google Scholar] [CrossRef] [Green Version]
  295. Chatterjee, K.; Das, P.; Chattopadhyay, N.R.; Mal, S.; Choudhuri, T. The interplay between Epstein-Bar virus (EBV) with the p53 and its homologs during EBV associated malignancies. Heliyon 2019, 5, e02624. [Google Scholar] [CrossRef]
  296. Tornesello, M.L.; Annunziata, C.; Tornesello, A.L.; Buonaguro, L.; Buonaguro, F.M. Human Oncoviruses and p53 Tumor Suppressor Pathway Deregulation at the Origin of Human Cancers. Cancers 2018, 10, 213. [Google Scholar] [CrossRef] [Green Version]
  297. Fitzsimmons, L.; Kelly, G.L. EBV and Apoptosis: The Viral Master Regulator of Cell Fate? Viruses 2017, 9, 339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  298. Koriyama, C.; Akiba, S.; Itoh, T.; Kijima, Y.; Sueyoshi, K.; Corvalan, A.; Herrera-Goepfer, R.; Eizuru, Y. Prognostic significance of Epstein-Barr virus involvement in gastric carcinoma in Japan. Int. J. Mol. Med. 2002, 10, 635–639. [Google Scholar] [PubMed]
  299. Gulley, M.L.; Pulitzer, D.R.; Eagan, P.A.; Schneider, B.G. Epstein-Barr virus infection is an early event in gastric carcinogenesis and is independent of bcl-2 expression and p53 accumulation. Hum. Pathol. 1996, 27, 20–27. [Google Scholar] [CrossRef]
  300. Ojima, H.; Fukuda, T.; Nakajima, T.; Nagamachi, Y. Infrequent overexpression of p53 protein in Epstein-Barr virus-associated gastric carcinomas. Jpn. J. Cancer Res. 1997, 88, 262–266. [Google Scholar] [CrossRef]
  301. Leung, S.Y.; Chau, K.Y.; Yuen, S.T.; Chu, K.M.; Branicki, F.J.; Chung, L.P. p53 overexpression is different in Epstein-Barr virus-associated and Epstein-Barr virus-negative carcinoma. Histopathology 1998, 33, 311–317. [Google Scholar] [PubMed]
  302. Chapel, F.; Fabiani, B.; Davi, F.; Raphael, M.; Tepper, M.; Champault, G.; Guettier, C. Epstein-Barr virus and gastric carcinoma in Western patients: Comparison of pathological parameters and p53 expression in EBV-positive and negative tumours. Histopathology 2000, 36, 252–261. [Google Scholar] [CrossRef]
  303. Sivachandran, N.; Dawson, C.W.; Young, L.S.; Liu, F.F.; Middeldorp, J.; Frappier, L. Contributions of the Epstein-Barr virus EBNA1 protein to gastric carcinoma. J. Virol. 2012, 86, 60–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Saridakis, V.; Sheng, Y.; Sarkari, F.; Holowaty, M.N.; Shire, K.; Nguyen, T.; Zhang, R.G.; Liao, J.; Lee, W.; Edwards, A.M.; et al. Structure of the p53 binding domain of HAUSP/USP7 bound to Epstein-Barr nuclear antigen 1 implications for EBV-mediated immortalization. Mol. Cell 2005, 18, 25–36. [Google Scholar] [CrossRef] [PubMed]
  305. Sato, Y.; Kamura, T.; Shirata, N.; Murata, T.; Kudoh, A.; Iwahori, S.; Nakayama, S.; Isomura, H.; Nishiyama, Y.; Tsurumi, T. Degradation of phosphorylated p53 by viral protein-ECS E3 ligase complex. PLoS Pathog. 2009, 5, e1000530. [Google Scholar] [CrossRef] [Green Version]
  306. Yi, F.; Saha, A.; Murakami, M.; Kumar, P.; Knight, J.S.; Cai, Q.; Choudhuri, T.; Robertson, E.S. Epstein-Barr virus nuclear antigen 3C targets p53 and modulates its transcriptional and apoptotic activities. Virology 2009, 388, 236–247. [Google Scholar] [CrossRef] [Green Version]
  307. Zheng, X.; Wang, J.; Wei, L.; Peng, Q.; Gao, Y.; Fu, Y.; Lu, Y.; Qin, Z.; Zhang, X.; Lu, J.; et al. Epstein-Barr Virus MicroRNA miR-BART5-3p Inhibits p53 Expression. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [Green Version]
  308. Wang, J.; Zheng, X.; Qin, Z.; Wei, L.; Lu, Y.; Peng, Q.; Gao, Y.; Zhang, X.; Zhang, X.; Li, Z.; et al. Epstein-Barr virus miR-BART3-3p promotes tumorigenesis by regulating the senescence pathway in gastric cancer. J. Biol. Chem. 2019, 294, 4854–4866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  309. Gong, L.P.; Chen, J.N.; Dong, M.; Xiao, Z.D.; Feng, Z.Y.; Pan, Y.H.; Zhang, Y.; Du, Y.; Zhang, J.Y.; Bi, Y.H.; et al. Epstein-Barr virus-derived circular RNA LMP2A induces stemness in EBV-associated gastric cancer. EMBO Rep. 2020, e49689. [Google Scholar] [CrossRef]
  310. Li, L.; Li, W.; Xiao, L.; Xu, J.; Chen, X.; Tang, M.; Dong, Z.; Tao, Q.; Cao, Y. Viral oncoprotein LMP1 disrupts p53-induced cell cycle arrest and apoptosis through modulating K63-linked ubiquitination of p53. Cell Cycle. 2012, 11, 2327–2336. [Google Scholar] [CrossRef] [Green Version]
  311. Ribeiro, J.; Malta, M.; Galaghar, A.; Silva, F.; Afonso, L.P.; Medeiros, R.; Sousa, H. P53 deregulation in Epstein-Barr virus-associated gastric cancer. Cancer Lett. 2017, 404, 37–43. [Google Scholar] [CrossRef] [PubMed]
  312. Hagemeier, S.R.; Barlow, E.A.; Meng, Q.; Kenney, S.C. The cellular ataxia telangiectasia-mutated kinase promotes epstein-barr virus lytic reactivation in response to multiple different types of lytic reactivation-inducing stimuli. J. Virol. 2012, 86, 13360–13370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Kraus, R.J.; Cordes, B.A.; Sathiamoorthi, S.; Patel, P.; Yuan, X.; Iempridee, T.; Yu, X.; Lee, D.L.; Lambert, P.F.; Mertz, J.E. Reactivation of Epstein-Barr Virus by HIF-1alpha Requires p53. J. Virol. 2020, 94. [Google Scholar] [CrossRef] [PubMed]
  314. Shin, S.K.; Li, M.S.; Fuerst, F.; Hotchkiss, E.; Meyer, R.; Kim, I.T.; Goel, A.; Boland, C.R. Oncogenic T-antigen of JC virus is present frequently in human gastric cancers. Cancer 2006, 107, 481–488. [Google Scholar] [CrossRef] [PubMed]
  315. Delbue, S.; Comar, M.; Ferrante, P. Review on the role of the human Polyomavirus JC in the development of tumors. Infect. Agent Cancer 2017, 12, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Sermeus, A.; Michiels, C. Reciprocal influence of the p53 and the hypoxic pathways. Cell Death Dis. 2011, 2, e164. [Google Scholar] [CrossRef] [Green Version]
  317. Ioachim, E.; Goussia, A.; Stefanou, D.; Agnantis, N.J. Expression of p53 protein in gastric cancer: An immunohistochemical study with correlation to proliferative activity. Anticancer Res. 1997, 17, 513–517. [Google Scholar]
  318. Saito, H.; Tujitani, S.; Ikeguchi, M.; Maeta, M.; Kaibara, N. Neoangiogenesis and relationship to nuclear p53 accumulation and vascular endothelial growth factor expression in advanced gastric carcinoma. Oncology 1999, 57, 164–172. [Google Scholar] [CrossRef] [PubMed]
  319. Huang, S.P.; Wu, M.S.; Shun, C.T.; Wang, H.P.; Lin, J.T. Tumor angiogenesis increases with nuclear p53 accumulation in gastric carcinoma. Hepatogastroenterology 2002, 49, 1453–1456. [Google Scholar]
  320. Xue, Y.; Bi, F.; Zhang, X.; Zhang, S.; Pan, Y.; Liu, N.; Shi, Y.; Yao, X.; Zheng, Y.; Fan, D. Role of Rac1 and Cdc42 in hypoxia induced p53 and von Hippel-Lindau suppression and HIF1alpha activation. Int. J. Cancer 2006, 118, 2965–2972. [Google Scholar] [CrossRef]
  321. Rohwer, N.; Dame, C.; Haugstetter, A.; Wiedenmann, B.; Detjen, K.; Schmitt, C.A.; Cramer, T. Hypoxia-inducible factor 1alpha determines gastric cancer chemosensitivity via modulation of p53 and NF-kappaB. PLoS ONE 2010, 5, e12038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  322. Song, W.; Yang, H.B.; Chen, P.; Wang, S.M.; Zhao, L.P.; Xu, W.H.; Fan, H.F.; Gu, X.; Chen, L.Y. Apoptosis of human gastric carcinoma SGC-7901 induced by deoxycholic acid via the mitochondrial-dependent pathway. Appl. Biochem. Biotechnol. 2013, 171, 1061–1071. [Google Scholar] [CrossRef] [PubMed]
  323. Yang, H.B.; Song, W.; Cheng, M.D.; Fan, H.F.; Gu, X.; Qiao, Y.; Lu, X.; Yu, R.H.; Chen, L.Y. Deoxycholic acid inhibits the growth of BGC-823 gastric carcinoma cells via a p53mediated pathway. Mol. Med. Rep. 2015, 11, 2749–2754. [Google Scholar] [CrossRef] [PubMed]
  324. Zhu, B.S.; Xing, C.G.; Lin, F.; Fan, X.Q.; Zhao, K.; Qin, Z.H. Blocking NF-kappaB nuclear translocation leads to p53-related autophagy activation and cell apoptosis. World J. Gastroenterol. WJG 2011, 17, 478–487. [Google Scholar] [CrossRef] [PubMed]
  325. Zhao, K.; Zhu, B.S.; Gong, W.; Zhu, M.L.; Gao, Z.T.; Wu, Y.Y.; Chen, Q.; Yang, X.D.; Xing, C.G. SN50 enhances the effects of LY294002 on cell death induction in gastric cancer cell line SGC7901. Arch. Med. Sci. 2013, 9, 990–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  326. Cao, H.; Jiang, S.; Yuan, R.; Zhang, W.; Liu, Y.; Shao, C.; Shao, S. Inhibition of IkappaB Kinase 2 Attenuated the Proliferation and Induced Apoptosis of Gastric Cancer. Dig. Dis. Sci. 2019, 64, 1204–1216. [Google Scholar] [CrossRef] [PubMed]
  327. Huang, S.; Chen, M.; Shen, Y.; Shen, W.; Guo, H.; Gao, Q.; Zou, X. Inhibition of activated Stat3 reverses drug resistance to chemotherapeutic agents in gastric cancer cells. Cancer Lett. 2012, 315, 198–205. [Google Scholar] [CrossRef] [PubMed]
  328. Endo, F.; Nishizuka, S.S.; Kume, K.; Ishida, K.; Katagiri, H.; Ishida, K.; Sato, K.; Iwaya, T.; Koeda, K.; Wakabayashi, G. A compensatory role of NF-kappaB to p53 in response to 5-FU-based chemotherapy for gastric cancer cell lines. PLoS ONE 2014, 9, e90155. [Google Scholar] [CrossRef] [Green Version]
  329. Luo, L.; Liu, H.; Xi, Q. Trastuzumab induces PUMA-dependent apoptosis and inhibits tumor growth in gastric cancer. FEBS Open Bio 2018, 8, 1911–1919. [Google Scholar] [CrossRef]
  330. Chae, M.; Kim, K.; Park, S.M.; Jang, I.S.; Seo, T.; Kim, D.M.; Kim, I.C.; Lee, J.H.; Park, J. IRF-2 regulates NF-kappaB activity by modulating the subcellular localization of NF-kappaB. Biochem. Biophys. Res. Commun. 2008, 370, 519–524. [Google Scholar] [CrossRef]
  331. Chen, Y.J.; Wu, H.; Zhu, J.M.; Li, X.D.; Luo, S.W.; Dong, L.; Liu, T.T.; Shen, X.Z. MicroRNA-18a modulates P53 expression by targeting IRF2 in gastric cancer patients. J. Gastroenterol. Hepatol. 2016, 31, 155–163. [Google Scholar] [CrossRef]
  332. Zheng, Y.; Pan, D. The Hippo Signaling Pathway in Development and Disease. Dev. Cell 2019, 50, 264–282. [Google Scholar] [CrossRef] [PubMed]
  333. Zhou, G.X.; Li, X.Y.; Zhang, Q.; Zhao, K.; Zhang, C.P.; Xue, C.H.; Yang, K.; Tian, Z.B. Effects of the hippo signaling pathway in human gastric cancer. Asian Pac. J. Cancer Prev. 2013, 14, 5199–5205. [Google Scholar] [CrossRef] [Green Version]
  334. Li, X.J.; Park, E.S.; Park, M.H.; Kim, S.M. 3,3′-Diindolylmethane suppresses the growth of gastric cancer cells via activation of the Hippo signaling pathway. Oncol. Rep. 2013, 30, 2419–2426. [Google Scholar] [CrossRef]
  335. Lin, Z.; Zhang, C.; Zhang, M.; Xu, D.; Fang, Y.; Zhou, Z.; Chen, X.; Qin, N.; Zhang, X. Targeting cadherin-17 inactivates Ras/Raf/MEK/ERK signaling and inhibits cell proliferation in gastric cancer. PLoS ONE 2014, 9, e85296. [Google Scholar] [CrossRef] [Green Version]
  336. Lv, H.; Liu, R.; Fu, J.; Yang, Q.; Shi, J.; Chen, P.; Ji, M.; Shi, B.; Hou, P. Epithelial cell-derived periostin functions as a tumor suppressor in gastric cancer through stabilizing p53 and E-cadherin proteins via the Rb/E2F1/p14ARF/Mdm2 signaling pathway. Cell Cycle. 2014, 13, 2962–2974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Liu, Y.; Tavana, O.; Gu, W. p53 modifications: Exquisite decorations of the powerful guardian. J. Mol. Cell Biol. 2019, 11, 564–577. [Google Scholar] [CrossRef] [Green Version]
  338. Suzuki, T.; Yokozaki, H.; Kuniyasu, H.; Hayashi, K.; Naka, K.; Ono, S.; Ishikawa, T.; Tahara, E.; Yasui, W. Effect of trichostatin A on cell growth and expression of cell cycle- and apoptosis-related molecules in human gastric and oral carcinoma cell lines. Int. J. Cancer 2000, 88, 992–997. [Google Scholar] [CrossRef]
  339. Terui, T.; Murakami, K.; Takimoto, R.; Takahashi, M.; Takada, K.; Murakami, T.; Minami, S.; Matsunaga, T.; Takayama, T.; Kato, J.; et al. Induction of PIG3 and NOXA through acetylation of p53 at 320 and 373 lysine residues as a mechanism for apoptotic cell death by histone deacetylase inhibitors. Cancer Res. 2003, 63, 8948–8954. [Google Scholar]
  340. Feng, L.; Pan, M.; Sun, J.; Lu, H.; Shen, Q.; Zhang, S.; Jiang, T.; Liu, L.; Jin, W.; Chen, Y.; et al. Histone deacetylase 3 inhibits expression of PUMA in gastric cancer cells. J. Mol. Med. 2013, 91, 49–58. [Google Scholar] [CrossRef]
  341. Sun, M.Y.; Zhang, H.; Tao, J.; Ni, Z.H.; Wu, Q.X.; Tang, Q.F. Expression and biological function of rhotekin in gastric cancer through regulating p53 pathway. Cancer Manag. Res. 2019, 11, 1069–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  342. Ma, Y.; Yue, Y.; Pan, M.; Sun, J.; Chu, J.; Lin, X.; Xu, W.; Feng, L.; Chen, Y.; Chen, D.; et al. Histone deacetylase 3 inhibits new tumor suppressor gene DTWD1 in gastric cancer. Am. J. Cancer Res. 2015, 5, 663–673. [Google Scholar]
  343. Spaety, M.E.; Gries, A.; Badie, A.; Venkatasamy, A.; Romain, B.; Orvain, C.; Yanagihara, K.; Okamoto, K.; Jung, A.C.; Mellitzer, G.; et al. HDAC4 Levels Control Sensibility toward Cisplatin in Gastric Cancer via the p53-p73/BIK Pathway. Cancers. 2019, 11, 1747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Noguchi, A.; Kikuchi, K.; Zheng, H.; Takahashi, H.; Miyagi, Y.; Aoki, I.; Takano, Y. SIRT1 expression is associated with a poor prognosis, whereas DBC1 is associated with favorable outcomes in gastric cancer. Cancer Med. 2014, 3, 1553–1561. [Google Scholar] [CrossRef] [PubMed]
  345. Hirai, S.; Endo, S.; Saito, R.; Hirose, M.; Ueno, T.; Suzuki, H.; Yamato, K.; Abei, M.; Hyodo, I. Antitumor effects of a sirtuin inhibitor, tenovin-6, against gastric cancer cells via death receptor 5 up-regulation. PLoS ONE 2014, 9, e102831. [Google Scholar] [CrossRef] [Green Version]
  346. Zhang, X.; Yashiro, M.; Ren, J.; Hirakawa, K. Histone deacetylase inhibitor, trichostatin A, increases the chemosensitivity of anticancer drugs in gastric cancer cell lines. Oncol. Rep. 2006, 16, 563–568. [Google Scholar] [CrossRef]
  347. Sasaki, Y.; Negishi, H.; Idogawa, M.; Suzuki, H.; Mita, H.; Toyota, M.; Shinomura, Y.; Imai, K.; Tokino, T. Histone deacetylase inhibitor FK228 enhances adenovirus-mediated p53 family gene therapy in cancer models. Mol. Cancer Ther. 2008, 7, 779–787. [Google Scholar] [CrossRef] [Green Version]
  348. Qiu, H.; Yashiro, M.; Shinto, O.; Matsuzaki, T.; Hirakawa, K. DNA methyltransferase inhibitor 5-aza-CdR enhances the radiosensitivity of gastric cancer cells. Cancer Sci. 2009, 100, 181–188. [Google Scholar] [CrossRef]
  349. Shin, D.Y.; Sung Kang, H.; Kim, G.Y.; Kim, W.J.; Yoo, Y.H.; Choi, Y.H. Decitabine, a DNA methyltransferases inhibitor, induces cell cycle arrest at G2/M phase through p53-independent pathway in human cancer cells. Biomed. Pharmacother. 2013, 67, 305–311. [Google Scholar] [CrossRef]
  350. Shin, H.; Lee, Y.S.; Lee, Y.C. Sodium butyrate-induced DAPK-mediated apoptosis in human gastric cancer cells. Oncol. Rep. 2012, 27, 1111–1115. [Google Scholar] [CrossRef]
  351. Liu, J.; Xie, Y.S.; Wang, F.L.; Zhang, L.J.; Zhang, Y.; Luo, H.S. Cytotoxicity of 5-Aza-2′-deoxycytidine against gastric cancer involves DNA damage in an ATM-P53 dependent signaling pathway and demethylation of P16(INK4A). Biomed. Pharmacother. 2013, 67, 78–87. [Google Scholar] [CrossRef] [PubMed]
  352. Cheng, L.L.; Itahana, Y.; Lei, Z.D.; Chia, N.Y.; Wu, Y.; Yu, Y.; Zhang, S.L.; Thike, A.A.; Pandey, A.; Rozen, S.; et al. TP53 genomic status regulates sensitivity of gastric cancer cells to the histone methylation inhibitor 3-deazaneplanocin A (DZNep). Clin. Cancer Res. 2012, 18, 4201–4212. [Google Scholar] [CrossRef] [Green Version]
  353. Bai, J.; Chen, J.; Ma, M.; Cai, M.; Xu, F.; Wang, G.; Tao, K.; Shuai, X. Inhibiting enhancer of zeste homolog 2 promotes cellular senescence in gastric cancer cells SGC-7901 by activation of p21 and p16. DNA Cell Biol. 2014, 33, 337–344. [Google Scholar] [CrossRef]
  354. Li, W.; Zhao, L.; Zang, W.; Liu, Z.; Chen, L.; Liu, T.; Xu, D.; Jia, J. Histone demethylase JMJD2B is required for tumor cell proliferation and survival and is overexpressed in gastric cancer. Biochem. Biophys. Res. Commun. 2011, 416, 372–378. [Google Scholar] [CrossRef]
  355. Xu, L.; Wu, W.; Cheng, G.; Qian, M.; Hu, K.; Yin, G.; Wang, S. Enhancement of Proliferation and Invasion of Gastric Cancer Cell by KDM5C Via Decrease in p53 Expression. Technol. Cancer Res. Treat. 2017, 16, 141–149. [Google Scholar] [CrossRef]
  356. Lee, K.H.; Park, J.W.; Sung, H.S.; Choi, Y.J.; Kim, W.H.; Lee, H.S.; Chung, H.J.; Shin, H.W.; Cho, C.H.; Kim, T.Y.; et al. PHF2 histone demethylase acts as a tumor suppressor in association with p53 in cancer. Oncogene 2015, 34, 2897–2909. [Google Scholar] [CrossRef] [PubMed]
  357. Lee, J.H.; Yoo, N.J.; Kim, M.S.; Lee, S.H. Histone Demethylase Gene PHF2 Is Mutated in Gastric and Colorectal Cancers. Pathol. Oncol. Res. 2017, 23, 471–476. [Google Scholar] [CrossRef]
  358. Ohashi, M.; Kanai, F.; Ueno, H.; Tanaka, T.; Tateishi, K.; Kawakami, T.; Koike, Y.; Ikenoue, T.; Shiratori, Y.; Hamada, H.; et al. Adenovirus mediated p53 tumour suppressor gene therapy for human gastric cancer cells in vitro and in vivo. Gut 1999, 44, 366–371. [Google Scholar] [CrossRef] [Green Version]
  359. Takimoto, R.; Kato, J.; Terui, T.; Takada, K.; Kuroiwa, G.; Wu, J.; Ohnuma, H.; Takahari, D.; Kobune, M.; Sato, Y.; et al. Augmentation of antitumor effects of p53 gene therapy by combination with HDAC inhibitor. Cancer Biol. Ther. 2005, 4, 421–428. [Google Scholar] [CrossRef] [Green Version]
  360. Xie, Y.S.; Zhang, Y.H.; Liu, S.P.; Liu, S.Q.; Peng, C.W.; Wu, L.; Luo, H.S.; Li, Y. Synergistic gastric cancer inhibition by chemogenetherapy with recombinant human adenovirus p53 and epirubicin: An in vitro and in vivo study. Oncol. Rep. 2010, 24, 1613–1620. [Google Scholar]
  361. Chen, G.X.; Zheng, L.H.; Liu, S.Y.; He, X.H. rAd-p53 enhances the sensitivity of human gastric cancer cells to chemotherapy. World J. Gastroenterol. WJG 2011, 17, 4289–4297. [Google Scholar] [CrossRef] [PubMed]
  362. Zhang, Y.; Shi, Y.; Li, X.; Du, W.; Luo, G.; Gou, Y.; Wang, X.; Guo, X.; Liu, J.; Ding, J.; et al. Inhibition of the p53-MDM2 interaction by adenovirus delivery of ribosomal protein L23 stabilizes p53 and induces cell cycle arrest and apoptosis in gastric cancer. J. Gene Med. 2010, 12, 147–156. [Google Scholar] [CrossRef] [PubMed]
  363. Zhang, Y.F.; Zhang, B.C.; Zhang, A.R.; Wu, T.T.; Liu, J.; Yu, L.F.; Wang, W.X.; Gao, J.F.; Fang, D.C.; Rao, Z.G. Co-transduction of ribosomal protein L23 enhances the therapeutic efficacy of adenoviral-mediated p53 gene transfer in human gastric cancer. Oncol. Rep. 2013, 30, 1989–1995. [Google Scholar] [CrossRef]
  364. Tsunemitsu, Y.; Kagawa, S.; Tokunaga, N.; Otani, S.; Umeoka, T.; Roth, J.A.; Fang, B.; Tanaka, N.; Fujiwara, T. Molecular therapy for peritoneal dissemination of xenotransplanted human MKN-45 gastric cancer cells with adenovirus mediated Bax gene transfer. Gut 2004, 53, 554–560. [Google Scholar] [CrossRef] [PubMed]
  365. Dvory-Sobol, H.; Sagiv, E.; Liberman, E.; Kazanov, D.; Arber, N. Suppression of gastric cancer cell growth by targeting the beta-catenin/T-cell factor pathway. Cancer 2007, 109, 188–197. [Google Scholar] [CrossRef]
  366. Kunisaki, R.; Ikawa, S.; Maeda, T.; Nakazaki, Y.; Kurita, R.; Harata, M.; Shutoh, Y.; Bai, Y.S.; Soda, Y.; Tanabe, T.; et al. p51/p63, a novel p53 homologue, potentiates p53 activity and is a human cancer gene therapy candidate. J. Gene Med. 2006, 8, 1121–1130. [Google Scholar] [CrossRef]
  367. Endo, S.; Yamato, K.; Hirai, S.; Moriwaki, T.; Fukuda, K.; Suzuki, H.; Abei, M.; Nakagawa, I.; Hyodo, I. Potent in vitro and in vivo antitumor effects of MDM2 inhibitor nutlin-3 in gastric cancer cells. Cancer Sci. 2011, 102, 605–613. [Google Scholar] [CrossRef] [PubMed]
  368. Yi, H.; Yan, X.; Luo, Q.; Yuan, L.; Li, B.; Pan, W.; Zhang, L.; Chen, H.; Wang, J.; Zhang, Y.; et al. A novel small molecule inhibitor of MDM2-p53 (APG-115) enhances radiosensitivity of gastric adenocarcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 97. [Google Scholar] [CrossRef] [PubMed]
  369. Subhash, V.V.; Yeo, M.S.; Wang, L.; Tan, S.H.; Wong, F.Y.; Thuya, W.L.; Tan, W.L.; Peethala, P.C.; Soe, M.Y.; Tan, D.S.P.; et al. Anti-tumor efficacy of Selinexor (KPT-330) in gastric cancer is dependent on nuclear accumulation of p53 tumor suppressor. Sci. Rep. 2018, 8, 12248. [Google Scholar] [CrossRef]
  370. Liu, X.; Wilcken, R.; Joerger, A.C.; Chuckowree, I.S.; Amin, J.; Spencer, J.; Fersht, A.R. Small molecule induced reactivation of mutant p53 in cancer cells. Nucleic Acids Res. 2013, 41, 6034–6044. [Google Scholar] [CrossRef]
  371. Miller, J.J.; Orvain, C.; Jozi, S.; Clarke, R.M.; Smith, J.R.; Blanchet, A.; Gaiddon, C.; Warren, J.J.; Storr, T. Multifunctional Compounds for Activation of the p53-Y220C Mutant in Cancer. Chemistry 2018, 24, 17734–17742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  372. Miller, J.J.; Blanchet, A.; Orvain, C.; Nouchikian, L.; Reviriot, Y.; Clarke, R.M.; Martelino, D.; Wilson, D.; Gaiddon, C.; Storr, T. Bifunctional ligand design for modulating mutant p53 aggregation in cancer. Chem. Sci. 2019, 10, 10802–10814. [Google Scholar] [CrossRef] [Green Version]
  373. Wei, X.W.; Yuan, J.M.; Huang, W.Y.; Chen, N.Y.; Li, X.J.; Pan, C.X.; Mo, D.L.; Su, G.F. 2-Styryl-4-aminoquinazoline derivatives as potent DNA-cleavage, p53-activation and in vivo effective anticancer agents. Eur. J. Med. Chem. 2020, 186, 111851. [Google Scholar] [CrossRef]
  374. Wischhusen, J.; Naumann, U.; Ohgaki, H.; Rastinejad, F.; Weller, M. CP-31398, a novel p53-stabilizing agent, induces p53-dependent and p53-independent glioma cell death. Oncogene 2003, 22, 8233–8245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  375. Zhang, G.; Li, L.; Bi, J.; Wu, Y.; Li, E. Targeting DNA and mutant p53 by a naphthalimide derivative, NA20, exhibits selective inhibition in gastric tumorigenesis by blocking mutant p53-EGFR signaling pathway. Eur. J. Pharmacol. 2020, 887, 173584. [Google Scholar] [CrossRef] [PubMed]
  376. Benosman, S.; Meng, X.; Von Grabowiecki, Y.; Palamiuc, L.; Hritcu, L.; Gross, I.; Mellitzer, G.; Taya, Y.; Loeffler, J.P.; Gaiddon, C. Complex regulation of p73 isoforms after alteration of amyloid precursor polypeptide (APP) function and DNA damage in neurons. J. Biol. Chem. 2011, 286, 43013–43025. [Google Scholar] [CrossRef] [Green Version]
  377. Gaiddon, C.; Lokshin, M.; Ahn, J.; Zhang, T.; Prives, C. A Subset of Tumor-Derived Mutant Forms of p53 Down-Regulate p63 and p73 through a Direct Interaction with the p53 Core Domain. Mol. Cell Biol. 2001, 21, 1874–1887. [Google Scholar] [CrossRef] [Green Version]
  378. Han, S.; Semba, S.; Abe, T.; Makino, N.; Furukawa, T.; Fukushige, S.; Takahashi, H.; Sakurada, A.; Sato, M.; Shiiba, K.; et al. Infrequent somatic mutations of the p73 gene in various human cancers. Eur. J. Surg. Oncol. 1999, 25, 194–198. [Google Scholar] [CrossRef] [PubMed]
  379. Pilozzi, E.; Talerico, C.; Platt, A.; Fidler, C.; Ruco, L. p73 gene mutations in gastric adenocarcinomas. Mol. Pathol. 2003, 56, 60–62. [Google Scholar] [CrossRef]
  380. Hamajima, N.; Matsuo, K.; Suzuki, T.; Nakamura, T.; Matsuura, A.; Hatooka, S.; Shinoda, M.; Kodera, Y.; Yamamura, Y.; Hirai, T.; et al. No associations of p73 G4C14-to-A4T14 at exon 2 and p53 Arg72Pro polymorphisms with the risk of digestive tract cancers in Japanese. Cancer Lett. 2002, 181, 81–85. [Google Scholar] [CrossRef]
  381. Shirai, O.; Ohmiya, N.; Taguchi, A.; Nakamura, M.; Kawashima, H.; Miyahara, R.; Itoh, A.; Hirooka, Y.; Watanabe, O.; Ando, T.; et al. P53, p21, and p73 gene polymorphisms in gastric carcinoma. Hepatogastroenterology 2010, 57, 1595–1601. [Google Scholar] [PubMed]
  382. Liu, Y.; Dong, W.; Mou, Q.; Leng, Y.; Zhang, L.; Duan, L. Impact of p73 gene polymorphism on cancer susceptibility: A meta analysis. Int. J. Clin. Exp. Pathol. 2014, 7, 6820–6825. [Google Scholar] [PubMed]
  383. Cheng, C.; Lingyan, W.; Yi, H.; Cheng, Z.; Huadan, Y.; Xuting, X.; Leiting, X.; Meng, Y.; Shiwei, D. Association between TLR2, MTR, MTRR, XPC, TP73, TP53 genetic polymorphisms and gastric cancer: A meta-analysis. Clin. Res. Hepatol. Gastroenterol. 2014, 38, 346–359. [Google Scholar] [CrossRef] [PubMed]
  384. Shao, A.; Zheng, L.; Chen, S.; Gu, H.; Jing, H. p21, p53, TP53BP1 and p73 polymorphisms and the risk of gastric cardia adenocarcinoma in a Chinese population. Biomarkers 2015, 20, 109–115. [Google Scholar] [CrossRef] [PubMed]
  385. De Feo, E.; Persiani, R.; La Greca, A.; Amore, R.; Arzani, D.; Rausei, S.; D’Ugo, D.; Magistrelli, P.; van Duijn, C.M.; Ricciardi, G.; et al. A case-control study on the effect of p53 and p73 gene polymorphisms on gastric cancer risk and progression. Mutat. Res. 2009, 675, 60–65. [Google Scholar] [CrossRef] [PubMed]
  386. Tannapfel, A.; Schmelzer, S.; Benicke, M.; Klimpfinger, M.; Kohlhaw, K.; Mossner, J.; Engeland, K.; Wittekind, C. Expression of the p53 homologues p63 and p73 in multiple simultaneous gastric cancer. J. Pathol. 2001, 195, 163–170. [Google Scholar] [CrossRef]
  387. Carrasco, G.; Diaz, J.; Valbuena, J.R.; Ibanez, P.; Rodriguez, P.; Araya, G.; Rodriguez, C.; Torres, J.; Duarte, I.; Aravena, E.; et al. Overexpression of p73 as a tissue marker for high-risk gastritis. Clin. Cancer Res. 2010, 16, 3253–3259. [Google Scholar] [CrossRef] [Green Version]
  388. Saavedra, K.; Valbuena, J.; Olivares, W.; Marchant, M.J.; Rodriguez, A.; Torres-Estay, V.; Carrasco-Avino, G.; Guzman, L.; Aguayo, F.; Roa, J.C.; et al. Loss of Expression of Reprimo, a p53-induced Cell Cycle Arrest Gene, Correlates with Invasive Stage of Tumor Progression and p73 Expression in Gastric Cancer. PLoS ONE 2015, 10, e0125834. [Google Scholar] [CrossRef]
  389. Tomkova, K.; Belkhiri, A.; El-Rifai, W.; Zaika, A.I. p73 isoforms can induce T-cell factor-dependent transcription in gastrointestinal cells. Cancer Res. 2004, 64, 6390–6393. [Google Scholar] [CrossRef] [Green Version]
  390. Vilgelm, A.; Wei, J.X.; Piazuelo, M.B.; Washington, M.K.; Prassolov, V.; El-Rifai, W.; Zaika, A. DeltaNp73alpha regulates MDR1 expression by inhibiting p53 function. Oncogene 2008, 27, 2170–2176. [Google Scholar] [CrossRef] [Green Version]
  391. Vilgelm, A.E.; Hong, S.M.; Washington, M.K.; Wei, J.; Chen, H.; El-Rifai, W.; Zaika, A. Characterization of DeltaNp73 expression and regulation in gastric and esophageal tumors. Oncogene 2010, 29, 5861–5868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  392. Slade, N.; Zaika, A.I.; Erster, S.; Moll, U.M. DeltaNp73 stabilises TAp73 proteins but compromises their function due to inhibitory hetero-oligomer formation. Cell Death Differ. 2004, 11, 357–360. [Google Scholar] [CrossRef] [PubMed]
  393. Zaika, E.; Bhardwaj, V.; Wei, J.; Washington, M.K.; Souza, R.; El-Rifai, W.; Zaika, A. Proinflammatory cytokines and bile acids upregulate DeltaNp73 protein, an inhibitor of p53 and p73 tumor suppressors. PLoS ONE 2013, 8, e64306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. Kang, M.J.; Park, B.J.; Byun, D.S.; Park, J.I.; Kim, H.J.; Park, J.H.; Chi, S.G. Loss of imprinting and elevated expression of wild-type p73 in human gastric adenocarcinoma. Clin. Cancer Res. 2000, 6, 1767–1771. [Google Scholar] [PubMed]
  395. Ushiku, T.; Chong, J.M.; Uozaki, H.; Hino, R.; Chang, M.S.; Sudo, M.; Rani, B.R.; Sakuma, K.; Nagai, H.; Fukayama, M. p73 gene promoter methylation in Epstein-Barr virus-associated gastric carcinoma. Int. J. Cancer 2007, 120, 60–66. [Google Scholar] [CrossRef]
  396. Okada, T.; Nakamura, M.; Nishikawa, J.; Sakai, K.; Zhang, Y.; Saito, M.; Morishige, A.; Oga, A.; Sasaki, K.; Suehiro, Y.; et al. Identification of genes specifically methylated in Epstein-Barr virus-associated gastric carcinomas. Cancer Sci. 2013, 104, 1309–1314. [Google Scholar] [CrossRef] [PubMed]
  397. Wei, J.; O’Brien, D.; Vilgelm, A.; Piazuelo, M.B.; Correa, P.; Washington, M.K.; El-Rifai, W.; Peek, R.M.; Zaika, A. Interaction of Helicobacter pylori with gastric epithelial cells is mediated by the p53 protein family. Gastroenterology 2008, 134, 1412–1423. [Google Scholar] [CrossRef] [Green Version]
  398. Luo, J.; Chen, X.Q.; Li, P. The Role of TGF-beta and Its Receptors in Gastrointestinal Cancers. Transl. Oncol. 2019, 12, 475–484. [Google Scholar] [CrossRef] [PubMed]
  399. Ma, G.F.; Miao, Q.; Zeng, X.Q.; Luo, T.C.; Ma, L.L.; Liu, Y.M.; Lian, J.J.; Gao, H.; Chen, S.Y. Transforming growth factor-beta1 and -beta2 in gastric precancer and cancer and roles in tumor-cell interactions with peripheral blood mononuclear cells in vitro. PLoS ONE 2013, 8, e54249. [Google Scholar] [CrossRef]
  400. Yamamura, Y.; Lee, W.L.; Goh, M.X.; Ito, Y. Role of TAp73alpha in induction of apoptosis by transforming growth factor-beta in gastric cancer cells. FEBS Lett. 2008, 582, 2663–2667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  401. Yuan, Z.M.; Shioya, H.; Ishiko, T.; Sun, X.; Gu, J.; Huang, Y.Y.; Lu, H.; Kharbanda, S.; Weichselbaum, R.; Kufe, D. p73 is regulated by tyrosine kinase c-Abl in the apoptotic response to DNA damage. Nature 1999, 399, 814–817. [Google Scholar] [CrossRef]
  402. Pediconi, N.; Ianari, A.; Costanzo, A.; Belloni, L.; Gallo, R.; Cimino, L.; Porcellini, A.; Screpanti, I.; Balsano, C.; Alesse, E.; et al. Differential regulation of E2F1 apoptotic target genes in response to DNA damage. Nat. Cell Biol. 2003, 5, 552–558. [Google Scholar] [CrossRef]
  403. Urist, M.; Tanaka, T.; Poyurovsky, M.V.; Prives, C. p73 induction after DNA damage is regulated by checkpoint kinases Chk1 and Chk2. Genes Dev. 2004, 18, 3041–3054. [Google Scholar] [CrossRef] [Green Version]
  404. Qiang, L.; Ji, Z.; Wang, X. Expression of TAp73alpha affects the therapy effect of chemotherapy drugs in gastric cancer. Oncol. Res. 2018. [Google Scholar] [CrossRef] [PubMed]
  405. Ji, Z.P.; Qiang, L.; Zhang, J.L. Transcription activated p73-modulated cyclin D1 expression leads to doxorubicin resistance in gastric cancer. Exp. Ther. Med. 2018, 15, 1831–1838. [Google Scholar] [CrossRef] [Green Version]
  406. Tang, S.; Yuan, Y.; Liu, Z.; He, Y.; Pan, D. Casein kinase 2 inhibitor CX-4945 elicits an anti-Warburg effects through the downregulation of TAp73 and inhibits gastric tumorigenesis. Biochem. Biophys. Res. Commun. 2020, 530, 686–691. [Google Scholar] [CrossRef] [PubMed]
  407. Li, L.; Li, L.; Li, W.; Chen, T.; Bin, Z.; Zhao, L.; Wang, H.; Wang, X.; Xu, L.; Liu, X.; et al. TAp73-induced phosphofructokinase-1 transcription promotes the Warburg effect and enhances cell proliferation. Nat. Commun. 2018, 9, 4683. [Google Scholar] [CrossRef]
  408. Wang, H.; Liu, Z.; Li, J.; Zhao, X.; Wang, Z.; Xu, H. DeltaNp63alpha mediates proliferation and apoptosis in human gastric cancer cells by the regulation of GATA-6. Neoplasma 2012, 59, 416–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  409. Song, Y.; Liu, D.; He, G. TKTL1 and p63 are biomarkers for the poor prognosis of gastric cancer patients. Cancer Biomark. 2015, 15, 591–597. [Google Scholar] [CrossRef]
  410. Glickman, J.N.; Yang, A.; Shahsafaei, A.; McKeon, F.; Odze, R.D. Expression of p53-related protein p63 in the gastrointestinal tract and in esophageal metaplastic and neoplastic disorders. Hum. Pathol. 2001, 32, 1157–1165. [Google Scholar] [CrossRef]
  411. Ye, F.; Tang, C.; Shi, W.; Qian, J.; Xiao, S.; Gu, M.; Dang, Y.; Liu, J.; Chen, Y.; Shi, R.; et al. A MDM2-dependent positive-feedback loop is involved in inhibition of miR-375 and miR-106b induced by Helicobacter pylori lipopolysaccharide. Int. J. Cancer 2015, 136, 2120–2131. [Google Scholar] [CrossRef] [PubMed]
  412. Hoebe, E.; Wille, C.; Hagemeier, S.; Kenney, S.; Greijer, A.; Middeldorp, J. Epstein-Barr Virus Gene BARF1 Expression is Regulated by the Epithelial Differentiation Factor DeltaNp63alpha in Undifferentiated Nasopharyngeal Carcinoma. Cancers. 2018, 10, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Histological and molecular subgroups of gastric cancer. Gastric cancer has two major histological subtypes: the intestinal subtype (upper left) that presents differentiated cancer cell structures in glands, and the diffuse subtype (lower left) that displays undifferentiated cancer cells dispersed within the stroma. There are also four molecular subgroups defined by the TCGA: the Chromosome instable (CIN), the Genetic Stable (GS), the Microsatellites Instable (MSI), and the Epstein–Barr Virus (EBV) subgroups. Each are characterized by specific deregulated mechanisms. Of note is the two molecular subgroups that tend to respond the best to immunotherapy, the MSI and the EBV subgroups.
Figure 1. Histological and molecular subgroups of gastric cancer. Gastric cancer has two major histological subtypes: the intestinal subtype (upper left) that presents differentiated cancer cell structures in glands, and the diffuse subtype (lower left) that displays undifferentiated cancer cells dispersed within the stroma. There are also four molecular subgroups defined by the TCGA: the Chromosome instable (CIN), the Genetic Stable (GS), the Microsatellites Instable (MSI), and the Epstein–Barr Virus (EBV) subgroups. Each are characterized by specific deregulated mechanisms. Of note is the two molecular subgroups that tend to respond the best to immunotherapy, the MSI and the EBV subgroups.
Cancers 13 00916 g001
Figure 2. The isoforms of the p53 family. The p53 family has 3 genes: TP53, TP63, and TP73, that express multiple isoforms via alternative promoters (P1, P1′, and P2), generating full-length proteins with N-terminus transactivation domains (e.g., TA) or without (e.g., ∆N) and alternative splicing generating variations in the C-terminus (e.g., α, β, and γ). The encoded proteins contain transactivation domains (TAD1, 2, and 3); a proline-rich region (PRD); a DNA-binding domain (DBD), tetramerization domain (TD), sterile α domain (SAM), and an inhibitor domain (ID).
Figure 2. The isoforms of the p53 family. The p53 family has 3 genes: TP53, TP63, and TP73, that express multiple isoforms via alternative promoters (P1, P1′, and P2), generating full-length proteins with N-terminus transactivation domains (e.g., TA) or without (e.g., ∆N) and alternative splicing generating variations in the C-terminus (e.g., α, β, and γ). The encoded proteins contain transactivation domains (TAD1, 2, and 3); a proline-rich region (PRD); a DNA-binding domain (DBD), tetramerization domain (TD), sterile α domain (SAM), and an inhibitor domain (ID).
Cancers 13 00916 g002
Figure 3. Deregulation affecting the isoforms of the p53 family in gastric cancer. Mutations in TP53 or alterations in the expression of TP53, TP63, and TP73 isoforms are observed during gastric tumorigenesis, evolving from a normal stomach epithelium toward adenocarcinomas via gastritis, metaplasia, and dysplasia. Images taken 20X.
Figure 3. Deregulation affecting the isoforms of the p53 family in gastric cancer. Mutations in TP53 or alterations in the expression of TP53, TP63, and TP73 isoforms are observed during gastric tumorigenesis, evolving from a normal stomach epithelium toward adenocarcinomas via gastritis, metaplasia, and dysplasia. Images taken 20X.
Cancers 13 00916 g003
Figure 4. Signals and mechanisms affecting the expression and activity of the isoforms of the p53 family in gastric cancer. Activators are indicated in red and repressors in blue. Underlines indicate existing clinically relevant data.
Figure 4. Signals and mechanisms affecting the expression and activity of the isoforms of the p53 family in gastric cancer. Activators are indicated in red and repressors in blue. Underlines indicate existing clinically relevant data.
Cancers 13 00916 g004
Figure 5. Therapeutic strategies targeting the p53 family that have been investigated in gastric cancer.
Figure 5. Therapeutic strategies targeting the p53 family that have been investigated in gastric cancer.
Cancers 13 00916 g005
Table 1. Molecular alterations in the p53 family and related pathways present in gastric cancer.
Table 1. Molecular alterations in the p53 family and related pathways present in gastric cancer.
Histological SubgroupIntestinalDiffuseMixND
Molecular subgroupCINGSMSIEBV
Infectious factorH. pylori (HP)
(CagA; ROS; AID; CHAC1)
Epstein Barr Virus
(EBNA1; EBNA3C; miR-BART5-3p; LMP1)
TP53 mutation
  • 41% missense + 23% truncating
  • Li-Fraumeni mutations (ex. R175H, E287X; 5% penetrance)
  • 11% missense + 2% truncating
  • Li-Fraumeni mutations (ex. R175H, E287X; 5% penetrance)
  • 24% missense + 9% truncating
  • mutation frequency increase in late stage (33%)
TP53 elevated expression
  • 62%
  • ∆133p53
  • 48%
MDM2 alterations
  • 29% amplification or gain
  • polymorphism SNP309 G/G
  • 13% amplification or gain
  • polymorphism SNP309 G/G
  • 12% gain
  • 6% missense or truncation
  • 38% amplification or gain
MDM2 activatorsCTD2 MDMX; ENIGMA; AURKA
MDM2 inhibitors FHIT SMAD1; PICT1; UBTB1
p53 activators PAX5; OSR; PKNOX2; BCL6B; TIP30; PHF2; miR-27b; miR100; MEG3 lncRNA
p53 inhibitors TRIM59; CREP; HDAC4; SIRT1; EZH2; JMJD2B; KDM5C; miR-3174; miR-370; OnclncRNA-626; VCAN-AS1 lncRNA; lncRNA ZFPM2-AS; Hascirc0001546
P53 target genes BTG4; DR5
TP73
  • elevated expression of TAp73 upon HP infection
  • elevated ∆Np73
promoter methylation
  • 1% missense mutation
TP63
  • decreased ∆Np63 in HP infected cells
  • Elevated p63 staining
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Blanchet, A.; Bourgmayer, A.; Kurtz, J.-E.; Mellitzer, G.; Gaiddon, C. Isoforms of the p53 Family and Gastric Cancer: A Ménage à Trois for an Unfinished Affair. Cancers 2021, 13, 916. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13040916

AMA Style

Blanchet A, Bourgmayer A, Kurtz J-E, Mellitzer G, Gaiddon C. Isoforms of the p53 Family and Gastric Cancer: A Ménage à Trois for an Unfinished Affair. Cancers. 2021; 13(4):916. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13040916

Chicago/Turabian Style

Blanchet, Anais, Agathe Bourgmayer, Jean-Emmanuel Kurtz, Georg Mellitzer, and Christian Gaiddon. 2021. "Isoforms of the p53 Family and Gastric Cancer: A Ménage à Trois for an Unfinished Affair" Cancers 13, no. 4: 916. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13040916

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop