Abstract
Currently being the seventh most prevalent form of cancer worldwide, and the fifth most common cause of cancer-related death, based on GLOBOCAN 2020 data, gastric cancer is still an important public health problem, despite its dropping incidence. Regions around the world are still at high-risk, mostly in populations with a high prevalence of Helicobacter pylori infection or a carcinogenic favorable diet. Gastric cardia cancer incidence is on the rise in some areas. Great steps were made in the last decades in understanding the pathogenesis of gastric cancer and its risk factors. Host genetic polymorphisms play a quintessential role in disease outcome. Helicobacter pylori eradication and endoscopic surveillance are the most effective options to further decrease gastric cancer incidence. Surgery is required for a curative treatment in most cases.
This review summarizes the latest worldwide epidemiological data of gastric cancer and aims to provide an accessible and credible source of evidence for physicians who assess risk factors for gastric cancer.
Keywords: epidemiology, prevalence, mortality, gastric cancer
Introduction
Gastric cancer (GC) is the 7th most common type of cancer worldwide, approximately 1.9 million patients being currently diagnosed with this disease, 3.6% of all cancer diagnoses, and the 4th most common cause of cancer deaths globally. A total of 1,089,103 new cases of GC were reported in 2020 (Table I) [1].
Table I.
Worldwide 5-year incidence and prevalence of cancers, both sexes, all ages [1].
| Cancer | Incidence (No of cases/5 years) | Prevalence (No of cases/5 years) |
|---|---|---|
| Breast | 2 261 419 | 7 790 717 |
| Non-melanoma skin cancer | 1 198 073 | 6 458 885 |
| Colorectum | 1 931 590 | 5 253 335 |
| Prostate | 1 414 259 | 4 956 901 |
| Lung | 2 206 771 | 2 604 791 |
| Thyroid | 586 202 | 1 984 927 |
| Stomach | 1 089 103 | 1 805 968 |
| Cervix uteri | 604 127 | 1 495 211 |
| Liver | 905 677 | 994 539 |
| Oesophagus | 604 100 | 666 388 |
The stomach is the most dilated part of the digestive system and it is composed by 4 main parts: cardia, fundus, body and the pylorus. Its primary functions are the temporary storage of food and the initiation of food digestion [2]. The gastric mucosa is primarily formed of parietal cells, chief cells, mucus cells and neuroendocrine cells [3]. The gastric mucosal turnover is a dynamic process, defined by continuous cell proliferation which is counterbalanced by apoptosis. The cell’s turnover times vary from 3 to 60 days [4]. Having this high turnover rate, the gastric epithelial cells are prone to mutations because they are rapidly dividing. Both environmental and genetic factors influence the cancer development process.
Although GC was the leading cause of cancer deaths until the mid 1990s, its mortality and incidence have considerably declined around the world. Many reasons were linked to this decline including the introduction of commercially available refrigeration which by itself reduced other factors that influence the development of GC including the consumption of smoked and salted preserved foods and increasing the consumption of fresh fruits and vegetables, a greater use of antibiotics which reduced the Helicobacter pylori (HP) infection, and a drop in smoking incidence in the western countries [5]. HP is considered the most important factor in the development of GC, but the GC pathogenesis cannot be induced by HP infection alone, because only 2–5% of people with this infection develop GC [6].
World Health Organization guidelines of GC include the following subtypes, adenocarcinoma, signet ring-cell carcinoma and undifferentiated carcinoma.
The more commonly used classification of GC is based on Lauren’s histo-clinical classification, which include the intestinal type, diffuse and mixed. More commonly found is the intestinal type, named because of the similar etiopathogenesis with the other digestive tract adenocarcinomas. It is more frequently seen in high-risk areas, males and older patients. The less common type is the diffuse type, defined by the loss of intracellular adhesions, which leads to a lack of glandular structures. It is more common in younger patients and has a worse prognosis [7–8]. The worldwide decline of the GC overall was counterbalanced by a rise in the incidence of cancer of the gastric cardia [8].
Epidemiology
Approximately one million patients are diagnosed with GC each year around the globe. GC is the 7th most commonly prevalent cancer and the 6th most commonly diagnosed in the world [1]. Countries with the highest incidence rates (>15 new cases/100000 patients) include Mongolia, Japan, Republic of Korea, Tajikistan, Kyrgyzstan etc (Table II). Higher than average rates countries include Eastern-European countries, Turkey, Mali, Portugal, Peru, Ecuador and Columbia. Lower rates (<5 new cases/100000 patients) are seen in western counties including France, United States, Canada, United Kingdom, Norway, Sweden, Australia and also in Central-African countries, although the latter conclusion can be partially attributed to the lower accessibility to medical care (Table III) [1]. 66% of the new cases of GC diagnosed in 2020 were in men. The men to women ratio was 2:1. No country currently has a higher of GC in women than in men and also no country currently has an increasing trend in incidence [1,9].
Table II.
Estimated age-standardized highest incidence rates in 2020, both sexes, all ages [1].
| Population | Value (per 100.000) |
|---|---|
| Mongolia | 32.5 |
| Japan | 31.6 |
| Republic of Korea | 27.9 |
| Tajikistan | 23.4 |
| China | 20.6 |
| Kyrgyzstan | 19.7 |
| Cabo Verde | 18.4 |
| Bhutan | 17.7 |
| Islamic Republic of Iran | 17.5 |
| Vietnam | 15.5 |
Table III.
Estimated number of prevalent cases in all continents in 2020 [1].
| Populations | 5-year | Proportions (per 100000) |
|---|---|---|
| Asia | 1397478 | 30.1 |
| Europe | 213013 | 28.4 |
| Africa | 44194 | 3.3 |
| Northern America | 50387 | 13.7 |
| Oceania | 5389 | 12.6 |
| Latin America and the Caribbean | 95507 | 14.6 |
The cumulative risk of GC is highest in Eastern Asia (2.64%) and lowest in southern Africa (0.42%) [9].
High-HDI (Human Development Index) countries have a 2 to 3-fold increase in average incidence rates when compared to low and middle HID countries. Families with high-middle income are currently at the highest risk of developing GC, most probably correlated with the western diet [1].
GC is also the 4th most common cause of cancer death worldwide, accounting for 783,793 deaths in 2020 alone [1], out of which 502,788 were in men. Asia had the highest mortality in GC related death (575,206 people), followed by Europe (96,997 people) and Latin America and Caribbean (53,392 people) [1]. The annual percent change (APC) in gastric cancer mortality decreased between 3 to 4 percent in European counties, Korea, Japan and Australia between 1980 to 2005 [10]. In Latin America the APC was lower but still constant: Brazil and Chile (−1.6%); Argentina and Mexico (−2.3%). Currently, the only reported country to have an increasing trend in mortality in men is Thailand (AAPC, 3.92; 95% CI, 2.14–5.74; P = .001) [9].
In the United States, the 5-year survival rate for GC is 31%. Most diagnosed cases are already metastatic, which lowers survival rates. When the diagnosis is made in the pre-metastatic phase, the 5-year survival rate rises to 67% [11]. Asian patients tend to have a better prognosis than Caucasians, having higher survival rates [12]. UK’s 5-year survival rates currently stands at 20.8%, while Europe’s is slightly higher, at 26% [13].
The 5-year survival rates of gastric cancer according to TNM staging system are mentioned in table IV.
Table IV.
5-year survival rates of GC – TNM staging system [14].
| AJCC TNM staging system of GC- 8th edition | 5-year overall survival rates |
|---|---|
| IA | 94.7% |
| IB | 89.9% |
| IIA | 80.7% |
| IIB | 72.5% |
| IIIA | 58.4% |
| IIIB | 40.8% |
| IIIC | 20.2% |
| IV | 8.8% |
Risk factors
Risk factors can be further classified as exogenous and endogenous. Due to the fact that the pathogenesis of GC is a multifactorial and multi-step process, there is a considerable interdependence of these risk factors for gastric cancer (Table V).
Table V.
Risk factors – GC.
| Exogenous | Endogenous | |
|---|---|---|
| HP infection | Preexistent gastric disease | Hereditary factors |
| Nitroso-compound rich diet | Chronic atrophic gastritis | Family history of GC |
| Smoking | Ménétrier disease | ‘A’ Blood group |
| Epstein-Barr virus infection | Gastric ulcer | Lynch Syndrome |
| Socioeconomic status | Subtotal gastrectomy | |
| Obesity | Gastric adenomatous polyps | |
| Gastroesophageal Reflux Disease | ||
1. Genetics
The TP53 gene is currently described as the highest mutated gene in GC, over 50% of cases presenting abnormalities. Its role as a tumor suppressor gene is well-documented [15]. Important pathogenically mutations have also been observed in KRAS, CTNBB1, PIK3CA oncogenes and in SMAD4, APC tumor suppresor genes.
Somatic copy number alterations (SCNAs) is a major mechanism of activating oncogenes or inactivating tumor suppressor genes and is seen in GC as well. RTK/RAS/MAPK signaling, including HER2, EGFR, MET, FGFR2 şi RAS, is a pathway altered in 30–40% of cases [16,17]. CCND1, CCNE1 şi CDK6 are genes that regulate cell-cycle and can be amplyfied in GC [18].
DNA methylation is another pathogenical factor that can induce genetic alterations. Mehylation of CDH1, RUNX3, p16, and hMLH1 is described in GC [19,20]. It was also confirmed that the way that Epstein-Barr virus induces GC is by DNA methylation [21].
Recent studies described mutations in the ARID1A tumor suppressor gene, which encodes SW1/SNF chromatin remodeling complex [22]. IL-17 and IL-10 mutations were described in Asian populations.
2. Preexisting conditions
Chronic gastric atrophy and intestinal metaplasia are considered as the most relevant processes in GC pathogenesis. Mucosal atrophy is characterized by the loss of the glandular elements, thus being replaced by areas of metaplasia and areas of fibrosis. Intestinal metaplasia is considered a precancerous condition, with an abundance of goblet and mucosal, similar to those found in the intestinal mucosa. The chronic inflammation induces the transcription factor NF-κB, one of the key mediators of inflammation. It also induces oxidative stress, reactive oxygen species and nitrosamines being generated by leukocytes and macrophages [23–26].
Ménétrier disease is a hypertrophic gastropathy, characterized by massive growth of mucous cells in the gastric mucosa. While the cause is currently not well-understood, it is linked with a 10% risk of malignant transformation [27–29].
Gastric stump cancer is defined as a primary GC that arises at a minimum of five years after a partial gastrectomy. The five-year mark was appointed in case an inappropriate initial diagnosis was made. Post-procedural hypochlorhydria causes bacterial overgrowth which leads to the accumulation of nitroso compounds. A 1.66-fold increase of the risk of developing GC compared to general population has been attributed to gastric stump cancer. Clinical and endoscopic surveillance are the best strategies for prevention and early detection [30].
Gastric polyps are most-often incidental findings during upper G.I. tract endoscopy. Histology is required to exclude malignancy. Hyperplastic polyps have malignant potential through a dysplasia/carcinoma sequence and the risk of malignancy is increased in polyps >1cm and pedunculated. An estimate of 8–59% of gastric adenomas occur at the same time with GC. Size, villous contour and degree of dysplasia are associated with the presence of invasive carcinoma [31,32].
3. Helicobacter pylori infection
In 1995, HP infection was classified by the International Agency for Research on Cancer as a group 1 carcinogen. Although the causality is certain, there are important interpersonal variation in the course of infection.
The complex interaction between environmental, genetic and bacterial factors suggests an unpredictable evolution [33,34]. The classical carcinogenesis sequence – superficial gastritis, chronic atrophic gastritis, intestinal metaplasia, dysplasia, carcinoma – is pathognomonic for the intestinal type. The diffuse subtype is also linked with GC [35].
Neutrophil migration is triggered by HP. One at the mucosal site, the neutrophils induce the nitric oxide synthase and reactive oxygen species which in turn leads to DNA damage [36]. Severe DNA damage triggers apoptosis, represented as gastric atrophy. It was shown that treating the infection normalizes the rate of apoptosis [36,37].
HP eradication reduces GC risk. A meta-analysis of 27 studies, with 48,606 HP positive patients, of which 715 developed GC, has shown that patients that were cured of HP infection had lower incidence of GC [38–40]. Gastric cancer prevention can be achieved by H. pylori screen-and-treat strategies with selection of the most effective timing for intervention [40]. No consensus has yet been reached regarding the population screening. Some guidelines suggest that it may be beneficial in populations that have an increased risk.
4. Diet
Excess salt was proven to have a direct carcinogenic effect in GC. There is also synergistic effect of salt consumption and HP infection in GC patients. The decline in GC incidence is attributed to the changes made in food preservation in the last 50 years, commercially available refrigeration replacing salt as the main preserving method [41–44].
Nitrates are chemical compounds which contain the -NO group and are not only taken from diet and smoking, but also endogenous sources. Processed meats and dairy products are rich in nitrates. Once absorbed in the stomach, the nitrates react with amines, amides or aminoacids to form N-Nitro compounds. High gastric N-Nitro levels were associated with the presence of advanced premalignant lesions. Processed meat was classified in 2015 by the World Health Organization as a group 1 carcinogen [43,45–47].
Multiple studies have showed the association between smoking and GC. A meta-analysis showed a 1.53-fold increased risk of GC in smokers compared to non-smokers. The European EPIC study showed a similar risk, which decreases after 10 years of quitting. Currently, approximately 18% of GC cases are linked with smoking [48].
Non-steroidal anti-inflammatory drugs were proven to decrease the risk of GC. The effect is even more favorable in HP positive patients [49,50].
Conclusions
Although a significant decrease in GC incidence, prevalence and mortality in GC patients was seen in the last thirty years, gastric cancer still remains an important public health problem. This process is the final consequence of a complex interaction between environmental, hereditary and host factors.
HP infection and the western diet are important risk factors, especially for non-cardia cancers, that are difficult to control at a global scale. Smoking is also important in carcinogenesis. Host genetic polymorphisms are quintessential in the pathogenic process.
Management of HP infection should be prioritized in every patient with a positive test. Endoscopic surveillance in patients with chronic atrophic gastritis and intestinal metaplasia would increase survival of patients if early gastric cancer is discovered. Structuring a genetic carcinogenic sequence may be the key to discovering a gene therapy.
References
- 1.International Agency for the Research on Cancer (WHO) Global Cancer Observatory - Globocan. Available from: http://globocan.iarc.fr/Default.aspx)
- 2.Chaudhry SR, Liman MNP, Peterson DC. StatPearls [Internet] Treasure Island (FL): StatPearls Publishing; 2022. Anatomy, Abdomen and Pelvis, Stomach. [PubMed] [Google Scholar]
- 3.Hsu M, Safadi AO, Lui F. StatPearls [Internet] Treasure Island (FL): StatPearls Publishing; 2022. Physiology, Stomach. [PubMed] [Google Scholar]
- 4.Karam SM. Lineage commitment and maturation of epithelial cells in the gut. Front Biosci. 1999;4:D286–D298. doi: 10.2741/karam. [DOI] [PubMed] [Google Scholar]
- 5.Roder DM. The epidemiology of gastric cancer. Gastric Cancer. 2002;5(Suppl 1):5–11. doi: 10.1007/s10120-002-0203-6. [DOI] [PubMed] [Google Scholar]
- 6.Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S, Yamakido M, et al. Helicobacter pylori infection and the development of gastric cancer. N Engl J Med. 2001;345:784–789. doi: 10.1056/NEJMoa001999. [DOI] [PubMed] [Google Scholar]
- 7.Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965;64:31–49. doi: 10.1111/apm.1965.64.1.31. [DOI] [PubMed] [Google Scholar]
- 8.Chan AOO, Wong B. Epidemiology of gastric cancer. In: Post TW, editor. UpToDate. UpToDate; Waltham, MA: [Google Scholar]
- 9.Wong MCS, Huang J, Chan PSF, Choi P, Lao XQ, Chan SM, et al. Global Incidence and Mortality of Gastric Cancer, 1980–2018. JAMA Netw Open. 2021;4:e2118457. doi: 10.1001/jamanetworkopen.2021.18457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bertuccio P, Chatenoud L, Levi F, Praud D, Ferlay J, Negri E, et al. Recent patterns in gastric cancer: a global overview. Int J Cancer. 2009;125:666–673. doi: 10.1002/ijc.24290. [DOI] [PubMed] [Google Scholar]
- 11.Howlader NA, Krapcho M, Miller D, et al. SEER Cancer Statistics Review, 1975–2014. National Cancer Institute; Bethesda, MD: based on November 2016 SEER data submission, posted to the SEER web site, April 2017. Available from: https://seer.cancer.gov/csr/1975_2014/ [Google Scholar]
- 12.Wang J, Sun Y, Bertagnolli MM. Comparison of gastric cancer survival between Caucasian and Asian patients treated in the United States: results from the Surveillance Epidemiology and End Results (SEER) database. Ann Surg Oncol. 2015;22:2965–2971. doi: 10.1245/s10434-015-4388-4. [DOI] [PubMed] [Google Scholar]
- 13.Cancer Research UK. Available fromL https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/stomach-cancer/survival.
- 14.Kim SG, Seo HS, Lee HH, Song KY, Park CH. Comparison of the Differences in Survival Rates between the 7th and 8th Editions of the AJCC TNM Staging System for Gastric Adenocarcinoma: a Single-Institution Study of 5,507 Patients in Korea. J Gastric Cancer. 2017;17:212–219. doi: 10.5230/jgc.2017.17.e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lei Z, Tan IB, Das K, Deng N, Zouridis H, Pattison S, et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology. 2013;145:554–565. doi: 10.1053/j.gastro.2013.05.010. [DOI] [PubMed] [Google Scholar]
- 16.Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, 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. doi: 10.1136/gutjnl-2011-301839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dulak AM, Schumacher SE, van Lieshout J, Imamura Y, Fox C, Shim B, et al. Gastrointestinal adenocarcinomas of the esophagus, stomach, and colon exhibit distinct patterns of genome instability and oncogenesis. Cancer Res. 2012;72:4383–4393. doi: 10.1158/0008-5472.CAN-11-3893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Scaltriti M, Eichhorn PJ, Cortés J, Prudkin L, Aura C, Jiménez J, et al. Cyclin E amplification/overexpression is a mechanism of trastuzumab resistance in HER2+ breast cancer patients. Proc Natl Acad Sci U S A. 2011;108:3761–3766. doi: 10.1073/pnas.1014835108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Nakamura J, Tanaka T, Kitajima Y, Noshiro H, Miyazaki K. Methylation-mediated gene silencing as biomarkers of gastric cancer: a review. World J Gastroenterol. 2014;20:11991–12006. doi: 10.3748/wjg.v20.i34.11991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Niwa T, Tsukamoto T, Toyoda T, Mori A, Tanaka H, Maekita T, et al. Inflammatory processes triggered by Helicobacter pylori infection cause aberrant DNA methylation in gastric epithelial cells. Cancer Res. 2010;70:1430–1440. doi: 10.1158/0008-5472.CAN-09-2755. [DOI] [PubMed] [Google Scholar]
- 21.Stanland LJ, Luftig MA. The Role of EBV-Induced Hypermethylation in Gastric Cancer Tumorigenesis. Viruses. 2020;12:1222. doi: 10.3390/v12111222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ashizawa M, Saito M, Min AKT, Ujiie D, Saito K, Sato T, et al. Prognostic role of ARID1A negative expression in gastric cancer. Sci Rep. 2019;9:6769. doi: 10.1038/s41598-019-43293-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Correa P. Human gastric carcinogenesis: a multistep and multifactorial process--First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res. 1992;52:6735–6740. [PubMed] [Google Scholar]
- 24.Eaton KA, Logan SM, Baker PE, Peterson RA, Monteiro MA, Altman E. Helicobacter pylori with a truncated lipopolysaccharide O chain fails to induce gastritis in SCID mice injected with splenocytes from wild-type C57BL/6J mice. Infect Immun. 2004;72:3925–3931. doi: 10.1128/IAI.72.7.3925-3931.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Almeida R, Silva E, Santos-Silva F, Silberg DG, Wang J, De Bolós C, et al. Expression of intestine-specific transcription factors, CDX1 and CDX2, in intestinal metaplasia and gastric carcinomas. J Pathol. 2003;199:36–40. doi: 10.1002/path.1246. [DOI] [PubMed] [Google Scholar]
- 26.Goldenring JR, Nomura S. Differentiation of the gastric mucosa III. Animal models of oxyntic atrophy and metaplasia. Am J Physiol Gastrointest Liver Physiol. 2006;291:G999–G1004. doi: 10.1152/ajpgi.00187.2006. [DOI] [PubMed] [Google Scholar]
- 27.Rich A, Toro TZ, Tanksley J, Fiske WH, Lind CD, Ayers GD, et al. Distinguishing Ménétrier’s disease from its mimics. Gut. 2010;59:1617–1624. doi: 10.1136/gut.2010.220061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Scharschmidt BF. The natural history of hypertrophic gastrophy (Ménétrier’s disease). Report of a case with 16 year follow-up and review of 120 cases from the literature. Am J Med. 1977;63:644–652. doi: 10.1016/0002-9343(77)90210-8. [DOI] [PubMed] [Google Scholar]
- 29.Musumba C, Jorgensen A, Sutton L, Van Eker D, Moorcroft J, Hopkins M, et al. The relative contribution of NSAIDs and Helicobacter pylori to the aetiology of endoscopically-diagnosed peptic ulcer disease: observations from a tertiary referral hospital in the UK between 2005 and 2010. Aliment Pharmacol Ther. 2012;36:48–56. doi: 10.1111/j.1365-2036.2012.05118.x. [DOI] [PubMed] [Google Scholar]
- 30.Tersmette AC, Offerhaus GJA, Tersmette KW, Giardiello FM, Moore GW, Tytgat GN, et al. Meta-analysis of the risk of gastric stump cancer: detection of high risk patient subsets for stomach cancer after remote partial gastrectomy for benign conditions. Cancer Res. 1990;50:6486–6489. [PubMed] [Google Scholar]
- 31.Hirota WK, Zuckerman MJ, Adler DG, Davila RE, Egan J, Leighton JA, et al. ASGE guideline: the role of endoscopy in the surveillance of premalignant conditions of the upper GI tract. Gastrointest Endosc. 2006;63:570–580. doi: 10.1016/j.gie.2006.02.004. [DOI] [PubMed] [Google Scholar]
- 32.ASGE Standards of Practice Committee. Sharaf RN, Shergill AK, Odze RD, Krinsky ML, Fukami N, et al. Endoscopic mucosal tissue sampling. Gastrointest Endosc. 2013;78:216–224. doi: 10.1016/j.gie.2013.04.167. [DOI] [PubMed] [Google Scholar]
- 33.Eslick GD, Lim LL, Byles JE, Xia HH, Talley NJ. Association of Helicobacter pylori infection with gastric carcinoma: a meta-analysis. Am J Gastroenterol. 1999;94:2373–2379. doi: 10.1111/j.1572-0241.1999.01360.x. [DOI] [PubMed] [Google Scholar]
- 34.Hansson LR, Engstrand L, Nyrén O, Lindgren A. Prevalence of Helicobacter pylori infection in subtypes of gastric cancer. Gastroenterology. 1995;109:885–888. doi: 10.1016/0016-5085(95)90398-4. [DOI] [PubMed] [Google Scholar]
- 35.Parsonnet J, Vandersteen D, Goates J, Sibley RK, Pritikin J, Chang Y. Helicobacter pylori infection in intestinal- and diffuse-type gastric adenocarcinomas. J Natl Cancer Inst. 1991;83:640–643. doi: 10.1093/jnci/83.9.640. [DOI] [PubMed] [Google Scholar]
- 36.Garza-González E, Bosques-Padilla FJ, El-Omar E, Hold G, Tijerina-Menchaca R, Maldonado-Garza HJ, et al. Role of the polymorphic IL-1B, IL-1RN and TNF-A genes in distal gastric cancer in Mexico. Int J Cancer. 2005;114:237–241. doi: 10.1002/ijc.20718. [DOI] [PubMed] [Google Scholar]
- 37.Xia HH, Talley NJ. Apoptosis in gastric epithelium induced by Helicobacter pylori infection: implications in gastric carcinogenesis. Am J Gastroenterol. 2001;96:16–26. doi: 10.1111/j.1572-0241.2001.03447.x. [DOI] [PubMed] [Google Scholar]
- 38.Parsonnet J, Harris RA, Hack HM, Owens DK. Modelling cost-effectiveness of Helicobacter pylori screening to prevent gastric cancer: a mandate for clinical trials. Lancet. 1996;348:150–154. doi: 10.1016/s0140-6736(96)01501-2. [DOI] [PubMed] [Google Scholar]
- 39.Liou JM, Lee YC, El-Omar EM, Wu MS. Efficacy and Long-Term Safety of H. pylori Eradication for Gastric Cancer Prevention. Cancers (Basel) 2019;11:593. doi: 10.3390/cancers11050593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Malfertheiner P. Helicobacter pylori Treatment for Gastric Cancer Prevention. N Engl J Med. 2018;378:1154–1156. doi: 10.1056/NEJMe1800147. [DOI] [PubMed] [Google Scholar]
- 41.Shikata K, Kiyohara Y, Kubo M, Yonemoto K, Ninomiya T, Shirota T, et al. A prospective study of dietary salt intake and gastric cancer incidence in a defined Japanese population: the Hisayama study. Int J Cancer. 2006;119:196–201. doi: 10.1002/ijc.21822. [DOI] [PubMed] [Google Scholar]
- 42.Peleteiro B, Lopes C, Figueiredo C, Lunet N. Salt intake and gastric cancer risk according to Helicobacter pylori infection, smoking, tumour site and histological type. Br J Cancer. 2011;104:198–207. doi: 10.1038/sj.bjc.6605993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tatematsu M, Takahashi M, Fukushima S, Hananouchi M, Shirai T. Effects in rats of sodium chloride on experimental gastric cancers induced by N-methyl-N-nitro-N-nitrosoguanidine or 4-nitroquinoline-1-oxide. J Natl Cancer Inst. 1975;55:101–106. doi: 10.1093/jnci/55.1.101. [DOI] [PubMed] [Google Scholar]
- 44.Takahashi M, Kokubo T, Furukawa F, Kurokawa Y, Hayashi Y. Effects of sodium chloride, saccharin, phenobarbital and aspirin on gastric carcinogenesis in rats after initiation with N-methyl-N′-nitro-N-nitrosoguanidine. Gan. 1984;75:494–501. [PubMed] [Google Scholar]
- 45.Tricker AR. N-nitroso compounds and man: sources of exposure, endogenous formation and occurrence in body fluids. Eur J Cancer Prev. 1997;6:226–268. [PubMed] [Google Scholar]
- 46.You WC, Zhang L, Yang CS, Chang YS, Issaq H, Fox SD, et al. Nitrite, N-nitroso compounds, and other analytes in physiological fluids in relation to precancerous gastric lesions. Cancer Epidemiol Biomarkers Prev. 1996;5:47–52. [PubMed] [Google Scholar]
- 47.Ladeiras-Lopes R, Pereira AK, Nogueira A, Pinheiro-Torres T, Pinto I, Santos-Pereira R, et al. Smoking and gastric cancer: systematic review and meta-analysis of cohort studies. Cancer Causes Control. 2008;19:689–701. doi: 10.1007/s10552-008-9132-y. [DOI] [PubMed] [Google Scholar]
- 48.González CA, Pera G, Agudo A, Palli D, Krogh V, Vineis P, et al. Smoking and the risk of gastric cancer in the European Prospective Investigation Into Cancer and Nutrition (EPIC) Int J Cancer. 2003;107:629–634. doi: 10.1002/ijc.11426. [DOI] [PubMed] [Google Scholar]
- 49.Wu CY, Wu MS, Kuo KN, Wang CB, Chen YJ, Lin JT. Effective reduction of gastric cancer risk with regular use of nonsteroidal anti-inflammatory drugs in Helicobacter pylori-infected patients. J Clin Oncol. 2010;28:2952–2957. doi: 10.1200/JCO.2009.26.0695. [DOI] [PubMed] [Google Scholar]
- 50.Epplein M, Nomura AM, Wilkens LR, Henderson BE, Kolonel LN. Nonsteroidal antiinflammatory drugs and risk of gastric adenocarcinoma: the multiethnic cohort study. Am J Epidemiol. 2009;170:507–514. doi: 10.1093/aje/kwp162. [DOI] [PMC free article] [PubMed] [Google Scholar]
