Open Access

Gastric adenocarcinoma with high‑level microsatellite instability: A case report

  • Authors:
    • Alejandro Alfaro
    • Daniel Zanabria
    • Alfredo Aguilar
    • Sergio A. Jimenez‑Solano
    • Alejandra Zevallos
    • Williams Fajardo
  • View Affiliations

  • Published online on: January 26, 2023     https://doi.org/10.3892/mco.2023.2612
  • Article Number: 16
  • Copyright : © Alfaro et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) ranks fifth on the list of the most common malignancies worldwide. In Peru, gastric neoplasms are considered the second leading cause of mortality among males. Among the molecular subgroups of GC, microsatellite instability presents a favorable prognosis due to its hypermutated phenotype, which activates immunosurveillance. The present study describes the case of a 75‑year‑old patient, who was admitted in the hospital with a history of upper gastrointestinal bleeding and recurrent hospital admission, due to severe anemia. The patient presented with pale skin, normal vital functions, slight swelling of the lower extremities, and abdominal distention and bloating upon a physical examination. An endoscopic examination revealed an infiltrating circular ulcerated lesion. The histopathological analysis identified a moderately differentiated intestinal‑type adenocarcinoma with pathological stage T3N0M0. Tumor genomic profiling demonstrated alterations in 15 different genes with a tumor mutational burden of 28 mutations/Mb. Finally, the patient underwent a partial gastrectomy without pre‑operative chemotherapy. After 4 days, the patient presented with post‑operative complications for which he was re‑operated on. The patient did not survive. To the best of our knowledge, in the present case, pernicious anemia was an early sign of GC and a gastroscopy had to be performed. Furthermore, MutS homolog 3 alterations probably conditioned the presence of multiple frame‑shift mutations.

Introduction

Gastric cancer (GC) is one of the most common neoplasms worldwide, affecting mainly males at a later age (1,2). In Peru, GC is considered the main cause of cancer-related mortality in the general population and the second most frequent type of cancer among males (3). Furthermore, one of the main risk factors for GC in patients of Peruvian descent is Hellicobacter pylori infection. A study performed at the National Institute of Neoplastic Diseases (INEN) revealed that up to 60% of patients with GC presented with Hellicobacter pylori infection (4).

By contrast, the incidence of GC increases considerably with age. Of note, the 30-day post-operative mortality rate is higher in patients who are >70 years of age following a gastrectomy (5). Likewise, other studies have demonstrated that patients with GC who are >65 years of age have a higher risk of mortality following surgical resection, including a gastrectomy (6,7).

Finally, the molecular subgroup of GC with microsatellite instability (MSI) represents <10% of GC cases. However, MSI is considered a favorable prognostic biomarker, since its hypermutation phenotype activates immunosurveillance, establishing this as a promising candidate for immunotherapy (8). The present study describes the case of an older patient with recurrent genomic mutations in a moderately differentiated gastric adenocarcinoma with a torpid evolution.

Case report

A 75-year-old male patient from Huaral (Peru) visited the Emergency Department of the Dos de Mayo National Hospital (Lima, Peru) on September 15, 2020 with symptoms of severe anemia and melena. The patient had a history of tuberculosis, upper gastrointestinal bleeding and gonorrhea. Likewise, with a significant family history, his father was deceased due to prostate cancer and he also had two siblings who had died due to malignant neoplasia in the stomach. The patient had visited another hospital 18 months prior to this, due to the presence of peripheral edema associated with mild fatigue. He was diagnosed with severe anemia (hemoglobin, 7 mg/dl) and was prescribed with drinkable iron. After 8 months, the patient returned to another hospital for type I feces on the Bristol scale, mild constipation, mild fatigue and increased volume in the lower extremities, and was again diagnosed with severe anemia (hemoglobin, 8 mg/dl). Ferrous sulfate treatment was administered. Following treatment, the patient reported that no improvement and continued to have intermittent dark stools.

Upon a physical examination, the patient exhibited pale skin, normal vital functions, slight swelling of the lower limbs, and abdominal distention and bloating. Laboratory tests revealed a hemoglobin level lower than normal (4 g/dl) and a normal biochemical profile.

The endoscopic analysis demonstrated a large circular ulcerated lesion, measuring 5x3 cm in size, which was detected in the lesser curvature of the antrum. The observable tumor border was irregular with edges that do not appear smooth, with encompassing fibrin deposition (Fig. 1). Furthermore, following the clinical practice guidelines of the Dos de Mayo Hospital, a biopsy of the stomach lining proband was obtained and this was found to be positive for Helicobacter pylori. Based on these findings, the patient underwent an open subtotal gastrectomy. The patient was treated according to the American College of Surgeons National Surgical Quality Improvement Program/American Geriatrics Society Best Practices guidelines (9).

Hematoxylin and eosin (H&E) staining was conducted on formalin-fixed and paraffin-embedded tumor tissue as described by Feldman and Wolfe (10). The stained tissues were cut into 5-mm-thick slices, dewaxed, mounted with neutral balsam and then imaged using a Leika DM100 microscope (Leica Microsystems, GmbH). This test revealed a moderately differentiated intestinal-type adenocarcinoma according to the Lauren classification (Fig. 2). Venous vascular invasion and surgical margins free of tumor involvement were observed. A total of 18 lymph nodes were examined with no evidence of neoplasia. According to the clinicopathological assessment by a pathologist (AAl), the pathological stage was defined as T3N0M0, and the clinical stage as IIB.

After signing a written informed consent, the patient was enrolled in a GC observational study (unpublished data); therefore, a genomic analysis was performed with targeted NGS using the platform FoundationOne CDx® (Foundation Medicine, Inc.) as previously described by Frampton et al (11). Since the observational study was multicentric, the Via Libre Ethics Committee that is duly registered and accredited by the National Institute of Health in Peru was addressed. The genomic profile revealed a high microsatellite instability with a tumor mutational burden (TMB) of 28 mutations/Mb. Alterations were observed in 15 different genes [F-box and WD repeat domain containing 7 gene (FBXW7), T-rich interactive domain-containing protein 1A gene (ARID1A), KRAS, ring finger protein 43 gene (RNF43), ATR serine/threonine kinase gene (ATR), bromodomain-containing protein 4 gene (BRD4), caspase 8 gene (CASP8), cyclic adenosine monophosphate response element binding protein binding protein (CREBBP), folliculin gene (FLCN), Janus kinase gene (JAK1), MAP3K1, histone-lysine N-methyltransferase 2D gene (MLL2), MutS homolog 3 (MSH3), SMAD4, SOX9], 14 of which are related to frameshift mutations and one related to an amino acid substitution (KRAS) (Table I).

Table I

Patient microsatellite instability analysis.

Table I

Patient microsatellite instability analysis.

No.GeneAlterationVAF (%)
1FBXW7C46fs*1417.4
2ARID1AY551fs*6818.6
3KRASG13D17.3
4RNF43G659fs*4139.6
5ATRI774fs*517.8
6BRD4P475fs*10918.1
7CASP8K490fs*7317.3
8CREBBPI1084fs*1518.3
9FLCNH429fs*3920.5
10JAK1P430fs*218.4
11MAP3K1L920fs*1019.1
12MLL2G1235fs*9516.7
13MSH3K383fs*3230.1
14SMAD4S32fs*116.9
15  SOX9  P350fs*33, R264fs*3225.1, 23.5

[i] VAF, variant allele frequency.

Finally, 4 days after the subtotal gastrectomy, the patient presented with post-operative complications and underwent two additional surgeries. The first surgery was performed due to an early small bowel obstruction caused by fibrous bands of tissue in the abdomen, forming after surgical procedures. The second surgery was performed due to wound dehiscence and stump infection. A peritoneal dissemination was produced by an anastomotic leakage, which led to an abdominal sepsis. After the second surgery, the patient developed septic shock and a multisystemic organ failure ultimately, leading to mortality.

Discussion

Overall, >6 and <24% of patients with gastroesophageal cancer present with MSI (12-15), with an increase trend towards 48% in adults >40 years of age (16). Microsatellites are short and repetitive DNA sequences that are abundant in the human genome, particularly in non-coding DNA regions (17,18). Deficiencies in the mismatch repair (MMR) mechanism are caused by germline or sporadic mutations, which may result in nucleotide insertion or deletion in microsatellite regions during DNA replication. This phenomenon is known as MSI (18,19).

Individuals with mutations in the protein complexes that cause MMR have a high predisposition to develop neoplasms (20). In the patient treated in the present study, it was observed that MSH3 presented alterations. MSH3 is a member of the MMR system (21). Several studies have demonstrated its importance as a tumor suppressor gene (20-23). In the present study, the patient presented several frameshift mutations that may be related to the MSH3 mutation. To date, studies have revealed that MSH3 deficiencies are related to changes in the reading frame of microsatellite regions and may contribute to tumorigenesis (26,27). In total, >2 and <7.7% of patients with GC have mutations in MSH3 (28,29). Similarly, the MSH3 deletion has been found to be associated with increased chromosomal instability in p53-deficient tumors (25).

Within the classification of GC, the MSI-high (MSI-H) subtype predominates in elderly patients (>65 years of age), and is related to the intestinal type of the Lauren classification, being usually located in the distal part of the stomach, particularly the pylorus and gastric body, and presenting with a better prognosis (28-30). In this sense, MSI-H tumors are characterized by high levels of CD8+ T cell infiltration, thus being good candidates for immunotherapeutic treatments (33,34).

Several studies have revealed that TMB may be a promising predictive biomarker for immunotherapy in various types of cancer (35-40). To date, it has been revealed in the literature that patients with advanced-stage GC with a high TMB have a better clinical response to immunotherapy and demonstrate an improved overall survival (41). However, it is unclear whether TMB has clinical relevance for patients with advanced or metastatic GC (38,42,43). In addition, TMB usually is determined by a whole genome sequencing, thus limiting its clinical use, due to its increased cost and long turnaround times. In the case presented herein, the FoundationOne CDx panel was used to obtain the molecular profiling of the patient. Recently, two studies have demonstrated the feasibility and utility of this panel on clinical application with Japanese cohorts. The panel presented an assay success rate of 97,3% and physicians were able to give targeted therapy ≤14% of their population sample (44,45). Personalized medicine has not yet been implemented in routine oncological practice in Peru. However, there is an urgent need for the improvement of cancer diagnosis and prognosis, since this type of technology not only permits the provision of an efficient therapeutic regimen to all patients, but also increases drug accessibility to regional healthcare establishments (46).

By contrast, in the case in the present study, severe anemia was diagnosed. The study by Medrano-Guzmán et al (47) revealed that ≤2/3 of patients with GC had anemia as part of the initial symptoms. Furthermore, anemia was an adverse prognostic factor for survival (OR, 3.62; P<0.001; CI, 1.4-13.8) (47). Another study involving cancer patients revealed that anemia was a factor associated with mortality (HR, 3.04; P=0.002; CI:1.51-6.09), increasing the risk of mortality by ≤3-fold (P=0.008; CI, 1.35-7.05) (48). Likewise, anemia contributes to a hypoxic state of the tumor, which promotes angiogenesis; consequently, the attenuation or elimination of anemia improves survival and response to treatment (49). Similarly, Helicobacter pylori infection is a critical factor to consider for patients, as this has also been shown to be associated with MMR deficiencies and the presence of MSI (50,51). The study by Machado et al (52) demonstrated in vitro and in vivo that Helicobacter pylori infection reduced MMR activity, thus rendering gastric epithelial cells vulnerable to genetic instability, further contributing to gastric carcinogenesis in Helicobacter pylori-infected individuals.

The evolution of surgical technology has made habitual for physicians to perform surgical treatments in gastrointestinal cancer patients with advanced age. However, geriatric patients tend to present a compromised health state due to several factors, including comorbidities, frailty, geriatric syndromes, infections and transfusions, which increase their risk of post-operative complications and death (53). Likewise, it is important to highlight that patients with malnutrition present a higher risk of post-surgical complications including infections and other events (54-56). In the present case report, the patient died due to post-operative complications; a previous meta-analysis by Xue et al (57) demonstrated that the incidence of post-operative complications in geriatric patients with gastrointestinal cancer was between 24 to 76.3%. Early post-operative small bowel obstruction (ESBO) is present in ~1 to 12% of abdominal operations (58-61). Of note, the study by Nakamura et al (62) revealed that 10% of patients who underwent open surgery developed ESBO; thus, this type of surgery represents an independent risk factor for the develoment of ESBO (odds ratio, 5.621; P=0.015). By contrast, the literature reports that malnutrition and especially hypoalbuminemia are associated with poor healing processes, decreased collagen synthesis in surgical wounds, and decreased immune response. These factors determine the higher prevalence of surgical site infections and intestinal anastomosis leakage (63-65). Additionally, it should be noted that low hemoglobin counts, and hypoalbuminemia are common findings in patients undergoing surgery in public hospitals in Peru, since a number of these patients present chronic malnutrition (66-68).

Additionally, the barriers for the management of cancer patients in Peru should be highlighted. In the present case report, the patient was referred from other hospitals of primary care. The first symptom of the patient was a persistent anemia which lead to a misdiagnosis by previous physicians, contributing to the health deterioration of the patient. However, in the public sector there are delays not just in the counter-referral of patients to hospitals of more complexity, but also in the access to specialized cancer care, once a patient is referred to the new hospital (69). Literature reports that anemia is a common symptom in patients with GC (70). In that regard, it is important to develop and strengthen GC screening programs at the primary care centers.

Lastly, high-level microsatellite instability has been widely associated in the literature with better prognosis in GC patients and its improvement with immunotherapy has been described (18,30,71). The case described in the present study highlights the importance of carefulness concerning the treatment and management of geriatric patients with GC, particularly when it is necessary to perform radical treatments, such as gastrectomy. The probabilities of post-operative complications were also underlined, particularly in the Peruvian population, where patients generally present with malnutrition.

Acknowledgements

Not applicable.

Funding

Funding: The molecular profile was funded by a research grant from Roche-Peru (GSD039-20).

Availability of data and materials

The data from the patient's molecular profile presented in the presesnt case report are available from the Figshare platform (https://doi.org/10.6084/m9.figshare.20522820).

Authors' contributions

All authors (AAl, DZ, AAg, SAJS, AZ and WF) contributed to the conception and design of the present case report. Material preparation and data collection was performed by AA, WF and DZ. The first draft of the manuscript was written by AZ and SAJS. All authors commented on previous versions of the manuscript. WF and AZ confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present case report was part of an observational study named Influencia del nivel socioeconómico en la genómica del cancer gástrico which recruited patients from three hospitals that are mentioned in the informed consent (Hospital Dos de Mayo, Hospital Regional de Ica and Clinica Oncosalud-AUNA). This observational study involving human tissue samples was approved by the Medical Ethics Committee Via Libre (reference no. AUNA-20-03). Written informed consent was obtained from the patient.

Patient consent for publication

The patient was enrolled in a GC genomic study. The patient provided consent for the publication of his clinical data.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rawla P and Barsouk A: Epidemiology of gastric cancer: Global trends, risk factors and prevention. Prz Gastroenterol. 14:26–38. 2019.PubMed/NCBI View Article : Google Scholar

2 

International Agency for Research on Cancer; World Health Organization: World Source: Globocan 2020. Available from: https://gco.iarc.fr/today/data/factsheets/populations/900-wolrd-fact-sheet.pdf. Accessed on February 20, 2022.

3 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar

4 

Castaneda CA, Castillo M, Chavez I, Barreda F, Suarez N, Nieves J, Bernabe LA, Valdivia D, Ruiz E, Dias-Neto E, et al: Prevalence of helicobacter pylori infection, its virulent genotypes, and Epstein-Barr virus in peruvian patients with chronic gastritis and gastric cancer. J Glob Oncol. 2019:1–9. 2019.PubMed/NCBI View Article : Google Scholar

5 

Nelen SD, Bosscha K, Lemmens VEPP, Hartgrink HH, Verhoeven RHA and de Wilt JHW: Morbidity and mortality according to age following gastrectomy for gastric cancer. Br J Surg. 105:1163–1170. 2018.PubMed/NCBI View Article : Google Scholar

6 

Jeong JR, Choi JW, Ryu SY and Choe YR: Relationship between frailty and mortality after gastrectomy in older patients with gastric cancer. J Geriatr Oncol. 13:67–73. 2022.PubMed/NCBI View Article : Google Scholar

7 

Lee KG, Lee HJ, Yang JY, Oh SY, Bard S, Suh YS, Kong SH and Yang HK: Risk factors associated with complication following gastrectomy for gastric cancer: Retrospective analysis of prospectively collected data based on the Clavien-Dindo system. J Gastrointest Surg. 18:1269–1277. 2014.PubMed/NCBI View Article : Google Scholar

8 

David H and Ilson MP: Microsatellite instability: A biomarker in gastric cancer. NEJM J Watch: Sep 12, 2019 (Epub ahead of print).

9 

Mohanty S, Rosenthal RA, Russell MM, Neuman MD, Ko CY and Esnaola NF: Optimal perioperative management of the geriatric patient: A best practices guideline from the American college of surgeons NSQIP and the American geriatrics society. J Am Coll Surg. 222:930–947. 2016.PubMed/NCBI View Article : Google Scholar

10 

Feldman AT and Wolfe D: Tissue processing and hematoxylin and eosin staining. Methods Mol Biol. 1180:31–43. 2014.PubMed/NCBI View Article : Google Scholar

11 

Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, Schnall-Levin M, White J, Sanford EM, An P, et al: Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 31:1023–1031. 2013.PubMed/NCBI View Article : Google Scholar

12 

Choi YY, Kim H, Shin SJ, Kim HY, Lee J, Yang HK, Kim WH, Kim YW, Kook MC, Park YK, et al: Microsatellite instability and programmed cell death-ligand 1 expression in stage II/III gastric cancer: Post hoc analysis of the classic randomized controlled study. Ann Surg. 270:309–316. 2019.PubMed/NCBI View Article : Google Scholar

13 

Haag GM, Czink E, Ahadova A, Schmidt T, Sisic L, Blank S, Heger U, Apostolidis L, Berger AK, Springfeld C, et al: Prognostic significance of microsatellite-instability in gastric and gastroesophageal junction cancer patients undergoing neoadjuvant chemotherapy. Int J Cancer. 144:1697–1703. 2019.PubMed/NCBI View Article : Google Scholar

14 

Hashimoto T, Kurokawa Y, Takahashi T, Miyazaki Y, Tanaka K, Makino T, Yamasaki M, Nakajima K, Ikeda JI, Mori M and Doki Y: Predictive value of MLH1 and PD-L1 expression for prognosis and response to preoperative chemotherapy in gastric cancer. Gastric Cancer. 22:785–792. 2019.PubMed/NCBI View Article : Google Scholar

15 

Kim SY, Choi YY, An JY, Shin HB, Jo A, Choi H, Seo SH, Bang HJ, Cheong JH, Hyung WJ and Noh SH: The benefit of microsatellite instability is attenuated by chemotherapy in stage II and stage III gastric cancer: Results from a large cohort with subgroup analyses. Int J Cancer. 137:819–825. 2015.PubMed/NCBI View Article : Google Scholar

16 

Smyth EC, Wotherspoon A, Peckitt C, Gonzalez D, Hulkki-Wilson S, Eltahir Z, Fassan M, Rugge M, Valeri N, Okines A, et al: Mismatch repair deficiency, microsatellite instability, and survival: An exploratory analysis of the medical research council adjuvant gastric infusional chemotherapy (MAGIC) trial. JAMA Oncol. 3:1197–1203. 2017.PubMed/NCBI View Article : Google Scholar

17 

Ellegren H: Microsatellites: Simple sequences with complex evolution. Nat Rev Genet. 5:435–445. 2004.PubMed/NCBI View Article : Google Scholar

18 

Ratti M, Lampis A, Hahne JC, Passalacqua R and Valeri N: Microsatellite instability in gastric cancer: Molecular bases, clinical perspectives, and new treatment approaches. Cell Mol Life Sci. 75:4151–4162. 2018.PubMed/NCBI View Article : Google Scholar

19 

Hudler P: Genetic aspects of gastric cancer instability. ScientificWorldJournal. 2012(761909)2012.PubMed/NCBI View Article : Google Scholar

20 

Harfe BD and Jinks-Robertson S: DNA mismatch repair and genetic instability. Annu Rev Genet. 34:359–399. 2000.PubMed/NCBI View Article : Google Scholar

21 

Genschel J, Littman SJ, Drummond JT and Modrich P: Isolation of MutSbeta from human cells and comparison of the mismatch repair specificities of MutSbeta and MutSalpha. J Biol Chem. 273:19895–19901. 1998.PubMed/NCBI View Article : Google Scholar

22 

De Wind N, Dekker M, Claij N, Jansen L, van Klink Y, Radman M, Riggins G, van der Valk M, van't Wout K and te Riele H: HNPCC-like cancer predisposition in mice through simultaneous loss of Msh3 and Msh6 mismatch-repair protein functions. Nat Genet. 23:359–362. 1999.PubMed/NCBI View Article : Google Scholar

23 

Miao HK, Chen LP, Cai DP, Kong WJ, Xiao L and Lin J: MSH3 rs26279 polymorphism increases cancer risk: A meta-analysis. Int J Clin Exp Pathol. 8:11060–11067. 2015.PubMed/NCBI

24 

Ni H, Jiang B, Zhou Z, Yuan X, Cao X, Huang G and Li Y: Inactivation of MSH3 by promoter methylation correlates with primary tumor stage in nasopharyngeal carcinoma. Int J Mol Med. 40:673–678. 2017.PubMed/NCBI View Article : Google Scholar

25 

Van Oers JMM, Edwards Y, Chahwan R, Zhang W, Smith C, Pechuan X, Schaetzlein S, Jin B, Wang Y, Bergman A, et al: The MutSβ complex is a modulator of p53-driven tumorigenesis through its functions in both DNA double strand break repair and mismatch repair. Oncogene. 33:3939–3946. 2014.PubMed/NCBI View Article : Google Scholar

26 

Haugen AC, Goel A, Yamada K, Marra G, Nguyen TP, Nagasaka T, Kanazawa S, Koike J, Kikuchi Y, Zhong X, et al: Genetic instability caused by loss of MutS homologue 3 in human colorectal cancer. Cancer Res. 68:8465–8472. 2008.PubMed/NCBI View Article : Google Scholar

27 

Lee S, Chung H, Devaraj B, Iwaizumi M, Han HS, Hwang DY, Seong MK, Jung BH and Carethers JM: Microsatellite alterations at selected tetranucleotide repeats are associated with morphologies of colorectal neoplasias. Gastroenterology. 139:1519–1525. 2010.PubMed/NCBI View Article : Google Scholar

28 

Wang K, Yuen ST, Xu J, Lee SP, Yan HHN, Shi ST, Siu HC, Deng S, Chu KM, Law S, et al: Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet. 46:573–582. 2014.PubMed/NCBI View Article : Google Scholar

29 

Guo YA, Chang MM, Huang W, Ooi WF, Xing M, Tan P and Skanderup AJ: Mutation hotspots at CTCF binding sites coupled to chromosomal instability in gastrointestinal cancers. Nat Commun. 9(1520)2018.PubMed/NCBI View Article : Google Scholar

30 

Zhu L, Li Z, Wang Y, Zhang C, Liu Y and Qu X: Microsatellite instability and survival in gastric cancer: A systematic review and meta-analysis. Mol Clin Oncol. 3:699–705. 2015.PubMed/NCBI View Article : Google Scholar

31 

Seo JY, Jin EH, Jo HJ, Yoon H, Shin CM, Park YS, Kim N, Jung HC and Lee DH: Clinicopathologic and molecular features associated with patient age in gastric cancer. World J Gastroenterol. 21:6905–6913. 2015.PubMed/NCBI View Article : Google Scholar

32 

Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014.PubMed/NCBI View Article : Google Scholar

33 

Angell HK, Lee J, Kim KM, Kim K, Kim ST, Park SH, Kang WK, Sharpe A, Ogden J, Davenpor A, et al: PD-L1 and immune infiltrates are differentially expressed in distinct subgroups of gastric cancer. Oncoimmunology. 8(e1544442)2019.PubMed/NCBI View Article : Google Scholar

34 

van Velzen MJM, Derks S, van Grieken NCT, Mohammad NH and van Laarhoven HWM: MSI as a predictive factor for treatment outcome of gastroesophageal adenocarcinoma. Cancer Treat Rev. 86(102024)2020.PubMed/NCBI View Article : Google Scholar

35 

Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, et al: Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 372:2018–2028. 2015.PubMed/NCBI View Article : Google Scholar

36 

Gogas H, Dréno B, Larkin J, Demidov L, Stroyakovskiy D, Eroglu Z, Ferrucci PF, Pigozzo J, Rutkowski P, Mackiewicz J, et al: Cobimetinib plus atezolizumab in BRAFV600 wild-type melanoma: Primary results from the randomized phase III IMspire170 study. Ann Oncol. 32:384–394. 2021.PubMed/NCBI View Article : Google Scholar

37 

Barroso-Sousa R, Keenan TE, Pernas S, Exman P, Jain E, Garrido-Castro AC, Hughes M, Bychkovsky B, Umeton R, Files JL, et al: Tumor mutational burden and PTEN alterations as molecular correlates of response to PD-1/L1 blockade in metastatic triple-negative breast cancer. Clin Cancer Res. 26:2565–2572. 2020.PubMed/NCBI View Article : Google Scholar

38 

Kim J, Kim B, Kang SY, Heo YJ, Park SH, Kim ST, Kang WK, Lee J and Kim KM: Tumor mutational burden determined by panel sequencing predicts survival after immunotherapy in patients with advanced gastric cancer. Front Oncol. 10(314)2020.PubMed/NCBI View Article : Google Scholar

39 

Rizzo A, Ricci AD and Brandi G: PD-L1, TMB, MSI, and other predictors of response to immune checkpoint inhibitors in biliary tract cancer. Cancers (Basel). 13(558)2021.PubMed/NCBI View Article : Google Scholar

40 

Kim JY, Kronbichler A, Eisenhut M, Hong SH, van der Vliet HJ, Kang J, Shin JI and Gamerith G: Tumor mutational burden and efficacy of immune checkpoint inhibitors: A systematic review and meta-analysis. Cancers (Basel). 11(1798)2019.PubMed/NCBI View Article : Google Scholar

41 

Wang F, Wei XL, Wang FH, Xu N, Shen L, Dai GH, Yuan XL, Chen Y, Yang SJ, Shi JH, et al: Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann Oncol. 30:1479–1486. 2019.PubMed/NCBI View Article : Google Scholar

42 

Guo X, Liang X, Wang Y, Cheng A, Zhang H, Qin C and Wang Z: Significance of tumor mutation burden combined with immune infiltrates in the progression and prognosis of advanced gastric cancer. Front Genet. 12(642608)2021.PubMed/NCBI View Article : Google Scholar

43 

Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, Liu XQ, Sher X, Jung H, Lee M, et al: Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. 24:1449–1458. 2018.PubMed/NCBI View Article : Google Scholar

44 

Takeda M, Takahama T, Sakai K, Shimizu S, Watanabe S, Kawakami H, Tanaka K, Sato C, Hayashi H, Nonagase Y, et al: Clinical application of the foundationone CDx assay to therapeutic decision-making for patients with advanced solid tumors. Oncologist. 26:e588–e596. 2021.PubMed/NCBI View Article : Google Scholar

45 

Sunami K, Ichikawa H, Kubo T, Kato M, Fujiwara Y, Shimomura A, Koyama T, Kakishima H, Kitami M, Matsushita H, et al: Feasibility and utility of a panel testing for 114 cancer-associated genes in a clinical setting: A hospital-based study. Cancer Sci. 110:1480–1490. 2019.PubMed/NCBI View Article : Google Scholar

46 

Bilkey GA, Burns BL, Coles EP, Mahede T, Baynam G and Nowak KJ: Optimizing precision medicine for public health. Front Public Health. 7(42)2019.PubMed/NCBI View Article : Google Scholar

47 

Medrano-Guzmán R, Valencia-Mercado D, Luna-Castillo M, García-Ríos LE and González-Rodríguez D: Prognostic factors for survival in patients with resectable advanced gastric adenocarcinoma. Cir Cir. 84:469–476. 2016.PubMed/NCBI View Article : Google Scholar : (In Spanish).

48 

Garcia-Arispe JE, Loo-Valverde M, Chanduví W, Vela-Ruiz JM, Guillen-Ponce R and Cruz-Vargas DL: Anemia y supervivencia en pacientes con cáncer, un estudio peruano con seguimiento de 5 años. Rev Fac Med Hum Enero. 21:90–100. 2021.(In Spain).

49 

Littlewood T: The impact of hemoglobin levels on treatment outcomes in patients with cancer. Semin Oncol. 28:49–53. 2001.PubMed/NCBI View Article : Google Scholar

50 

Kim JJ, Tao H, Carloni E, Leung WK, Graham DY and Sepulveda AR: Helicobacter pylori impairs DNA mismatch repair in gastric epithelial cells. Gastroenterology. 123:542–553. 2002.PubMed/NCBI View Article : Google Scholar

51 

Silva-Fernandes IJDL, de Oliveira ES, Santos JC, Ribeiro ML, Ferrasi AC, Pardini MI, Burbano RMR and Rabenhorst SHB: The intricate interplay between MSI and polymorphisms of DNA repair enzymes in gastric cancer H.pylori associated. Mutagenesis. 32:471–478. 2017.PubMed/NCBI View Article : Google Scholar

52 

Machado AMD, Figueiredo C, Touati E, Máximo V, Sousa S, Michel V, Carneiro F, Nielsen FC, Seruca R and Rasmussen LJ: Helicobacter pylori infection induces genetic instability of nuclear and mitochondrial DNA in gastric cells. Clin Cancer Res. 15:2995–3002. 2009.PubMed/NCBI View Article : Google Scholar

53 

Koh FH, Chua JM, Tan JL, Foo FJ, Tan WJ, Sivarajah SS, Ho LML, Teh BT and Chew MH: Paradigm shift in gastrointestinal surgery-combating sarcopenia with prehabilitation: Multimodal review of clinical and scientific data. World J Gastrointest Surg. 13:734–755. 2021.PubMed/NCBI View Article : Google Scholar

54 

Kanda M: Preoperative predictors of postoperative complications after gastric cancer resection. Surg Today. 50:3–11. 2020.PubMed/NCBI View Article : Google Scholar

55 

Kanda M, Tanaka C, Murotani K, Kobayashi D, Ito S, Mochizuki Y, Ishigure K, Ishiyama A, Teramoto H, Murai T, et al: <Editors' Choice> Efficacy of enteral nutrients containing β-hydroxy-β-methylbutyrate, glutamine, and arginine for the patients with anastomotic leakage after gastrectomy: Study protocol of a multicenter phase II clinical trial. Nagoya J Med Sci. 80:351–355. 2018.PubMed/NCBI View Article : Google Scholar

56 

Sasahara M, Kanda M, Ito S, Mochizuki Y, Teramoto H, Ishigure K, Murai T, Asada T, Ishiyama A, Matsushita H, et al: The preoperative prognostic nutritional index predicts short-term and long-term outcomes of patients with stage II/III gastric cancer: Analysis of a multi-institution dataset. Dig Surg. 37:135–144. 2020.PubMed/NCBI View Article : Google Scholar

57 

Xue DD, Cheng Y, Wu M and Zhang Y: Comprehensive geriatric assessment prediction of postoperative complications in gastrointestinal cancer patients: A meta-analysis. Clin Interv Aging. 13:723–736. 2018.PubMed/NCBI View Article : Google Scholar

58 

Suwa K, Ushigome T, Ohtsu M, Narihiro S, Ryu S, Shimoyama Y, Okamoto T and Yanaga K: Risk factors for early postoperative small bowel obstruction after anterior resection for rectal cancer. World J Surg. 42:233–238. 2018.PubMed/NCBI View Article : Google Scholar

59 

Barmparas G, Branco BC, Schnüriger B, Oliver M, Konstantinidis A, Lustenberger T, Eberle BM, Inaba K and Demetriades D: In-hospital small bowel obstruction after exploratory laparotomy for trauma. J Trauma. 71:486–490. 2011.PubMed/NCBI View Article : Google Scholar

60 

Ellozy SH, Harris MT, Bauer JJ, Gorfine SR and Kreel I: Early postoperative small-bowel obstruction: A prospective evaluation in 242 consecutive abdominal operations. Dis Colon Rectum. 45:1214–1217. 2002.PubMed/NCBI View Article : Google Scholar

61 

Khoraki J, Mazzini GS, Shah AS, Del Prado PAR, Wolfe LG and Campos GM: Early small bowel obstruction after laparoscopic gastric bypass: A surgical emergency. Surg Obes Relat Dis. 14:1118–1125. 2018.PubMed/NCBI View Article : Google Scholar

62 

Nakamura T, Sato T, Naito M, Ogura N, Yamanashi T, Miura H, Tsutsui A, Yamashita K and Watanabe M: Laparoscopic surgery is useful for preventing recurrence of small bowel obstruction after surgery for postoperative small bowel obstruction. Surg Laparosc Endosc Percutan Tech. 26:e1–e4. 2016.PubMed/NCBI View Article : Google Scholar

63 

Badia-Tahull MB, Llop-Talaveron J, Fort-Casamartina E, Farran-Teixidor L, Ramon-Torrel JM and Jódar-Masanés R: Preoperative albumin as a predictor of outcome in gastrointestinal surgery. E Spen Eur E J Clin Nutr Metab. 4:e248–e251. 2009.

64 

Alberda C, Graf A and McCargar L: Malnutrition: Etiology, consequences, and assessment of a patient at risk. Best Pract Res Clin Gastroenterol. 20:419–439. 2006.PubMed/NCBI View Article : Google Scholar

65 

Morse BC, Simpson JP, Jones YR, Johnson BL, Knott BM and Kotrady JA: Determination of independent predictive factors for anastomotic leak: Analysis of 682 intestinal anastomoses. Am J Surg. 206:950–956. 2013.PubMed/NCBI View Article : Google Scholar

66 

Meza JN, Ortiz FM and Segura ER: Hospital malnutrition in a low-resources country: A survey and critique of the status of the published available literature. Clin Nutr. 34:330–331. 2015.PubMed/NCBI View Article : Google Scholar

67 

Baca DA, Rego FR, Lazo MS, Rique IA and Kano PL: Estado nutricional y morbi-mortalidad en pacientes con anastomosis gastrointestinales en el hospital nacional hipolito unanue (HNHU). Rev Gastroenterol Perú. 32:273–280. 2012.PubMed/NCBI

68 

Robles Valcárcel P and Seclén MPT: Nutritional status morbility and mortality in patients with gastrointestinal anastomosis in the ‘Hospital Nacional Hipolito Unanue’ (HNHU). Rev Gastroenterol Peru. 32:273–280. 2012.PubMed/NCBI(In Spanish).

69 

Pinto JA, Pinillos L, Villarreal-Garza C, Morante Z, Villarán MV, Mejía G, Caglevic C, Aguilar A, Fajardo W, Usuga F, et al: Barriers in Latin America for the management of locally advanced breast cancer. Ecancermedicalscience. 13(897)2019.PubMed/NCBI View Article : Google Scholar

70 

Li WH, Zhang JY, Liu WH and Chen XX: Role of the initial degree of anaemia and treatment model in the prognosis of gastric cancer patients treated by chemotherapy: A retrospective analysis. BMC Cancer. 20(414)2020.PubMed/NCBI View Article : Google Scholar

71 

Puliga E, Corso S, Pietrantonio F and Giordano S: Microsatellite instability in gastric cancer: Between lights and shadows. Cancer Treat Rev. 95(102175)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2023
Volume 18 Issue 3

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Alfaro A, Zanabria D, Aguilar A, Jimenez‑Solano SA, Zevallos A and Fajardo W: Gastric adenocarcinoma with high‑level microsatellite instability: A case report. Mol Clin Oncol 18: 16, 2023
APA
Alfaro, A., Zanabria, D., Aguilar, A., Jimenez‑Solano, S.A., Zevallos, A., & Fajardo, W. (2023). Gastric adenocarcinoma with high‑level microsatellite instability: A case report. Molecular and Clinical Oncology, 18, 16. https://doi.org/10.3892/mco.2023.2612
MLA
Alfaro, A., Zanabria, D., Aguilar, A., Jimenez‑Solano, S. A., Zevallos, A., Fajardo, W."Gastric adenocarcinoma with high‑level microsatellite instability: A case report". Molecular and Clinical Oncology 18.3 (2023): 16.
Chicago
Alfaro, A., Zanabria, D., Aguilar, A., Jimenez‑Solano, S. A., Zevallos, A., Fajardo, W."Gastric adenocarcinoma with high‑level microsatellite instability: A case report". Molecular and Clinical Oncology 18, no. 3 (2023): 16. https://doi.org/10.3892/mco.2023.2612