Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2019 Jun 1.
Published in final edited form as: JAMA Oncol. 2018 Dec 1;4(12):1698–1699. doi: 10.1001/jamaoncol.2018.2803

MUC16 mutations and prognosis in gastric cancer; a little goes a long way

Elizabeth C Smyth 1, Rebecca C Fitzgerald 2
PMCID: PMC6292505  EMSID: EMS79380  PMID: 30098141

Gastric cancer is a global health problem; although incidence rates are declining, it remains the third most common cause of cancer death worldwide.1 Gastric cancer patients with advanced disease have limited treatment options available, and most will live for less than two years, therefore exploration of gastric cancer disease biology is warranted to identify new targets.2 Recent comprehensive molecular analyses have identified distinct subgroups of gastric cancer which may have therapeutic relevance, however with the exception of microsatellite unstable tumours the potential for genomically guided therapy has not been realised.35

In this edition of JAMA Oncology, Li and colleagues investigate the relationship between MUC16 mutation, tumour mutation load (TML) and survival in patients with gastric adenocarcinoma.6 Using data from the TCGA gastric cancer dataset and a second, smaller, Asian validation cohort, the investigators demonstrate that tumours which are MUC16 mutant are more likely to have a higher tumour mutation burden (p<0.001). This association was independent of age, gender, and the presence of mutations in genes affecting genomic stability such as BRCA1/2 and POLE. Independence from microsatellite instability (MSI) status could not be determined as MSI results were not available for every TCGA patient; however, the association between MUC16 mutation and TML also appeared to be independent of derived mismatch repair deficiency signatures. Li et al then proceed to demonstrate that patients who have MUC16 mutant tumours have longer median overall survival than patients with a MUC16 wildtype tumour genotype. In TCGA patients with resected gastric cancer the median overall survival was 26.7 months for MUC16 wildtype versus 46.9 months for MUC16 mutant tumours respectively (p=0.007); similar results were demonstrated in the Asian dataset. This prognostic effect of MUC16 mutation on overall survival also remained statistically significant when adjusted for confounding factors.

The findings presented by Li et al are interesting because if MUC16 mutation is truly predictive of high TML this could have clinical implications. The most readily apparent use of MUC16 mutation as a surrogate for TML would be to identify gastric cancer patients who might benefit from immune checkpoint blockade. As only one in six patients with PD-L1 selected gastroesophageal cancer respond to anti-PD-1 therapy, better enrichment biomarkers are a priority.7 Retrospective analysis across multiple cancer types and clinical trials demonstrates a significant correlation between tumour mutational load and response to PD-1 inhibition.8 Prospectively in the Checkmate 577 study, non-small cell lung cancer patients with a high tumour mutation load (≥10 mutations per megabase) had significantly superior progression free survival when treated with nivolumab and ipilimumab compared to chemotherapy, independent of PD-L1 expression.9 The authors’ transcriptomic results support the suggestion that MUC16 mutation could predict for sensitivity to anti-PD-1 therapy; MUC16 mutant gastric tumours are immunologically “hot”. Most recently, tumour mutation load has been assessed using a commercially available next generation sequencing (NGS) panel.8,9 However, if TML assessment were to be reduced to MUC16 sequencing only as a surrogate for a larger NGS panel, precise quantification of the relationship between MUC16 mutation and mutation rate per megabase in gastric cancer would be required to inform biomarker development.

Clinical limitations of the current work include the small size, restricted ethnic make-up, and heterogeneous nature of the validation cohort. The first major challenge to the scientific validity of the current work is the risk of a false positive association between MUC16 mutation and high TML because of characteristics associated with MUC16. MUC16 is a very large gene sited in an area with low replication timing. Regions of low replication timing are more likely to accumulate frequent mutations. Considering other relevant mutational co-variates, it is possible that MUC16 has a mutation rate which is in line with what might be expected by chance. Secondly, correlation between MUC16 mutation and high TML does not necessarily imply causation; a functional explanation for the link between MUC16 mutation and high TML is absent. An alternative explanation for the improved long-term survival of MUC16 mutant patients in this study may not relate directly to tumour mutation load. In an assessment of the tumour biology of long term survivors following pancreatic tumour resection, neoantigens (but not non-antigenic mutations) in MUC16 were identified four times more frequently in long term survivors than in patients who had shorter survival.10 In vitro, MUC16 expression is associated with proliferation and metastatic potential in tumour cells, and MUC16 is also known to inhibit natural killer cell mediated lysis of tumours.11,12 Thus, elimination of MUC16 neoantigen presenting cancer cells via immune “pruning” could hypothetically leave a residual population of MUC16 non-expressing cells with reduced metastatic and immunosuppressive potential, which could be associated with longer survival; this could present an alternative explanation for the authors’ transcriptomic results on immune infiltrate. Data on MUC16 neoantigen presentation are not provided in the current manuscript; this should be a priority for future research.

There is a paucity of evidence linking MUC16 gene function to cancer, therefore it is unlikely that MUC16 mutations are positively selected oncogenic drivers. This makes the hypothesis that MUC16 mutations act through neoantigen presentation and immunoediting more plausible. Despite the challenges in understanding the relationship between MUC16 mutation and TML, the relationship with prognosis appears to be robust. Although the results presented by Li and colleagues are preliminary and require further validation, they could provide a signpost for clinically relevant research in gastric cancer, or in view of the ubiquity of MUC16 mutations, in other tumour types.

Acknowledgements

ECS acknowledges honoraria for advisory role from Five Prime Therapeutics, BMS, Gritstone Oncology, Servier and Celgene.

RCF acknowledges licensed Cytosponge IP to Medtronic.

The authors would like to acknowledge Gift Nyamundanda and Alex M Frankell for their help with the review of this manuscript.

References

  • 1.Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
  • 2.Wagner AD, Syn NL, Moehler M, et al. Chemotherapy for advanced gastric cancer. The Cochrane Library. 2017 doi: 10.1002/14651858.CD004064.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513(7517):202. doi: 10.1038/nature13480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cristescu R, Lee J, Nebozhyn M, Kim KM, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nature Medicine. 2015;21(5):449. doi: 10.1038/nm.3850. [DOI] [PubMed] [Google Scholar]
  • 5.Smyth EC, Wotherspoon A, Peckitt C, et al. Mismatch Repair Deficiency, Microsatellite Instability, and Survival : An Exploratory Analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) Trial. JAMA oncology. 2017 doi: 10.1001/jamaoncol.2016.6762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Li X, Pasche B, Zhang W. MUC16/CA125 mutation is associated with high tumor mutation load and outcomes in gastric cancer. JAMA Oncology. 2018 doi: 10.1001/jamaoncol.2018.2805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Fuchs CS, Doi T, Jang RW, et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients With Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA oncology. 2018 doi: 10.1001/jamaoncol.2018.0013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N Engl J Med. 2017;377(25):2500–2501. doi: 10.1056/NEJMc1713444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hellmann MD, Callahan MK, Awad MM, et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell. 2018;33(5):853–861.e854. doi: 10.1016/j.ccell.2018.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Balachandran VP, Luksza M, Zhao JN, et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature. 2017;551(7681):512–516. doi: 10.1038/nature24462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Muniyan S, Haridas D, Chugh S, et al. MUC16 contributes to the metastasis of pancreatic ductal adenocarcinoma through focal adhesion mediated signaling mechanism. Genes & cancer. 2016;7(3–4):110–124. doi: 10.18632/genesandcancer.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gubbels JA, Felder M, Horibata S, et al. MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells. Molecular cancer. 2010;9:11. doi: 10.1186/1476-4598-9-11. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES