Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
Gastric cancer remains a major cause of cancer-related mortality in the world, despite declining rates of incidence in many industrialized countries. In this chapter, we mainly discuss gastric adenocarcinoma, which makes up most of gastric malignancies.
Gastric cancer is the third leading cause of cancer mortality in the world, although the overall incidence is declining. In Western countries, the incidence of gastric cancer has decreased dramatically over the past century; in the USA, gastric cancer mortality has decreased 87% since 1950 with a similar trend being reported in Europe. In the USA, the incidence of gastric cancer has diminished to approximately 7.6 cases per 100,000 people, whereas as recently as 1945, gastric cancer was the leading cause of cancer mortality in men.
There is great geographic variation in gastric cancer incidence, with the highest incidence rates in the Far East ( Fig. 54.1 ). Eastern Europe and Central and South America also have high incidence rates, with the lowest incidence rates observed in North America, North Africa, South Asia, and Australia. Although gastric cancer was common in industrialized countries in the past, the latest epidemiologic data indicate that more than 70% of new cases of gastric cancer are in developing countries, reflecting a more rapid decline in developed countries.
In the USA, the median age of diagnosis is 70 years. In Japan, a country with a high incidence of gastric cancer, the mean age of diagnosis is roughly a decade earlier, perhaps reflecting lead-time bias due to widespread screening. The incidence of gastric cancer in males is approximately twice that in females ( Table 54.1 ). The incidence of gastric cancer in blacks in the USA is nearly double that in whites. Native Americans and Hispanics also have a higher risk of developing gastric cancer than whites. In contrast to the pattern seen with nonjunctional gastric cancers, the incidence rates of adenocarcinomas at the esophagogastric junction (EGJ, formerly “cardia cancer”) are rising, According to the US Surveillance, Epidemiology, and Ends Results (SEER) database, these junctional cancers now represent 27% of gastric cancers in the USA, up from just 10% in 1975.
Incidence | Mortality | |||
---|---|---|---|---|
Male | Female | Male | Female | |
Developed countries | 15.6 | 6.7 | 9.2 | 4.2 |
Developing countries | 18.1 | 7.8 | 14.4 | 6.5 |
There are numerous dietary, environmental, and genetic risk factors for gastric adenocarcinoma ( Box 54.1 ). The dominant risk factor remains, however, infection with Hp and the associated chronic-active inflammation of the gastric mucosa (see Chapter 52 ).
High salt intake
History of gastric ulcer
Obesity (adenocarcinoma of the cardia only)
Pernicious anemia ∗
Regular aspirin or other NSAID use (protective)
Snuff tobacco use
Diet high in nitrates
Heavy alcohol use
High ascorbate intake (protective)
High intake of fresh fruits and vegetables (protective)
Low socioeconomic status
Ménétrier disease
Statin use (protective)
High green tea consumption (protective)
Hyperplastic and fundic gland polyps
Gastric cancer can be subdivided using the Laurén classification into distinct histologic subtypes with different epidemiologic and prognostic features ( Fig. 54.2 ). The intestinal type of cancer is characterized by the formation of gland-like tubular structures with features reminiscent of intestinal glands. This type of gastric cancer is more closely linked to environmental and dietary risk factors, tends to be the predominant form in regions with a high incidence of gastric cancer, and is the form of cancer that is now declining worldwide. The diffuse type of cancer lacks glandular structure and consists of poorly cohesive cells that infiltrate the wall of the stomach. It is found at the same frequency throughout the world, occurs at a younger age, and is associated with a worse prognosis than the intestinal form. Extensive involvement of the stomach by the diffuse type can result in a rigid and thickened stomach, a condition referred to as linitis plastic ( ). Another key feature of diffuse type cancers are signet-ring cells, special mucin-filled cells that are not present in intestinal type adenocarcinomas. There are also mixed phenotypes that contain heterogenous areas that feature predominantly either intestinal- or diffuse-type characteristics.
Video 54-1 EUS of a gastric lipoma.
Adenocarcinoma of the stomach is also classified into proximal tumors (EGJ and gastric cardia) and distal or nonjunctional tumors (fundus, body, and antrum of the stomach). Junctional cancers can be further classified according to the Siewert classification by the location of the main tumor mass into Type I (1 to 5 cm above the EGJ), Type II (from 1 cm above to 2 cm below the junction), and Type III (2 to 5 cm below the junction) tumors. There is no clear distinction between the genetic and cellular origin of adenocarcinomas of the distal esophagus, the EGJ, and a subgroup of nonjunctional distal gastric cancers. Interestingly, with the decreasing incidence of Hp infection, nonjunctional tumors have been declining while more proximal tumors have been increasing. In a mouse model, it has even been postulated that Barrett esophagus-related esophageal cancer and cancer of the EGJ have their origins in the gastric cardia. Emerging data from gene expression profiling suggest that differences in pathologic appearance and clinical behavior may be due to the presence of unique molecular phenotypes. Characterization of the gastric cancer genomic landscape reveals the presence of multiple alterations in the expression of tyrosine kinase receptors, which in conjunction with their ligands and downstream effector molecules represent potential pathways for future drug development.
The Cancer Genome Atlas (TCGA) consortium suggested gastric cancer subtypes based on the genomic profile of about 300 gastric cancers. This classification correlated well with the clustering of high throughput data of different platforms including epigenome, transcriptome, and proteome analysis. So far, there are only little data to support the biological relevance of this proposed classification. Previous transcriptome analyses of gastric cancers, on the other hand, have demonstrated phenotypic clusters with either distinct prognostic outcomes or different response to systemic treatment. ,
It is believed that the development of intestinal-type gastric cancer occurs through a multistep process in which the normal mucosa is sequentially transformed into a hyperproliferative epithelium, followed by metaplastic processes leading to glandular atrophy, dysplasia, and then carcinoma. In colon cancer, the evidence is strong that each step in the transition is associated with a specific gene mutation, but the evidence that gastric cancer follows a comparable sequence of genetic events has been lacking. However, in both the intestinal-type gastric cancer and colorectal cancer, it does appear that DNA mutations are established over time in stem cells in the normal human stomach, and that in intestinal metaplasia these mutations spread through the stomach through a process involving crypt fission and monoclonal conversion of glands. The contention that the pathogenesis of intestinal-type gastric cancer is a multistep process is supported mainly by the observation that both chronic atrophic gastritis and intestinal metaplasia are found in higher incidences in patients with intestinal-type cancer and in countries with a high incidence of gastric cancer (see Chapter 52 ). ,
This multistep model of intestinal-type gastric cancer, developed in large part by Pelayo Correa and colleagues, postulates that there is a temporal sequence of preneoplastic changes that eventually lead to the development of gastric cancer. A common feature of the initiation and progression to intestinal-type gastric cancer is chronic inflammation of the gastric mucosa. Hp infection is the primary cause of gastric inflammation and the leading etiologic agent for gastric cancer (see Chapter 52 ). In a subset of patients, the inflammatory process leads to the development of atrophic gastritis (with loss of glandular tissue) followed by progression to intestinal metaplasia, dysplasia, early gastric cancer, and, eventually, advanced gastric cancer ( Fig. 54.3 ). Although animal models suggested that all stages prior to the development of high-grade dysplasia are potentially reversible, there is still ongoing debate what defines the “point of no return” for humans from which further progression of neoplasia can no longer be prevented. , Eradication of Hp has the potential to prevent gastric cancer as shown in recent meta-analyses. , The preventive effect of eradication is more evident if there are no preneoplastic conditions of the gastric mucosa (glandular atrophy, intestinal metaplasia) at the time of intervention. Hp eradication can prevent further progression of preneoplastic conditions, and even a certain degree of regression can be documented. Although it is currently assumed that presence of intestinal metaplasia is most likely to mark the point of no return , there is even an effect of Hp eradication if advanced lesions are present (e.g., after endoscopic resection of an early gastric cancer).
Unlike the situation observed with colon cancer, the precise genes involved in each step of this progression are still not defined. Nevertheless, next-generation sequencing techniques have shown that there is more heterogeneity in genetic alterations in gastric cancer and cancer of the EGJ than in colon cancer. , Furthermore, the premalignant stages of gastric cancer are not as readily identifiable during endoscopy as those of colon cancer, and many gastric carcinomas are very heterogeneous, containing a large percentage of stromal cells. These stromal cells, which also include cancer-associated fibroblasts known to promote tumor growth, have been reported to show distinct genetic and epigenetic changes that may confound tumor analysis. , This feature makes characterization of the timing of specific gene mutations in gastric cancer difficult at best. Currently, the role of chronic inflammation in the diffuse type of gastric cancer, as well as the similarities if any to the proposed pathway in Fig. 54.3 for the intestinal type of cancer, remain to be clarified. One common factor that is related to both histological subtypes is a strong association with Hp infection, which has shown to directly modify genes involved in DNA damage repair (DDR) pathways. Modifications of DDR-related genes are a common event in gastric carcinogenesis.
Hp is a gram-negative microaerophilic bacterium that infects nearly half the world’s population and is recognized as the primary etiologic agent for gastric cancer. Indeed, H. plyori has been classified as a class I (or definite) carcinogen by the International Agency for Research on Cancer, a branch of the WHO. Infection with Hp has been found in every population studied, although the prevalence is higher in developing countries and most parts of East Asia. ,
The natural history of chronic Hp infection includes 3 possible outcomes : (1) simple gastritis, where patients often remain asymptomatic; (2) duodenal ulcer phenotype, which occurs in 10% to 15% of infected subjects; and (3) gastric ulcer/gastric cancer phenotype. The risk for gastric cancer development varies with the type of background gastritis, but in general, corpus-dominant gastritis resulting in a low acid state is mainly associated with an increased risk. Hp -induced duodenal ulcer disease is associated with a high gastric acid output as well as a reduced risk for developing gastric cancer. Studies suggest that Hp -infected patients develop chronic atrophic gastritis at a rate of 1% to 3% per year of infection. , , Thus, those patients who are genetically predisposed to developing atrophic gastritis in response to Hp infection are likely to be also predisposed to gastric cancer. Although Hp infection is associated with both diffuse-type and intestinal-type adenocarcinomas, we focus in this chapter mainly on the mechanisms responsible for the formation of intestinal-type adenocarcinoma because they have been better studied. The association of Hp with mucosa-associated lymphoid tissue lymphoma is discussed in Chapter 32 .
The increased risk of development of gastric adenocarcinoma due to Hp infection depends on multiple factors including host genetic factors, the strain of bacteria (including bacterial virulence factors), the duration of infection, and the presence or absence of other environmental risk factors (e.g., poor diet, smoking). In a Japanese cohort, only those infected with Hp developed gastric adenocarcinoma during follow-up (2.9% vs. 0%; P <0.001). Additional cohort studies from China and Taiwan have reported similar findings. , In Western countries, the association between Hp and gastric cancer appears to be confined to nonjunctional tumors. There are data, however, that Hp is likely to be also associated with Siewert type III junctional cancers as well as potentially a subgroup of type II tumors. ,
A combination of a virulent bacterial strain, a genetically permissive host, and a favorable gastric environment may be necessary for cancer to occur. Currently, genetic susceptibility factors of the human host are studied based on individual genes, but new technologies such as next-generation sequencing will enhance the identification of host genetic factors. Nevertheless, the most important factor appears to be the induction of chronic inflammation by Hp infection. This leads to an impairment of the epithelial barrier function of the gastric mucosa, thereby increasing the impact of other pathogenic factors (e.g., diet). Several aspects of the inflammatory milieu have been implicated as carcinogens; they include increased oxidative stress and the formation of oxygen free radicals, leading to DNA damage, increased CD4+ T cells and myeloid cells, and elevated proinflammatory cytokine production, all leading to accelerated cell turnover, reduced apoptosis, and the potential for faulty or incomplete DNA repair. Indeed, recent studies suggest that animals with deficient DNA repair mechanisms display more severe gastric dysplasia after chronic infection with Hp . As mentioned earlier, Hp is capable of directly modifying DDR-related genes and their function. Thus, evidence to date clearly indicates that the most important cofactor in the induction of Helicobacter -related disease is the host immune response. Indeed, chronic inflammation has been linked to a large number of nongastric cancers.
Chronic inflammation of the gastric mucosa appears necessary for the progression through atrophy to gastric cancer. Disease mechanisms are difficult to study in human infection, and therefore, much of our understanding of the immune response to Helicobacter organisms comes from work performed in a mouse model. Different inbred strains of mice respond to infection with varying degrees of disease susceptibility, and several knockout models have helped to elucidate the roles of individual components of the immune response in disease.
Genetic manipulation of the C57BL/6 susceptible murine strain has facilitated detailed study and has thus led to a deeper understanding of genetic factors that promote murine gastric cancer, and particularly, the role of the adaptive immune response. For example, gastric Helicobacter infection in mice deficient in lymphocytes does not result in tissue damage, cell lineage alterations, or the metaplasia-dysplasia-carcinoma sequence. , In contrast, infection in B cell-deficient mice (which retain a normal T cell response) results in severe atrophy and metaplasia identical to that seen in infected wild-type mice. Taken together, these studies underscore the crucial role of CD4+ T lymphocytes in orchestrating gastric neoplasia.
Susceptible mouse strains, such as C57BL/6, mount a strong Th1 (T helper cell type 1, expressing interferon [IFN]-γ and interleukin [IL]-12) type of immune response, whereas resistant strains, such as the BALB/c, have a polarized Th2 response (expressing IL-4 and IL-5). A Th2 response is associated with protection from mucosal damage despite the inability to eliminate bacterial colonization and, in fact, is often associated with higher bacterial colonization rates. Mouse strains such as the C3H, which has a mixed Th1/Th2 cytokine profile, show intermediate disease, suggesting that cytokines within an immune response interact to form a continuum of disease rather than discrete disease states. More recently, Th17 cells (expressing IL-17), have been shown to be an important component of Hp -induced gastritis.
Although the composite immune milieu most likely dictates disease manifestations, there may be a role for individual cytokines in both the predisposition to and protection from disease. During Helicobacter infection, the Th1 cytokine IFN-γ can promote or inhibit inflammation-driven cancer of the stomach, suggesting that a more specific immune response is responsible for cancer promotion or surveillance. Although studies in the past have suggested that IFN-γ might promote the development of gastric preneoplasia, IFN-γ overexpression in the stomach at low levels was recently shown to be able to suppress gastric cancer in models of IL-1β– and Helicobacter felis –dependent carcinogenesis. In addition, IFN-γ was shown to counteract the development of Th17 cells. Thus, different composition of the same cells and cytokines in the tumor microenvironment can contribute to a constellation that favors or inhibits carcinogenesis. On the other hand, mice lacking IL-10, a cytokine that acts to dampen an immune response, demonstrate severe atrophic gastritis in response to infection. More recently, genetic murine models have illustrated the importance of the IL-6/IL-11 family of cytokines in the development of gastric cancer.
Manipulation of the immune response within wild-type strains confirms the central role of the Th1/Th2 response in producing disease. For example, infection with the intestinal helminth Heligmosomoides polygyrus skews the immune response toward Th2 polarization and protects the C57BL/6 host from Helicobacter -induced atrophy and metaplasia. This mouse model mimics both the parasitic infection status and the paradoxical low gastric cancer-high Hp infection rates seen in areas of Africa, potentially explaining this apparent inconsistency. These observations in mice led to human studies in Africa and Latin America that confirmed that geographic regions with low gastric cancer rates had much higher Th2/Th1 immune responses to Hp . , In general, the increased Th2 type responses were found in areas where serum IgE levels were high and the prevalence of intestinal parasitism by helminthes is above 50%. These findings further stress the importance of the host response to infection and suggest the possibility that manipulation of the genetically predetermined host cytokine profile in response to environmental challenges may lessen or exacerbate the disease process.
Studies on human tissue have also demonstrated that the degree of colonization with Hp depends on various factors, such as the presence and activity of regulatory T-cells (Treg) or the initial (naive) parietal cell mass (which reflects the acid-secreting capacity of the gastric body). Tregs are associated with increasing bacterial colonization, chronic inflammatory changes, and the expression of immunosuppressive cytokines like IL10, IL17, and TGF-β. , In case of gastric cancer, Tregs are increased both in the gastric mucosa and the peripheral blood. The ratio of Th1/Th2-derived cytokines is the highest in asymptomatic gastritis, showing a steady decrease in gastric atrophy, intestinal metaplasia and intraepithelial neoplasia progression to gastric adenocarcinoma. This is associated with a concomitant increase of the Treg cell compartment in the peripheral blood as well as persistence of CagA positive strains that favors a Treg-mediated chronic inflammation. Whereas Hp infection has been unequivocally linked to gastric cancer, the development of dysplasia and invasive cancer tends to occur at a time when Hp colonization has either dramatically declined or, in some cases, has disappeared from the stomach altogether. Gastric cancer almost always occurs in the setting of prolonged gastric atrophy and hypochlorhydria, a condition that predisposes to enteric bacterial overgrowth. Although antibiotic eradication therapy targeting Hp delays and inhibits development of gastric cancer in mice, , antibiotics eradicate not only Hp but also other microorganisms that colonize the atrophic, hypochlorhydric stomach. Indeed, infection of otherwise germ-free INS-GAS mice with Hp resulted in delayed progression to gastric cancer compared to Hp -infected INS-GAS mice colonized with conventional flora. Thus, Hp may represent simply the initial, or the most prevalent, microbial factor responsible for gastric cancer progression. Ferreira et al. reported recently that the gastric microbiota of patients with gastric adenocarcinomas is significantly different from patients with chronic gastritis. The dominant gastric dysbiosis is characterized by reduced microbial diversity and reduced Hp abundance as well as an overrepresentation of bacterial genera that include intestinal commensals. With regard to these findings, it is also of interest that Hp —when present—dominates the otherwise much more diverse gastric microbiome in humans, as well as mice, prior to cancer development.
There is a great deal of genetic diversity between strains of Hp owing to point mutations, insertions, deletions, and base-pair substitutions within the genome. Several strains may infect a single individual, and existing strains can undergo mutations and change over time. , Despite this genetic diversity, several genes are recognized as risk factors for gastric carcinoma, including the cag pathogenicity island, the vacA gene, and the babA2 gene, being the most relevant and most extensively studied thus far among other factors.
The Hp genome is 1.65 million base pairs and codes for approximately 1500 genes, two thirds of which have been assigned biological roles. The function of the remaining one third of the genome remains obscure, but genome-wide analyses using DNA microarray or whole-genome sequencing technology will give a broad view of the genome of Hp in the near future. Factors that contribute to carcinogenesis include those that enable the bacteria to effectively colonize the gastric mucosa, those that incite a more aggressive host immune response, and those that affect host cell-growth signaling pathways.
Motility toward epithelial cells of the stomach is a vital feature of Hp survival tactics. This is ensured by several factors. Spiraling movement is mediated by the FlaA and FlaB proteins, which are designed to navigate the thick gastric mucus. Additionally, Hp produces HP 1069, a putative collagenase, which modifies the extracellular matrix and mucus layer, thus decreasing viscosity and allowing bacterial penetration. , In addition, Hp expresses a variety of genes that contribute to buffering of stomach acid in order to maintain a relatively neutral pH. This includes a urease gene cluster consisting of 7 genes, of which UreA/UreB complex (comprising the urease enzyme) codes for 10% of the protein of Hp and is vital for its survival.
The cag pathogenicity island is approximately 40 kb and contains 31 genes. The terminal gene of this island, cagA , is often used as a marker for the entire cag locus. Compared with cagA-negative (cagA−) strains, cag-positive (cagA+) strains are associated with more severe inflammation, higher degrees of atrophy, and a greater chance of progressing to gastric adenocarcinoma. The estimated relative risk has ranged from 2 to as high as 28.4. However, many of the genes adjacent to cagA code for a type 4 secretion system (TFSS), often viewed as a molecule needle that injects bacterial proteins (e.g., cagA) into host cells. The remarkable finding that CagA is injected into host cells, where it is phosphorylated by Src- and c-Abl kinases, has raised the possibility that CagA could directly promote growth, migration, and transformation. Indeed, transgenic expression of Hp CagA induces both GI and hematopoietic neoplasms in mice. Other genes within the pathogenicity island are also believed to be important for disease ( cagE or picB, cagG, cagH, cagI, cagL, cagM ) because they appear to be required for in vitro epithelial cell cytokine release, although they do not seem to have as great an effect on immune cell cytokine activation. These findings may explain the attenuated inflammatory response and lower cancer risk with cagA− strains in vivo.
Intracellular phosphorylation of CagA occurs at certain glutamate-proline-isoleucine-tyrosine-alanine (EPIYA) motifs. Four distinct EPIYA motifs are described (EPIYA-A, -B, -C, and -D), whose prevalence varies by geographical region. , The motifs further influence the CagA-induced immune response as well as the related cancer risk. The odds ratio for gastric cancer is close to 7.3 in the case of 1 EPIYA-C segment and can be up to 51 in case of 2 or more segments. , Also, genetic variations in further cagPAI-related genes have been demonstrated to be associated with gastric cancer.
All strains of Hp carry the vacA gene, which codes for a pore-forming, vacuolating toxin, but expression differs according to allelic variation. Approximately 50% of Hp strains express the vacA protein, which has been shown to be a very powerful inhibitor of T cell activation in vitro. Although and cagA map to different loci within the Hp genome, the vacA protein is commonly expressed in cagA+ strains. There are various forms of vacA, and the s1m1 strains are highly toxigenic. Other bacterial virulence factors, such as cagE , may play a role in the modulation of apoptosis and the host inflammatory response, thereby contributing to disease manifestations. Indeed, “virulent strains” (cagA+, cagE+, and VacA+ s1m1) appear to be more potent inducers of proinflammatory mediators than “nonvirulent strains” (cagA−, cagE−, and VacA-), possibly explaining the higher association of cagA+ strains with gastric cancer.
Numerous dietary factors have been implicated as risk factors for gastric cancer. The decline in gastric cancer rates has coincided with the widespread use of refrigeration and the concomitant higher intake of fresh fruits and vegetables and lower intake of pickled and salted foods. Use of refrigeration for more than 10 to 20 years has been associated with a decreased risk of gastric cancer. Lower temperatures reduce the rate of bacterial, fungal, and other contaminants of fresh food, as well as the bacterial formation of nitrites. Additionally, high intake of highly preserved foods may be associated with increased gastric cancer risk, potentially due to higher contents of salt, nitrates, and polycyclic aromatic amines.
Much attention has been given to the effects of high nitrate intake. When nitrates are reduced to nitrite by bacteria or macrophages, they can react with other nitrogenated substances to form N -nitroso compounds that are known mitogens and carcinogens . In rats, N -nitroso compounds have been shown to cause gastric cancer. However, studies trying to link N -nitroso exposure to gastric cancer risk have been inconclusive, perhaps reflecting the fact that nitrate intake does not necessarily correlate with nitrosation levels. A Swedish cohort study found a nearly 2-fold increased risk of gastric cancer associated with high dietary nitrate intake. However, separate large cohort studies from Europe did not demonstrate an association between nitrate intake and risk of gastric cancer. ,
Another factor implicated in the development of gastric cancer is a diet high in salt (pickled foods, soy sauce, dried and salted fish, and meat). High salt intake has been associated with higher rates of atrophic gastritis in humans and animals in the setting of Helicobacter infection and increases the mutagenicity of nitrosated food in animal models. , High-salt diets are associated with a roughly a 1.5- to 2-fold increased risk of gastric cancer. Cohort and case-control studies have also found an increased risk of gastric cancer associated with processed meat intake. , Possible mechanisms include higher bacterial loads, up-regulation of Hp cagA expression, and increased cell proliferation and p21 expression. , , There is a clear interaction between Hp infection and dietary risk factors for gastric cancer leading to a disproportional risk increase for Hp -positive subjects with high intake of red or processed meat compared to individuals in whom only one of these risk factors is present.
Epidemiologic studies have had inconsistent findings with regard to fruit and vegetable consumption and risk of gastric cancer. Other foods or dietary factors that have been implicated as potential risk factors for gastric cancer are high intake of fried food, foods high in fat, high intake of red meat, and aflatoxins. , Diets with a high intake of fresh fish and antioxidants may be protective (also see later). , However, there are insufficient data to make definitive conclusions regarding these factors.
Tobacco has long been established as a carcinogen, and numerous epidemiologic studies have demonstrated an association between cigarette smoking and gastric cancer. Several large cohort studies from Europe and Asia have reported a significantly increased risk of gastric cancer among smokers. A recent meta-analysis found that, compared to never smokers, current smokers had a 1.5- to 2-fold increased risk of gastric cancer, both for the cardia and noncardia region. The authors also reported an increased association with greater amounts of smoking.
Moist snuff is a smokeless tobacco product promoted as an alternative to cigarettes that has reportedly reduced levels of carcinogenic nitrosamines. Nevertheless, results of a Swedish cohort study demonstrated a 1.4-fold increased risk of noncardia gastric cancer among regular snuff users. Snuff exposure also increases the rate of gastric carcinogenesis in Hp -infected mice.
Most epidemiologic studies have failed to demonstrate an association between alcohol consumption and cardia or noncardia gastric cancer. , , However, several meta-analyses suggest a small but significant association between heavy alcohol use and gastric cancer risk (RR, 1.16-1.87 in selected subgroups). Although some of these analyses suggest that the risk is higher for junctional cancers than more distal gastric cancer, some authors state the opposite effect. Overall, the risk increase seems to be moderate and influenced by multiple factors (including tobacco consumption and physical activity). Interestingly, alcohol intake may increase the risk of gastric cancer in patients with certain polymorphisms of the alcohol dehydrogenase gene.
Obesity is a recognized risk factor for numerous GI malignancies. Increased BMI is associated with a mild to moderate increased risk of gastric cardia cancer, but not noncardia cancer. Results of the National Institutes of Health-American Association of Retired Persons (NIH-AARP) Diet and Health Cohort Study demonstrated that morbid obesity (defined as a BMI ≥35) as well as large waist circumference were associated with a 2- to 3-fold increased risk of gastric cardia cancer, but not noncardia cancer. A separate cohort study from the Netherlands also found an increased risk of cardia cancer with increasing BMI. In a recent analysis of the EPIC cohort data from 391,456 individuals with 124 incident esophageal and gastric adenocarcinomas, neither 193 cardia cancers nor 224 noncardia gastric cancers were associated with BMI. The possible association between obesity and cardia cancer risk is likely mediated by proinflammatory cytokines and adipokines produced by intra-abdominal visceral fat.
As is true for most malignancies, both genetic and environmental factors play important roles in the pathogenesis of gastric cancer. Generally, intestinal-type gastric cancer is considered largely due to environmental causes (i.e., Hp infection), whereas diffuse gastric cancer is considered a primarily genetic malignancy. In the case of intestinal-type gastric cancer, however, assigning relative values to environmental and genetic contributions is complex, given that the major environmental factor, Hp , also tends to exhibit familial clustering. Nevertheless, in the future, gastric cancer types might rather be classified by genetic alterations and grouped to molecular subgroups with distinct carcinogenic mechanisms as well as clinical behavior, than to a histologic phenotype.
Overall, 10% of cases of gastric cancer appear to exhibit familial clustering, and family history is likely an independent risk factor even after controlling for Hp status. , In a cohort study of relatives of patients with gastric cancer, siblings had a 2-fold increased risk of gastric cancer, adjusted for Hp infection. In a case-control study from Japan, a positive family history was associated with a significantly increased odds of gastric cancer in women (OR, 5.10), but not in men. A study from Scandinavia showed that having a twin with gastric cancer conferred a markedly higher relative risk for the disease (RR, 9.9 for monozygotic twins and 6.6 for dizygotic twins), leading the researchers to calculate that heritable factors accounted for 28% of gastric cancers, compared with 10% for shared environmental factors and 62% for nonshared environmental factors. A recent meta-analysis of 32 studies including more than 80,000 individuals reported an increased pooled relative risk of 2.35 (95% CI: 1.96 to 2.81) for subjects with a positive family history of gastric cancer; the risk was even higher (RR 2.71; 95% CI: 2.08 to 3.53) if first-degree relatives had a gastric cancer diagnosis.
Some of the familial clustering seen with intestinal-type gastric cancer may be related to genetic factors that play a role in the host immune response to Hp infection. Data from South Korea indicate that individuals with a family history of gastric cancer more frequently have both Hp infection and associated atrophic gastritis or intestinal metaplasia. In a case-control study from Scotland, relatives of patients with gastric cancer had a higher prevalence of atrophy and hypochlorhydria, but a similar prevalence of Hp infection, compared with controls. The greater prevalence of atrophy was confined to those patients with Hp infection, suggesting the possibility these individuals were perhaps exhibiting a more vigorous immune response to Hp . In a number of model systems, the development of gastric atrophy has been linked to a strong Th1 immune response. , , Thus, it was postulated that candidate disease-susceptibility genes for gastric atrophy and cancer might be genes that participate in the innate and adaptive immune responses to Hp infection. Inflammation is modulated by an array of pro- and anti-inflammatory cytokines, and several genetic polymorphisms have been described that influence cytokine response. With the recently started next-generation sequencing approaches, we may be able to determine whether families with increased gastric cancer incidence have a genetic predisposition for a more carcinogenic immune response.
One such factor is IL-1β, an important proinflammatory cytokine and a powerful inhibitor of acid secretion. Indeed, there is an association between proinflammatory IL-1 gene cluster polymorphisms ( IL-1B encoding IL-1β, and IL-1RN encoding its naturally occurring receptor antagonist, IL-1RA) and neoplastic progression in the setting of Hp infection. Individuals with the IL-1β -31 ∗ C or -511 ∗ T and IL-1RN ∗ 2/ ∗ 2 genotypes were shown in the study to be at higher risk for developing Hp -dependent hypochlorhydria and gastric cancer. The increased risk of progression to cancer with these genotypes was in the 2- to 3-fold range compared with noninflammatory genotypes. The initial report was confirmed in other studies. Subsequently, Hwang and colleagues demonstrated that carriers of the IL-1B-511T/T genotype or the IL-1RN ∗ 2 allele had higher mucosal IL-1β levels than noncarriers and also confirmed the association between the -511T/T genotype and severe gastric inflammation and atrophy. The importance of IL-1β carcinogenesis has now been demonstrated in a transgenic study, where stomach-specific expression of human IL-1β in transgenic mice led to spontaneous gastric inflammation and cancer that correlated with early recruitment of myeloid-derived suppressor cells to the stomach. Of note, in a mouse model of Barrett esophagus and esophageal and EGJ tumors, IL-1β expression in the esophageal squamous epithelium also led to esophagitis and expansion of cardia stem cells forming gastroesophageal tumors, supporting the hypothesis that Barrett’s-associated adenocarcinoma arises from the gastric cardia resembling a gastric cancer phenotype.10
Additional associations with gastric cancer risk have been reported for genetic polymorphisms in TNF-α and IL-10. Proinflammatory genotypes of TNF-α and IL-10 were each associated with a 2-fold higher risk of noncardia gastric cancer. When combined with proinflammatory genotypes of IL-1B and IL-1RN , patients with 3 or 4 high-risk genotypes showed a 27-fold greater risk of gastric cancer. Additional studies have shown that polymorphisms of the Toll-like receptor-4 (TLR-4) gene also increases the risk of gastric cancer. Carriers of the TLR4+ 896G polymorphism had an 11-fold increased odds ratio for hypochlorhydria, and significantly more severe gastric atrophy and inflammation. Accumulated evidence suggests that the genetic predisposition to gastric cancer may be largely determined by the TLR and cytokine responses to chronic Helicobacter infection. Polymorphisms in the TLR1 seem to protect against gastric cancer development (OR 0.4; 95% CI: 0.22 to 0.72) and are furthermore related to alterations of downstream cytokine signaling. ,
The best described form of hereditary gastric cancer is the diffuse type gastric cancer that is seen in the presence of a germline mutation in the gene CDH1 , which encodes the cell adhesion molecule E-cadherin. A large New Zealand kindred was found to have a germline mutation in the E-cadherin gene, and similar mutations have been reported in several additional kindreds, all with diffuse-type gastric cancer. The age of onset of gastric cancer in individuals with CDH1 mutations is less than 40 years but can be highly variable, and the estimated lifetime risk of gastric cancer is close to 70%. , Germline CDH1 mutations are also associated with familial lobular breast cancer. ,
A small part of the familial clustering of gastric cancer can be attributed to other cancer syndromes. Patients with familial adenomatous polyposis have a prevalence of gastric adenomas ranging from 35% to 100%, and their risk of gastric cancer is close to 10-fold higher than that of the general population. Dysplastic lesions in this specific group of patients frequently arise from fundic gland polyps ( ) and develop at an early age. , Fundic gland polyps do not otherwise tend to progress toward dysplasia. Patients with hereditary nonpolyposis colorectal cancer (HNPCC) syndrome have an approximately 11% risk of developing gastric cancer, predominantly of the intestinal type, with a mean age at diagnosis of 56 years. Patients with juvenile polyposis also have a 12% to 20% incidence of gastric cancer. ,
Video 54.2 EGD of multiple fundic gland polyps in FAP.
In addition to the germline genetic alterations described earlier, next-generation sequencing techniques such as exome sequencing have led to the detection of new molecules and mechanisms that are involved in gastric carcinogenesis. In 8% to 10% of the gastric cancer patients, somatic mutations were identified in the ARID1A gene (also called BAF250a, SMARCF1 , or OSA1 ), an accessory subunit of the SWI-SNF chromatin remodeling complex that is involved in processes of DNA repair, differentiation, and development. Notably, cancers with EBV infection showed mutations of ARID1A in 73% of the cases. Additionally, ARID1A mutations were negatively associated with mutations in TP53 and occurred together with PIK3CA mutations. Patients with ARID1A alterations had longer recurrence-free survival, suggesting that these cancers belong to a molecular subgroup with distinct carcinogenic mechanisms as well as clinical behavior. Analysis of somatic copy number aberrations have additionally shown significantly amplified genes, including therapeutically targetable kinases such as ERBB2 , FGFR1 , FGFR2 , EGFR , and MET in gastric and gastroesophageal cancers.
Although atrophy and intestinal metaplasia correlate with gastric cancer risk, direct cell progression through these stages has not been conclusively shown. Indeed, gastric cancer most likely arises from stem or progenitor cells present within the gastric mucosa rather than directly from terminally differentiated metaplastic cells. Investigators have for several decades sought to unravel the mutations responsible for gastric cancer initiation and progression to uncover a logical progression of acquired mutations akin to what is seen in colorectal cancer. However, gastric cancer does not follow a pattern like colorectal carcinoma progression, there is no clear-cut linear sequence of mutations in gastric cancers, and there is an even greater heterogeneity in genetic alterations.
Although initial studies on large high throughput data focused mainly on transcriptome analysis, the advent of more advanced genomic sequencing enables genome-wide analyses of the mutational landscape of gastric cancer. The combined efforts of multiple research groups in international consortia enabled a more comprehensive integrative analysis of multilevel omics -data from large cohorts. , , TCGA consortium presented comprehensive data on 5 different platforms for about 300 gastric cancers. They demonstrated a good correlation of clustering between different levels of genomic data as well as epigenetic changes and transcriptome and even proteome analyses. Based on their cluster analysis, the authors suggested a 4-group classification of gastric cancers, with the first group (EBV) being related to EBV infection, showing a dominant epigenetic hypermethylation profile (EBV). The second group (MSI) was positive for microsatellite instability (MSI), similar to the MSI subgroup of colorectal cancers. The remaining tumors were divided into a group (GS) with a low mutation rate and low frequency for copy number aberrations, called the “genomically stable” subtype and a group (CIN) with high mutation rates and further related genomic changes, called the “chromosomally instable” type. It is of note the GS group comprised predominantly diffuse-type tumors, whereas CIN tumors represented more intestinal-type cancers. There were some differences in the distribution of these subtypes with regard to location, with CIN tumors showing higher proportions with more proximal location, which was later also confirmed for esophageal adenocarcinomas.9 The clinical relevance of this classification was suggested by the pattern of dominant changes in genes encoding for signaling pathways utilized in targeted treatment approaches. The Asian Cancer Research Group followed a similar approach in their cohort of 3000 gastric cancers, although putting more focus on transcriptome data than the TCGA did.13 Thus, Cristescu et al. presented a similar 4-group classification, also reporting properties as prognostic predictors for their groups. This classification was validated in several independent cohorts, including the group of TCGA patients. The actual genomic changes that have been described in these comprehensive studies are in line with results that have been published previously.
Aneuploidy is common in gastric cancer (60% to 75%), but cytogenetic studies have failed to identify any consistent chromosomal abnormality. Comparative genomic hybridization studies have shown that chromosome arms 4q, 5q, 9p, 17p, and 18q exhibit frequent decreases in DNA copy number, whereas chromosomes 8q, 17q, and 20q often have increased DNA copy number.
There is a general consensus that TP53 is the most commonly mutated gene in gastric cancer (60% to 70% of gastric cancers) and that mutations in Ras , APC , and Myc are rare. , Loss of heterozygosity at the APC locus occurs more commonly. Another genetic abnormality found at high frequency (≈60%) is the deletion or suppression of the fragile histidine triad gene (FHIT) , a tumor suppressor locus on chromosome 3p. Genes that inhibit entry into the cell cycle, such as p16 and p27 , show diminished expression in nearly one half of gastric cancers. Absence of p27 expression is associated with a poorer prognosis. , Absence of p16 expression is seen most commonly in poorly differentiated carcinomas but has no measurable impact on prognosis. Diminished expression of p16 and p27 occurs in the absence of detectable mutations and is believed to be secondary to hypermethylation. Many of these cancers show hypermethylation of a number of promoter regions, including the MLH1 promoter region, and show the high-level microsatellite instability (MSI-H) phenotype (see Chapter 1 ). Multiple tumor suppressor genes have been shown to be methylated in gastric cancers. Emerging evidence suggests that these epigenetic changes, including global hypomethylation and promoter hypermethylation, occurs quite early in gastric carcinogenesis. In addition, it appears that DNA methylation changes also occur in the tumor-associated stromal fibroblasts, suggesting an important role for the tumor microenvironment.
Overexpressions or amplifications of a number of growth factor pathways has been described, including COX-2 (70%), hepatocyte growth factor/scatter factor ( HGF/SF ) (60%), vascular endothelial growth factor ( VEGF ) (50%), c-met (50%), amplified in breast cancer-1 ( AIB-1 ) (40%), and β-catenin (25%) ( Table 54.2 ). Approximately 15% of gastric cancers have been reported to overexpress both EGF and EGF receptor (EGFR), consistent with an autocrine mechanism. Mutations in PIC3A , a gene that codes for a catalytic subunit of phosphatidylinositol 3-kinase (PI3K), has been found in up to 25% of gastric cancers analyzed. In addition, mutations in genes encoding human protein tyrosine phosphatases (PTPs) were found by the same laboratory in 17% of gastric cancers, with the protein tyrosinase phosphatase receptor type the most frequently altered.
Abnormalities | Approximate Gene Frequency (%) |
---|---|
Microsatellite instability | 15-50 |
DNA aneuploidy | 60-75 |
Deletion/Suppression | |
p53 | 60-70 |
FHIT (fragile histidine triad gene) | 60 |
APC (adenomatous polyposis coli gene) loss of heterozygosity | 50 |
DCC (deleted in colorectal cancer gene) loss of heterozygosity | 50 |
Decreased Expression Due to Hypermethylation | |
p16 | ≈50 |
TFF1 (human trefoil factor 1 gene) | ≈50 |
p27 | <50 |
MLH1 (human mutL homolog 1 gene) | 15-20 |
E-cadherin | 50 |
Amplification/Overexpression | |
COX-2 | 70 |
HGF (hepatocyte growth factor) | 60 |
VEGF (vascular endothelial growth factor) | 50 |
c-met | 50 |
AIB-1 (amplified in breast cancer-1) | 40 |
Beta-catenin | 25 |
EGFR (EGF receptor gene) | 15 |
Mutations | |
PI3K (phosphotidylinositol 3-kinase gene) | 25 |
PTPRT (protein-tyrosine phosphatase receptor type gene) | 17 |
Gastric-specific tumor suppressor genes TFF1 (Trefoil factor 1) and RUNX3 (Runt-related transcription factor 3), which have now been identified and may represent “gatekeepers” of the gastric cancer pathway, are logical targets for further study. , Loss of TFF1 has been described in around 50% of gastric carcinomas, and TFF1 knockout mice develop spontaneous gastric antral tumors. Mutations of TFF1 have also been described, and these enhance gastric cancer cell invasion through signaling pathways that include PI3-kinase and phospholipase-C. TFF1 expression is repressed by STAT-3, and activation of STAT-3 is also emerging as a key pathway that leads to gastric cancer.54 RUNX3 most likely suppresses gastric epithelial growth by inducing p21 and Bim, attenuates Wnt signaling, and is altered in 82% of gastric cancers. Investigations into these genes and their contributions to the gastric cancer phenotype will prove valuable to our understanding of disease progression.
MSI in dinucleotide repeats secondary to defects in DNA mismatch repair genes, such as MLH1 and MLH2 (mutL homologs 1 and 2), have been mainly implicated in the development of colorectal cancer, and in particular the HNPCC syndrome. Patients with HNPCC have an 11% incidence of gastric cancer, suggesting that MSI may also play a role in the development of gastric cancer. MSI is found in 15% to 50% of sporadic gastric cancers, with a higher prevalence in intestinal type of cancers. Low-level microsatellite activity (e.g., MSI-low) can be found in 40% of areas of intestinal metaplasia in patients with gastric cancer and in 14% to 20% of adenomatous polyps. , , MSI-H occurs in only 10% to 16% of gastric cancers. MSI is associated with the less frequent occurrence of TP53 mutations, well-to-moderately well−differentiated histology, and distal location in the stomach. Studies that have examined the effect of MSI on patient survival have shown inconsistent results. , When the findings are taken together, it would appear that MSI does play a role in the pathogenesis of gastric cancer, likely before the development of intestinal metaplasia (see Fig. 54.3 ), and is most commonly due to methylation of the MLH1 promoter.
There are increasing data regarding the genetics of diffuse-type gastric cancer. , Several families with hereditary diffuse gastric cancer have been found to carry a germline mutation in the E-cadherin gene ( CDH1 ). , , However, mutations in CDH1 have also been described as a dominant feature in sporadic diffuse-type gastric cancer. Further evidence supporting a role for E-cadherin in the pathogenesis of gastric cancer comes from studies showing that suppression of E-cadherin expression occurs in 51% of gastric cancers, with a higher percentage found in diffuse-type cancers. Furthermore, E-cadherin under-expression is associated with higher rates of lymph node metastases and reduced survival. , The overall rates of CDH1 mutations in gastric cancer are low. Thus, the decreased expression of E-cadherin seen in gastric cancer is likely secondary to hypermethylation of the CDH1 promoter, which occurs in 50% of gastric cancers and 83% of diffuse-type gastric cancers. E-cadherin is a transmembrane protein that connects to the actin cytoskeleton through α- and β-catenins to establish cell polarity and mediates homophilic cellular interactions. , Decreased expression of E-cadherin is believed to promote dissociation of cancer cells from their cell matrix, enhancing the migration and invasion of gastric cancer cells. Expression of α-catenin is also decreased or absent in 68% of gastric cancers. Therefore, E-cadherin appears to act as a tumor suppressor gene that may be important in the pathogenesis of diffuse gastric cancer. Other alterations that commonly occur in diffuse type gastric carcinoma are alterations in Wnt-related genes as well as changes in the Ras homolog gene family, Member A gene ( RHOA ), which seems to be exclusive to this histological subtype.
Perhaps as important as the genetic alterations acquired during the progression to gastric adenocarcinoma is the question, “In what target cells do these changes occur?” For a cell to accumulate the quantity of genetic changes necessary for autonomous growth, it must be long lived. For these reasons, the current thinking is that a resident tissue stem cell is the target of genetic mutations and becomes the “cancer stem cell”—capable of autonomous growth and with metastatic potential. Recently, several elegant genetic lineage-tracing studies in mice established markers that allow the distinction of 2 different types of GI stem cells. Crypt base columnar cells (CBC) are fast-cycling stem cells expressing Lgr5 and CD (Prom-1). , A villin transgene has allowed the identification of a multipotent progenitor located in the lower third of a subset of antral gastric glands, whereas multiple intestinal stem cell markers could also be identified in the antrum. Interestingly, Lgr5 shows lineage labeling in some antral gastric glands and in the gastric cardia. Slower cycling cells, which are usually found at the +4 position of the crypts of the antrum (i.e., the fourth epithelial cell in the crypt, counting from the bottom of the crypt upward), are characterized by a pronounced expression of Bmi1 and Tert. , Although these 2 types of cells are functionally interconnected, their exact hierarchical relationship remains to be identified. Sigal and colleagues demonstrated a direct CagA-dependent activation of Lgr5-positive gastric stem cells, thus reporting a further mechanism by which H. pylori infection induces gastric carcinogenesis. The same group also suggested orchestration of epithelial hyperproliferation and gland hyperplasia as a response to the infection by cells of the stromal compartment, mainly myofibroblasts involving Hp -induced Wnt signaling.
In the gastric oxyntic glands, the proliferative zone with the gastric stem cell has been localized to the isthmus, the middle portion of the tubule, and cells are thought to migrate bidirectionally to supply gastric surface mucus cells that coat the gastric pits, and gastric parietal and zymogenic cells that comprise the base of the gland. The gastric corpus stem cell has not yet been identified; none of the markers discussed earlier labels any specific cells within the gastric isthmus. Recently, progenitor cells (e.g., Krt19+ and TFF2+ cells) have been shown through lineage tracing studies to label different gastric progenitor cells. , Typically, columnar metaplasia is positive for TFF2 and Krt19. Given that intestinal metaplasia arises in the gastric mucosa and in the esophagus, it is plausible that a similar stem cell gives rise to both. Regardless of their localization (CBC or +4 position) or their function, GI stem cells depend on signals from the stem cell niche, such as pericryptal myofibroblasts and neighboring differentiated epithelia. Important signaling pathways required for stem cell maintenance and proliferation comprise the Wnt, Notch, bone morphogenetic proteins, and Hedgehog pathways.
There is increasing knowledge on the interaction of the local stromal microenvironment with the epithelium. A regulator of the biological behavior of gastric cancer cells are cancer-associated fibroblasts, that have been shown to modify TGFβ-dependent signaling, increasing cellular motility, and, therefore, invasiveness. , The density of tumor-infiltrating lymphocytes and their vicinity also modifies the tumor’s aggressiveness and therefore has an impact on the prognostic outcome in patients with adenocarcinomas in the stomach or at the EGJ. , This factor is partly reflected by the level of systemic inflammation. Systemic inflammation can be partly mediated by the visceral adipose tissue and it has been demonstrated that omental adipocytes enhance the invasiveness of gastric cancer by activation of PI3K-Akt signaling in the tumor cells.
Besides the valuable data from in vitro and ex vivo models, approaches for “virtual microdissection” of next generation high throughput and sequencing data are promising to enhance our understanding of the network of different cellular components and its impact on tumor initiation, promotion, and progression (including invasive and metastasizing behavior) ( Fig. 54.4 ).
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here