Gastric cancer (GC) represents a serious health problem on a global scale. It is the second leading cause of cancer-related death worldwide.1 In the United States, GC is relatively less frequent, with 22,220 new cases and 13,730 cancer deaths occurring in 2014.2 Between 2002 and 2008, the 5-year relative survival rate was only 27% according to the SEER database.3
Despite some recent advances in the field, the prognosis in metastatic GC remains poor. Even in localized disease, the adjunctive therapies improve overall survival (OS) by only approximately 10%.4–6 Patients often succumb because of the complications of metastases rather than from the primary tumor. A better understanding of molecular biology, which is likely to lead to improved treatment options, is needed and is the basis for this review.
Currently, only 2 molecular subtypes have been identified that are relevant in the clinic. This is based on the overexpression of HER2 protein and/or the amplification of its gene ERBB2, thus allowing some advantage from the use of trastuzumab in the first-line setting.7 This article focuses on the multitude of novel molecular markers with prognostic implications in GC. Table 1 highlights molecular biomarker differences between intestinal and diffuse-type GC; for more detailed discussion, refer to the article by Wadhwa et al.8
Potential Novel Prognostic Biomarkers in GC
Aldehyde Dehydrogenase 1
Aldehyde dehydrogenases (ALDHs) are generally regarded as detoxification enzymes that are critical for protecting organisms against various aldehydes that could otherwise be harmful.9–11 The deficiencies and polymorphisms in various ALDH enzymes can lead
Molecular Biomarker Differences Between Intestinal- and Diffuse-Type Gastric Cancer


SOX9
SOX9, a high-mobility group box transcription factor, is required for development and lineage commitment. SOX9 has been reported to be a direct target for Notch signaling15 and is a documented stem cell marker.16 Notch signaling and SOX9 have been implicated in cancer development, and most recently our group has shown that SOX9 also plays an important role in esophageal carcinoma.17 In our study, the loss of an important tumor growth factor (TGF)-β adaptor β2SP in esophageal adenocarcinoma and MEF cells led to activation of Notch signaling and increased expression of SOX9; high levels of nuclear SOX9 expression were associated with poor survival and adverse disease status (lymph node metastasis) in patients with esophageal adenocarcinoma. Using immunohistochemistry, nuclear SOX9 expression was detected in 64 (34.6%) of 185 GCs; however, the authors found no significant relationship between altered expression of SOX9 protein and clinicopathologic parameters, including OS.18 The role of SOX9 has also been evaluated in terms of its effects on promoting invasion and metastasis in gastric adenocarcinomas.19 The authors were able to show that higher SOX9 expression was associated with higher stage (P<.0005), higher positive lymph node stage (P<.0005), and higher tumor stage (P<.0005). These results suggest that SOX9 expression is related to tumor progression in gastric adenocarcinoma. Importantly, SOX9 can serve as a therapeutic target through YAP (Figure 1).
YAP and Hippo Pathway
The Hippo pathway is known to control organ size in multiple species (Figure 1). Two kinase cascades participate in this pathway and are formed by MST1/2 and LATS1/2.20 The activation of these kinases leads to phosphorylation of the downstream transcriptional coactivator YAP/TAZ, thus preventing its interaction with, and therefore transactivation of, the DNA-binding transcriptional factor TEADs/TEF.21,22 Inactivation of the Hippo pathway leads to an increased cancer risk through increased proliferation and antiapoptotic effects.23–25
YAP is a downstream effector in this pathway, which plays an important role in cell proliferation. It has been mostly reported to be an oncoprotein, and its elevated expression and nuclear localization have been frequently observed in human cancers (ie, lung, liver, colon, pancreatic, and breast cancers).25–27 YAP does not contain any DNA-binding domains. It functions through interaction with TEADs, resulting in the induction of target gene expression, in turn promoting cell proliferation and inhibiting apoptosis. Similar to YAP, the mammalian Vestigial-like proteins VGLL1 though VGLL4 do not contain a DNA-binding domain, and they also exert their transcriptional regulatory functions through pairing with TEADs via their Tondu (TDU) domains.28 Recently, VGLL4 in Drosophila and humans has been shown to be a transcriptional repressor that inhibits YAP-induced overgrowth and tumorigenesis.29–31
Increased YAP mRNA levels in GC samples were previously reported to correlate with lymphatic metastasis and tumor TNM stage.32 In a more recent study,33 YAP mRNA levels were found to be associated with not only tumor size, differentiation, and stage, but also Helicobacter pylori infectious status (P<.05). YAP-positive rates were much higher in GC and dysplasia than in normal tissues (P<.01). Consistently, the protein levels of YAP also tend to increase in higher-grade tumors. This same article found that the average ratio of YAP to VGLL4 at mRNA levels in higher-grade tumor samples is significantly higher than in lower-grade tumor samples (P<.05). Moreover, the YAP/VGLL4 ratio at mRNA level is strongly correlated with GC clinical factors, including lymphatic invasion, tumor size, TNM stage, and H pylori infectious status. Immunohistochemical analysis showed that among patients with YAP-positive GC, those with VGLL4-positive expression have a better clinical outcome (survival time, 39 months) when compared with VGLL4-negative expression (survival time, 16 months).33 These new results suggest that the YAP/VGLL4 ratio can be used as a prognostic marker in GC. Interestingly, the investigators found that a peptide mimicking this function of VGLL4 potently suppressed tumor growth in vitro and in vivo, which may lead to new therapeutic targets in GC.33
SHH
The SHH signaling pathway plays a critical role in stem cell maintenance and specifying patterns of cell growth and differentiation during embryonic development.34 SHH activates its signaling by binding to the receptor, Patched (Ptch), which relieves Ptch-mediated repression of a downstream membrane protein related to G protein–coupled receptors, Smoothened (Smo). On activation, Smo promotes nuclear translocation of a family of transcription factors (Ci

The Hippo/YAP pathway and the crosstalk with epidermal growth factor receptor (EGFR) and G-protein–coupled receptor (GPCR) signaling. Two kinase cascades participate in this pathway and are formed: MST1/2 and LATS1/2. The activation of these kinases leads to phosphorylation of the downstream transcriptional coactivator YAP/TAZ, thus preventing its interaction with and therefore transactivation of the DNA-binding transcriptional factor TEADs/TEF. Inactivation of the Hippo pathway leads to an increased cancer risk through increased proliferation and antiapoptotic effects.
YAP functions through interaction with TEADs, resulting in the induction of target gene expression, in turn promoting cell proliferation and inhibiting apoptosis. Similar to YAP, the mammalian vestigial-like proteins VGLL1 through VGLL4 do not contain DNA-binding domain and they also exert their transcriptional regulatory functions through pairing with TEADs via their TONDU (TDU) domains. YAP/TAZ binding to TEADs leads to the increased expression of amphiregulin, which in turn activates EGFR leading to PI3K/mTOR and Ras/Raf pathway activation. Activation of EGFR and RAS activates the Yorkie homologue YAP, and EGFR-RAS-MAPK signaling promotes phosphorylation of the Ajuba family protein WTIP, and also enhances WTIP binding to the Warts and LATS. The YAP pathway can also be activated through interaction of GPCR and activation of Rho GTPase, leading to increased assembly of F-actin. PTEN is regulated by miR-29, and YAP-TEAD binds to the promoter of mir-29 and promotes its transcription. miR29, through its inhibition of PTEN, inhibits the PI3K/Akt pathway.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 13, 4; 10.6004/jnccn.2015.0064

The Hippo/YAP pathway and the crosstalk with epidermal growth factor receptor (EGFR) and G-protein–coupled receptor (GPCR) signaling. Two kinase cascades participate in this pathway and are formed: MST1/2 and LATS1/2. The activation of these kinases leads to phosphorylation of the downstream transcriptional coactivator YAP/TAZ, thus preventing its interaction with and therefore transactivation of the DNA-binding transcriptional factor TEADs/TEF. Inactivation of the Hippo pathway leads to an increased cancer risk through increased proliferation and antiapoptotic effects.
YAP functions through interaction with TEADs, resulting in the induction of target gene expression, in turn promoting cell proliferation and inhibiting apoptosis. Similar to YAP, the mammalian vestigial-like proteins VGLL1 through VGLL4 do not contain DNA-binding domain and they also exert their transcriptional regulatory functions through pairing with TEADs via their TONDU (TDU) domains. YAP/TAZ binding to TEADs leads to the increased expression of amphiregulin, which in turn activates EGFR leading to PI3K/mTOR and Ras/Raf pathway activation. Activation of EGFR and RAS activates the Yorkie homologue YAP, and EGFR-RAS-MAPK signaling promotes phosphorylation of the Ajuba family protein WTIP, and also enhances WTIP binding to the Warts and LATS. The YAP pathway can also be activated through interaction of GPCR and activation of Rho GTPase, leading to increased assembly of F-actin. PTEN is regulated by miR-29, and YAP-TEAD binds to the promoter of mir-29 and promotes its transcription. miR29, through its inhibition of PTEN, inhibits the PI3K/Akt pathway.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 13, 4; 10.6004/jnccn.2015.0064
The Hippo/YAP pathway and the crosstalk with epidermal growth factor receptor (EGFR) and G-protein–coupled receptor (GPCR) signaling. Two kinase cascades participate in this pathway and are formed: MST1/2 and LATS1/2. The activation of these kinases leads to phosphorylation of the downstream transcriptional coactivator YAP/TAZ, thus preventing its interaction with and therefore transactivation of the DNA-binding transcriptional factor TEADs/TEF. Inactivation of the Hippo pathway leads to an increased cancer risk through increased proliferation and antiapoptotic effects.
YAP functions through interaction with TEADs, resulting in the induction of target gene expression, in turn promoting cell proliferation and inhibiting apoptosis. Similar to YAP, the mammalian vestigial-like proteins VGLL1 through VGLL4 do not contain DNA-binding domain and they also exert their transcriptional regulatory functions through pairing with TEADs via their TONDU (TDU) domains. YAP/TAZ binding to TEADs leads to the increased expression of amphiregulin, which in turn activates EGFR leading to PI3K/mTOR and Ras/Raf pathway activation. Activation of EGFR and RAS activates the Yorkie homologue YAP, and EGFR-RAS-MAPK signaling promotes phosphorylation of the Ajuba family protein WTIP, and also enhances WTIP binding to the Warts and LATS. The YAP pathway can also be activated through interaction of GPCR and activation of Rho GTPase, leading to increased assembly of F-actin. PTEN is regulated by miR-29, and YAP-TEAD binds to the promoter of mir-29 and promotes its transcription. miR29, through its inhibition of PTEN, inhibits the PI3K/Akt pathway.
Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 13, 4; 10.6004/jnccn.2015.0064
Prognostic Biomarkers/Molecules Undergoing or Tested in Clinical Trials
Fibroblast Growth Factor Receptor
Fibroblast growth factor (FGF) triggers the autophosphorylation of FGF receptor (FGFR) at a key tyrosine residue in an activation loop of the tyrosine kinase domain, resulting in a structural change of the tyrosine kinase domain from an inactive to an active form.40 The activated tyrosine kinase domain of FGFR then phosphorylates other tyrosine residues, ultimately leading to catalysis of phosphatidylinositol diphosphate (PIP2) to diacylglycerol (DAG) and inositol triphosphate (IP3).41 Activated FGFR phosphorylates FGFR substrate 2 (FRS2) to recruit the GRB2 adaptor molecule.42 FGF signals are transduced to the RAS-MAPK or PI3K/Akt signaling cascades. SRC tyrosine kinases are also activated by FGF signals.43 FGF-induced RAS-MAPK activation is involved in cellular proliferation, whereas FGF-induced PI3K/Akt activation is involved in cellular survival.44
In particular, somatic FGFR2 mutations have been reported in lung, gastric, and ovarian cancers.45–47 FGFR2 amplification has been associated with tumor cell proliferation and survival of GC cell lines.48 Furthermore, FGFR2 amplification may correlate with a poor prognosis for patients with GC.49 In a recent genomic survey of GC using high-resolution single nucleotide polymorphism (SNP) arrays, FGFR2 copy number gain was found in 9.3% of tumors and was more common than EGFR (7.7%), HER2 (7.2%), or MET (4.3%) copy number gains.50 Another large screening study, including Caucasian (n=408) and Korean (n=356) GC samples,51 found that 5.9% of the samples presented FGFR2 amplification. The FGFR2 amplification was significantly associated with lymph node status (P<.0007). FGFR2 amplification was found to be a prognostic biomarker of shorter OS in both cohorts. Jung et al52 also showed the presence of FGFR2 amplification in Korean GC samples (4.5%), and its association with a higher TNM stage and a shorter OS by univariate analysis.52 FGFR is currently a target of interest in the treatment of GC, and FGFR inhibitors have been developed (dovitinib and AZD4547).50,53 These inhibitors have demonstrated efficacy in GC cell lines and xenograft models in vivo. Whether this will translate into clinical efficacy remains to be seen and is being tested in an ongoing phase II trial comparing AZD4547 with paclitaxel in the second-line setting in patients with FGFR2 polysomy or amplification (ClinicalTrials.gov identifier: NCT01457846).
HER2
In GC, ERBB2 amplification or HER2 overexpression has been reported in 7% to 34% of the tumors,54–56 particularly in the gastroesophageal junction carcinomas (proximal) and in intestinal-type GC.54,56,57 The prognostic value of HER2-positivity in advanced GC is, however, a controversial issue. Reports and a meta-analysis have indicated that ERBB2 amplification is associated with poor prognosis and aggressive disease,55,56,58–60 whereas other reports show no difference in prognosis when compared with HER2-negative tumors.61–63 Two recent studies showed that HER2 status is not an independent prognostic biomarker in early gastroesophageal adenocarcinoma.64,65
Although the prognostic value of HER2 expression remains in question, a modest survival advantage in patients with HER2-positive tumors treated with trastuzumab has been established.7,66,67 The Trastuzumab for Gastric Cancer (ToGA) trial (Table 2) randomized 584 patients whose tumors overexpressed HER2 to receive a fluoropyrimidine (5-FU or capecitabine) plus cisplatin with or without trastuzumab.8 Addition of trastuzumab to chemotherapy increased the OS from 11.1 to 13.8 months (hazard ratio [HR], 0.74; 95% CI, 0.60, 0.91; P=.0046). On extended follow-up, the HR of OS for the addition of trastuzumab decreased to 0.80,68 indicating that, although real, the response to trastuzumab may be short-lived. Based on the results of this trial, the addition of trastuzumab to chemotherapy has become the standard of care in patients whose tumors overexpress HER2. Neither the LoGIC nor the TYTAN trials (in the first- and the second-line settings, respectively) evaluating the use of lapatinib (dual epidermal growth factor receptor [EGFR] and HER2 tyrosine kinase inhibitor) met their primary end points of OS.57,69
Currently, 2 ongoing phase III trials are testing the benefit of alternative agents to HER2 in GC. Pertuzumab (JACOB trial), a monoclonal antibody that binds to a different epitope of HER2 than trastuzumab, is being studied,70 and has already proven to be effective in HER2-positive breast cancer.71 An ongoing randomized trial is also investigating the predictive value of trastuzumab emtansine (T-DM1),
Major Phase III Trials Using Targeted Therapy in Advanced Gastric Cancer


Epidermal Growth Factor Receptor
EGFR (encoded by EGFR) is another member of the HER family that, unlike HER2, is activated by the binding of specific ligands, including EGF and TGF-α. In a study using fluorescence in situ hybridization (FISH) assays, 365 GC samples were analyzed and the incidence of EGFR amplification was found to be 4.9%. A second study, using SNP assays, found that 7.7% of GCs harbored EGFR amplification.50 A larger study examined 511 GC samples for EGFR overexpression by immunohistochemistry and FISH and reported that 27.4% of the samples were positive for EGFR protein expression by immunohistochemistry, but only 2.3% were amplified when detected by FISH.73 EGFR overexpression has been shown to be a prognostic indicator of worse outcome in GC.74,75 Although EGFR immunohistochemical or FISH positivity has been associated with the presence of lymph node metastasis, higher stage, and poor survival, after multivariate analysis, only EGFR immunohistochemical positivity remained a poor prognostic factor.73 However, a recent study has also found conflicting data, in that EGFR expression using immunohistochemistry was correlated with improved progression-free survival and OS,76 whereas a meta-analysis found that EGFR was not an independent predictor for survival in patients with GC.77
EGFR inhibitors have been tested in multiple clinical trials; however, the results in GC have been disappointing. The EXPAND trial did not achieve its primary end point, with the median progression-free survival for the capecitabine-cisplatin plus cetuximab arm being 4.4 months compared with 5.6 months for the capecitabine-cisplatin arm (HR, 1.09; 95% CI, 0.92–1.29; P=.32).78 The REAL-3 study was terminated prematurely because a statistically significantly lower OS was noted in patients treated with modified epirubicin/oxaliplatin/capecitabine (EOC) and panitumumab. The final analysis of this study79 showed a median OS of 11.3 months in patients allocated EOC versus 8.8 months in those allocated EOC and panitumumab (HR, 1.37; 95% CI, 1.07–1.76; P=.013). It is possible that these trials were negative because they did not appropriately enrich the patient population likely to benefit from this treatment.
MET (Hepatocyte Growth Factor Receptor)
MET (encoded by MET) belongs to the hepatocyte growth factor receptor (HGFR) family. MET amplification and/or overexpression of its protein product has long been implicated in the pathogenesis of GC, with many reports based on gene copy number, RNA expression, and/or protein expression, supporting its role as a poor prognostic factor.80–84 Nevertheless, the prevalence of MET amplification in GC varies widely in the literature, from 0%85 to 21%.81 This discrepancy is greatly attributed to the methodology used to detect gene amplification/copy number gain and/or protein expression. Depending on the method used, different MET amplification prevalences have been further reported.80,81,83,84 Nonetheless, in all of these studies, the patients with GC with polysomic and/or amplified MET demonstrated poorer disease-free survival and OS compared with the nonpolysomic MET. In a small study with crizotinib, 2 of 4 patients with 5 or more MET copy number gains had a longer response duration than those with fewer than 5 copies of the gene.83 Furthermore, rilotumumab (AMG 102), a fully human monoclonal antibody, demonstrated longer OS for patients whose tumors had high total c-MET expression.86 The RILOMET-1 phase III study (ClinicalTrials.gov identifier: NCT01697072), testing the efficacy of rilotumumab in combination with chemotherapy as a first-line treatment of metastatic gastric and gastroesophageal junction adenocarcinoma, and the MetGastric study (ClinicalTrials.gov identifier: NCT01662869), testing the efficacy of onartuzumab versus placebo combined with chemotherapy in MET-positive metastatic GC, were recently terminated because they did not meet safety/efficacy end points and the protocol-defined futility criteria would likely have been met at the planned interim analysis in March 2015. An interesting study of single-agent AMG 337 (oral MET tyrosine kinase inhibitor) in 90 patients with advanced solid tumors included 13 individuals with MET-amplified gastroesophageal adenocarcinomas. Of these 13 patients, 8 had partial or near-complete responses, indicating that small molecule tyrosine kinase inhibitors still hold some promise in this disease.87
Angiogenic Pathway Markers
Angiogenesis, the growth of new blood vessels, is an important aspect of tumorigenesis. It is primarily modulated by the vascular endothelial growth factor A (VEGF-A) and its receptors VEGFRs, particularly types 1 and 2. In GC, expression of VEGF-A and VEGFR was reported in 40% and 36 % of cases, respectively.74 VEGF-A expression in GC and serum has been associated with poor prognosis, lymph node involvement, and metastasis, in both resectable and advanced disease.88 Increased VEGF levels were also correlated with decreased OS (P=.009), and in a multivariate analysis were found to be an independent prognostic factor for OS.89 A recent meta-analysis also confirmed that VEGF-A overexpression indicates a poor prognosis for OS and disease-free survival in patients with GC.90 The GC trials testing the usefulness of VEGF therapy have been somewhat conflicting. Bevacizumab failed to demonstrate an OS benefit in patients with advanced gastric and gastroesophageal junction adenocarcinoma.91 A subsequent retrospective biomarker analysis of the AVAGAST trial showed that patients with high baseline plasma VEGF-A levels and with low baseline expression of neuropilin-1 seemed to have an improved OS.92 It is important to note that neither of these biomarkers has been validated. The recently published REGARD trial demonstrated a marginal improvement in median OS in the second-line setting with the addition of ramucirumab (anti-VEGFR2 antibody).93 Subsequently, the RAINBOW trial showed that the addition of ramucirumab to paclitaxel significantly prolonged the primary end point of OS from a median of 7.36 to 9.63 months (P=.0169).94 At the 2015 ASCO Gastrointestinal Cancers Symposium, Tabernero et al95 presented the findings regarding the exposure/response relationship of ramucirumab in patients from the REGARD and RAINBOW trials. In these 2 trials, longer OS was seen in patients with higher levels of ramucirumab exposure (concentration minimum at the end of dose one). This association also corresponded with progression-free survival. Clearly, more studies are needed to develop an optimized ramucirumab exposure strategy.
PI3K-mTOR Pathway
The phosphatidylinositol-3-kinase (PI3K)/mTOR pathway represents one common final convergence signalling pathway originated by the activation of several receptor tyrosine kinases (RTKs). Activation of the PI3K/mTOR pathway in GC has been demonstrated in preclinical studies,96,97 and its deregulation has been associated with increased lymph node metastasis and decreased survival in patients with GC.98,99 The GRANITE-1 study testing the efficacy of everolimus did not achieve its primary end point of OS (5.4 months with everolimus and 4.3 months with placebo; HR, 0.90; 95% CI, 0.75–1.08; P=.124).100
Oncogenic mutations in PIK3CA (gene encoding the alpha p110 catalytic subunit of PI3K) have been observed in GC, constitutively activating the PI3KA/mTOR pathway. Studies in GC reported a PIK3CA mutation frequency of 5% to 25%.101–105 One of these studies reported PIK3CA amplification in 67% of GCs104 and an association with poor prognosis, indicating that this is a major mechanism leading to activation of the PI3K/mTOR pathway in GC.
Future Directions
The future of GC treatment, which is a disease influenced by inflammation, likely lies in the exploitation of the immune system to fight disease. This can be achieved through reprogramming T cells, the use of antibody conjugates (such as T-DM1) to target tumor cells more specifically, gene editing, the use of cancer vaccines, and the inhibition of checkpoint receptors.
To this end, the other interesting targets/biomarkers are programmed death ligand (PDL) and programmed death-1 (PD-1). PD-1 is a potent immunoregulatory molecule expressed on activated T and B lymphocytes, natural killer T cells, and monocytes.106 It can attenuate activation of T cells and suppress cytokine secretion through delivering inhibitory signals, and may also induce immune tolerance to tumor after interaction with its 2 ligands, programmed death-1 ligands (PD-Ls) 1 and 2.107,108 PD-Ls expression has been reported extensively in various tumors, including esophageal cancer.109–112 In addition, upregulation of PD-Ls in several carcinomas can contribute to tumor evasion from the host immune system and is correlated with unfavorable clinical prognosis of many tumors, and gastric and esophageal cancer.112–115 Previous studies have shown that blocking the PD-1/PD-L pathway can result in efficient antitumor T-cell responses and a better control of tumor.116–118 Given that the immune system plays an important role in GC, these biomarkers are of interest in this disease.
Of 2 recent studies using whole genome and exome sequencing, one identified TP53, CDKN2A, SMAD4, ARID1A, PIK3CA, and chromatin-modifying factors as driver mutations,119 whereas the other identified TP53, ARID1A, CDH1, MUC6, CTNNA2, GLI3, and RNF34 as important.120 Interestingly, 2 studies found RHOA mutations in diffuse-type tumors but not in intestinal-type tumors.120,121 In one cohort, all of the tumors with RHOA mutations were HER2-negative; this finding may be useful in the treatment of patients with HER2-negative tumors for whom no targeted therapies are yet available.121 Recently, The Cancer Genome Atlas (TCGA) analysis uncovered 4 genotypes of gastroesophageal adenocarcinoma122: Epstein-Barr virus (EBV)–containing GC (10%), in which PIK3CA alterations are common; tumors showing microsatellite instability (20%); “chromosomally unstable” (50%; most common); and “genomically stable” (20% contain diffuse histology and 30% have RHOA signaling alterations). Unfortunately, these discoveries are not sufficient to change treatment strategies and more work is clearly needed.
With the advent of cancer genomics, many new mutations/potential biomarkers will likely be found to play a role in tumorigenesis and to subsequently affect prognosis. However, the sheer complexity of the genome is staggering, and genetic abnormalities do not always translate into functional/protein aberrations. In a recent article, Vogelstein et al123 proposed dthat the whole idea of eradicating cancer through targeting metastatic disease is impossible. What is known is that carcinogenesis is a process that takes decades to develop, and involves the accumulation of multiple genetic alterations that in turn affect the function of numerous pathways. What should be clear is that more work toward the discovery of “driver mutations” is necessary and that the cure of cancer is unlikely to come from increasing drug development, but rather that early cancer prevention and diagnosis (eg, blood biomarker testing) are likely to be much more fruitful. In patients for whom this is not possible and who develop metastatic disease, the exploration of predictive (rather than prognostic) biomarkers is necessary to better direct care.
Conclusions
Understanding of molecular mechanisms of diseases such as GC, which are prevalent in the East and Asia, lags behind that of diseases such as breast and colon cancer, which are common in the West. Better algorithms to identify driver mutations are needed, but, more importantly, prevention and the development of novel biomarkers for early diagnosis are the keys to managing GC.
Dr. Ajan is on the advisory committee for Amgen Inc.; Celgene Corporaqtion; Eli Lilly and Company; and Novartis. He has received research Corporation funding from Amgen Inc.; Genentech; Eli Lilly and Company; Novartis; Roche Laboratories, Inc.; and Taiho Pharmaceutical Co., LTD. The remaining authors have disclosed that they have no financial interests, arrangements, affiliations, or commercial interests with the manufacturers of any products discussed in this article or their competitors.
This work was supported in part from philanthropic donations received from the Caporella, Dallas, Sultan, Park, Smith, Frazier, Oaks, Vansteklenburg, and Cantu Families, and from the Schecter Private Foundation, Rivercreek Foundation, the Kevin Fund, the Myer Fund, the Dio Fund, the Milrod Fund, and the Multidisciplinary Research Grant provided by the University of Texas MD Anderson Cancer Center, Houston, Texas. It was also supported in part by CA129906 and CA172741 from the National Cancer Institute (J.A.A.).
References
- 3.↑
Howlader N, Ries LA, Stinchcomb DG, Edwards BK. The impact of underreported Veterans Affairs data on national cancer statistics: analysis using population-based SEER registries. J Natl Cancer Inst 2009;101:533–536.
- 4.↑
Macdonald JS, Smalley SR, Benedetti J et al.. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med 2001;345:725–730.
- 5.
Smalley SR, Benedetti JK, Haller DG et al.. Updated analysis of SWOG-directed intergroup study 0116: a phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol 2012;30:2327–2333.
- 6.↑
Cunningham D, Allum WH, Stenning SP et al.. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 2006;355:11–20.
- 7.↑
Bang YJ, Van Cutsem E, Feyereislova A et al.. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687–697.
- 8.↑
Wadhwa R, Song S, Lee JS et al.. Gastric cancer-molecular and clinical dimensions. Nat Rev Clin Oncol 2013;10:643–655.
- 9.↑
Marchitti SA, Brocker C, Stagos D, Vasiliou V. Non-P450 aldehyde oxidizing enzymes: the aldehyde dehydrogenase superfamily. Expert Opin Drug Metab Toxicol 2008;4:697–720.
- 10.
Vasiliou V, Nebert DW. Analysis and update of the human aldehyde dehydrogenase (ALDH) gene family. Hum Genomics 2005;2:138–143.
- 11.↑
Vasiliou V, Pappa A. Polymorphisms of human aldehyde dehydrogenases. Consequences for drug metabolism and disease. Pharmacology 2000;61:192–198.
- 12.↑
Ajani JA, Wang X, Song S et al.. ALDH-1 expression levels predict response or resistance to preoperative chemoradiation in resectable esophageal cancer patients. Mol Oncol 2014;8:142–149.
- 13.↑
Nishikawa S, Konno M, Hamabe A et al.. Aldehyde dehydrogenase high gastric cancer stem cells are resistant to chemotherapy. Int J Oncol 2013;42:1437–1442.
- 14.↑
Chen W, Zhang X, Chu C et al.. Identification of CD44+ cancer stem cells in human gastric cancer. Hepatogastroenterology 2013;60:949–954.
- 15.↑
Zong Y, Panikkar A, Xu J et al.. Notch signaling controls liver development by regulating biliary differentiation. Development 2009;136:1727–1739.
- 17.↑
Song S, Maru DM, Ajani JA et al.. Loss of TGF-beta adaptor beta2SP activates notch signaling and SOX9 expression in esophageal adenocarcinoma. Cancer Res 2013;73:2159–2169.
- 18.↑
Choi YJ, Song JH, Yoon JH et al.. Aberrant expression of SOX9 is associated with gastrokine 1 inactivation in gastric cancers. Gastric Cancer 2014;17:247–254.
- 19.↑
Zhou CJ, Guo JQ, Zhu KX et al.. Elevated expression of SOX9 is related with the progression of gastric carcinoma. Diagn Cytopathol 2011;39:105–109.
- 20.↑
Huang J, Wu S, Barrera J et al.. The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila Homolog of YAP. Cell 2005;122:421–434.
- 21.↑
Wu S, Liu Y, Zheng Y et al.. The TEAD/TEF family protein Scalloped mediates transcriptional output of the Hippo growth-regulatory pathway. Dev Cell 2008;14:388–398.
- 22.↑
Zhao B, Ye X, Yu J et al.. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev 2008;22:1962–1971.
- 24.
Harvey K, Tapon N. The Salvador-Warts-Hippo pathway: an emerging tumour-suppressor network. Nat Rev Cancer 2007;7:182–191.
- 26.
Avruch J, Zhou D, Bardeesy N. YAP oncogene overexpression supercharges colon cancer proliferation. Cell Cycle 2012;11:1090–1096.
- 27.↑
Hergovich A. YAP-Hippo signalling downstream of leukemia inhibitory factor receptor: implications for breast cancer. Breast Cancer Res 2012;14:326.
- 28.↑
Pobbati AV, Chan SW, Lee I et al.. Structural and functional similarity between the Vgll1-TEAD and the YAP-TEAD complexes. Structure 2012;20:1135–1140.
- 29.↑
Guo T, Lu Y, Li P et al.. A novel partner of Scalloped regulates Hippo signaling via antagonizing Scalloped-Yorkie activity. Cell Res 2013;23:1201–1214.
- 30.
Koontz LM, Liu-Chittenden Y, Yin F et al.. The Hippo effector Yorkie controls normal tissue growth by antagonizing scalloped-mediated default repression. Dev Cell 2013;25:388–401.
- 31.↑
Zhang W, Gao Y, Li P et al.. VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex. Cell Res 2014;24:331–343.
- 32.↑
Zhou GX, Li XY, Zhang Q et al.. Effects of the hippo signaling pathway in human gastric cancer. Asian Pac J Cancer Prev 2013;14:5199–5205.
- 33.↑
Jiao S, Wang H, Shi Z et al.. A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell 2014;25:166–180.
- 35.↑
Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes Dev 2001;15:3059–3087.
- 36.↑
Kim JY, Ko GH, Lee YJ et al.. Prognostic value of sonic hedgehog protein expression in gastric cancer. Jpn J Clin Oncol 2012;42:1054–1059.
- 37.↑
Sims-Mourtada J, Izzo JG, Apisarnthanarax S et al.. Hedgehog: an attribute to tumor regrowth after chemoradiotherapy and a target to improve radiation response. Clin Cancer Res 2006;12:6565–6572.
- 38.↑
Sims-Mourtada J, Izzo JG, Ajani J, Chao KS. Sonic Hedgehog promotes multiple drug resistance by regulation of drug transport. Oncogene 2007;26:5674–5679.
- 39.↑
Yoo YA, Kang MH, Lee HJ et al.. Sonic hedgehog pathway promotes metastasis and lymphangiogenesis via activation of Akt, EMT, and MMP-9 pathway in gastric cancer. Cancer Res 2011;71:7061–7070.
- 40.↑
Bae JH, Schlessinger J. Asymmetric tyrosine kinase arrangements in activation or autophosphorylation of receptor tyrosine kinases. Mol Cells 2010;29:443–448.
- 41.↑
Dailey L, Ambrosetti D, Mansukhani A, Basilico C. Mechanisms underlying differential responses to FGF signaling. Cytokine Growth Factor Rev 2005;16:233–247.
- 42.↑
Beenken A, Mohammadi M. The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov 2009;8:235–253.
- 43.↑
Cunningham DL, Sweet SM, Cooper HJ, Heath JK. Differential phosphoproteomics of fibroblast growth factor signaling: identification of Src family kinase-mediated phosphorylation events. J Proteome Res 2010;9:2317–2328.
- 44.↑
Katoh M, Nakagama H. FGF receptors: cancer biology and therapeutics. Med Res Rev 2014;34:280–300.
- 45.↑
Jang JH, Shin KH, Park JG. Mutations in fibroblast growth factor receptor 2 and fibroblast growth factor receptor 3 genes associated with human gastric and colorectal cancers. Cancer Res 2001;61:3541–3543.
- 46.
Davies H, Hunter C, Smith R et al.. Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res 2005;65:7591–7595.
- 47.↑
Stephens P, Edkins S, Davies H et al.. A screen of the complete protein kinase gene family identifies diverse patterns of somatic mutations in human breast cancer. Nat Genet 2005;37:590–592.
- 48.↑
Bai A, Meetze K, Vo NY et al.. GP369, an FGFR2-IIIb-specific antibody, exhibits potent antitumor activity against human cancers driven by activated FGFR2 signaling. Cancer Res 2010;70:7630–7639.
- 49.↑
Su X, Zhan P, Gavine PR et al.. FGFR2 amplification has prognostic significance in gastric cancer: results from a large international multicentre study. Br J Cancer 2014;110:967–975.
- 50.↑
Deng N, Goh LK, Wang H et al.. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut 2012;61:673–684.
- 51.↑
Kilgour E, Su X, Zhan P et al.. Prevalence and prognostic significance of FGF receptor 2 (FGFR2) gene amplification in Caucasian and Korean gastric cancer cohorts [abstract]. J Clin Oncol 2012;30(Suppl):Abstract 4124.
- 52.↑
Jung EJ, Jung EJ, Min SY et al.. Fibroblast growth factor receptor 2 gene amplification status and its clinicopathologic significance in gastric carcinoma. Hum Pathol 2012;43:1559–1566.
- 53.↑
Xie L, Su X, Zhang L et al.. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin Cancer Res 2013;19:2572–2583.
- 54.↑
Hofmann M, Stoss O, Shi D et al.. Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology 2008;52:797–805.
- 55.↑
Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol 2008;19:1523–1529.
- 56.↑
Tanner M, Hollmen M, Junttila TT et al.. Amplification of HER-2 in gastric carcinoma: association with Topoisomerase IIalpha gene amplification, intestinal type, poor prognosis and sensitivity to trastuzumab. Ann Oncol 2005;16:273–278.
- 57.↑
Bang YJ. A randomized, open-label, phase III study of lapatinib in combination with weekly paclitaxel versus weekly paclitaxel alone in the second-line treatment of HER2 amplified advanced gastric cancer (AGC) in Asian population: Tytan study [abstract]. J Clin Oncol 2013;31(Suppl):Abstract 11.
- 58.↑
Bar-Sela G, Hershkovitz D, Haim N et al.. The incidence and prognostic value of HER2 overexpression and cyclin D1 expression in patients with gastric or gastroesophageal junction adenocarcinoma in Israel. Oncol Lett 2013;5:559–563.
- 59.
Barros-Silva JD, Leitao D, Afonso L et al.. Association of ERBB2 gene status with histopathological parameters and disease-specific survival in gastric carcinoma patients. Br J Cancer 2009;100:487–493.
- 60.↑
Chen C, Yang JM, Hu TT et al.. Prognostic role of human epidermal growth factor receptor in gastric cancer: a systematic review and meta-analysis. Arch Med Res 2013;44:380–389.
- 61.↑
Aizawa M, Nagatsuma AK, Kitada K et al.. Evaluation of HER2-based biology in 1,006 cases of gastric cancer in a Japanese population. Gastric Cancer 2014;17:34–42.
- 62.
Gordon MA, Gundacker HM, Benedetti J et al.. Assessment of HER2 gene amplification in adenocarcinomas of the stomach or gastroesophageal junction in the INT-0116/SWOG9008 clinical trial. Ann Oncol 2013;24:1754–1761.
- 63.↑
Grabsch H, Sivakumar S, Gray S et al.. HER2 expression in gastric cancer: rare, heterogeneous and of no prognostic value—conclusions from 924 cases of two independent series. Cell Oncol 2010;32:57–65.
- 64.↑
Oh HS, Eom DW, Kang GH et al.. Prognostic implications of EGFR and HER-2 alteration assessed by immunohistochemistry and silver in situ hybridization in gastric cancer patients following curative resection. Gastric Cancer 2014;17:402–411.
- 65.↑
Okines AF, Thompson LC, Cunningham D et al.. Effect of HER2 on prognosis and benefit from peri-operative chemotherapy in early oesophago-gastric adenocarcinoma in the MAGIC trial. Ann Oncol 2013;24:1253–1261.
- 66.↑
Smith I, Procter M, Gelber RD et al.. 2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial. Lancet 2007;369:29–36.
- 67.↑
Slamon DJ, Leyland-Jones B, Shak S et al.. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:783–792.
- 68.↑
Office of Medical Products and Tobacco. Trastuzumab. U.S. Food and Drug Adminstration Web site. Available at: http://www.fda.gov/AboutFDA/CentersOffices/OfficeofMedicalProductsandTobacco/CDER/ucm230418.htm. Accessed March 12, 2015.
- 69.↑
Hecht JR, Bang YJ, Qin S et al.. Lapatinib in combination with capecitabine plus oxaliplatin (CapeOx) in HER2-positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma (AC): the TRIO-013/LOGiC trial [abstract]. J Clin Oncol 2013;31(Suppl):Abstract LBA4001.
- 70.↑
Tabernero J, Hoff PM, Shen L et al.. Pertuzumab (P) with trastuzumab (T) and chemotherapy (CTX) in patients (pts) with HER2-positive metastatic gastric or gastroesophageal junction (GEJ) cancer: an international phase III study (JACOB) [abstract]. J Clin Oncol 2013;31(Suppl):Abstract TPS4150.
- 71.↑
Baselga J, Cortes J, Kim SB et al.. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med 2012;366:109–119.
- 72.↑
Verma S, Miles D, Gianni L et al.. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med 2012;367:1783–1791.
- 73.↑
Kim MA, Lee HS, Lee HE et al.. EGFR in gastric carcinomas: prognostic significance of protein overexpression and high gene copy number. Histopathology 2008;52:738–746.
- 74.↑
Lieto E, Ferraraccio F, Orditura M et al.. Expression of vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) is an independent prognostic indicator of worse outcome in gastric cancer patients. Ann Surg Oncol 2008;15:69–79.
- 75.↑
Kandel C, Leclair F, Bou-Hanna C et al.. Association of HER1 amplification with poor prognosis in well differentiated gastric carcinomas. J Clin Pathol 2014;67:307–312.
- 76.↑
Aydin K, Okutur SK, Bozkurt M et al.. Effect of epidermal growth factor receptor status on the outcomes of patients with metastatic gastric cancer: a pilot study. Oncol Lett 2014;7:255–259.
- 77.↑
Hong L, Han Y, Yang J et al.. Prognostic value of epidermal growth factor receptor in patients with gastric cancer: a meta-analysis. Gene 2013;529:69–72.
- 78.↑
Lordick F, Kang YK, Chung HC et al.. Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:490–499.
- 79.↑
Waddell T, Chau I, Cunningham D et al.. Epirubicin, oxaliplatin, and capecitabine with or without panitumumab for patients with previously untreated advanced oesophagogastric cancer (REAL3): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:481–489.
- 80.↑
Kiyose S, Nagura K, Tao H et al.. Detection of kinase amplifications in gastric cancer archives using fluorescence in situ hybridization. Pathol Int 2012;62:477–484.
- 81.↑
Lee J, Seo JW, Jun HJ et al.. Impact of MET amplification on gastric cancer: possible roles as a novel prognostic marker and a potential therapeutic target. Oncol Rep 2011;25:1517–1524.
- 82.
Graziano F, Galluccio N, Lorenzini P et al.. Genetic activation of the MET pathway and prognosis of patients with high-risk, radically resected gastric cancer. J Clin Oncol 2011;29:4789–4795.
- 83.↑
Lennerz JK, Kwak EL, Ackerman A et al.. MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J Clin Oncol 2011;29:4803–4810.
- 84.↑
Lee HE, Kim MA, Lee HS et al.. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer 2012;107:325–333.
- 85.↑
Janjigian YY, Tang LH, Coit DG et al.. MET expression and amplification in patients with localized gastric cancer. Cancer Epidemiol Biomarkers Prev 2011;20:1021–1027.
- 86.↑
Oliner KS, Tang R, Anderson A et al.. Evaluation of MET pathway biomarkers in a phase II study of rilotumumab (R, AMG 102) or placebo (P) in combination with epirubicin, cisplatin, and capecitabine (ECX) in patients (pts) with locally advanced or metastatic gastric (G) or esophagogastric junction (EGJ) cancer [abstract]. J Clin Oncol 2012;30(Suppl):Abstract 4005.
- 87.↑
Kwak EL, LoRusso P, Hamid O et al.. Clinical activity of AMG 337, an oral MET kinase inhibitor, in adult patients (pts) with MET-amplified gastroesophageal junction (GEJ), gastric (G), or esophageal (E) cancer [abstract]. J Clin Oncol 2015;33(Suppl):Abstract 1.
- 88.↑
Yoshikawa T, Tsuburaya A, Kobayashi O et al.. Plasma concentrations of VEGF and bFGF in patients with gastric carcinoma. Cancer Lett 2000;153:7–12.
- 89.↑
Park do J, Yoon C, Thomas N et al.. Prognostic significance of targetable angiogenic and growth factors in patients undergoing resection for gastric and gastroesophageal junction cancers. Ann Surg Oncol 2014;21:1130–1137.
- 90.↑
Ji YN, Wang Q, Li Y, Wang Z. Prognostic value of vascular endothelial growth factor A expression in gastric cancer: a meta-analysis. Tumour Biol 2014;35:2787–2793.
- 91.↑
Ohtsu A, Shah MA, Van Cutsem E et al.. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 2011;29:3968–3976.
- 92.↑
Van Cutsem E, de Haas S, Kang YK et al.. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol 2012;30:2119–2127.
- 93.↑
Fuchs CS, Tomasek J, Yong CJ et al.. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014;383:31–39.
- 94.↑
Wilke H, Van Cutsem E, Oh SC et al.. RAINBOW: a global, phase III, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastroesophageal junction (GEJ) and gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy rainbow IMCL CP12-0922 (I4T-IE-JVBE) [abstract]. J Clin Oncol 2014;32(Suppl):Abstract LBA7.
- 95.↑
Tabernero J, Ohtsu A, Muro K et al.. Exposure-response (E-R) relationship of ramucirumab (RAM) from two global, randomized, double-blind, phase 3 studies of patients (Pts) with advanced second-line gastric cancer [abstract]. J Clin Oncol 2015;33(Suppl):Abstract 121.
- 96.↑
Taguchi F, Kodera Y, Katanasaka Y et al.. Efficacy of RAD001 (everolimus) against advanced gastric cancer with peritoneal dissemination. Invest New Drugs 2011;29:1198–1205.
- 97.↑
Lang SA, Gaumann A, Koehl GE et al.. Mammalian target of rapamycin is activated in human gastric cancer and serves as a target for therapy in an experimental model. Int J Cancer 2007;120:1803–1810.
- 98.↑
Xu DZ, Geng QR, Tian Y et al.. Activated mammalian target of rapamycin is a potential therapeutic target in gastric cancer. BMC Cancer 2010;10:536.
- 99.↑
Yu G, Wang J, Chen Y et al.. Overexpression of phosphorylated mammalian target of rapamycin predicts lymph node metastasis and prognosis of chinese patients with gastric cancer. Clin Cancer Res 2009;15:1821–1829.
- 100.↑
Ohtsu A, Ajani JA, Bai YX et al.. Everolimus for previously treated advanced gastric cancer: results of the randomized, double-blind, phase III GRANITE-1 study. J Clin Oncol 2013;31:3935–3943.
- 101.↑
Samuels Y, Wang Z, Bardelli A et al.. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004;304:554.
- 102.
Velho S, Oliveira C, Ferreira A et al.. The prevalence of PIK3CA mutations in gastric and colon cancer. Eur J Cancer 2005;41:1649–1654.
- 103.
Lee J, van Hummelen P, Go C et al.. High-throughput mutation profiling identifies frequent somatic mutations in advanced gastric adenocarcinoma. PLoS One 2012;76:e38892.
- 104.↑
Shi J, Yao D, Liu W et al.. Highly frequent PIK3CA amplification is associated with poor prognosis in gastric cancer. BMC Cancer 2012;12:50.
- 105.↑
Barbi S, Cataldo I, De Manzoni G et al.. The analysis of PIK3CA mutations in gastric carcinoma and metanalysis of literature suggest that exon-selectivity is a signature of cancer type. J Exp Clin Cancer Res 2010;29:32.
- 106.↑
Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol 2007;8:239–245.
- 107.↑
James ES, Harney S, Wordsworth BP et al.. PDCD1: a tissue-specific susceptibility locus for inherited inflammatory disorders. Genes Immun 2005;6:430–437.
- 108.↑
Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008;26:677–704.
- 109.↑
Gao Q, Wang XY, Qiu SJ et al.. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res 2009;15:971–979.
- 110.
Karim R, Jordanova ES, Piersma SJ et al.. Tumor-expressed B7-H1 and B7-DC in relation to PD-1+ T-cell infiltration and survival of patients with cervical carcinoma. Clin Cancer Res 2009;15:6341–6347.
- 111.
Hua D, Sun J, Mao Y et al.. B7-H1 expression is associated with expansion of regulatory T cells in colorectal carcinoma. World J Gastroenterol 2012;18:971–978.
- 112.↑
Ohigashi Y, Sho M, Yamada Y et al.. Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clin Cancer Res 2005;11:2947–2953.
- 113.
Zeng Z, Shi F, Zhou L et al.. Upregulation of circulating PD-L1/PD-1 is associated with poor post-cryoablation prognosis in patients with HBV-related hepatocellular carcinoma. PLoS One 2011;6:e23621.
- 114.
Nomi T, Sho M, Akahori T et al.. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res 2007;13:2151–2157.
- 115.↑
Saito H, Kuroda H, Matsunaga T et al.. Increased PD-1 expression on CD4+ and CD8+ T cells is involved in immune evasion in gastric cancer. J Surg Oncol 2013;107:517–522.
- 116.↑
Blank C, Mackensen A. Contribution of the PD-L1/PD-1 pathway to T-cell exhaustion: an update on implications for chronic infections and tumor evasion. Cancer Immunol Immunother 2007;56:739–745.
- 117.
Westin JR, Chu F, Zhang M et al.. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. Lancet Oncol 2014;15:69–77.
- 118.↑
Brahmer JR, Tykodi SS, Chow LQ et al.. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455–2465.
- 119.↑
Dulak AM, Stojanov P, Peng S et al.. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet 2013;45:478–486.
- 120.↑
Wang K, Yuen ST, Xu J et al.. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet 2014;46:573–582.
- 121.↑
Kakiuchi M, Nishizawa T, Ueda H et al.. Recurrent gain-of-function mutations of RHOA in diffuse-type gastric carcinoma. Nat Genet 2014;46:583–587.
- 122.↑
Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513:202–209.
- 123.↑
Vogelstein B, Papadopoulos N, Velculescu VE et al.. Cancer genome landscapes. Science 2013;339:1546–1558.
- 124.
Fornaro L, Lucchesi M, Caparello C et al.. Anti-HER agents in gastric cancer: from bench to bedside. Nat Rev Gastroenterol Hepatol 2011;8:369–383.
- 125.
Liu Z, Liu L, Li M et al.. Epidermal growth factor receptor mutation in gastric cancer. Pathology 2011;43:234–238.
- 126.
Mammano E, Belluco C, Sciro M et al.. Epidermal growth factor receptor (EGFR): mutational and protein expression analysis in gastric cancer. Anticancer Res 2006;26:3547–3550.
- 127.
Takahashi Y, Cleary KR, Mai M et al.. Significance of vessel count and vascular endothelial growth factor and its receptor (KDR) in intestinal-type gastric cancer. Clin Cancer Res 1996;2:1679–1684.
- 128.
Kitoh T, Yanai H, Saitoh Y et al.. Increased expression of matrix metalloproteinase-7 in invasive early gastric cancer. J Gastroenterol 2004;39:434–440.
- 129.
Corso S, Ghiso E, Cepero V et al.. Activation of HER family members in gastric carcinoma cells mediates resistance to MET inhibition. Mol Cancer 2010;9:121.
- 130.
Komuro A, Yashiro M, Iwata C et al.. Diffuse-type gastric carcinoma: progression, angiogenesis, and transforming growth factor beta signaling. J Natl Cancer Inst 2009;101:592–604.
- 131.
Fukaya M, Isohata N, Ohta H et al.. Hedgehog signal activation in gastric pit cell and in diffuse-type gastric cancer. Gastroenterology 2006;131:14–29.
- 132.
Song S, Ajani JA. The role of microRNAs in cancers of the upper gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2013;10:109–118.
- 133.
Wong H, Yau T. Targeted therapy in the management of advanced gastric cancer: are we making progress in the era of personalized medicine? Oncologist 2012;17:346–358.
- 134.
Pereira PS, Teixeira A, Pinho S et al.. E-cadherin missense mutations, associated with hereditary diffuse gastric cancer (HDGC) syndrome, display distinct invasive behaviors and genetic interactions with the Wnt and Notch pathways in Drosophila epithelia. Hum Mol Genet 2006;15:1704–1712.
- 135.
Zhou Y, Li G, Wu J et al.. Clinicopathological significance of E-cadherin, VEGF, and MMPs in gastric cancer. Tumour Biol 2010;31:549–558.
- 136.
Guo CY, Xu XF, Wu JY, Liu SF. PCR-SSCP-DNA sequencing method in detecting PTEN gene mutation and its significance in human gastric cancer. World J Gastroenterol 2008;14:3804–3811.
- 137.
Byun DS, Cho K, Ryu BK et al.. Frequent monoallelic deletion of PTEN and its reciprocal associatioin with PIK3CA amplification in gastric carcinoma. Int J Cancer 2003;104:318–327.