An integrative in-silico analysis discloses a novel molecular subset of colorectal cancer possibly eligible for immune checkpoint immunotherapy.
Colorectal cancer
Immunoinformatics
Immunotherapy
Meta-analysis
Multi-omics
Journal
Biology direct
ISSN: 1745-6150
Titre abrégé: Biol Direct
Pays: England
ID NLM: 101258412
Informations de publication
Date de publication:
09 05 2022
09 05 2022
Historique:
received:
16
02
2022
accepted:
23
04
2022
entrez:
9
5
2022
pubmed:
10
5
2022
medline:
12
5
2022
Statut:
epublish
Résumé
Historically, the molecular classification of colorectal cancer (CRC) was based on the global genomic status, which identified microsatellite instability in mismatch repair (MMR) deficient CRC, and chromosomal instability in MMR proficient CRC. With the introduction of immune checkpoint inhibitors, the microsatellite and chromosomal instability classification regained momentum as the microsatellite instability condition predicted sensitivity to immune checkpoint inhibitors, possibly due to both high tumor mutation burden (TMB) and high levels of infiltrating lymphocytes. Conversely, proficient MMR CRC are mostly resistant to immunotherapy. To better understand the relationship between the microsatellite and chromosomal instability classification, and eventually discover additional CRC subgroups relevant for therapeutic decisions, we developed a computational pipeline that include molecular integrative analysis of genomic, epigenomic and transcriptomic data. The first step of the pipeline was based on unsupervised hierarchical clustering analysis of copy number variations (CNVs) versus hypermutation status that identified a first CRC cluster with few CNVs enriched in Hypermutated and microsatellite instability samples, a second CRC cluster with a high number of CNVs mostly including non-HM and microsatellite stable samples, and a third cluster (7.8% of the entire dataset) with low CNVs and low TMB, which shared clinical-pathological features with Hypermutated CRCs and thus defined Hypermutated-like CRCs. The mutational features, DNA methylation profile and base substitution fingerprints of these tumors revealed that Hypermutated-like patients are molecularly distinct from Hypermutated and non-Hypermutated tumors and are likely to develop and progress through different genetic events. Transcriptomic analysis highlighted further differences amongst the three groups and revealed an inflamed tumor microenvironment and modulation Immune Checkpoint Genes in Hypermutated-like CRCs. Therefore, our work highlights Hypermutated-like tumors as a distinct and previously unidentified CRC subgroup possibly responsive to immune checkpoint inhibitors. If further validated, these findings can lead to expanding the fraction of patients eligible to immunotherapy.
Sections du résumé
BACKGROUND
Historically, the molecular classification of colorectal cancer (CRC) was based on the global genomic status, which identified microsatellite instability in mismatch repair (MMR) deficient CRC, and chromosomal instability in MMR proficient CRC. With the introduction of immune checkpoint inhibitors, the microsatellite and chromosomal instability classification regained momentum as the microsatellite instability condition predicted sensitivity to immune checkpoint inhibitors, possibly due to both high tumor mutation burden (TMB) and high levels of infiltrating lymphocytes. Conversely, proficient MMR CRC are mostly resistant to immunotherapy. To better understand the relationship between the microsatellite and chromosomal instability classification, and eventually discover additional CRC subgroups relevant for therapeutic decisions, we developed a computational pipeline that include molecular integrative analysis of genomic, epigenomic and transcriptomic data.
RESULTS
The first step of the pipeline was based on unsupervised hierarchical clustering analysis of copy number variations (CNVs) versus hypermutation status that identified a first CRC cluster with few CNVs enriched in Hypermutated and microsatellite instability samples, a second CRC cluster with a high number of CNVs mostly including non-HM and microsatellite stable samples, and a third cluster (7.8% of the entire dataset) with low CNVs and low TMB, which shared clinical-pathological features with Hypermutated CRCs and thus defined Hypermutated-like CRCs. The mutational features, DNA methylation profile and base substitution fingerprints of these tumors revealed that Hypermutated-like patients are molecularly distinct from Hypermutated and non-Hypermutated tumors and are likely to develop and progress through different genetic events. Transcriptomic analysis highlighted further differences amongst the three groups and revealed an inflamed tumor microenvironment and modulation Immune Checkpoint Genes in Hypermutated-like CRCs.
CONCLUSION
Therefore, our work highlights Hypermutated-like tumors as a distinct and previously unidentified CRC subgroup possibly responsive to immune checkpoint inhibitors. If further validated, these findings can lead to expanding the fraction of patients eligible to immunotherapy.
Identifiants
pubmed: 35534873
doi: 10.1186/s13062-022-00324-y
pii: 10.1186/s13062-022-00324-y
pmc: PMC9082922
doi:
Substances chimiques
Immune Checkpoint Inhibitors
0
Types de publication
Journal Article
Research Support, Non-U.S. Gov't
Langues
eng
Sous-ensembles de citation
IM
Pagination
10Informations de copyright
© 2022. The Author(s).
Références
Nature. 2006 Nov 30;444(7119):638-42
pubmed: 17136094
Nat Med. 2015 Nov;21(11):1350-6
pubmed: 26457759
Brief Bioinform. 2021 May 20;22(3):
pubmed: 32814346
Nucleic Acids Res. 2015 Apr 20;43(7):e47
pubmed: 25605792
Genome Med. 2017 Apr 19;9(1):34
pubmed: 28420421
Nature. 2020 Feb;578(7793):94-101
pubmed: 32025018
Mol Cancer Ther. 2017 Nov;16(11):2598-2608
pubmed: 28835386
Science. 2017 Jul 28;357(6349):409-413
pubmed: 28596308
Cancers (Basel). 2019 Jan 27;11(2):
pubmed: 30691222
Genome Biol. 2011;12(4):R41
pubmed: 21527027
Lancet. 2018 May 26;391(10135):2128-2139
pubmed: 29754777
Cell. 1997 Mar 7;88(5):593-602
pubmed: 9054499
Gut. 2019 Mar;68(3):465-474
pubmed: 29382774
N Engl J Med. 2020 Dec 3;383(23):2207-2218
pubmed: 33264544
Genome Res. 2018 Nov;28(11):1747-1756
pubmed: 30341162
BMC Genomics. 2012 Nov 04;13:591
pubmed: 23442169
Br J Cancer. 2021 Nov;125(10):1341-1349
pubmed: 34302062
Clin Cancer Res. 2008 Oct 1;14(19):6005-13
pubmed: 18829479
Gastroenterology. 2010 Jun;138(6):2059-72
pubmed: 20420946
Clin Cancer Res. 2021 Mar 1;27(5):1236-1241
pubmed: 33199494
Genome Res. 2012 Feb;22(2):271-82
pubmed: 21659424
Sci Rep. 2019 Jun 21;9(1):9020
pubmed: 31227763
Cancer Cell. 2019 Feb 11;35(2):315-328.e6
pubmed: 30753828
Clin Epigenetics. 2016 Mar 02;8:25
pubmed: 26941852
Front Oncol. 2020 May 07;10:560
pubmed: 32457828
Nat Med. 2020 Apr;26(4):566-576
pubmed: 32251400
Science. 2017 Oct 13;358(6360):234-238
pubmed: 28912133
Oncogenesis. 2018 Jul 22;7(7):55
pubmed: 30032163
Nat Rev Gastroenterol Hepatol. 2019 Jun;16(6):361-375
pubmed: 30886395
Gastroenterology. 2010 Jun;138(6):2073-2087.e3
pubmed: 20420947
J Immunother Cancer. 2019 Nov 21;7(1):313
pubmed: 31753019
Nat Immunol. 2002 Jul;3(7):611-8
pubmed: 12087419
J Mol Diagn. 2006 Nov;8(5):582-8
pubmed: 17065427
Cancer Immunol Res. 2018 Nov;6(11):1388-1400
pubmed: 30266715
Stat Appl Genet Mol Biol. 2005;4:Article17
pubmed: 16646834
Nature. 2012 Jul 18;487(7407):330-7
pubmed: 22810696
BMC Bioinformatics. 2008 Dec 29;9:559
pubmed: 19114008
Cell Immunol. 2020 Jan;347:104008
pubmed: 31733822
Oncol Lett. 2018 Jul;16(1):9-18
pubmed: 29928381
Oncol Lett. 2019 Dec;18(6):5977-5985
pubmed: 31788072
N Engl J Med. 2009 Dec 17;361(25):2449-60
pubmed: 20018966
Oncotarget. 2016 Aug 9;7(32):50875-50882
pubmed: 27248473
Cancer Cell. 2018 Jan 8;33(1):125-136.e3
pubmed: 29316426
Nat Rev Genet. 2014 Sep;15(9):585-98
pubmed: 24981601