Background:Signet ring cell colorectal cancer (SRCCa) has a bleak prognosis. Employing molecular pathology techniques we investigated the potential of precision medicine in this disease.Methods:Using test (n=26) and validation (n=18) cohorts, analysis of mutations, DNA methylation and transcriptome was carried out. Microsatellite instability (MSI) status was established and immunohistochemistry (IHC) was used to test for adaptive immunity (CD3) and the immune checkpoint PDL1.Results:DNA methylation data split the cohorts into hypermethylated (n=18, 41%) and hypomethylated groups (n=26, 59%). The hypermethylated group predominant in the proximal colon was enriched for CpG island methylator phenotype (CIMP), BRAF V600E mutation and MSI (P<0.001). These cases also had a high CD3+ immune infiltrate (P<0.001) and expressed PDL1 (P=0.03 in intra-tumoural lymphoid cells). The hypomethylated group predominant in the distal colon did not show any characteristic molecular features. We also detected a common targetable KIT mutation (c.1621A>C) across both groups. No statistically significant difference in outcome was observed between the two groups.Conclusions:Our data show that SRCCa phenotype comprises two distinct genotypes. The MSI+/CIMP+/BRAF V600E+/CD3+/PDL1+ hypermethylated genotype is an ideal candidate for immune checkpoint inhibitor therapy. In addition, one fourth of SRCCa cases can potentially be targeted by KIT inhibitors.
Small bowel accounts for only 0.5% of cancer cases in the US but incidence rates have been rising at 2.4% per year over the past decade. One-third of these are adenocarcinomas but little is known about their molecular pathology and no molecular markers are available for clinical use.Using a retrospective 28 patient matched normal-tumor cohort, next-generation sequencing, gene expression arrays and CpG methylation arrays were used for molecular profiling.Next-generation sequencing identified novel mutations in IDH1, CDH1, KIT, FGFR2, FLT3, NPM1, PTEN, MET, AKT1, RET, NOTCH1 and ERBB4. Array data revealed 17% of CpGs and 5% of RNA transcripts assayed to be differentially methylated and expressed respectively (p < 0.01). Merging gene expression and DNA methylation data revealed CHN2 as consistently hypermethylated and downregulated in this disease (Spearman −0.71, p < 0.001). Mutations in TP53 which were found in more than half of the cohort (15/28) and Kazald1 hypomethylation were both were indicative of poor survival (p = 0.03, HR = 3.2 and p = 0.01, HR = 4.9 respectively).By integrating high-throughput mutational, gene expression and DNA methylation data, this study reveals for the first time the distinct molecular profile of small bowel adenocarcinoma and highlights potential clinically exploitable markers.
Around 12-15% of patients with locally advanced rectal cancer undergo a pathologically complete response (tumor regression grade 4) to long-course preoperative chemoradiotherapy; the remainder exhibit a spectrum of tumor regression (tumor regression grade 1-3). Understanding therapy-related transcriptional alterations may enable better prediction of response as measured by progression-free and overall survival, in addition to aiding the development of improved strategies based on the underlying biology of the disease. To this end, we performed high-throughput gene expression profiling in 40 pairs of formalin-fixed paraffin-embedded rectal cancer biopsies and matched resections following long-course preoperative chemoradiotherapy (discovery cohort). Differential gene expression analysis was performed contrasting tumor regression grades in resections. Enumeration of the tumor microenvironment cell population was undertaken using in silico analysis of the transcriptional data, and real-time PCR validation of NCR1 undertaken. Immunohistochemistry and survival analysis was used to measure CD56+ cell populations in an independent cohort (n=150). Gene expression traits observed following long-course preoperative chemoradiotherapy in the discovery cohort suggested an increased abundance of natural killer cells in tumors that displayed a clinical response to CRT in a tumor regression grade-dependent manner. CD56+ natural killer-cell populations were measured by immunohistochemistry and found to be significantly higher in tumor regression grade 3 patients compared with tumor regression grade 1-2 in the validation cohort. Furthermore, it was observed that patients positive for CD56 cells after therapy had a better overall survival (HR=0.282, 95% CI=0.109-0.729, χ=7.854, P=0.005). In conclusion, we have identified a novel post-therapeutic natural killer-like transcription signature in patients responding to long-course preoperative chemoradiotherapy. Furthermore, patients with a higher abundance of CD56-positive natural killer cells post long-course preoperative chemoradiotherapy had better overall survival. Therefore, harnessing a natural killer-like response after therapy may improve outcomes for locally advanced rectal cancer patients. Finally, we hypothesize that future assessment of this natural killer-like response in on-treatment biopsy material may inform clinical decision-making for treatment duration.
Small bowel accounts for only 0.5% of cancer cases in the US; a third of which are adenocarcinomas. But incidence rates have been rising at a rate of 2.4% per year over the last decade. Because of the rarity of this cancer, little is known about its molecular pathology and there are no molecular markers for diagnosis, predicting prognosis or therapeutic intervention. The aim of this study was therefore to look into this disease at a molecular level to better understand its biology and identify biomarkers and potential points of therapeutic intervention. Using a retrospective 28 patient matched normal-tumor cohort next generation sequencing (NGS) was performed using a 50 gene cancer hotspot panel, gene expression arrays were used to profile ∼29,000 RNA transcripts and 450k CpG methylation arrays were used to carry out DNA methylation analysis. We also looked at microsatellite instability (MSI), HER2 and p53 expression. NGS identified novel mutations in IDH1, CDH1, KIT, NRAS, FGFR2, FLT3, NPM1, PTEN, MET, AKT1, RET, NOTCH1 and ERBB4. Previously known mutations such as high-frequency KRAS and TP53 and low-frequency HER2 were also confirmed in our cohort. The average patient had 2.6 mutations with eight patients having only a single mutation to one having seven. Array data revealed 17% of CpGs and 5% of RNA transcripts assayed to be differentially methylated and expressed respectively (p<0.01). Gene expression and DNA methylation data was merged to reveal 266 transcripts where DNA methylation was inversely correlated with gene expression (Spearman < -0.5). Almost 90% of these CpGs were found to be located inside gene bodies, the first exons and 5′ and 3′ UTRs. Almost 60% of the overall genome-wide changes in methylation (hyper and hypo) also occurred inside the gene bodies, the first exons and 5′ and 3′ UTRs. CHN2 was identified as one of the genes consistently hypermethylated and downregulated in tumor compared to normal (Spearman -0.71, p<0.001). This trend was validated using immunohistochemistry and pyrosequencing. Finally mutations in TP53 which were found in more than half of the cohort (15/28) and Kazald1 hypomethylation were both were indicative of poor survival (p = 0.0345, HR = 3.2 and p = 0.0079, HR = 4.9 respectively). Sanger sequencing was used to confirm TP53 mutations and immunohistochemistry analysis confirmed protein overexpression in all mutant cases. Six cases were positive for MSI and three for HER2 overexpression. This study has for the first time highlighted the extent of molecular changes taking place in SBA. The clinical potential of TP53 mutations and Kazald1 hypomethylation as prognostic biomarkers and CHN2 as a diagnostic biomarker are focus areas for further research by our group. Citation Format: Muhammad A. Alvi, Darragh G. McArt, Paul Kelly, Marc-Aurel Fuchs, Matthew Alderdice, Clare M. McCabe, Victoria Bingham, Claire McGready, Stephen McQuaid, Perry Maxwell, Peter Hamilton, Jacqueline A. James, Richard Wilson, Manuel Salto-Tellez. Comprehensive molecular pathology analysis of small bowel adenocarcinoma reveals novel targets with clinical utility. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4792. doi:10.1158/1538-7445.AM2015-4792
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.