Next Article in Journal
Overview of the Ultrasound Classification Systems in the Field of Thyroid Cytology
Next Article in Special Issue
Gastrointestinal Adenocarcinoma Incidence and Survival Trends in South Australia, 1990–2017
Previous Article in Journal
Deregulated miRNAs Contribute to Silencing of B-Cell Specific Transcription Factors and Activation of NF-κB in Classical Hodgkin Lymphoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Clinical, Molecular and Genetic Characteristics of Early Onset Gastric Cancer: Analysis of a Large Multicenter Study

1
Gastroenterology Department, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain
2
Gastroenterology Department, Hospital del Mar, 08003 Barcelona, Spain
3
Gastroenterology Department, Hospital Universitari de Bellvitge, 08097 Barcelona, Spain
4
Department Pathology, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain
5
Gastroenterology Department, Biodonostia Health Research Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(13), 3132; https://doi.org/10.3390/cancers13133132
Submission received: 17 March 2021 / Revised: 18 June 2021 / Accepted: 18 June 2021 / Published: 23 June 2021
(This article belongs to the Special Issue Young-Onset GI Cancer)

Abstract

:

Simple Summary

Gastric cancer is one of the most common cancers worldwide, showing high mortality rates. A small portion of gastric cancer patients, known as early onset gastric cancer (EOGC) patients, develop the disease before age 50, and their characteristics are poorly described. Thus, our main objective was to describe the clinical, molecular, and genetic characteristics of EOGC in a large multicenter cohort of patients. We were able to identify that most EOGC cases have similar characteristics: diagnosed at advanced stage, diffuse type, and infrequent DNA mismatch repair somatic deficiency. Although familial aggregation of gastric cancer was uncommon, a germline genetic mutation was identified in 25% of the patients tested. Our results show that EOGC has a marked genetic heterogeneity. Thus, it is essential to consider familial history of tumors, not only GC, in order to select adequate patients to perform a suitable genetic counseling and enhance the emerging use of multigene panels.

Abstract

Gastric adenocarcinoma (GC) is a common tumor with high morbidity and mortality. Only 7% of patients with GC are diagnosed before age 50 (early onset gastric cancer (EOGC)), and their characteristics have been poorly described. We aimed to describe clinical, molecular, and genetic characteristics of EOGC. A total of 309 patients with EOGC were retrospectively studied in four Spanish centers. Personal information, family history, and tumor information were registered. Germinal genetic analysis was performed in patients who met current criteria of a hereditary syndrome at the time of diagnosis. The median age at diagnosis was 44 years. The majority (73.3%) of tumors were diffuse, and 78.3% were diagnosed in an advanced stage. Familial aggregation of GC was present in 18/117 (15.4%) cases, and 5/117 (4.3%) met criteria for familial GC. MMR-IHC was performed in 126/309 (40.7%) tumors: 4/126 (3.1%) had loss of expression in MLH1/PMS2, without an associated germline mutation. Sixteen germline genetic analyses were performed, detecting a pathogenic variant in four (25%) cases: one in BRCA2, one in TP53, and two in CDH1. Most EOGC are diffuse and diagnosed in an advanced stage. In these patients, DNA MMR system deficiency is uncommon. Although familial aggregation was observed in only 15% of cases, a germline mutation was found in 25% of patients tested with clinical criteria. This demonstrates that EOGC has a marked genetic heterogeneity, reinforcing the importance of an accurate genetic counseling and enhancing the emerging use of multigene panels.

1. Introduction

Gastric cancer (GC) is the fifth most common and the third most deadly cancer in the world [1], representing a worldwide health problem [2]. The average age at diagnosis is 60 years, only 7% occur before age 50 and 2% before age 40 [3]. The etiology of GC is multifactorial, with Helicobacter pylori, diet factors, and tobacco being the main environmental agents implicated in its pathogenesis [4]. Although most GC cases are sporadic, a familial aggregation is observed in approximately 10% of cases, with an underlying genetic cause identified in up to 5% of all GC [5]. Familial characteristics that suggest a hereditary predisposition include the existence of several affected family members, an autosomal dominant pattern of inheritance, disease presentation at young ages, and association with other extra-gastric neoplasms [6].
In terms of these assumptions, there are mainly three clinical situations where familial predisposition to GC may be found [7]. First, hereditary syndromes with higher risk for GC, including two entities: gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS) [8], and the most common inherited GC syndrome, hereditary diffuse gastric cancer (HDGC). It is characterized by two or more cases of GC at any age in first or second relatives, with at least one confirmed diffuse gastric cancer (DGC); or personal history of DGC before the age of 50; or personal or family history (first- or second-degree relatives) of DGC and lobular breast cancer, with one being diagnosed before 70 years [9]. This syndrome is mainly caused by CDH1 germline mutations, which encode the tumor suppressor protein E-cadherin. However, during the last years, another gene, CTNNA1, has also been identified in HDGC families [10].
Other clinical situations are hereditary syndromes with higher risk for GC and other tumors, including Lynch syndrome and, less commonly, familial adenomatous polyposis (FAP), Peutz–Jeghers syndrome, juvenile polyposis, Li–Fraumeni syndrome, and Cowden syndrome [11], with germline mutations in different genes. The last clinical situation is familial intestinal gastric cancer (FIGC) characterized by familial aggregation of intestinal GCs without an identified inherited cause. FIGC is defined as two or more cases of GC in first-degree (FDR) or second-degree relatives (SDR), with at least one confirmed case of intestinal histology in someone younger than 50 years, or three or more confirmed cases of intestinal GC in FDR or SDR, regardless of age [12,13].
In spite of this, the genetic cause is not identified in a high percentage of GC patients [14,15]. This fact is especially important in early-onset gastric cancer cases (EOGC) because 90% of these young patients do not have a family history, hampering identification and early diagnosis [16]. The remaining 10% of EOGC cases that have a family history are explained by the previously mentioned hereditary syndromes.
The definition of early onset gastric cancer (EOGC) varies across studies, but one of the most accepted definitions includes those diagnosed at the age of 50 or younger. Although the incidence of GC is declining globally, EOGC is increasing [17]. In fact, a recent study has reported that nowadays EOGC comprises up to 30% of all cases of GC in the United States [18]. EOGC has been associated with some clinical and pathological characteristics, such as predomination of diffuse histology and infrequent association with intestinal metaplasia [19,20]. Moreover, EOGC is usually diagnosed in an advanced stage, associated with a high mortality. However, clinical and molecular features of EOGC have been poorly described [16].
Identification of individuals at high risk of GC allows us to establish preventive measures, early diagnosis, and personalized treatments; thus, we aimed to describe the clinical, molecular, and genetic characteristics of EOGC (≤50 years) in order to identify high-risk forms of GC.

2. Materials and Methods

2.1. Study Population

Patients with GC diagnosed before 51 years old were retrospectively studied at four centers in Spain between 1999 and 2018. The study was approved by the Institutional Review Board (or Ethics Committee) of Hospital Clínic in Barcelona (register number 2015/0153, date of approval 22/04/2015).
Clinical and demographic data were evaluated through electronic clinical reports, including environmental risks factors such as tobacco consumption, alcohol intake, and Helicobacter pylori infection.
Personal and family history of GC and other tumors related with hereditary syndromes (i.e., HDGC, Peutz–Jeghers syndrome, Lynch syndrome, familial adenomatous polyposis, HBOC, juvenile polyposis, and Li–Fraumeni syndrome) were registered (including FDRs and SDRs). Patients who met criteria of familial GC were also identified.

2.2. Tumor Characteristics

The tumor characteristics were analyzed by histological report. The location, diagnostic stage (TNM), histologic features (intestinal, diffuse, or mixed), and grade of differentiation were considered.
Tumor mismatch repair (MMR) deficiency was evaluated by immunostaining including analysis of MLH1, MSH2, MSH6, and PMS2 protein expression, as previously described [21].

2.3. Germline Genetic Analysis

Germline genetic testing was performed on genomic DNA isolated from peripheral blood leukocytes by both multiple ligation probe amplification analysis and direct sequencing. The analysis was performed through a commercial multigene panel (Trusight Cancer v1, Illumina Inc., San Diego, CA, USA) involving the most frequent genes related to GC germline predisposition (MLH1, MSH2, MSH6, PMS2, CDH1, EPCAM, BRCA1, BRCA2, PALB2, TP53, APC, MUTYH, STK11, SMAD4/BMPR1A, PTEN).
The genetic test was performed in patients with available germline DNA who fulfilled the diagnostic criteria of a hereditary syndrome related to GC at the time of diagnosis or in whom the tumoral analysis of DNA mismatch repair proteins was altered (i.e., loss of protein expression of MLH1, MSH2, MSH6, or PMS2) [13].

2.4. Statistical Analysis

All data were analyzed using the 22.0 SPSS software package (IBM SPSS Statistics for Window, Version 22.0. Armonk, NY: IBM Corp.).
Baseline characteristics were described in percentages for categorical data, using median, range, and interquartile range (IQR). When information was missing, the denominator was accordingly to patients with available data. Univariate binary logistic regression was performed for selection of variables associated with the diagnosis of a hereditary cancer. For multivariable logistic regression analyses, only candidate variables with p-values of ≤0.05 on univariate analysis were used in the final multivariate model. Odds ratios (ORs) with 95% confidence intervals (CIs) were included to quantify the magnitude of the association.

3. Results

3.1. General Characteristics

Three hundred and nine patients with EOGC were included. Clinico-pathological features of patients included in the study are summarized in Table 1.
The median age at diagnosis was 44 years old (IQR 40-48, range 33), with a predominance of men, with 191 (61.8%) cases. Related to environmental risk factors, 77/169 (45%) were smokers and 21/105 (20%) had chronic alcohol consumption, whereas in 24/82 (29%) cases, a Helicobacter pylori infection was detected. Most of the patients were from Spain; however, 8/309 (2.6%) were from South America, 4/309 (1.3%) were from Asia, and 4/309 (1.3%) were South African.
Out of the 309 (2.9%) patients, 9 had previously developed other tumors, including breast and/or ovarian in 5/309 (1.6%), lung in 1/309 (0.3%), cervix in 1/309 (0.3%), thyroid in 1/309 (0.3%), and Hodgkin lymphoma in 1/309 (0.3%).
A total of 18/117 (15.4%) patients presented familial aggregation of GC (≥1 FDR or SDR affected), and 5/117 (4.3%) met criteria for FIGC. A total of 67/117 (57.2%) patients had family history of cancers related with a hereditary syndrome, mainly ovarian and/or breast in 15/117 (12.8%), followed by colorectal cancer in 8/117 (6.8%) cases and other types of tumors in 39/117 (34%). Detailed characteristics are described in Table 1.

3.2. Tumor Characteristics

The predominant tumor histology was diffuse, observed in 118/161 (73.3%) of the cases, and the signet ring cell subtype was detected in 38/118 (32%). Among patients with diffuse GC and H. pylori information available, in 8/45 (17.8%), the infection was present. Among patients with intestinal GC subtype with H. pylori status available, 3/14 (21.4%) were infected. No statistically significant differences regarding H. pylori infection and histology subtype were found (p = 0.09). According to the WHO classification (2019), the degree of differentiation only applies to the intestinal GC, and thus within this subtype, 9/43 (39.1%) were poorly differentiated tumors. Regarding tumor location, the most common sites were the body in 111/203 (55%) and antrum in 50/203 (25%). An advanced stage (III/IV) at diagnosis was present in 166/212 (78.3%) cases, and only in 44/212 (20.8%) was the diagnosis at an early stage (I/II).
In 122/205 (59.5%) cases, surgery with or without chemotherapy was performed; 67/205 (32.7%) patients were treated with chemotherapy (CT) +/− radiotherapy (RT) alone, and 16/205 (7.8%) patients did not receive a specific oncological treatment.
The immunohistochemistry of DNA mismatch repair proteins (MMR-IHC) was performed in 126 out of 309 (40.7%) tumors, and only 4/126 (3.1%) showed loss of protein expression, specifically MLH1/PMS2 (Figure 1). The tumor and MMR-IHC characteristics are shown in Table 2 and Figure 2, respectively.
Regarding survival, with a median follow up of 7.6 years (IQR 17-38), a 5-year survival rate of 32.6% was observed, with a significant difference based on clinical stage (stage I–II 87% vs. stage III–IV 11.3%, p = 0.0001; Figure 3), and diffuse histology was associated with worse prognosis (p = 0.019); no differences in gender, age, family history of GC, H. pylori infection, smoking or alcohol consumption, tumor differentiation grade, or MMR-IHC were detected.

3.3. Germline Genetic Analysis

Genetic analysis was performed in the 16 patients with available germline DNA out of the 44 patients that fulfilled clinical criteria of germline testing. Among them, in 11/16 (68.7%) patients, the analysis was performed due to the fulfillment of criteria for HDGC, in 2/16 patients because they met criteria of HBOC; and in 3/16 cases, the genetic analysis was performed on the basis of a somatic loss of expression of MLH1/PMS2 proteins at IHC. In the remaining 28/44 patients, the analysis was not performed because the DNA was not available.
A germline genetic mutation was identified in 4/16 (25%) cases, one at 49 years old (with personal history of GC and breast cancer) and another three with GC younger than 41 years old. The mutated genes detected were BRCA2 and TP53, and in two cases, CDH1 (Table 3). Integrative Genomics Viewer was used for visualization of these variants; Figure S1 shows an example of two of these variants. None of those patients had tumors with loss of expression in DNA mismatch repair protein (MMR), and only one of them reported family history of GC. Within four patients with an altered MMR-IHC, the germline genetic analysis performed in three patients did not identify any pathogenic variant, and in the remaining patient, the germline analysis could not be performed because he died before the MMR-IHC was done.

3.4. Factors Associated with High Risk of Gastric Cancer

Personal characteristics as well as family history were analyzed in order to identify risk factors of a hereditary GC syndrome (germline mutation identified). No statistically significant factor associated with the presence of a germline mutation was identified. However, family history of other neoplasms showed a trend towards statistical significance with p = 0.057.

4. Discussion

It is well known that EOGC has clinical and pathological differences with older onset GC [22], although their clinical and molecular characteristics have been poorly reported [16]. In the present study, we describe clinical, molecular, and genetic characteristics of 309 EOGC patients. Our study shows that most of EOGC are histologically diffuse (73%, in comparison with 32% in older patients), poorly differentiated, and diagnosed at an advanced stage, supporting what has been previously described in other studies [18,19,20,23]. Moreover, as the already reported trend of increasing rate in general population [24] of proximal GC over the distal location, we observed in our study that more than half of the tumors were located in the gastric body. This could be explained not only due to the diffuse histology, but also because the low incidence of Helicobacter pylori infection (less than 30%) and a high proportion (almost 60%) of patients with positive oncological family history, suggesting a different carcinogenesis process.
Although there are known characteristics of EOGC, there are some inconsistent data between studies, i.e., some studies reported a female predominance [23], while in others there was an increasing trend for males or without a significant difference between genders [25,26]. In the present study, a male predominance was identified.
On the basis of different features, our study attempted to deepen in the clinical and molecular characterization of EOGC with the ultimate goal of being able to identify high-risk individuals and establish preventive measures, early diagnosis, and personalized treatments.
Lynch syndrome (LS), one of the most common cancer hereditary syndromes, carries a cumulative risk of GC of 11–19% [27,28]. However, DNA mismatch repair deficiency is exceptional in GC [29]. In order to consider the diagnoses of possible LS, we analyzed the MMR system deficiency, observing that loss of protein expression was an infrequent event, and only 4/126 (3.1%) patients displayed it. The low incidence of MMR deficiency in EOGC is probably related to the high proportion of diffuse tumors, wherein MSI is less common [30] and also related to the fact that genomically stable tumors are usually diagnosed at an earlier age [31]. Moreover, in cases with MMR deficiency, we did not find a correlation with a germline mutation, suggesting a somatic loss origin due to hypermethylation of the MLH1 promoter gene [32]. Thus, based upon our results, in this subgroup of patients, systematically analysis of MMR deficiency to rule out Lynch syndrome through IHC is likely not very useful.
Germline analysis was performed in 16 patients, representing 36% of patients who met clinical criteria for genetic testing according with the current guideline at that moment of the study (i.e., 2018–2019) [13]. Despite the fact that genetic testing could not be performed in the whole cohort of patients (due to loss of follow up or death), we found a germline mutation in 25% of tested patients. These mutations were located on BRCA2, CDH1, and TP53 genes. Both patients with CDH1 germline mutations displayed familial history of GC, while patients with BRCA2 and TP53 germline mutations showed familial history of ovarian and breast cancer, and breast and colorectal cancer, respectively. Thus, although familial aggregation of GC was present in only 15% of cases and the majority of patients with a germline mutation did not have familial aggregation of GC, family history of other tumors related with a hereditary syndrome was common. Therefore, this observation reinforces that, although family history of GC is poorly predictive, a very accurate medical personal and family history including any tumor type is mandatory in order to select the appropriate candidates for genetic testing.
Analyzing other studies, Tedaldi et al. focused on 96 patients that fulfilled different criteria such as HDGC criteria, suspected Lynch syndrome, familial aggregation, or patients with polyps and family history of GC. They sequenced 94 genes involved in cancer predisposition, identifying eight different CDH1 pathogenic/likely patogenic mutations in nine different patients with DGC and a mean age of almost 40 years. Although they identified more variants in other genes, their carriers were patients with more than 50 years old, and therefore they cannot be considered as an early onset cohort [33]. Moreover, a Canadian cohort was studied using single-site and multi-gene panels. The authors identified mutations in CDH1 and BRCA2 in five and two patients with DGC before the age of 50, respectively. Comparing with the present study, similar results were obtained, although the TP53 gene was not identified in this cohort [34]. However, a study performed by Vogelaar et al. did not identify any mutation neither in CDH1 nor in CTNNA1 in a cohort of 54 GC EOGC patients [35].
The relevance of the diagnosis of a hereditary syndrome is the opportunity to establish prevention and early diagnosis measures. For example, specifically in the context of CDH1 mutation carriers, most asymptomatic individuals do not have macroscopic lesions on endoscopic examinations; however, intramucosal foci of gastric cancer, usually multiple, are observed in the surgical specimens. Therefore, it is recommended that one perform prophylactic total gastrectomy in carriers of a pathogenic variant who are older than 20 years [36,37]. Annual endoscopic screening is reserved for individuals who do not accept prophylactic gastrectomy, patients with a variant of uncertain significance, and patients in whom the germline mutation has not been identified. This point is reflected in our cohort, wherein the two patients with a CDH1 mutation were diagnosed at stage IV (metastatic). In one case, two relatives were carriers of the mutation, with normal upper gastroscopy but with multiple focuses of diffuse adenocarcinoma, both in early stages (T1a and T1b, N0, M0). In the second patient, no additional CDH1 mutation carriers were identified.
This study has some limitations, most of them because the data obtained were collected retrospectively, which implies potential inclusion biases and the difficulty to obtain the information of some variables: Epstein–Barr virus infection; associated gastritis; HER-2 or PDL1 status; and, as mentioned previously, germline testing, which was only evaluated in 16/44 (36%) of patients who met clinical criteria for genetic testing, and CTNNA1 was not performed. Moreover, it is important to mention that H. pylori status was available in only 82 cases, being positive in 24 of them (29%); however, no difference regarding H. pylori prevalence among diffuse and intestinal histology was detected. In this context, other studies have been focused on the role of H. pylori infection and EOGC development, suggesting that it is important for tumor development [24], but to a lesser extent than in older GC patients [38]. Moreover, Rugge, et al. confirmed that H. pylori infection was significantly associated with both diffuse and intestinal histotypes [39]. In this sense, the low prevalence of H. pylori in our cohort, and the lack of differences in H. pylori status between histotypes, suggest that the implication of this infection in the carcinogenesis process is less relevant in EOGC, although prospective and large cohorts are needed to deepen in this observation.
The main strength of our study is that it describes not only clinical but also histological and molecular data of a large cohort of more than 300 patients with EOGC, although we are aware of the limitations associated to an observational and retrospective study.

5. Conclusions

Our results show that that most early onset GC cases are diagnosed in advanced stage, have diffuse histology, and have infrequent DNA mismatch repair somatic deficiency. Moreover, early onset GC has a marked genetic heterogeneity. Thus, it is essential to consider familial history of tumors, not only GC, but also and more importantly other tumors related with hereditary syndromes (such as colorectal, breast, and ovarian cancer), in order to select adequate patients to perform a suitable genetic counseling and enhance the emerging use of multigene panels.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cancers13133132/s1, Figure S1: Example of visualization using Integrative Genomics Viewer of two of the germline variants identified. (a) CDH1 c.2164+5G>C (splicing variant); (b) TP53 c.365_366delTG (p.Val122fs; frameshift variant).

Author Contributions

Conceptualization, L.M., A.P., S.C., C.H.-P.; methodology, L.M., A.P., S.C., C.H.-P.; formal analysis, A.P., C.H.-P., L.M.; investigation, A.P., S.C., L.C., J.M.B., M.C., T.O., L.M., C.H.-P., A.S., J.L., L.B.; data curation, L.M., A.P., S.C., C.H.-P., F.B., L.B.; writing—original draft preparation, L.M., A.P., C.H.-P.; writing—review and editing, A.P., S.C., L.B., F.B., L.M.; supervision, L.M.; project administration, L.M.; funding acquisition, L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by CIBEREHD (grant 18PI03). CIBEREHD is funded by the Instituto de Salud Carlos III.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board (or Ethics Committee) of Hospital Clínic in Barcelona (register number 2015/0153, date of approval 22/04/2015).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer Incidence and Mortality Worldwide: Sources, Methods and Major Patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef]
  2. Gerdner, L.A. Individualized Music for Dementia: Evolution and Application of Evidence-Based Protocol. World J. Psychiatry 2012, 2, 26. [Google Scholar] [CrossRef] [PubMed]
  3. Kluijt, I.; Sijmons, R.H.; Hoogerbrugge, N.; Plukker, J.T.; De Jong, D.; Van Krieken, J.H.; Van Hillegersberg, R.; Ligtenberg, M.; Bleiker, E.; Cats, A. Familial Gastric Cancer: Guidelines for Diagnosis, Treatment and Periodic Surveillance. Fam. Cancer 2012, 11, 363–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yusefi, A.R.; Lankarani, K.B.; Bastani, P.; Radinmanesh, M.; Kavosi, Z. Risk Factors for Gastric Cancer: A Systematic Review. Asian Pac. J. Cancer Prev. 2018, 19, 591–603. [Google Scholar] [PubMed]
  5. Lott, P.C.; Carvajal-Carmona, L.G. Resolving Gastric Cancer Aetiology: An Update in Genetic Predisposition. Lancet Gastroenterol. Hepatol. 2018, 3, 874–883. [Google Scholar] [CrossRef]
  6. Llach, J.; Moreno, L.; Sánchez, A.; Herrera-Pariente, C.; Ocaña, T.; Cuatrecasas, M.; Rivero-Sánchez, L.; Moreira, R.; Díaz, M.; Jung, G.; et al. Genetic Counseling for Hereditary Gastric and Pancreatic Cancer in High-Risk Gastrointestinal Cancer Clinics: An Effective Strategy. Cancers 2020, 12, 2386. [Google Scholar] [CrossRef]
  7. Corso, G.; Roncalli, F.; Marrelli, D.; Carneiro, F.; Roviello, F. History, Pathogenesis, and Management of Familial Gastric Cancer: Original Study of John XXIII’s Family. Biomed. Res. Int. 2013, 2013, 385132. [Google Scholar] [CrossRef]
  8. Carneiro, F. Hereditary Gastric Cancer. Pathologe 2012, 33 (Suppl. 2), 231–234. [Google Scholar] [CrossRef]
  9. Blair, V.R.; McLeod, M.; Carneiro, F.; Coit, D.G.; D’Addario, J.L.; van Dieren, J.M.; Harris, K.L.; Hoogerbrugge, N.; Oliveira, C.; van der Post, R.S.; et al. Hereditary Diffuse Gastric Cancer: Updated Clinical Practice Guidelines. Lancet Oncol. 2020, 21, e386–e397. [Google Scholar] [CrossRef]
  10. Majewski, I.J.; Kluijt, I.; Cats, A.; Scerri, T.S.; De Jong, D.; Kluin, R.J.C.; Hansford, S.; Hogervorst, F.B.L.; Bosma, A.J.; Hofland, I.; et al. An α-E-Catenin (CTNNA1) Mutation in Hereditary Diffuse Gastric Cancer. J. Pathol. 2013, 229, 621–629. [Google Scholar] [CrossRef]
  11. Leoz, M.L.; Sánchez, A.; Carballal, S.; Ruano, L.; Ocaña, T.; Pellisé, M.; Castells, A.; Balaguer, F.; Moreira, L. Síndromes de Predisposición a Cáncer Gástrico y Cáncer Pancreático. Gastroenterol. Hepatol. 2016, 39, 481–493. [Google Scholar] [CrossRef]
  12. Oliveira, C.; Pinheiro, H.; Figueiredo, J.; Seruca, R.; Carneiro, F. Familial Gastric Cancer: Genetic Susceptibility, Pathology, and Implications for Management. Lancet Oncol. 2015, 16, e60–e70. [Google Scholar] [CrossRef]
  13. Stjepanovic, N.; Moreira, L.; Carneiro, F.; Balaguer, F.; Cervantes, A.; Balmaña, J.; Martinelli, E. Hereditary Gastrointestinal Cancers: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2019, 30, 1558–1571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Hansford, S.; Kaurah, P.; Li-Chang, H.; Woo, M.; Senz, J.; Pinheiro, H.; Schrader, K.A.; Schaeffer, D.F.; Shumansky, K.; Zogopoulos, G.; et al. Hereditary Diffuse Gastric Cancer Syndrome: CDH1 Mutations and Beyond. JAMA Oncol. 2015, 1, 23–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Herrera-Pariente, C.; Capó-García, R.; Díaz-Gay, M.; Carballal, S.; Muñoz, J.; Llach, J.; Sánchez, A.; Bonjoch, L.; Arnau-Collell, C.; de Lima, Y.S.; et al. Identification of New Genes Involved in Germline Predisposition to Early-Onset Gastric Cancer. Int. J. Mol. Sci. 2021, 22, 1310. [Google Scholar] [CrossRef]
  16. Milne, A.N.; Offerhaus, G.J.A. Early-Onset Gastric Cancer: Learning Lessons from the Young. World J. Gastrointest. Oncol. 2010, 2, 59–64. [Google Scholar] [CrossRef] [PubMed]
  17. Anderson, W.F.; Camargo, M.C.; Fraumeni, J.F.; Correa, P.; Rosenberg, P.S.; Rabkin, C.S. Age-Specific Trends in Incidence of Noncardia Gastric Cancer in US Adults. JAMA J. Am. Med. Assoc. 2010, 303, 1723–1728. [Google Scholar] [CrossRef] [Green Version]
  18. Bergquist, J.R.; Leiting, J.L.; Habermann, E.B.; Cleary, S.P.; Kendrick, M.L.; Smoot, R.L.; Nagorney, D.M.; Truty, M.J.; Grotz, T.E. Early-Onset Gastric Cancer Is a Distinct Disease with Worrisome Trends and Oncogenic Features. Surgery 2019, 166, 547–555. [Google Scholar] [CrossRef]
  19. Medina-Franco, H.; Heslin, M.J.; Cortes-Gonzalez, R. Clinicopathological Characteristics of Gastric Carcinoma in Young and Elderly Patients: A Comparative Study. Ann. Surg. Oncol. 2000, 7, 515–519. [Google Scholar] [CrossRef]
  20. Schildberg, C.W.; Abba, M.; Merkel, S.; Agaimy, A.; Dimmler, A.; Schlabrakowski, A.; Croner, R.; Leupold, J.H.; Hohenberger, W.; Allgayer, H. Gastric Cancer Patients Less than 50 Years of Age Exhibit Significant Downregulation of E-Cadherin and CDX2 Compared to Older Reference Populations. Adv. Med. Sci. 2014, 59, 142–146. [Google Scholar] [CrossRef] [PubMed]
  21. Pérez-Carbonell, L.; Ruiz-Ponte, C.; Guarinos, C.; Alenda, C.; Payá, A.; Brea, A.; Egoavil, C.M.; Castillejo, A.; Barberá, V.M.; Bessa, X.; et al. Comparison between Universal Molecular Screening for Lynch Syndrome and Revised Bethesda Guidelines in a Large Population-Based Cohort of Patients with Colorectal Cancer. Gut 2012, 61, 865–872. [Google Scholar] [CrossRef] [PubMed]
  22. Kim, H.S.; Kim, J.W.; Hwang, I.G.; Lee, H.S.; Kim, W.H. Expression of DNA Damage Response Markers in Early-Onset or Familial Gastric Cancers. Asian Pac. J. Cancer Prev. 2019, 20, 1369–1376. [Google Scholar] [CrossRef] [Green Version]
  23. Giryes, A.; Oweira, H.; Mannhart, M.; Decker, M.; Abdel-Rahman, O. Exploring the Differences between Early-Onset Gastric Cancer and Traditional-Onset Gastric Cancer. J. Gastrointest. Oncol. 2018, 9, 1157–1163. [Google Scholar] [CrossRef] [PubMed]
  24. Machlowska, J.; Baj, J.; Sitarz, M.; Maciejewski, R.; Sitarz, R. Gastric Cancer: Epidemiology, Risk Factors, Classification, Genomic Characteristics and Treatment Strategies. Int. J. Mol. Sci. 2020, 21, 4012. [Google Scholar] [CrossRef] [PubMed]
  25. Merchant, S.J.; Kim, J.; Choi, A.H.; Sun, V.; Chao, J.; Nelson, R. A Rising Trend in the Incidence of Advanced Gastric Cancer in Young Hispanic Men. Gastric Cancer 2017, 20, 226–234. [Google Scholar] [CrossRef]
  26. Li, S.; Rexin, P.; Qin, Z.; Changbo, C.; Guanghui, C.; Luyao, W.; Voelker, H.U.; Stauch, G. Different Incidence of Early-Onset Gastric Carcinoma Depending on Ethnicity: Preliminary Results of a Hospital in Liangshan. Sci. World J. 2020, 1–5. [Google Scholar] [CrossRef] [Green Version]
  27. Adar, T.; Friedman, M.; Rodgers, L.H.; Shannon, K.M.; Zukerberg, L.R.; Chung, D.C. Gastric Cancer in Lynch Syndrome Is Associated with Underlying Immune Gastritis. J. Med. Genet. 2019, 56, 844–845. [Google Scholar] [CrossRef]
  28. Kim, J.; Braun, D.; Ukaegbu, C.; Dhingra, T.G.; Kastrinos, F.; Parmigiani, G.; Syngal, S.; Yurgelun, M.B. Clinical Factors Associated With Gastric Cancer in Individuals With Lynch Syndrome. Clin. Gastroenterol. Hepatol. 2020, 18, 830–837.e1. [Google Scholar] [CrossRef]
  29. Zhang, Q.; Wang, L.; Ni, S.; Tan, C.; Cai, X.; Huang, D.; Sheng, W. Clinicopathological Features and Prognostic Value of Mismatch Repair Protein Deficiency in Gastric Cancer. Int. J. Clin. Exp. Pathol. 2018, 11, 2579–2587. [Google Scholar]
  30. Hayden, J.D.; Cawkwell, L.; Sue-Ling, H.; Johnston, D.; Dixon, M.F.; Quirke, P.; Martin, I.G. Assessment of Microsatellite Alterations in Young Patients with Gastric Adenocarcinoma. Cancer 1997, 79, 684–687. [Google Scholar] [CrossRef]
  31. Bass, A.J.; Thorsson, V.; Shmulevich, I.; Reynolds, S.M.; Miller, M.; Bernard, B.; Hinoue, T.; Laird, P.W.; Curtis, C.; Shen, H.; et al. Comprehensive Molecular Characterization of Gastric Adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar]
  32. Moreira, L.; Muñoz, J.; Cuatrecasas, M.; Quintanilla, I.; Leoz, M.L.; Carballal, S.; Ocaña, T.; Lõpez-Cerõn, M.; Pellise, M.; Castellví-Bel, S.; et al. Prevalence of Somatic Mutl Homolog 1 Promoter Hypermethylation in Lynch Syndrome Colorectal Cancer. Cancer 2015, 121, 1395–1404. [Google Scholar] [CrossRef] [PubMed]
  33. Tedaldi, G.; Pirini, F.; Tebaldi, M.; Zampiga, V.; Cangini, I.; Danesi, R.; Arcangeli, V.; Ravegnani, M.; Khouzam, R.A.; Molinari, C.; et al. Multigene Panel Testing Increases the Number of Loci Associated with Gastric Cancer Predisposition. Cancers 2019, 11, 1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Aronson, M.; Swallow, C.; Govindarajan, A.; Semotiuk, K.; Cohen, Z.; Kaurah, P.; Velsher, L.; Ambus, I.; Buckley, K.; Forster-Gibson, C.; et al. Germline Variants and Phenotypic Spectrum in a Canadian Cohort of Individuals with Diffuse Gastric Cancer. Curr. Oncol. 2020, 27, e182–e190. [Google Scholar] [CrossRef] [PubMed]
  35. Vogelaar, I.P.; Van Der Post, R.S.; Van Krieken, J.H.J.M.; Spruijt, L.; Van Zelst-Stams, W.A.G.; Kets, C.M.; Lubinski, J.; Jakubowska, A.; Teodorczyk, U.; Aalfs, C.M.; et al. Unraveling Genetic Predisposition to Familial or Early Onset Gastric Cancer Using Germline Whole-Exome Sequencing. Eur. J. Hum. Genet. 2017, 25, 1246–1252. [Google Scholar] [CrossRef] [PubMed]
  36. Vos, E.L.; Salo-Mullen, E.E.; Tang, L.H.; Schattner, M.; Yoon, S.S.; Gerdes, H.; Markowitz, A.J.; Mandelker, D.; Janjigian, Y.; Offitt, K.; et al. Indications for Total Gastrectomy in CDH1 Mutation Carriers and Outcomes of Risk-Reducing Minimally Invasive and Open Gastrectomies. JAMA Surg. 2020, 155, 1050–1057. [Google Scholar] [CrossRef]
  37. Gamble, L.A.; Heller, T.; Davis, J.L. Hereditary Diffuse Gastric Cancer Syndrome and the Role of CDH1: A Review. JAMA Surg. 2021, 156, 387–392. [Google Scholar] [CrossRef]
  38. Milne, A.N.; Sitarz, R.; Carvalho, R.; Carneiro, F.; Offerhaus, G.J.A. Early Onset Gastric Cancer: On the Road to Unraveling Gastric Carcinogenesis. Curr. Mol. Med. 2007, 7, 15–28. [Google Scholar] [CrossRef]
  39. Rugge, M.; Busatto, G.; Cassaro, M.; Shiao, Y.H.; Russo, V.; Leandro, G.; Avellini, C.; Fabiano, A.; Sidoni, A.; Covacci, A. Patients Younger than 40 Years with Gastric Carcinoma: Helicobacter Pylori Genotype and Associated Gastritis Phenotype. Cancer 1999, 85, 2506–2511. [Google Scholar] [CrossRef]
Figure 1. Immunohistochemistry of DNA mismatch repair proteins (MMR-IHC) in gastric cancer tissue loss of protein expression of MLH1 and PMS2, and normal protein expression of MSH2 and MSH6.
Figure 1. Immunohistochemistry of DNA mismatch repair proteins (MMR-IHC) in gastric cancer tissue loss of protein expression of MLH1 and PMS2, and normal protein expression of MSH2 and MSH6.
Cancers 13 03132 g001
Figure 2. Scheme followed summarizing the results of IHC-MMR and germline genetic testing.
Figure 2. Scheme followed summarizing the results of IHC-MMR and germline genetic testing.
Cancers 13 03132 g002
Figure 3. Overall survival rate based on diagnostic stage (stage I–II vs. stage III–IV). There is a significant difference in the 5-year survival rate: stage I–II 87% vs. stage III–IV 11.3%, p = 0.0001.
Figure 3. Overall survival rate based on diagnostic stage (stage I–II vs. stage III–IV). There is a significant difference in the 5-year survival rate: stage I–II 87% vs. stage III–IV 11.3%, p = 0.0001.
Cancers 13 03132 g003
Table 1. Clinical characteristics of the patients (N = 309).
Table 1. Clinical characteristics of the patients (N = 309).
EOGC
Age (years) at diagnosis; median (IQR)44 (40–48)
Gender, number (%):
Man
191 (61.8)
Environmental risk factors; number (%)
Helicobacter pylori infection24/82 (29.3)
Smokers77/169 (45.6)
Moderate or high alcohol consumers21/105 (20)
Personal history of extra-gastric cancer (n = 309); number (%)
Breast and/or ovarian5 (1.6)
Lung1 (0.3)
Thyroid1 (0.3)
Hodgkin lymphoma1 (0.3)
Cervix1 (0.3)
Familial history of cancer (n = 117); number (%)
GC18 (15.4)
Colorectal8 (6.8)
Ovarian and/or breast15 (12.8)
Others39 (33.3)
Criteria of familial GC5 (4.3)
IQR, interquartile range; EOGC, early onset gastric cancer; GC, gastric cancer.
Table 2. Characteristics of the tumors (N = 309).
Table 2. Characteristics of the tumors (N = 309).
EOGC
Gastric location (N = 203); number (%)
Cardias3 (1.5)
Fundus18 (8.9)
Body111 (54.7)
Antrum50 (24.6)
Extensive21 (10.3)
Stage (N = 212); number (%)
I/II44 (20.8)
III/IV166 (78.3)
Histology (N = 161); number (%)
(a) Diffuse
Signet ring cell subset
118 (73.3)
38/118 (32.2)
(b) Intestinal30 (18.6)
(c) Mixed13 (8.1)
Tumor differentiation grade (N = 23/43) number (%) *
High grade (poorly differentiated)9 (39.1)
Low-grade (well/moderately differentiated)14 (60.9)
Treatment (N = 205) number (%)
Surgery40 (19.5)
Surgery + chemotherapy58 (31.2)
Chemotherapy64 (28.3)
Surgery + chemotherapy + radiotherapy24 (11.7)
Chemotherapy + radiotherapy3 (1.5)
Palliative16 (7.8)
EOGC, early onset gastric cancer. * According to the WHO classification (2019), the degree of differentiation only applies to the intestinal type.
Table 3. Characteristics in patients with hereditary syndromes.
Table 3. Characteristics in patients with hereditary syndromes.
PatientAgeGenderTumor CharacteristicsH. pylori InfectionPersonal History of Other TumorsFamilial History of GCFamilial History of Other TumorsMMR-IHCGene (Pathogenic Variant)
149WomanIntestinal hist. Stage IINot availableBreastNoOvarian and breastMMR+
(normal)
BRCA2
(c.3166C>T; p.Gln1056 *; nonsense)
238ManDiffuse hist.
Multifocal (plastic linitis)
Stage IV
NoNoYesNoMMR+
(normal)
CDH1
(c.2164+5G>C; splicing)
334ManDiffuse hist.
Multifocal (plastic linitis)
Stage IV
NoNoNoBreast and colorectalMMR+
(normal)
TP53
(c.365_366delTG; p.Val122fs; frameshift)
440ManDiffuse hist.
Multifocal (plastic linitis)
Stage IV
NoNoYesNoMMR+
(normal)
CDH1
(c.187C>T; p.Arg63 *; nonsense)
hist., histology; GC, gastric cancer; MMR-IHC, immunohistochemistry of DNA mismatch repair proteins; MMR, tumor mismatch repair; *, stop codon
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pocurull, A.; Herrera-Pariente, C.; Carballal, S.; Llach, J.; Sánchez, A.; Carot, L.; Botargues, J.M.; Cuatrecasas, M.; Ocaña, T.; Balaguer, F.; et al. Clinical, Molecular and Genetic Characteristics of Early Onset Gastric Cancer: Analysis of a Large Multicenter Study. Cancers 2021, 13, 3132. https://doi.org/10.3390/cancers13133132

AMA Style

Pocurull A, Herrera-Pariente C, Carballal S, Llach J, Sánchez A, Carot L, Botargues JM, Cuatrecasas M, Ocaña T, Balaguer F, et al. Clinical, Molecular and Genetic Characteristics of Early Onset Gastric Cancer: Analysis of a Large Multicenter Study. Cancers. 2021; 13(13):3132. https://doi.org/10.3390/cancers13133132

Chicago/Turabian Style

Pocurull, Anna, Cristina Herrera-Pariente, Sabela Carballal, Joan Llach, Ariadna Sánchez, Laura Carot, Josep María Botargues, Miriam Cuatrecasas, Teresa Ocaña, Francesc Balaguer, and et al. 2021. "Clinical, Molecular and Genetic Characteristics of Early Onset Gastric Cancer: Analysis of a Large Multicenter Study" Cancers 13, no. 13: 3132. https://doi.org/10.3390/cancers13133132

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop