ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Gastric cancer screening

Gastric cancer screening
Literature review current through: Jan 2024.
This topic last updated: Dec 04, 2023.

INTRODUCTION — Gastric cancer is one of the most common cancers worldwide [1]. However, there are significant differences in the incidence of gastric cancer by region [2]. The value of screening asymptomatic individuals for gastric cancer is controversial even in areas with a relatively high incidence of gastric cancer [3]. This topic will review the screening of gastric cancer. The epidemiology, risk factors, pathology, pathogenesis, clinical features, diagnosis, and management of gastric cancer are discussed in detail, separately. (See "Epidemiology of gastric cancer" and "Risk factors for gastric cancer" and "Clinical features, diagnosis, and staging of gastric cancer" and "Early gastric cancer: Epidemiology, clinical manifestations, diagnosis, and staging" and "Early gastric cancer: Treatment, natural history, and prognosis" and "Surgical management of invasive gastric cancer" and "Adjuvant and neoadjuvant treatment of gastric cancer" and "Gastric cancer: Pathology and molecular pathogenesis".)

SCREENING MODALITIES — The two main modalities for gastric cancer screening are upper endoscopy and contrast radiography.

Upper endoscopy — Upper endoscopy allows for direct visualization of the gastric mucosa and for biopsies to be obtained for diagnosing precancerous lesions such as gastric atrophy, intestinal metaplasia, or gastric dysplasia in addition to gastric cancer. Although it is more invasive and has a higher cost, upper endoscopy is also more sensitive for diagnosing a variety of gastric lesions as compared with alternative diagnostic strategies. (See 'Test performance' below.)

Contrast radiography — Double-contrast barium radiographs with photofluorography or digital radiography can identify malignant gastric ulcers, infiltrating lesions, and some early gastric cancers. However, false-negative barium studies can occur in as many as 50 percent of cases [4]. In early gastric cancer, the sensitivity of a barium study may be as low as 14 percent [5]. The one scenario in which a barium study may be superior to upper endoscopy is in patients with linitis plastica. The decreased distensibility of the stiff, "leather-flask" appearing stomach is more obvious on the radiographic study, and the endoscopic appearance may be relatively normal. (See "Early gastric cancer: Epidemiology, clinical manifestations, diagnosis, and staging", section on 'Upper endoscopy' and "Clinical features, diagnosis, and staging of gastric cancer".)

Other tests — While other modalities of screening for gastric cancer or its precursors have been proposed, further studies are needed to support their use.

Serum pepsinogen — A low serum pepsinogen I concentration and a low serum pepsinogen I/II ratio are suggestive of the presence of atrophic gastritis, a risk factor for gastric cancer. Serum pepsinogen testing has therefore been proposed to identify higher risk individuals who could benefit from gastric cancer screening with upper endoscopy [6-11]. However, in a pooled meta-analysis of 27 population-based screening studies and 15 studies in selected high-risk groups (eg, atrophic gastritis) that included approximately 300,000 individuals, the pooled sensitivity and specificity of serum pepsinogen (pepsinogen I level ≤70 ng/mL and serum pepsinogen I/II ratio <3) for gastric cancer were only 77 and 73 percent in population-based studies and 57 and 80 percent in selected high-risk groups [6].

Serum trefoil factor 3 — Serum trefoil factor 3 (TFF3) is a small stable protein expressed in the goblet cells of the small and large intestine and in gastric intestinal metaplasia. In one study, the sensitivity and specificity of serum TFF3 for detection of gastric cancer were both 81 percent compared with 45 and 88 percent, respectively, for serum pepsinogen (pepsinogen I level ≤70 ng/mL and serum pepsinogen I/II ratio <3) [12]. The combination of pepsinogen and TFF3 may provide even higher sensitivity for gastric cancer [13]. Prospective studies are needed to compare the performance of TFF3 with upper endoscopy and to establish its diagnostic utility.

MicroRNAs — At least three microRNAs (miRNAs), miRNA-421, miRNA 18a, and miR-106a, are highly expressed in gastric cancers and are detectable in peripheral blood and gastric aspirates [14-16]. Assays for multiple miRNAs may further improve diagnostic accuracy [17]. However, additional studies are needed to define the role of these miRNAs as biomarkers for gastric cancer.

Multianalyte blood tests — An early study examining combinations of tumor-specific circulating proteins and mutations in cell-free DNA in the blood (CancerSEEK) suggests promise for early detection of potentially resectable gastric cancer (sensitivity 75 percent with a high degree of specificity) [18]. However, to establish clinical utility and demonstrate that early diagnosis of gastric cancer using this assay saves lives, prospective trials in large populations will be required.

COMPARISON OF SCREENING METHODS

Test performance — Although upper endoscopy and contrast radiography have not been directly compared, studies suggest that endoscopic screening may be a more sensitive test for screening for gastric cancer [19-23]. A population-based study in South Korea included 2,690,731 individuals who underwent screening for gastric cancer with either upper endoscopy or an upper gastrointestinal (GI) series [24]. Gastric cancer detection rates were 2.61 and 0.68 per 1000 screenings, respectively. The sensitivity rates for upper endoscopy versus upper GI series in detecting gastric cancer were 69 and 37 percent, respectively. Both studies had a specificity of 96 percent. The sensitivity of upper endoscopy in detecting a localized gastric cancer was also significantly higher as compared with upper GI series (68 versus 32 percent). In total, 2067 interval cancers occurred within one year of a negative upper GI series and 1083 cancers occurred after a negative upper endoscopy, but there was no difference in interval cancer rates (1.2 per 1000 screenings for both groups).

Effectiveness — Although some observational studies suggest that screening in areas of high gastric cancer incidence has contributed to the detection of cancer in early stages and an overall decline in gastric cancer mortality, there are no data from large randomized trials demonstrating lower gastric cancer-related mortality in screened populations [19,25-32]. In addition, lead time bias, length bias, and selection bias must be considered when appraising the overall effectiveness of screening demonstrated in observational studies. In a retrospective cohort study of 19,168 gastric cancer patients in Korea, endoscopy-screened patients and patients screened with upper GI series were significantly more likely to be diagnosed with localized gastric cancer as compared with never-screened patients (odds ratio 2.1, 95% CI 1.9-2.3 and 1.2, 95% CI 1.1-1.4, respectively) [32]. However, this study did not include data as to which patients were symptomatic or had undergone evaluation outside of the screening program for evaluation of symptoms.

Screening for gastric cancer may be cost-effective for high-risk subgroups, but not low-risk populations [33,34]. A cost-effectiveness analysis found that in a high-risk group of Chinese men ages 50 to 70 years (standardized incidence of gastric cancer of 25.9 per 100,000 population), screening with upper endoscopy every two years was highly cost-effective ($28,836 per quality-adjusted life-years saved [33]). By contrast, averting one gastric cancer death in men in the United States, assuming an incidence of gastric cancer of <10 per 100,000 population, would cost approximately $247,600, which does not compare favorably with other generally accepted cancer screening interventions. (See "A short primer on cost-effectiveness analysis".)

SCREENING STRATEGIES — Screening for gastric cancer is controversial, and recommendations for screening differ based on the incidence of gastric cancer.

Universal screening — Universal or population-based screening for gastric cancer has been implemented in some countries with a high incidence of gastric cancer (eg, Japan, Korea, Venezuela, and Chile) [19-21]. However, the recommended screening modality and intervals vary. As examples:

In Japan, population-based screening for gastric cancer is recommended for individuals older than 50 years with conventional double-contrast barium radiograph with photofluorography every year or upper endoscopy every two to three years [22,35,36].

In Korea, upper endoscopy is recommended every two years for individuals aged 40 to 75 years [36-38].

The optimal interval for screening has not been established in randomized trials. A two-year interscreen interval is supported by at least one study that evaluated the mean sojourn time (MST) for gastric cancer (ie, the asymptomatic period during which a cancer can be detected through screening tests before typical symptoms develop) in a cohort of 61,000 Korean men voluntarily attending a cancer screening program and rescreened by endoscopy [39]. A total of 91 incident cases were found during 19,598,598 person-years of follow-up, and the MST for gastric cancer was 2.4 years (95% CI 1.9-3.0). Of note, the MST was shorter in individuals 40 to 49 years of age (1.3 years, 95% CI 1.0-1.7) than the MST in those 50 to 59 years of age or 60 to 69 (3.2 and 3.7, respectively).  

At least some data suggest that the screening interval may be widened to a three-year rather than a two-year interval without significantly decreasing the proportion of gastric neoplasms that can be adequately treated by endoscopic methods [40-42]. However, intervals longer than three years may be associated with a greater risk of more advanced stage cancer at diagnosis. As an example, in a retrospective cohort study of 2485 patients with gastric adenocarcinoma in Korea, as compared with individuals who underwent annual screening for gastric cancer, the risk of advanced cancer was higher in individuals who underwent screening at four- or five-year intervals (four-year interval odds ratio [OR] 2.5, 95% CI 1.4-4.5, five-year interval OR 2.2, 95% CI 1.3-3.7), but not in individuals who underwent screening at two- or three-year intervals [40]. In subgroup analysis, individuals with a family history of gastric cancer and individuals in their 60s were more likely to be diagnosed with a higher stage of gastric cancer if upper endoscopies were performed every three years as compared with annually (family history of gastric cancer OR 2.68, 95% CI 1.3-5.7, gastric cancer in 60s OR 2.09, 95% CI 1.0-4.3).

Selective screening of high-risk subgroups — In areas of low gastric cancer incidence, screening for gastric cancer with upper endoscopy should be reserved for specific high-risk subgroups [43-54]. The intensity of screening should be based upon an appraisal of the magnitude of risk in each patient, their suitability for treatment should a lesion be detected, and their willingness to accept the uncertain benefits and risks of a screening program. Whether eradication of H. pylori can reverse or reduce the risk that precursor lesions will progress is unclear [55-62].

Individuals at increased risk for gastric cancer include those with the following:

Gastric adenomas

Pernicious anemia

Gastric intestinal metaplasia

Familial adenomatous polyposis

Lynch syndrome

Peutz-Jeghers syndrome

Juvenile polyposis syndrome

Specific recommendations for gastric cancer screening in these individuals are discussed separately. (See "Gastric polyps", section on 'Management' and "Gastric intestinal metaplasia", section on 'Endoscopic surveillance in selected patients' and "Clinical manifestations and diagnosis of vitamin B12 and folate deficiency", section on 'Determining the underlying cause of vitamin B12 deficiency' and "Juvenile polyposis syndrome" and "Familial adenomatous polyposis: Screening and management of patients and families", section on 'Upper gastrointestinal tumors' and "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Cancer screening and management", section on 'Gastric cancer'.)

Importantly, high-risk patients from families with hereditary diffuse gastric cancer are not good candidates for screening because of the propensity of these tumors to arise beneath an intact mucosa and elude radiographic and endoscopic detection. Instead, prophylactic gastrectomy should be strongly considered in these individuals. (See "Hereditary diffuse gastric cancer" and "Surgical management of hereditary diffuse gastric cancer".)

PREVENTION

Helicobacter pylori eradication — In several regions of high gastric cancer incidence, routine screening and eradication for H. pylori has been implemented or is being evaluated to decrease rates of gastric cancer [63]. There are limited data that suggest that screening for H. pylori in asymptomatic healthy individuals in areas of high gastric cancer incidence may decrease the risk of gastric cancer [64-67]. Successful eradication of H. pylori infection in first-degree relatives with gastric cancer has been demonstrated to significantly reduce the risk of a subsequent gastric cancer in areas of high cancer incidence [68]. This was illustrated in a randomized trial in South Korea in which 1838 individuals with H. pylori infection and a first-degree relative with gastric cancer were assigned to receive either eradication therapy or placebo. During a median follow-up of 9.2 years, gastric cancer developed in significantly fewer patients in the H. pylori treatment group as compared with the placebo group (1.2 versus 2.7 percent; HR 0.45).

In areas of low gastric cancer incidence, there is no role for routine screening of asymptomatic, average risk, healthy individuals for H. pylori in order to decrease the risk of gastric cancer. There may be a future role for screening selected asymptomatic individuals for H. pylori (eg, individuals who are both first-generation immigrants from areas of high gastric cancer incidence and have a first-degree relative with gastric cancer) [68]. However, further studies are needed before it can be recommended in any asymptomatic population. (See "Risk factors for gastric cancer", section on 'Importance of Helicobacter pylori infection' and "Clinical features, diagnosis, and staging of gastric cancer", section on 'Issues related to helicobacter pylori infection'.)

Other indications for testing and eradication of H. pylori are discussed in detail, separately. (See "Indications and diagnostic tests for Helicobacter pylori infection in adults".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Gastric cancer".)

SUMMARY AND RECOMMENDATIONS

Gastric cancer is one of the most common cancers worldwide. However, there are significant differences in the incidence of gastric cancer by region. Screening for gastric cancer is controversial even in areas with a relatively high incidence of gastric cancer. (See 'Introduction' above.)

The two main modalities for screening for gastric cancer are contrast radiography and upper endoscopy. Although upper endoscopy and contrast radiography have not been directly compared, studies suggest that endoscopic screening may be a more sensitive test for screening for gastric cancer. While other modalities of screening for gastric cancer or its precursors have been proposed, there are limited data to support their use. (See 'Screening modalities' above and 'Test performance' above.)

Although screening for gastric cancer may be cost-effective in high-risk subgroups, whether screening improves clinical outcomes (ie, gastric cancer-related mortality) is unclear. While some observational studies suggest that the screening has contributed to detection of cancer in early stages and an overall decline in gastric cancer mortality, there are no data from large controlled trials. (See 'Effectiveness' above.)

Recommendations for screening differ based on the endemic incidence of gastric cancer. Universal or population-based screening for gastric cancer has been implemented in some countries with a high incidence of gastric cancer (eg, Japan, Korea, Venezuela, and Chile). In areas of low gastric cancer incidence, screening for gastric cancer with upper endoscopy should be reserved for specific high-risk subgroups. Individuals at increased risk for gastric cancer include those with gastric adenomas, pernicious anemia, gastric intestinal metaplasia, familial adenomatous polyposis, and Lynch syndrome. There may be a future role for screening selected asymptomatic individuals for H. pylori (eg, individuals who are both first-generation immigrants from areas of high gastric cancer incidence and have a first-degree relative with gastric cancer). However, further studies are needed before it can be recommended in any asymptomatic population. (See 'Screening strategies' above.)

  1. Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin 2015; 65:87.
  2. Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat Rev Clin Oncol 2023; 20:338.
  3. Leung WK, Wu MS, Kakugawa Y, et al. Screening for gastric cancer in Asia: current evidence and practice. Lancet Oncol 2008; 9:279.
  4. Dooley CP, Larson AW, Stace NH, et al. Double-contrast barium meal and upper gastrointestinal endoscopy. A comparative study. Ann Intern Med 1984; 101:538.
  5. Longo WE, Zucker KA, Zdon MJ, Modlin IM. Detection of early gastric cancer in an aggressive endoscopy unit. Am Surg 1989; 55:100.
  6. Miki K. Gastric cancer screening using the serum pepsinogen test method. Gastric Cancer 2006; 9:245.
  7. Yoshida T, Kato J, Inoue I, et al. Cancer development based on chronic active gastritis and resulting gastric atrophy as assessed by serum levels of pepsinogen and Helicobacter pylori antibody titer. Int J Cancer 2014; 134:1445.
  8. Miki K, Fujishiro M, Kodashima S, Yahagi N. Long-term results of gastric cancer screening using the serum pepsinogen test method among an asymptomatic middle-aged Japanese population. Dig Endosc 2009; 21:78.
  9. Dinis-Ribeiro M, da Costa-Pereira A, Lopes C, et al. Validity of serum pepsinogen I/II ratio for the diagnosis of gastric epithelial dysplasia and intestinal metaplasia during the follow-up of patients at risk for intestinal-type gastric adenocarcinoma. Neoplasia 2004; 6:449.
  10. Väänänen H, Vauhkonen M, Helske T, et al. Non-endoscopic diagnosis of atrophic gastritis with a blood test. Correlation between gastric histology and serum levels of gastrin-17 and pepsinogen I: a multicentre study. Eur J Gastroenterol Hepatol 2003; 15:885.
  11. Yoshihara M, Hiyama T, Yoshida S, et al. Reduction in gastric cancer mortality by screening based on serum pepsinogen concentration: a case-control study. Scand J Gastroenterol 2007; 42:760.
  12. Aikou S, Ohmoto Y, Gunji T, et al. Tests for serum levels of trefoil factor family proteins can improve gastric cancer screening. Gastroenterology 2011; 141:837.
  13. Kaise M, Miwa J, Tashiro J, et al. The combination of serum trefoil factor 3 and pepsinogen testing is a valid non-endoscopic biomarker for predicting the presence of gastric cancer: a new marker for gastric cancer risk. J Gastroenterol 2011; 46:736.
  14. Cui L, Zhang X, Ye G, et al. Gastric juice MicroRNAs as potential biomarkers for the screening of gastric cancer. Cancer 2013; 119:1618.
  15. Zhou H, Guo JM, Lou YR, et al. Detection of circulating tumor cells in peripheral blood from patients with gastric cancer using microRNA as a marker. J Mol Med (Berl) 2010; 88:709.
  16. Su ZX, Zhao J, Rong ZH, et al. Diagnostic and prognostic value of circulating miR-18a in the plasma of patients with gastric cancer. Tumour Biol 2014; 35:12119.
  17. Wang R, Wen H, Xu Y, et al. Circulating microRNAs as a novel class of diagnostic biomarkers in gastrointestinal tumors detection: a meta-analysis based on 42 articles. PLoS One 2014; 9:e113401.
  18. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018; 359:926.
  19. Mizoue T, Yoshimura T, Tokui N, et al. Prospective study of screening for stomach cancer in Japan. Int J Cancer 2003; 106:103.
  20. Llorens P. Gastric cancer mass survey in Chile. Semin Surg Oncol 1991; 7:339.
  21. Pisani P, Oliver WE, Parkin DM, et al. Case-control study of gastric cancer screening in Venezuela. Br J Cancer 1994; 69:1102.
  22. Hamashima C, Shibuya D, Yamazaki H, et al. The Japanese guidelines for gastric cancer screening. Jpn J Clin Oncol 2008; 38:259.
  23. Hamashima C, Saito H, Nakayama T, et al. The standardized development method of the Japanese guidelines for cancer screening. Jpn J Clin Oncol 2008; 38:288.
  24. Choi KS, Jun JK, Park EC, et al. Performance of different gastric cancer screening methods in Korea: a population-based study. PLoS One 2012; 7:e50041.
  25. Murakami R, Tsukuma H, Ubukata T, et al. Estimation of validity of mass screening program for gastric cancer in Osaka, Japan. Cancer 1990; 65:1255.
  26. Hisamichi S, Sugawara N, Fukao A. Effectiveness of gastric mass screening in Japan. Cancer Detect Prev 1988; 11:323.
  27. Fukao A, Tsubono Y, Tsuji I, et al. The evaluation of screening for gastric cancer in Miyagi Prefecture, Japan: a population-based case-control study. Int J Cancer 1995; 60:45.
  28. Oshima A, Hirata N, Ubukata T, et al. Evaluation of a mass screening program for stomach cancer with a case-control study design. Int J Cancer 1986; 38:829.
  29. Tsuji I, Fukao A, Sugawara N, et al. Cost-effectiveness analysis of screening for gastric cancer in Japan. Tohoku J Exp Med 1991; 164:279.
  30. Kunisaki C, Ishino J, Nakajima S, et al. Outcomes of mass screening for gastric carcinoma. Ann Surg Oncol 2006; 13:221.
  31. Inaba S, Hirayama H, Nagata C, et al. Evaluation of a screening program on reduction of gastric cancer mortality in Japan: preliminary results from a cohort study. Prev Med 1999; 29:102.
  32. Choi KS, Jun JK, Suh M, et al. Effect of endoscopy screening on stage at gastric cancer diagnosis: results of the National Cancer Screening Programme in Korea. Br J Cancer 2015; 112:608.
  33. Dan YY, So JB, Yeoh KG. Endoscopic screening for gastric cancer. Clin Gastroenterol Hepatol 2006; 4:709.
  34. Yeh JM, Hur C, Ward Z, et al. Gastric adenocarcinoma screening and prevention in the era of new biomarker and endoscopic technologies: a cost-effectiveness analysis. Gut 2016; 65:563.
  35. Choi IJ. Endoscopic gastric cancer screening and surveillance in high-risk groups. Clin Endosc 2014; 47:497.
  36. Hamashima C, Kim Y, Choi KS. Comparison of guidelines and management for gastric cancer screening between Korea and Japan. Value Health 2015; 18:A272.
  37. Yoo KY. Cancer control activities in the Republic of Korea. Jpn J Clin Oncol 2008; 38:327.
  38. Park HA, Nam SY, Lee SK, et al. The Korean guideline for gastric cancer screening. J Korean Med Assoc 2015; 58:373.
  39. Bae JM, Shin SY, Kim EH. Mean sojourn time of preclinical gastric cancer in Korean men: a retrospective observational study. J Prev Med Public Health 2014; 47:201.
  40. Nam JH, Choi IJ, Cho SJ, et al. Association of the interval between endoscopies with gastric cancer stage at diagnosis in a region of high prevalence. Cancer 2012; 118:4953.
  41. Park CH, Kim EH, Chung H, et al. The optimal endoscopic screening interval for detecting early gastric neoplasms. Gastrointest Endosc 2014; 80:253.
  42. Hamashima C, Ogoshi K, Okamoto M, et al. A community-based, case-control study evaluating mortality reduction from gastric cancer by endoscopic screening in Japan. PLoS One 2013; 8:e79088.
  43. Hirota WK, Zuckerman MJ, Adler DG, et al. ASGE guideline: the role of endoscopy in the surveillance of premalignant conditions of the upper GI tract. Gastrointest Endosc 2006; 63:570.
  44. Lundegårdh G, Adami HO, Helmick C, et al. Stomach cancer after partial gastrectomy for benign ulcer disease. N Engl J Med 1988; 319:195.
  45. Tersmette AC, Goodman SN, Offerhaus GJ, et al. Multivariate analysis of the risk of stomach cancer after ulcer surgery in an Amsterdam cohort of postgastrectomy patients. Am J Epidemiol 1991; 134:14.
  46. MING SC, GOLDMAN H. GASTRIC POLYPS; A HISTOGENETIC CLASSIFICATION AND ITS RELATION TO CARCINOMA. Cancer 1965; 18:721.
  47. Huang JQ, Sridhar S, Chen Y, Hunt RH. Meta-analysis of the relationship between Helicobacter pylori seropositivity and gastric cancer. Gastroenterology 1998; 114:1169.
  48. Ohata H, Kitauchi S, Yoshimura N, et al. Progression of chronic atrophic gastritis associated with Helicobacter pylori infection increases risk of gastric cancer. Int J Cancer 2004; 109:138.
  49. Kokkola A, Sjöblom SM, Haapiainen R, et al. The risk of gastric carcinoma and carcinoid tumours in patients with pernicious anaemia. A prospective follow-up study. Scand J Gastroenterol 1998; 33:88.
  50. Alexander JR, Andrews JM, Buchi KN, et al. High prevalence of adenomatous polyps of the duodenal papilla in familial adenomatous polyposis. Dig Dis Sci 1989; 34:167.
  51. Aarnio M, Salovaara R, Aaltonen LA, et al. Features of gastric cancer in hereditary non-polyposis colorectal cancer syndrome. Int J Cancer 1997; 74:551.
  52. Yatsuya H, Toyoshima H, Tamakoshi A, et al. Individual and joint impact of family history and Helicobacter pylori infection on the risk of stomach cancer: a nested case-control study. Br J Cancer 2004; 91:929.
  53. Chen MJ, Wu DC, Ko YC, Chiou YY. Personal history and family history as a predictor of gastric cardiac adenocarcinoma risk: a case-control study in Taiwan. Am J Gastroenterol 2004; 99:1250.
  54. Boeing H. Epidemiological research in stomach cancer: progress over the last ten years. J Cancer Res Clin Oncol 1991; 117:133.
  55. de Jong D, van Dijk WC, van der Hulst RW, et al. CagA+ H. pylori strains and gastric lymphoma. Gastroenterology 1997; 113:2022.
  56. Forbes GM, Warren JR, Glaser ME, et al. Long-term follow-up of gastric histology after Helicobacter pylori eradication. J Gastroenterol Hepatol 1996; 11:670.
  57. Annibale B, Aprile MR, D'ambra G, et al. Cure of Helicobacter pylori infection in atrophic body gastritis patients does not improve mucosal atrophy but reduces hypergastrinemia and its related effects on body ECL-cell hyperplasia. Aliment Pharmacol Ther 2000; 14:625.
  58. Correa P, Fontham ET, Bravo JC, et al. Chemoprevention of gastric dysplasia: randomized trial of antioxidant supplements and anti-helicobacter pylori therapy. J Natl Cancer Inst 2000; 92:1881.
  59. Ohkusa T, Fujiki K, Takashimizu I, et al. Improvement in atrophic gastritis and intestinal metaplasia in patients in whom Helicobacter pylori was eradicated. Ann Intern Med 2001; 134:380.
  60. Sung JJ, Lin SR, Ching JY, et al. Atrophy and intestinal metaplasia one year after cure of H. pylori infection: a prospective, randomized study. Gastroenterology 2000; 119:7.
  61. Kokkola A, Sipponen P, Rautelin H, et al. The effect of Helicobacter pylori eradication on the natural course of atrophic gastritis with dysplasia. Aliment Pharmacol Ther 2002; 16:515.
  62. Ito M, Haruma K, Kamada T, et al. Helicobacter pylori eradication therapy improves atrophic gastritis and intestinal metaplasia: a 5-year prospective study of patients with atrophic gastritis. Aliment Pharmacol Ther 2002; 16:1449.
  63. Sugano K, Tack J, Kuipers EJ, et al. Kyoto global consensus report on Helicobacter pylori gastritis. Gut 2015; 64:1353.
  64. Ford AC, Forman D, Hunt RH, et al. Helicobacter pylori eradication therapy to prevent gastric cancer in healthy asymptomatic infected individuals: systematic review and meta-analysis of randomised controlled trials. BMJ 2014; 348:g3174.
  65. Uemura N, Mukai T, Okamoto S, et al. Effect of Helicobacter pylori eradication on subsequent development of cancer after endoscopic resection of early gastric cancer. Cancer Epidemiol Biomarkers Prev 1997; 6:639.
  66. Saito K, Arai K, Mori M, et al. Effect of Helicobacter pylori eradication on malignant transformation of gastric adenoma. Gastrointest Endosc 2000; 52:27.
  67. Wong BC, Lam SK, Wong WM, et al. Helicobacter pylori eradication to prevent gastric cancer in a high-risk region of China: a randomized controlled trial. JAMA 2004; 291:187.
  68. Choi IJ, Kim CG, Lee JY, et al. Family History of Gastric Cancer and Helicobacter pylori Treatment. N Engl J Med 2020; 382:427.
Topic 2618 Version 38.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟