Non-targeted biopsies in hereditary diffuse gastric cancer: Necessary, but not enough
Editorial Commentary

Non-targeted biopsies in hereditary diffuse gastric cancer: Necessary, but not enough

Kole H. Buckley1, Rashmi Tondon2, Parry Guilford3, Bryson W. Katona1

1Division of Gastroenterology and Hepatology, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA; 2Department of Pathology and Laboratory Medicine, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA; 3Cancer Genetics Laboratory, Centre for Translational Cancer Research (Te Aho Matatū), Department of Biochemistry, University of Otago, Dunedin, New Zealand

Correspondence to: Bryson W. Katona, MD, PhD. Division of Gastroenterology and Hepatology, Perelman School of Medicine at University of Pennsylvania, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA 19104, USA. Email: bryson.katona@pennmedicine.upenn.edu.

Comment on: Lee CYC, Olivier A, Honing J, et al. Endoscopic surveillance with systematic random biopsy for the early diagnosis of hereditary diffuse gastric cancer: a prospective 16-year longitudinal cohort study. Lancet Oncol 2023;24:107-16.


Keywords: Surveillance; CDH1; E-cadherin; hereditary diffuse gastric cancer (HDGC)


Submitted Mar 29, 2023. Accepted for publication Jun 09, 2023. Published online Jul 06, 2023.

doi: 10.21037/tcr-23-541


Hereditary diffuse gastric cancer (HDGC) is caused by germline pathogenic/likely pathogenic (P/LP) variants in the CDH1 and CTNNA1 genes (1-6). Carriers of P/LP CDH1 variants have an estimated 33–42% risk of diffuse gastric cancer by age 80 years old (7,8). Risk management strategies in HDGC are largely confined to two options: annual upper endoscopic surveillance or prophylactic total gastrectomy (9). While prophylactic total gastrectomy is currently the only definitive method to eliminate future gastric cancer risk, yearly endoscopic surveillance may offer value by informing the timing of total gastrectomy in patients who do not wish to immediately pursue surgery. Endoscopic surveillance may also offer value in carriers of CDH1 or CTNNA1 P/LP variants who do not have a family history of gastric cancer, a subgroup where future risk of diffuse gastric cancer remains particularly uncertain.

The principal goal of upper endoscopic surveillance is to use targeted and non-targeted biopsies to identify foci of signet ring cell carcinoma (SRCC) while still at an early stage (10). However, the reliability of this method is problematic given its high false negative rate, as demonstrated by gastrectomy specimens frequently showing SRCC despite negative endoscopic findings (11-13). The recent study by Lee et al., published in Lancet Oncology, aimed to determine whether non-targeted biopsies improve endoscopic detection of SRCC by evaluating the diagnostic yield of SRCC in targeted versus non-targeted biopsies and assessing the cumulative incidence of cancer during longitudinal follow-up (14). This study was conducted at the Cambridge University Hospitals National Health Service (NHS) Foundation Trust between June 1, 2005 and July 31, 2021. The patient cohort included 145 individuals from 76 families who fulfilled HDGC testing criteria according to the International Gastric Cancer Linkage Consortium (IGCLC) (9,14).

A major finding of this study was that non-targeted biopsies acquired during endoscopic surveillance provided a higher diagnostic yield of SRCC compared to targeted biopsies (14). Indeed, of 58 patients diagnosed with SRCC, 50% were first diagnosed via only non-targeted biopsies while 26% were first diagnosed via only targeted biopsies (14). Of the remaining patients, 10% were first diagnosed with SRCC observed by both non-targeted and targeted biopsies, 9% had macroscopic tumors that were directly visualized, and 5% were first diagnosed only after a total gastrectomy was performed (14). SRCC positive targeted biopsies were most frequently found in the antrum, whereas SRCC positive non-targeted biopsies were more often found in the cardia and fundus (14). This is likely due to better mucosal visualization in the antrum given the lack of rugae, therefore allowing better visualization of pale lesions of mucosa that are associated with SRCC (15,16). Importantly, when compared to targeted biopsies, the anatomical distribution of SRCC detected from non-targeted biopsies more closely mimicked the distribution of SRCC in total gastrectomy specimens (14). These findings provide support for the utilization of non-targeted biopsies during endoscopic surveillance in HDGC, a practice that has long been endorsed by the IGCLC (9).

Endoscopically detected flat pale lesions have previously been recognized to be associated with foci of SRCC (15,16). However, biopsy of pale lesions has a low positive predictive value, as demonstrated in this study where only 12% of pale lesions biopsied revealed foci of SRCC (14). While the characteristics of pale lesions that are suspected to contain SRCC can be challenging to define, it is important to establish such criteria as lesions with a similar appearance will accumulate with continued surveillance due to scarring of the gastric mucosa after repeated biopsy. Previous studies have indicated that pale patches with an irregular microvasculature and microsurface may be indicative of underlying SRCC (17,18). Building off this, the authors of the current study identified three additional criteria, including that pale lesions associated with SRCC are more round (rather than irregularly shaped), have well-defined borders, and are likely to be persistently observed from multiple different viewing angles as well as multiple viewing distances (Figure 1) (14). These new criteria were able to predict SRCC in pale flat lesions with a sensitivity of 67%, with only 16 of 386 (4%) non-suspicious lesions showing SRCC histology (14). As patients with a predisposition for HDGC may undergo multiple surveillance endoscopies, using these criteria to differentiate SRCC-associated pale lesions from previous biopsy scars or other non-malignant lesions could become increasingly important. This would be of particular importance when a high number of biopsies are taken annually, such as in the Bethesda protocol where four non-targeted biopsies are taken from each of 22 separate anatomic sites (11).

Figure 1 Proposed criteria for assessment of pale lesions.

Current IGCLC guidelines recommend annual endoscopic surveillance for carriers of P/LP CDH1 variants with a family history of diffuse gastric cancer who do not immediately pursue a prophylactic total gastrectomy (9). Of the longitudinal patient cohort in this study, 55 patients were diagnosed with SRCC via endoscopic surveillance. Between one and eight annual endoscopies were required to find SRCC in each of the 55 patients (14). Out of this group, 34/55 (62%) were diagnosed on the first endoscopy and 44/55 (80%) by the second endoscopy, demonstrating that SRCC are most likely to be identified within the first couple of surveillance endoscopies (14). However, these data do not take into account the other 43 CDH1 P/LP carriers in which SRCC were not identified (14). Given that SRCC are detected in gastrectomy specimens in greater than 95% of CDH1 P/LP carriers (12,19,20), it is likely that despite repeated upper endoscopies, there will always remain a subgroup of CDH1 P/LP carriers in whom foci of SRCC will not be detected. The authors of this study suggest that after five consecutive annual surveillance procedures with negative findings, the surveillance interval could be increased to every 2 years, particularly for individuals older than 35 years of age (14). However, without a better understanding of the time required for an undetected stage T1a SRCC to progress to ≥ stage T2 disease, such a decision might present substantial additional risk. This risk is amplified by the poor understanding we currently have of individual genetic or environmental risk factors for disease progression.

Regardless of endoscopic findings, the current IGCLC guidelines recommend total gastrectomy for CDH1 P/LP variant carrying patients with a family history of diffuse gastric cancer (9). While this study (14) provides evidence to support the use of non-targeted gastric biopsies during upper endoscopy, more reliable techniques and/or technologies are still needed to better detect and quantify SRCC in CDH1 carriers. Furthermore, and more importantly, further investigations to distinguish between foci of SRCC that are genuinely indolent and those with a higher likelihood of progression are needed (9). For P/LP variant carriers to avoid a total gastrectomy, treatments to prevent the development of and/or eliminate foci of SRCC are also needed. While no proven treatments are currently available, data from preclinical models suggest that E-cadherin-deficient cells may be susceptible to specific multikinase inhibitors and histone deacetylase (HDAC) inhibitors (21,22).

In conclusion, Lee et al. present data on the enhanced detection of early stage SRCC using targeted and non-targeted gastric biopsies in carriers of P/LP CDH1 variants. These data show the benefit of endoscopic surveillance for informing optimal decision-making around the timing of a total gastrectomy for individual carriers of P/LP CDH1 variants. This data is also an important component of the goal of reducing the need for prophylactic gastrectomies in HDGC families, a goal that will be further advanced by ongoing research on personalized risk stratification, chemoprevention and new endoscopic imaging modalities.


Acknowledgments

Funding: This work was supported by DeGregorio Family Foundation Grant Award to BWK.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Translational Cancer Research. The article has undergone external peer review.

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-541/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-541/coif). BWK reports that he receives grants from DeGregorio Family Foundation and American Gastroenterological Association. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Hansford S, Kaurah P, Li-Chang H, et al. Hereditary Diffuse Gastric Cancer Syndrome: CDH1 Mutations and Beyond. JAMA Oncol 2015;1:23-32. [Crossref] [PubMed]
  2. Majewski IJ, Kluijt I, Cats A, et al. An α-E-catenin (CTNNA1) mutation in hereditary diffuse gastric cancer. J Pathol 2013;229:621-9. [Crossref] [PubMed]
  3. Lobo S, Benusiglio PR, Coulet F, et al. Cancer predisposition and germline CTNNA1 variants. Eur J Med Genet 2021;64:104316. [Crossref] [PubMed]
  4. Guilford P, Hopkins J, Harraway J, et al. E-cadherin germline mutations in familial gastric cancer. Nature 1998;392:402-5. [Crossref] [PubMed]
  5. Clark DF, Michalski ST, Tondon R, et al. Loss-of-function variants in CTNNA1 detected on multigene panel testing in individuals with gastric or breast cancer. Genet Med 2020;22:840-6. [Crossref] [PubMed]
  6. Decourtye-Espiard L, Guilford P. Hereditary Diffuse Gastric Cancer. Gastroenterology 2023;164:719-35. [Crossref] [PubMed]
  7. Xicola RM, Li S, Rodriguez N, et al. Clinical features and cancer risk in families with pathogenic CDH1 variants irrespective of clinical criteria. J Med Genet 2019;56:838-43. [Crossref] [PubMed]
  8. Roberts ME, Ranola JMO, Marshall ML, et al. Comparison of CDH1 Penetrance Estimates in Clinically Ascertained Families vs Families Ascertained for Multiple Gastric Cancers. JAMA Oncol 2019;5:1325-31. [Crossref] [PubMed]
  9. Blair VR, McLeod M, Carneiro F, et al. Hereditary diffuse gastric cancer: updated clinical practice guidelines. Lancet Oncol 2020;21:e386-97. [Crossref] [PubMed]
  10. Gamble LA, Heller T, Davis JL. Hereditary Diffuse Gastric Cancer Syndrome and the Role of CDH1: A Review. JAMA Surg 2021;156:387-92. [Crossref] [PubMed]
  11. Curtin BF, Gamble LA, Schueler SA, et al. Enhanced endoscopic detection of occult gastric cancer in carriers of pathogenic CDH1 variants. J Gastroenterol 2021;56:139-46. [Crossref] [PubMed]
  12. Kumar S, Long JM, Ginsberg GG, et al. The role of endoscopy in the management of hereditary diffuse gastric cancer syndrome. World J Gastroenterol 2019;25:2878-86. [Crossref] [PubMed]
  13. Benesch MGK, Bursey SR, O'Connell AC, et al. CDH1 Gene Mutation Hereditary Diffuse Gastric Cancer Outcomes: Analysis of a Large Cohort, Systematic Review of Endoscopic Surveillance, and Secondary Cancer Risk Postulation. Cancers (Basel) 2021;13:2622. [Crossref] [PubMed]
  14. Lee CYC, Olivier A, Honing J, et al. Endoscopic surveillance with systematic random biopsy for the early diagnosis of hereditary diffuse gastric cancer: a prospective 16-year longitudinal cohort study. Lancet Oncol 2023;24:107-16. [Crossref] [PubMed]
  15. Pilonis ND, Tischkowitz M, Fitzgerald RC, et al. Hereditary Diffuse Gastric Cancer: Approaches to Screening, Surveillance, and Treatment. Annu Rev Med 2021;72:263-80. [Crossref] [PubMed]
  16. Mi EZ, Mi EZ, di Pietro M, et al. Comparative study of endoscopic surveillance in hereditary diffuse gastric cancer according to CDH1 mutation status. Gastrointest Endosc 2018;87:408-18. [Crossref] [PubMed]
  17. Muto M, Yao K, Kaise M, et al. Magnifying endoscopy simple diagnostic algorithm for early gastric cancer (MESDA-G). Dig Endosc 2016;28:379-93. Erratum in: Dig Endosc 2016;28:630. [Crossref] [PubMed]
  18. Yao K, et al. The magnified endoscopic finding of an irregular microvascular pattern is a very useful marker for differentiating between gastritis and gastric cancer: a prospective study. Gastrointest Endosc 2004;59:169. [Crossref]
  19. DiBrito SR, Blair AB, Prasath V, et al. Total Gastrectomy for CDH-1 Mutation Carriers: An Institutional Experience. J Surg Res 2020;247:438-44. [Crossref] [PubMed]
  20. Friedman M, Adar T, Patel D, et al. Surveillance Endoscopy in the Management of Hereditary Diffuse Gastric Cancer Syndrome. Clin Gastroenterol Hepatol 2021;19:189-91. [Crossref] [PubMed]
  21. Bougen-Zhukov N, Decourtye-Espiard L, Mitchell W, et al. E-Cadherin-Deficient Cells Are Sensitive to the Multikinase Inhibitor Dasatinib. Cancers (Basel) 2022;14:1609. [Crossref] [PubMed]
  22. Decourtye-Espiard L, Bougen-Zhukov N, Godwin T, et al. E-Cadherin-Deficient Epithelial Cells Are Sensitive to HDAC Inhibitors. Cancers (Basel) 2021;14:175. [Crossref] [PubMed]
Cite this article as: Buckley KH, Tondon R, Guilford P, Katona BW. Non-targeted biopsies in hereditary diffuse gastric cancer: Necessary, but not enough. Transl Cancer Res 2023;12(7):1883-1886. doi: 10.21037/tcr-23-541

Download Citation