ArticlePDF Available

The transition from primary colorectal cancer to isolated peritoneal malignancy is associated with an increased tumour mutational burden

Springer Nature
Scientific Reports
Authors:

Abstract and Figures

Colorectal Peritoneal metastases (CPM) develop in 15% of colorectal cancers. Cytoreductive surgery and heated intraperitoneal chemotherapy (CRS & HIPEC) is the current standard of care in selected patients with limited resectable CPM. Despite selection using known prognostic factors survival is varied and morbidity and mortality are relatively high. There is a need to improve patient selection and a paucity of research concerning the biology of isolated CPM. We aimed to determine the biology associated with transition from primary CRC to CPM and of patients with CPM not responding to treatment with CRS & HIPEC, to identify those suitable for treatment with CRS & HIPEC and to identify targets for existing repurposed or novel treatment strategies. A cohort of patients with CPM treated with CRS & HIPEC was recruited and divided according to prognosis. Molecular profiling of the transcriptome (n = 25), epigenome (n = 24) and genome (n = 21) of CPM and matched primary CRC was performed. CPM were characterised by frequent Wnt/ β catenin negative regulator mutations, TET2 mutations, mismatch repair mutations and high tumour mutational burden. Here we show the molecular features associated with CPM development and associated with not responding to CRS & HIPEC. Potential applications include improving patient selection for treatment with CRS & HIPEC and in future research into novel and personalised treatments targeting the molecular features identified here.
This content is subject to copyright. Terms and conditions apply.

Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports
The transition from primary
colorectal cancer to isolated
peritoneal malignancy is associated
with an increased tumour
mutational burden
Sally Hallam, Joanne Stockton, Claire Bryer, Celina Whalley, Valerie Pestinger,
Haney Youssef & Andrew D. Beggs*
Colorectal Peritoneal metastases (CPM) develop in 15% of colorectal cancers. Cytoreductive surgery
and heated intraperitoneal chemotherapy (CRS & HIPEC) is the current standard of care in selected
patients with limited resectable CPM. Despite selection using known prognostic factors survival is
varied and morbidity and mortality are relatively high. There is a need to improve patient selection
and a paucity of research concerning the biology of isolated CPM. We aimed to determine the biology
associated with transition from primary CRC to CPM and of patients with CPM not responding to
treatment with CRS & HIPEC, to identify those suitable for treatment with CRS & HIPEC and to
identify targets for existing repurposed or novel treatment strategies. A cohort of patients with CPM
treated with CRS & HIPEC was recruited and divided according to prognosis. Molecular proling of
the transcriptome (n = 25), epigenome (n = 24) and genome (n = 21) of CPM and matched primary CRC
was performed. CPM were characterised by frequent Wnt/ β catenin negative regulator mutations,
TET2 mutations, mismatch repair mutations and high tumour mutational burden. Here we show the
molecular features associated with CPM development and associated with not responding to CRS &
HIPEC. Potential applications include improving patient selection for treatment with CRS & HIPEC and
in future research into novel and personalised treatments targeting the molecular features identied
here.
Abbreviations
CRC Colorectal cancer
CPM Colorectal peritoneal metastasis
CRS & HIPEC Cytoreductive surgery and heated intraperitoneal chemotherapy
DFS Disease free survival
DMR Dierentially methylated regions
OS Overall survival
FFPE Formalin xed paran embedded
Background
Little is known about the biology of isolated colorectal peritoneal metastasis (CPM), which although a relatively
rare phenomenon is one with a high mortality rate1. Understanding tumour biology may identify which patients
with primary colorectal cancer (CRC) are at risk of developing CPM, and which are suitable for treatment with
cytoreductive surgery and heated intra-peritoneal chemotherapy (CRS & HIPEC). CRS & HIPEC (usually using
an agent such as mitomycin C or more recently, oxaliplatin) aims to achieve macroscopic tumour resection with
multiple visceral and peritoneal resections and ablation of microscopic disease. Five-year survival however varies
OPEN

*email: a.beggs@bham.ac.uk
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
widely, and morbidity and mortality are relatively high2. ere is a need therefore to improve patient selection,
allowing alternative existing or novel treatment strategies to be used for patients unlikely to respond.
Primary CRC research has identied markers of response to specic treatments, for example KRAS mutation
in selection for anti-EGFR mAb therapy3. Gene expression signatures have been developed and are in clinical
use for prognostication and therapeutic stratication in breast cancer47. Gene expression proling in primary
CRC has identied signatures associated with the development of metastasis6. One small study combining a small
number of CPM with a larger cohort of appendix adenocarcinoma identied a signature predictive of reduced
overall survival (OS) following CRS & HIPEC; these are however two biologically distinct tumours, appendix
having signicantly improved prognosis7.
e dysregulation of methylation is a key step in tumorigenesis CpG island promoter methylation (CIMP)
appears to be stable between matched primary CRC and hepatic metastasis suggesting an epigenetic methylation
programme is established prior to the development of metastasis810. Hypermethylation of KRAS, Wnt modula-
tors, tumour suppressor genes, CIMP and hypomethylation of oncogenes are associated with an unfavourable
response to chemotherapy and anti-EGFR antibodies as well as tumour recurrence and reduced OS in primary
and metastatic CRC
1116. Chromosomal instability is ubiquitous in cancer, increased copy number alteration,
indicative of chromosomal instability is found in metastatic CRC
17,18. Lopez-Garcia etal.19 demonstrated that
the evolution of chromosomal instability is depending on cellular tolerance, either via dysregulation of TP53 or
via alternate escape mechanisms such as dysfunction of BCL9L regulated caspase signalling.
CRC metastatic drivers are less clearly dened, apart from TP53 which is well characterised as being present
in metastatic cancer20. Some studies have found mutations exclusive to metastatic sites21,22, whereas others found
similar patterns of mutation between primary and metastasis23. Studies have examined the somatic mutations
in CPM and their prognostic implications. ese studies are limited to individual or small panels of mutations
routinely tested for in clinical practice with limited evidence to suggest which genes should be included in panel
sequencing in CPM. Schneider etal. examined the KRAS and BRAF mutation status of patients with CPM who
underwent CRS & HIPEC24. ey found mutations of RAS/RAF were associated with reduced OS independent
of the use of targeted anti-EGFR treatment24. Sasaki etal. examined the KRAS, BRAF and PIK3CA mutation
status of patients with metastatic CRC, with or without CPM25. ey found the incidence of BRAF mutation was
signicantly associated with the presence of CPM but not with prognosis25.
e landscape of metastatic colorectal cancer was studied by the MSK-IMPACT
20 group which undertook
panel based sequencing of 1134 metastatic colorectal cancers. Of these 39 patients were dened as “peritoneal”
malignancy, it is unclear whether these were isolated peritoneal metastasis. Only 14 of these patients had meta-
sectomy. 7 of these had peritonectomy suggesting isolated disease suitable for resection. ese tumours were
also not studied with matched primary tumour of origin.
ere is a need to improve the outcomes for patients with CPM and signicant variation in survival despite
patient selection for treatment using known prognostic factors. ere is a paucity of knowledge concerning CPM
tumour biology. Understanding tumour biology will identify patients with primary CRC at risk of developing
CPM, those suitable for treatment with CRS & HIPEC or alternative existing and novel treatment strategies. is
study aims to determine the landscape of gene expression, methylation, and somatic mutation prole associ-
ated with the transition from primary CRC to isolated CPM and determine the association between these and
prognosis following CRS & HIPEC in order to identify therapeutic targets.
Methods
Patient cohorts. is study obtained ethical approval from the North West Haydock Research Ethics Com-
mittee, (15/NW/0079), project ID (17/283). Participants gave informed consent. All experiments were per-
formed in accordance with relevant guidelines and regulations Consecutive retrospective patients were recruited
from an internally held database of all patients undergoing CRS & HIPEC at Good Hope hospital from 2011
to 2017. Patients with CPM (adenocarcinoma), no extra-abdominal metastasis, a complete resection (CC0)
and a peritoneal carcinomatosis index (PCI) of < 12 were eligible for inclusion. e completeness of cytoreduc-
tion score describes the degree of macroscopic tumour remaining aer CRS and the likelihood of benet from
intraperitoneal chemotherapy26. Patients with no residual tumour score CC0, residual tumour < 0.25cm, CC1,
residual tumour 0.25–2.5cm CC2. e extent of peritoneal metastasis is described by the PCI score. A PCI
of ≥ 12 is poor prognostic factor for patients undergoing CRS & HIPEC27. Patients were divided into two groups.
CRS & HIPEC is a long operation associated with a protracted inpatient and high dependency (HDU) or inten-
sive care (ITU) stay an associated mortality of 1–12% and morbidity of 7–63% and a prolonged post-operative
recovery2837.With palliative chemotherapy DFS is 11–13months and therefore patients post-treatment (CRS &
HIPEC) with disease free survival (DFS) < 12months were dened as “non-responders38. Patients undergoing
therapy with DFS > 12months were dened as “responders. Patients were imaged with CT which was reported
by an experienced CPM radiologist, diagnostic laparoscopy was not used, not all patients with recurrence are
suitable for iterative CRS & HIPEC and so this is not a standard procedure in their follow up. Adhesions follow-
ing primary excision and CRS & HIPEC may also preclude accurate assessment of peritoneal recurrence in all
areas with laparoscopy. Disease recurrence was determined when conrmed by CT and MDT review.
Demographic, tumour and treatment details were compared between the prognostic cohorts. For continuous
variables, the students T-test was applied to normally distributed data and Mann Whitney-U to non-normally
distributed data. Categorical variables were compared with the Chi-squared test or Fishers exact test. A p value
of < 0.05 was considered statistically signicant. DFS survival between the responders and non-responders was
compared using the Kaplan Meier method. Statistical analysis was performed in IBM SPSS Statistics for Win-
dows, Version 24.039.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
Nucleic acid extraction. DNA and RNA were extracted from histologically conrmed Formalin xed,
paran embedded (FFPE) scrolls using the Covaris E220 evolution focused-ultrasonicator and the truTRAC
FFPE total NA Kit. All peritoneal metastases samples were taken at the commencement of surgery. Nucleic acid
concentration was quantied using the Qubit 3.0 Fluorometer and Qubit RNA / DNA HS (high sensitivity)
assay kit. Nucleic acid quality was measured by electrophoresis using the Agilent 2200 TapeStation Nucleic Acid
System, Agilent 2200 TapeStation Soware A.01.05 and the Aligent High Sensitivity RNA / DNA ScreenTape
and reagents.
RNA library preparation, sequencing and bioinformatics. RNA library preparation was performed
using the Lexogen Quant Seq 3 mRNA-Seq Library Prep kit. RNA libraries were denatured, diluted, loaded onto
a 75-cycle High output ow cell and sequenced using the NextSeq500 at 2.5–5 million reads40.
Quality control, trimming and alignment to the reference genome, (NCBI build 37, hg19) was performed
with the Partek Flow genomics suite soware package (Partek, St Louis, MI, USA). e gene expression proles
of primary and CPM and responders and non-responders were compared using gene Specic Analysis (GSA)
Modelling using Partek ow with a false discovery rate (FDR) of < 0.1. Gene specic enrichment analysis (GSEA)
and gene expression pathway analysis was performed using Partek ow, a p value of ≤ 0.05 was considered sta-
tistically signicant.
CMS and CRIS classifications were performed using ‘CMScaller’ (v0.99.1) in the R package, version
2.10.238,41,42. Fishers exact test was used to compare contingency between primary and CPM and responders and
non-responders in IBM SPSS Statistics for Windows, Version 24.039. A p value of < 0.05 was considered signicant.
Methylation array and bioinformatics. DNA was treated with sodium bisulphite using the Zymo EZ-
DNA methylation kit, according to manufacturer’s instructions. Degraded FFPE DNA was restored prior to
methylation array with the Innium HD FFPE restore kit, according to manufacturer’s instructions. Methylation
array was performed according to the Innium MethylationEPIC BeadChip Kit manufacturer’s instructions.
BeadChips were imaged using the Illumina iScan system. Initial data quality was checked using GenomeStudio
Methylation Module Soware.
Raw data was loaded into the RStudio version 3.5.0 soware using the mini package. Bioinformatics analysis
was performed using the Chip Analysis Methylation Pipeline (ChAMP) R package, version 2.10.243,44. Probes
with signals from less than three functional beads, low condence with a detection p value > 0.01, covering
SNPs, non-CpG and those located on the X and Y chromosome where ltered. Beta-mixture quantile normali-
zation (BMIQ) was applied and a singular value decomposition (SVD) performed to identify batch eects. e
association between methylation and prognosis was determined using the Bioconductor R package limma and
bumphunter functions. Copy number alteration calling was performed using the CHAMP CNA function with
a signicance threshold of, p value < p < × 10–10.
Exome capture, high‑throughput sequencing and bioinformatics. DNA was sheared using the
Covaris E220 evolution focused-ultrasonicator to produce a uniform 150bp fragment size. Libraries were pre-
pared using the TruSeq Exome Kit then denatured, diluted, loaded onto a 150-cycle High output ow cell and
sequenced using the NextSeq500.
Sequencing reads were assessed using FastQC. Sequences with a Phred score of < 30 were removed giving
a base call accuracy of 99.9%. Sequence reads were aligned to the human reference genome, (hg19) using the
Burrows–Wheeler Aligner (BWA) package45. SAMTools was used to generate chromosomal coordinate-sorted
BAM les and Picard was used to remove PCR duplicates46. Somatic variants were called from matched tumour-
normal samples using Strelka2 in tumour/normal mode47. Somatic variants were viewed, ltered and annotated
in genomics workbench48. Mutations with a MAF of > 1% in known variant databases, (dbSNP and 100,000
genomes) were ltered. Mutations were annotated with information from known variant databases, (dbSNP and
100,000 genomes), PhastCons score and functional consequences. e prognostic groups were compared using
Fischer exact test to identify potential candidate driver mutations for non-responders. Somatic mutations were
entered into the IntOGen platform for further analysis49. e IntOGen-mutation platform incorporates a number
of pipelines to identify cancer driver mutations and activated pathways49. e OncodriveFM pipeline identies
mutations with a high functional impact using three scoring methods (Sorting Intolerant From Tolerant, (SIFT)50,
PolyPhen251, and Mutation Assessor scores)49,52, and assesses the likelihood that such mutations are cancer driv-
ers. e OncodriveCLUST pipeline assesses the clustering of mutations to identify relevant activated pathways49.
MSI assessment was carried out using MSI_classier_v3 (https ://rpubs .com/sigve n/msi_class ica tion_v3).
Ethics approval and consent to participate. North West Haydock Research Ethics Committee, (15/
NW/0079), project ID (17/283).
Results
Patient cohort. From 2011 to 2017 a total of n = 161 patients underwent CRS & HIPEC at University Hos-
pitals Birmingham, n = 88 patients for metachronous CPM.
Patients were excluded for the following reasons: other primary tumour (appendix, pseudomyxoma peritonei,
ovarian) n = 49, synchronous colorectal cancer n = 26, no primary tumour available n = 53 CC2 resection n = 826,
PCI of ≥ 12 n = 20, follow up period of 12months n = 27, leaving n = 28 patients. Complete information regard-
ing the primary CRC pathology and treatment was available for n = 26 patients who form the basis of this study.
Each patient had matched normal, primary CRC and CPM samples.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
irteen patients had a DFS of 24months (15–72 range) following CRS & HIPEC and formed the ‘responders
cohort, thirteen patients had a DFS of 6months (2–11 range) and formed the ‘non-responders. ere were no
signicant dierences between cohorts in demographics, primary CRC or CPM tumour, treatment or follow up
(Table1). No patients had neoadjuvant therapy for their primary tumour. ree patients (all in the responders
group) had poorly dierentiated, mucinous adenocarcinoma, one had signet ring adenocarcinoma (in the non-
responders group) and all the others had moderately dierentiated adenocarcinoma.
Following nucleic acid extraction all patients had adequate CPM RNA for RNAseq (n = 13 responders,
n = 13 non-responders), n = 25 had matched primary CRC samples. For methylation array n = 24 patients
(n = 12 responders, n = 12 non-responders) had adequate DNA. As the Innium methylation array comprises
a 32-prep kit, n = 4 responders and n = 4 non-responders primary tumours were matched to these. For exome
sequencing n = 24 patients (n = 12 responders, n = 12 non-responders) had adequate DNA from both the primary
and CPM samples, extraction of DNA from normal tissue resulted in n = 21 samples (n = 9 responders, n = 12
non-responders).
Exome sequencing. Across all six sequencing runs, we obtained a median of 60X coverage (42–166) with
a median uniformity of 88% (71–89).
Somatic mutations identied in the primary and matched CPM cohort. In the matched CPM
cohort, a total of n = 244,531 somatic SNV’s were identied (CPM-primary subtraction) signicantly more than
found in the matched primary cohort (n = 112,420).
Nine CPM samples, 9/24 (56%) had a high tumour mutational burden TMB 10 mut/Mb53 compared with
7/24 (30%) samples in the matched primary cohort. Mutations were identied in n = 69 of n = 95 known CRC
driver genes, n = 51 were shared between the primary and CPM, n = 13 were novel (supplementary tableS1)54.
Of the somatic variants identied in CPM, n = 58,958 (29%) were present in the primary CRC, n = 205,552 vari-
ants occurred exclusively in the CPM suggesting a signicant accumulation of mutations in the transition to
CPM (Fig.1). OncodriveFM identied n = 265 potential driver genes with high levels of functional mutation
(Q-value < 0.05) in the CPM cohort: FLNB, SPTB, PPL, TP53, PDE4DIP, RIOK2, CDC16, NUP98, CDC16 and
SVEP1 (supplementary tableS2), however these results must be treated with caution due to the bias of the hyper-
mutator phenotype. KEGG pathway analysis of mutations demonstrated enrichment in pathways concerning
the immune system, signalling, metabolism and cancer (supplementary tableS1). In the CPM group KRAS or
BRAF status was not signicantly associated with prognosis (chi2 p = 1.00).
Clonality analysis with SuperFreq showed signicant (Wilcoxon rank p = 0.007) dierences between the
responders and non-responders groups, with a median of 2 clones in the responders group of primary tumours
(range 1–4) and 3 clones in the non-responders group (range 2–7). In the peritoneal metastases there were a
Table 1. Comparison of responders and non-responders to CRS & HIPEC. N number value in parenthesis,
percentage, DFI disease free interval, time from primary CRC to metachronous CPM, PCI peritoneal
carcinomatosis index, CC score completeness of cytoreduction, DFS disease free survival, OS overall survival.
Log rank p < 0.0001.
Responders Non-responders p Val ue
Age, mean + /−SD 58 ± 13 58 ± 9 0.97
Gender, male n = 7 (54) n = 7 (54) 0.68
Tumour location
Right n = 9 (69) n = 6 (46)
Transverse n = 1 (8) n = 0 (0)
Le n = 3 (23) n = 7 (54) 0.33
T stage primary
3n = 3 (23) n = 3 (23)
4a n = 5 (38.5) n = 7 (54
4b n = 5 (38.5) n = 3 (23) 0.66
N stage primary
0n = 4 (31) n = 1 (8)
1n = 7 (54) n = 5 (38)
2n = 2 (15) n = 7 (54) 0.86
DFI months 25 ± 9 24 ± 12 0.83
PCI score, median (range) 5 (3–12) 8 (2–12) 0.019
CC score
CC0 n = 13 (100) n = 13 (100) 1
CC1 n = 0 (0) n = 0 (0)
CC2 n = 0 (0) n = 0 (0)
Follow up, months, median (range) 29 (19–72) 16 (5–55) 0.11
Adjuvant tre atment Yes n = 11 (85) n = 12 (92) 0.38
No n = 2 (15) n = 1 (8)
DFS, median (range) 24 (15–72) 6 (2–11) < 0.0001
OS, median (range) 29 (19–72) 16 (5–55) 0.12
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
median of 3 clones in both the responders (range 1–4) and non-responders (range 2–5) groups. Of note, in the
non-responders group during clonal expansion, the dominant clone in the peritoneal metastasis group arose
de-novo rather than being a prior clone that existed in the primary tumour (Supplementary Fig.1, S1e primary
tumours, 9/21 were MSI (47.4%) and 10/21 were MSS (52.6%) whereas in the isolated peritoneal metastasis
group, 4/21 (19.0%) were MSS and 17/21 MSI (81.0%) Demonstrating that there was a signicantly higher rate
of MSI in the isolated peritoneal metastasis group (p < 0.05, Chi2).
Non-responders had a higher frequency of somatic mutations: 60% of all mutations in CPM cohort vs. 40%.
Non-responders more commonly had a high tumour mutational burden, TMB ≥ 10 mut/Mb53, 56% vs. 44%. Of
the somatic mutations identied in non-responders, n = 35,461 (30%) were present in responders, n = 145,089
variants occurred exclusively in non-responders, suggesting a high tumour mutational burden was associated
with non-response to CRS & HIPEC (Fig.1). Mutational signature analysis of the MSI tumours demonstrated a
predominance of signature 5 (associated with mutational “clock” eects), signature 26 (associated with defective
mismatch repair) and signature 20 (associated with defective mismatch repair).
Comparison of somatic mutations in responders and non-responders identied two potential candidate genes
to identify non-responders, FAM13A and PIEZO2 (Fishers exact p < 0.05, FDR = 0.53) (Table2).
Dierentialene expression. Dierential gene expression between primary CRC and matched CPM. Pri-
mary CRC and matched CPM showed dierential expression of n = 65 genes with an FDR < 0.1. (Fig.2) Sixteen
genes showed signicantly decreased expression in CPM compared with primary CRC (Table3). Forty-nine
genes showed signicantly increased expression in CPM compared with primary CRC (Table3). A KEGG path-
way analysis was performed to identify the enriched biological functions among the dierentially expressed
genes (Supplementary Table1). e expression of FABP6, an intercellular bile acid transporter, was decreased
34.30-fold in CPM. OLFM4 is a target of the Wnt/β-catenin pathway, its expression was reduced 3.77-fold in
CPM. DCN and PTEN are able to initiate a number of signalling pathways including ERK and EGFR leading
to growth suppression, their expression was increased 3.3-fold and 3.25 fold in CPM, this was unexpected and
in contrast to the literature55. NF-κBIA expression was increased 3.24-fold in CPM, its upregulation may reect
increased NF-κB activity in the development of CPM56.
Gene specic enrichment analysis (GSEA) results are presented in supplementary table5 We identied 848
upregulated gene ontology categories in CPM and 14 upregulated gene pathways. which may contribute to the
pathogenesis of CPM: the mTOR pathway as well as immune pathways including the intestinal immune network
for IgA production, Leukocyte transendothelial migration and the actin cytoskeleton pathway.
Dierential gene expression between non-responders and responders to CRS & HIPEC. One hundred and forty-
nine genes showed increased expression in non-responders (Fig.3). Five genes showed decreased expression in
non-responders, however none had a fold change ≥ 1.5 suggesting minimal dierence in expression between the
responders and non-responders (Supplementary Table2). KEGG pathway analysis demonstrated enrichment in
endocytosis, metabolism, phagocytosis, cell movement and architecture, bacterial and viral cell infection, tran-
Figure1. Venn diagrams depicting the frequency of mutations exclusive to and shared between primary CRC
and matched CPM and responders and non-responders.
Table 2. Potential candidate variants, non-responders to CRS & HIPEC. CPM identied through Fisher exact
test, genomics workbench (Chr, chromosome, FDR, false discovery rate).
Chr Position Reference Allele p Va lu e FDR Sample frequency (case) Sample frequency (control) Gene ID
4 93,084,410 C G 0.007 0.53 62.5 0 FAM13A
18 11,552,313 G C 0.023 0.53 50 0 PIEZO2
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
BCYRN1
H19
MT2A
HBB
HBA2
MGP
TAGLN
HLA-DRB1
HLA-DRA
CD74
CTSB
TIMP1
FTL
COL1A1
COL1A2
SPARC
COL3A1
FN1
PSAP
COL6A1
ACTG1
RPS23P8
PPIA
FTH1
TPT1
EEF1A1
TMSB4X
TMSB10
B2M
S100A6
AC132217.4
RP11-543P15.1
ACTB
NEAT1
RNA5-8SP6
RP11-742N3.1
TFF3
CKB
PIGR
MTND1P23
PRSS3
GP2
CTRB1
CELA3B
AMY2A
PLA2G1B
CPB1
CTRB2
CLPS
PNLIP
PRSS1
CPA1
REG1A
SP100
IGHG4
IGHA1
IGJ
IGHG3
IGLC3
IGKC
IGLC2
IGLC1
MYL9
MALAT1
CRS
P
Z-score
Figure2.
Heatmap of dierential gene expression in 100 highest genes ranked by variance between primary CRC (P, red) and colorectal peritoneal metastasis (CRS, blue). Sample type is
indicated at the X axis of the heatmap with individual genes on the Y-axis. Individual IDs of each patient are below the indicators of primary or CRS sample. Gene expression as indicated by the
Z-score is displayed as colour ranging from green to black to red as shown in the legend. Created in Partek Flow.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
scription and the expression of genes controlling apoptosis, cell cycle, oxidative stress resistance and longevity
(Table3). e expression of CEACAM1, a member of the carcinoembryonic antigen (CEA) immunoglobulin
family, was increased 8.27-fold in non-responders57.
AXIN1 encodes a cytoplasmic protein which forms the ß-Catenin destruction complex, a negative regulator
of the WNT signalling pathway58. AXIN1 expression was increased 5.42-fold in non-responders59.
Gene specic enrichment analysis (GSEA) results are presented in supplementary table6. We identied 591
upregulated gene ontology categories in CPM and 15 upregulated gene pathways. which may contribute to the
pathogenesis of CPM: Endocytosis, the adherens junction pathway and immune pathways such as those regulat-
ing the bacterial invasion of epithelial cells.
Amongst the n = 51 primary CRC and CPM samples n = 29 were representative of each CMS subtype, the
remaining n = 22 samples did not have a consistent pattern (Fig.4). Comparison of the CMS subtypes in primary
and CPM and prognostic groups revealed an apparent transition from primary CRC to CPM. No primary CRC
samples were classied as CMS4 (mesenchymal subtype characterized by prominent transforming growth factor
activation, stromal invasion and angiogenesis) compared to 31% of CPM (p = 0.085). Secondly, non-responders
were more commonly CMS4, 46% vs. 15% (p = 0.005, Table4).
Methylation. Dierential methylation between primary CRC and matched CPM. irty-two samples in
total were hybridised successfully to the Illumina HumanMethylation EPIC microarrays. DMPs were called
between the primary CRC and CPM. e top ranked dierentially methylated probe was cg04146982, BF 34.5,
adjusted p value 5.67 × 10–16 (chr8:144,943,810–144,943,810, hg19 coordinates), which tags a CpG dinucleotide
3651bp upstream of the transcription start site of gene Epiplakin 1,(EPPK1)60. EPPK1 is part of the Plakin fam-
ily an important component of the cell cytoskeleton61. e other DMP was cg12209861, BF 7.1, adjusted p value
0.059 (chr4:37,459,078–37,459,078, hg19 coordinates), 3526bp upstream of the transcription start site of gene
Chromosome 4 Open Reading Frame 19, (C4orf19). DMRs were called between primary CRC and CPM via the
dmrLasso function of the CHAMP pipeline (Supplementary Table3). e top 10 most DMRs were in the region
of IGF2, ZNF461, RASGFR1, CELF4, ZSCAN18, EDNRB, ZBED9, VTRNA2-1, ZNF256 and EGFLAM. KEGG
pathway analysis did not reveal any signicantly enriched pathways.
Comparison of CNA between primary and CPM via methylation arrays did not identify and signicant dif-
ferences in CNA between primary and CPM at a stringent p value of < × 10–10 however a number of CNA were
identied at a lower signicance threshold, p = 2.78 × 10–07 (Supplementary Table4).Genes showing CNA gains
of known signicance in patients with CPM included; TRIM3, 5, 6, 21 and 22, MT1A, 2A, 3, 4 encode proteins
of the metallothionein family.
Table 3. e top 10 genes with signicantly altered expression (FDR < 0.1) in CPM samples compared with
primary CRC samples.
Rank Gene name Function Fold change FDR p value
Reduced expression CPM samples vs. primary CRC
1 FABP6 Intracellular bile acid transporter − 34.30 1.74 × 10–06
2 DEFA6 Cytotoxic peptide involved in host intestine defence − 8.15 8.55 × 10–06
3 DMBT1 Tumour suppressor − 6.06 2.43 × 10–04
4 TTC38 Protein coding gene − 4.56 5.80 × 10–05
5 OLFM4 Wnt/β-catenin pathway target − 3.77 1.01 × 10–04
6 IGHA1 Immune receptor − 3.66 4.23 × 10–05
7 CES2 Intestinal enzyme controlling drug clearance − 3.20 6.84 × 10–05
8 NDUFS6 Enzyme in electron transport chain of mitochondria − 2.70 7.74 × 10–05
9 P2RY11 G-protein coupled receptor − 2.53 6.37 × 10–04
10 MUC2 Encodes a mucinous intestinal coating − 2.34 7.22 × 10–04
Increased expression CPM samples vs. primary CRC
1 CD53 Tetraspanin 7.29 5.87 × 10–05
2 CYR61 Extracellular signalling protein 4.24 3.12 × 10–04
3 CXCL12 G-protein coupled receptor 3.64 9.25 × 10–04
4 NR2F1 Nuclear hormone receptor and transcriptional regulator 3.53 7.09 × 10–04
5 CTGF C onnective tissue growth factor 3.49 1.55 × 10–04
6 CSTB Cystatin 3.41 6.13 × 10–04
7 TSC22D3 Anti-inammatory protein glucocorticoid (GC)-induced leucine zipper 3.36 3.94 × 10–04
8 DCN Tumour suppressor gene 3.30 6.19 × 10–05
9 PTEN Tumour suppressor gene 3.25 9.28 × 10–04
10 NF-κBIA Inhibits the NF-κB transcription factor 3.24 1.06 × 10–04
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
NEAT1
RP11-742N3.1
RNA5-8SP6
MALAT1
MYL9
SP100
IGHG4
IGHA1
IGJ
IGKC
IGLC3
IGLC2
IGLC1
IGHG3
REG1A
GP2
CTRB1
PLA2G1B
CTRB2
CPB1
CPA1
PRSS1
PNLIP
CLPS
AMY2A
CELA3B
PRSS3
CKB
TFF3
PIGR
MTND1P23
BCYRN1
H19
MT2A
HBA2
HBB
MGP
TAGLN
HLA-DRB1
HLA-DRA
CD74
CTSB
AC132217.4
S100A6
PPIA
TPT1
FTH1
TMSB4X
EEF1A1
TMSB10
B2M
RPS23P8
ACTG1
COL6A1
TIMP1
FTL
FN1
COL1A2
SPARC
COL3A1
COL1A1
PSAP
RP11-543P15.1
ACTB
Resp
Non-r
Z-score
Figure3.
Heatmap dierential gene expression of top 100 genes as ranked by variance between responders (blue) and non-responders (red)Sample type is indicated at the transverse border
of the heatmap with individual genes on the longitudinal border. Gene expression as indicated by the Z-score is displayed as colour ranging from green to black to red as shown in the legend.
Created in Partek Flow.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
Figure4. Sankey diagram depicting the transition in consensus molecular subtypes (CMS) from primary to
CPM. CMS classications were performed using ‘CMScaller’ (v0.99.1) in the R /Bioconductor statistics package.
Classications include CMS1 to CMS4, non-consensus samples do not have a consistent pattern of subtype label
association. Primary CRC samples, classication and number are shown to the le of the diagram with CPM
samples, classication and number to the right of the diagram. Fishers exact p value 0.085, values in parenthesis
percentages.
Table 4. CMS classication responders vs. non-responders to CRS & HIPEC. CMS Fishers exact p value
0.005, CRIS Fischer’s exact p value 0.148, values in parenthesis percentages.
Non-consensus CMS1 CMS2 CMS3 CMS4 Tot a l
Responders 10 (77) 0 (0) 0 (0) 1 (8) 2 (15) 13
Non-responders 2 (15) 1 (8) 3 (23) 1 (8) 6 (46) 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved

Vol:.(1234567890)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
Dierential methylation between non-responders and responders to CRS & HIPEC. e top ranked dierentially
methylated probe was cg07951355, BF = 6, (chr1:40,123,717) which tags an intergenic region 1076bp before
gene NT5C1A. Cg25909064, BF 4 adjusted p value 0.47 (chr11:120,081,487–120,082,345) which tags an intron
of gene OAF and cg12977942, BF 4 adjusted p value 0.47 (chr5:92,839,309–92,839,309) which tags an intron of
gene NR2F1-AS160. Six signicant DMRs (Supplementary Table3) were identied in the regions of NKX6-2,
CHFR, GATA3, IRX5, HCK and BC019904. KEGG pathway analysis did not reveal any signicantly enriched
pathways.
Comparison of CNA between the CPM prognostic groups identied recurrent gene losses at chromosomes 3,
4, 14, 15, 17 and 19 (Supplementary Table4). CNA losses clustered in the RAS-MAPK-ERK signalling pathway
suggesting dysregulation in non-responders.
Comparison of CNA between the CPM prognostic groups identied n = 19 gene gains at chromosomes 9,
10 and 11. Genes showing CNA gains in non-responders included: SIT1, RNF38, MELK, PAX5, SHB, ZEB1,
DEAF1, ANTXR, EPS8L2 and PIDD1.
Discussion
is study determined the gene expression, CNA, methylation and somatic mutation prole of primary CRC and
matched isolated CPM to determine whether there were changes associated with the development of CPM or
predicting prognosis for patients with CPM. To our knowledge, this is the rst such analysis in a cohort of patients
with isolated CPM suitable for treatment with CRS & HIPEC. e MSKCC cohort of metastatic cancer20 had a
diverse range of metastatic cancer, none of whom overlapped with the type we have studied, which is isolated
colorectal peritoneal metastasis, with matched primary samples, suitable for cytoreduction.
Within this study responders and non-responders to CRS & HIPEC were well matched by demographics,
tumour stage, treatment and follow up. PCI varied between groups with responders having a median PCI of 5
(3–12) and non-responders a median PCI of 8 (2–12). A PCI of greater than 12 is associated with reduced survival
following CRS & HIPEC, no signicant dierence is consistently found at PCI levels below this27.
Comparison of patients with primary CRC and metachronous CPM identied biological changes associated
with the transition from primary CRC to CPM. Hypermethylation, CNA and hypermutation resulted in the
inactivation of tumour suppressors and oncogene activation in CPM, (TP53, VTRA2-1, TRIM proteins). ese
changes suggest a rapid rate of tumour growth unchecked by tumour suppressor or apoptotic mechanisms.
Increased MAPK and Wnt/β-catenin pathway activation was noted in CPM. Gene expression of negative
regulators of the Wnt pathway was reduced, (OLFM4, DEAFA6), negative Wnt regulators contained somatic
mutations, (APC, RNF43, FAM123B and TSC1), and the MAPK marker, RASFGFR1 was hypermethylated
suggesting persistent activation of MAPK and Wnt pathways. Multiple mutations of negative Wnt signalling
regulators make this an attractive therapeutic target. Porcupine inhibitors mediate the palmitoylation of Wnt
ligands, blocking Wnt signalling. e porcupine inhibitor LGK974 inhibits the upstream negative Wnt regulator
mutant RNF43 and is a potential therapeutic target in CPM62.
CPM contained a high proportion of MSH6 somatic mutations suggesting deciency in the mismatch repair
pathway and MSI. MSH6 mutations are commonly found in isolated peritoneal metastasis59. As expected for
tumours with mismatch repair deciency both the primary CRC and CPM cohort had a high tumour mutational
burden, crucially this suggests they may have a good response to treatment with immune checkpoint inhibitors
such as pembrolizumab63, a new therapeutic avenue for these dicult to treat patients. e frequency of hyper-
mutation seen in our study (48%) was considerably higher than that observed for both the MSKCC metastatic
disease cohort (5%) and the TCGA Colorectal64 cohort (10%). e expression of genes regulating innate immu-
nity however was downregulated, (DEFA6, DMBT1, MUC2) or altered via somatic mutations, (HLA-A antigen)
suggesting immune evasion in the transition to CPM which may reduce the likelihood of successful PD-1 therapy.
e expression of genes supressing invasion, migration and EMT was downregulated or hypermethylated,
(MUC2, MMP26, ILK, FLNB, SPTB, PPL, and SVEP1) and those triggering these processes upregulated, (CYR61,
CXCL12, CTGF, and CSTB). ese changes suggest a mechanism by which CPM cells metastasise from the
primary CRC. In keeping with changes in EMT regulators there appeared to be a transition in CMS subtypes
towards CMS4 from primary CRC to CPM. e CMS4 subtype is an interesting therapeutic target, TGFβ sig-
nalling inhibitors and targeted immunotherapies have been trialled with success in pre-clinical models to block
cross talk between the tumour microenvironment and halt disease progression of stromal rich CMS4 CRC
65,66.
Methylation appeared to be dysregulated in CPM with a bias towards a hypermethylator phenotype caused by
somatic mutation of the TET2 tumour suppressor and CDH7 chromatin regulator. Active DNA demethylation
by TET enzymes is an important tumour suppressor mechanism in a variety of cancers6769. Downregulation of
CES2, a gene known to activate the prodrug irinotecan, a chemotherapy used as part of the FOLFIRI regimen in
the UK in the adjuvant treatment of primary CRC and CPM was seen in this cohort. Resistance to the treatment
of primary CRC may in part explain the development of CPM.
CEACAM1 expression correlates with metastasis and reduced survival in CRC and was upregulated in this
cohort of patients70. Novel therapies in the form or CEA TCB IgG-based T-cell bispecic antibodies (Cibisa-
tamab) may therefore be of benet71. Additionally there was a downregulation of gene expression of negative
regulators of the Wnt pathway, (AXIN1) and somatic mutations of key Wnt regulators, (FAM13A) and hyper-
methylation of MAPK and TGF-β pathway markers, (RAB8A, RAB34, FGF5 and BMP3) suggesting persistent
activation of MAPK, TGF-β and Wnt in non-responders to CRS & HIPEC.
A recent randomised controlled trial has called into question the use of HIPEC in CPM, PRODIGE-7 treated
patients with CPM with CRS & HIPEC or CRS alone in addition to systemic chemotherapy. PRODIGE-7 suggests
no added benet from HIPEC however this study was not powered to stratify the impact of HIPEC according to
Content courtesy of Springer Nature, terms of use apply. Rights reserved

Vol.:(0123456789)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
PCI score, on subgroup analysis patients with a PCI of 11–15 had signicantly improved median survival with
the addition of HIPEC 41.6months vs. 32.7months p value 0.020972.
A relative weakness of this study is the small cohort of patients, the biological changes identied here form a
starting point in identifying the tumour biology associated with the development of CPM and predicting non-
responders to CRS & HIPEC. However, we have identied multiple potential targets for therapy, along with the
important nding that CPM appears to be a hypermutated, hypermethylated, immune evasive cancer which
allows it to be potentially targeted by emerging novel therapeutics. Our study ndings have implications for the
recent addition of oxaliplatin to HIPEC, as the FOXTROT study of neoadjuvant therapy in colorectal cancer
showed that oxaliplatin has no eect in dMMR tumours.
Conclusions
Patients with colorectal peritoneal metastasis (CPM) secondary to colorectal cancer have limited survival with the
best available treatments. Despite selection for treatment using known prognostic factors survival varies widely
and can be dicult to predict. ere is a paucity of knowledge concerning the biology of CPM, it is likely that
there are additional biological markers of response to currently available as well as novel or re-purposed alterna-
tive treatments. Here we have comprehensively proled a cohort of patients with isolated CPM and identied a
number of therapeutically targetable alterations including mutations in Wnt/β catenin regulators (via Porcupine
inhibitors), the mismatch repair pathway (via PD-1/CTLA-4 immunotherapy) and methylation regulators. We
suggest that these are urgently investigated in a larger cohort with the development of pre-clinical models as, in
particular, the nding that these patients may be sensitive to immunotherapy may radically change the therapy
options available for this dicult to treat group of patients.
Data availability
e data that support the ndings of this study are available from the corresponding author upon reasonable
request.
Received: 20 May 2020; Accepted: 13 October 2020
References
1. Mirnezami, R. et al. Cytoreductive surgery in combination with hyperthermic intraperitoneal chemotherapy improves survival in
patients with colorectal peritoneal metastases compared with systemic chemotherapy alone. Br. J. Cancer 111, 1500–1508. https
://doi.org/10.1038/bjc.2014.419 (2014).
2. Koumpa, F. S. et al. Colorectal peritoneal metastases: a systematic review of current and emerging trends in clinical and translational
research. Gastroenterol. Res. Pract. 2019, 5180895. h ttps ://doi.org/10.1155/2019/51808 95 (2019).
3. Lièvre, A. et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 66, 3992–
3995. https ://doi.org/10.1158/0008-5472.CAN-06-0191 (2006).
4. Cardoso, F. et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N. Engl. J. Med. 375, 717–729.
https ://doi.org/10.1056/NEJMo a1602 253 (2016).
5. van de Vijver, M. J. et al. A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 347, 1999–2009.
https ://doi.org/10.1056/NEJMo a0219 67 (2002).
6. Friederichs, J. et al. Gene expression proles of dierent clinical stages of colorectal carcinoma: toward a molecular genetic under-
standing of tumor progression. Int. J. Colorectal Dis. 20, 391–402. https ://doi.org/10.1007/s0038 4-004-0722-1 (2005).
7. Levine, E. A. et al. Gene expression proling of peritoneal metastases from appendiceal and colon cancer demonstrates unique
biologic signatures and predicts patient outcomes. J. Am. Coll. Surg. 214, 599–606; discussion 606–597. https ://doi.org/10.1016/j.
jamco llsur g.2011.12.028 (2012).
8. Ju, H. X. et al. Distinct proles of epigenetic evolution between colorectal cancers with and without metastasis. Am. J. Pathol. 178,
1835–1846. https ://doi.org/10.1016/j.ajpat h.2010.12.045 (2011).
9. Konishi, K. et al. DNA methylation proles of primary colorectal carcinoma and matched liver metastasis. PLoS ONE 6, e27889.
https ://doi.org/10.1371/journ al.pone.00278 89 (2011).
10. Murata, A. et al. Methylation levels of LINE-1 in primary lesion and matched metastatic lesions of colorectal cancer. Br. J. Cancer
109, 408–415. https ://doi.org/10.1038/bjc.2013.289 (2013).
11. Miranda, E. et al. Genetic and epigenetic changes in primary metastatic and nonmetastatic colorectal cancer. Br. J. Cancer 95,
1101–1107. https ://doi.org/10.1038/sj.bjc.66033 37 (2006).
12. Sharrard, R. M., Royds, J. A., Rogers, S. & Shorthouse, A. J. Patterns of methylation of the c-myc gene in human colorectal cancer
progression. Br. J. Cancer 65, 667–672 (1992).
13. Siemens, H. et al. Detection of miR-34a promoter methylation in combination with elevated expression of c-Met and β-catenin pre-
dicts distant metastasis of colon cancer. Clin. Cancer Res. O. J. Am. Assoc. Cancer Res. 19, 710–720. https ://doi.org/10.1158/1078-
0432.CCR-12-1703 (2013).
14. Tang, D. et al. Diagnostic and prognostic value of the methylation status of secreted frizzled-related protein 2 in colorectal cancer.
Clin. Invest. Med. 34, E88-95 (2011).
15. Zhang, X. et al. CpG island methylator phenotype is associated with the ecacy of sequential oxaliplatin- and irinotecan-based
chemotherapy and EGFR-related gene mutation in Japanese patients with metastatic colorectal cancer. Int. J. Clin. Oncol. 21,
1091–1101. https ://doi.org/10.1007/s1014 7-016-1017-6 (2016).
16. Kim, S. H. et al. CpG island methylator phenotype and methylation of Wnt pathway genes together predict survival in patients
with colorectal cancer. Yonsei Med. J. 59, 588–594. https ://doi.org/10.3349/ymj.2018.59.5.588 (2018).
17. Lee, S. Y. et al. Comparative genomic analysis of primary and synchronous metastatic colorectal cancers. PLoS ONE 9, e90459.
https ://doi.org/10.1371/journ al.pone.00904 59 (2014).
18. Turajlic, S. et al. Deterministic evolutionary trajectories inuence primary tumor growth: TRACERx renal. Cell 173, 595-610 e511.
https ://doi.org/10.1016/j.cell.2018.03.043 (2018).
19. Lopez-Garcia, C. et al. BCL9L dysfunction impairs caspase-2 expression permitting aneuploidy tolerance in colorectal cancer.
Cancer Cell 31, 79–93. https ://doi.org/10.1016/j.ccell .2016.11.001 (2017).
20. Yaeger, R. et al. Clinical sequencing denes the genomic landscape of metastatic colorectal cancer. Cancer Cell 33, 125-136.e123.
https ://doi.org/10.1016/j.ccell .2017.12.004 (2018).
Content courtesy of Springer Nature, terms of use apply. Rights reserved

Vol:.(1234567890)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
21. Goryca, K. et al. Exome scale map of genetic alterations promoting metastasis in colorectal cancer. BMC Genet. 19, 85. https ://doi.
org/10.1186/s1286 3-018-0673-0 (2018).
22. Huang, D. et al. Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metastasis Rev. 37, 173–187.
https ://doi.org/10.1007/s1055 5-017-9726-5 (2018).
23. Mlecnik, B. et al. e tumor microenvironment and immunoscore are critical determinants of dissemination to distant metastasis.
Sci. Transl. Med. 8, 327ra326. https ://doi.org/10.1126/scitr anslm ed.aad63 52 (2016).
24. Schneider, M. A. et al. Mutations of RAS/RAF proto-oncogenes impair survival aer cytoreductive surgery and HIPEC for peri-
toneal metastasis of colorectal origin. Ann. Surg. 268, 845–853. https ://doi.org/10.1097/SLA.00000 00000 00289 9 (2018).
25. Sasaki, Y. et al. Value of KRAS, BRAF, and PIK3CA mutations and survival benet from systemic chemotherapy in colorectal
peritoneal carcinomatosis. Asian Pac. J Cancer Prev. 17, 539–543 (2016).
26. Sugarbaker, P. H. Successful management of microscopic residual disease in large bowel cancer. Cancer Chemother. Pharmacol.
43(Suppl), S15-25 (1999).
27. Hallam, S., Tyler, R., Price, M., Beggs, A. & Youssef, H. Meta-analysis of prognostic factors for patients with colorectal perito-
neal metastasis undergoing cytoreductive surgery and heated intraperitoneal chemotherapy. BJS Open 3, 585–594. https ://doi.
org/10.1002/bjs5.50179 (2019).
28. Cashin, P. H., Graf, W., Nygren, P. & Mahteme, H. Cytoreductive surgery and intraperitoneal chemotherapy for colorectal peritoneal
carcinomatosis: prognosis and treatment of recurrences in a cohort study. Eur. J. Surg. Oncol. 38, 509–515. https ://doi.org/10.1016/j.
ejso.2012.03.001 (2012).
29. Glehen, O. et al. Intraperitoneal chemohyperthermia and attempted cytoreductive surgery in patients with peritoneal carcinoma-
tosis of colorectal origin. Br. J. Surg. 91, 747–754. https ://doi.org/10.1002/bjs.4473 (2004).
30. Shen, P. et al. Cytoreductive surgery and intraperitoneal hyperthermic chemotherapy with mitomycin C for peritoneal carcino-
matosis from nonappendiceal colorectal carcinoma. Ann. Surg. Oncol. 11, 178–186 (2004).
31. Cavaliere, F. et al. 120 peritoneal carcinomatoses from colorectal cancer treated with peritonectomy and intra-abdominal chemo-
hyperthermia: a SITILO multicentric study. Vivo 20, 747–750 (2006).
32. Vaira, M. et al. Treatment of peritoneal carcinomatosis from colonic cancer by cytoreduction, peritonectomy and hyperthermic
intraperitoneal chemotherapy (HIPEC). Experience of ten years. In Vivo 24, 79–84 (2010).
33. Hompes, D. et al. e treatment of peritoneal carcinomatosis of colorectal cancer with complete cytoreductive surgery and hyper-
thermic intraperitoneal peroperative chemotherapy (HIPEC) with oxaliplatin: a Belgian multicentre prospective phase II clinical
study. Ann. Surg. Oncol. 19, 2186–2194. https ://doi.org/10.1245/s1043 4-012-2264-z (2012).
34. Froysnes, I. S., Larsen, S. G., Spasojevic, M., Dueland, S. & Flatmark, K. Complete cytoreductive surgery and hyperthermic intra-
peritoneal chemotherapy for colorectal peritoneal metastasis in Norway: prognostic factors and oncologic outcome in a national
patient cohort. J. Surg. Oncol. 114, 222–227. https ://doi.org/10.1002/jso.24290 (2016).
35. Desantis, M. et al. Morbidity, mortality, and oncological outcomes of 401 consecutive cytoreductive procedures with hyperthermic
intraperitoneal chemotherapy (HIPEC). Langenbecks Arch. Surg. 400, 37–48. https ://doi.org/10.1007/s0042 3-014-1253-z (2015).
36. Simkens, G. A. et al. Predictors of severe morbidity aer cytoreductive surgery and hyperthermic intraperitoneal chemotherapy
for patients with colorectal peritoneal carcinomatosis. Ann. Surg. Oncol. 23, 833–841. https ://doi.org/10.1245/s1043 4-015-4892-6
(2016).
37. Simkens, G. A. et al. Treatment-related mortality aer cytoreductive surgery and HIPEC in patients with colorectal peritoneal
carcinomatosis is underestimated by conventional parameters. Ann. Surg. Oncol. 23, 99–105. https ://doi.org/10.1245/s1043 4-015-
4699-5 (2016).
38. Team, R. C. R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing (2020).
39. Corp, I. IBM SPSS Statistics for Windows, Version 24.0, Released 2016.
40. Liu, Y., Zhou, J. & White, K. P. RNA-seq dierential expression studies: more sequence or more replication?. Bioinformatics 30,
301–304. https ://doi.org/10.1093/bioin forma tics/btt68 8 (2014).
41. Rinaudo, P., Boudah, S., Junot, C. & évenot, E. A. biosigner: a new method for the discovery of signicant molecular signatures
from omics data. Front. Mol. Biosci. 3, 26. https ://doi.org/10.3389/fmolb .2016.00026 (2016).
42. Eide, P. W., Bruun, J., Lothe, R. A. & Sveen, A. CMScaller: an R package for consensus molecular subtyping of colorectal cancer
pre-clinical models. Sci. Rep. 7, 16618. https ://doi.org/10.1038/s4159 8-017-16747 -x (2017).
43. Morris, T. J. et al. ChAMP: 450k chip analysis methylation pipeline. Bioinformatics 30, 428–430. https ://doi.org/10.1093/bioin
forma tics/btt68 4 (2014).
44. Aryee, M. J. et al. Min: a exible and comprehensive bioconductor package for the analysis of innium DNA methylation micro-
arrays. Bioinformatics 30, 1363–1369. https ://doi.org/10.1093/bioin forma tics/btu04 9 (2014).
45. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595. https
://doi.org/10.1093/bioin forma tics/btp69 8 (2010).
46. Li, H. et al. e sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079. ht t ps ://doi.org/10.1093/bioin forma
tics/btp35 2 (2009).
47. Saunders, C. T. et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics
28, 1811–1817. https ://doi.org/10.1093/bioin forma tics/bts27 1 (2012).
48. Qiagen. CLC Genomics Workbench 11.0, https ://www.qiage nbioi nform atics .com.
49. Gonzalez-Perez, A. et al. IntOGen-mutations identies cancer drivers across tumor types. Nat. Methods 10, 1081–1082. https ://
doi.org/10.1038/nmeth .2642 (2013).
50. Ng, P. C. & Heniko, S. SIFT: predicting amino acid changes that aect protein function. Nucleic Acids Res. 31, 3812–3814 (2003).
51. Adzhubei, I., Jordan, D. M. & Sunyaev, S. R. Predicting functional eect of human missense mutations using PolyPhen-2. Cur r.
Protoc. Hum. Genet. https ://doi.org/10.1002/04711 42905 .hg072 0s76 (2013).
52. Reva, B., Antipin, Y. & Sander, C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic
Acids Res. 39, e118. https ://doi.org/10.1093/nar/gkr40 7 (2011).
53. Büttner, R. et al. Implementing TMB measurement in clinical practice: considerations on assay requirements. ESMO Open 4,
e000442. https ://doi.org/10.1136/esmoo pen-2018-00044 2 (2019).
54. IntOGen. IntOGen Colorectal cancer driver genes, https ://www.intog en.org/searc h.
55. Copija, A. et al. PTEN—clinical signicance in colorectal cancer. OncoReview 6, A86-90 (2016).
56. Perkins, N. e diverse and complex roles of NF-κB subunits in cancer. Nat. Rev. Cancer 12, 121–132 (2012).
57. Safran, M. et al. GeneCards Version 3: the human gene integrator. Database (Oxford) 2010, baq020. ht tps ://doi.org/10.1093/datab
ase/baq02 0 (2010).
58. Morin, P. et al. Activation of β-catenin-Tcf signaling in colon cancer by mutations in β-catenin or APC. Science 275, 1787–1790
(1997).
59. Kim, C. G. et al. Eects of microsatellite instability on recurrence patterns and outcomes in colorectal cancers. Br. J. Cancer 115,
25–33. https ://doi.org/10.1038/bjc.2016.161 (2016).
60. Kent, W. J. et al. e human genome browser at UCSC. Genome Res. 12, 996–1006. https ://doi.org/10.1101/gr.22910 2 (2002).
61. Hu, L., Huang, Z., Wu, Z., Ali, A. & Qian, A. Mammalian Plakins, giant cytolinkers: versatile biological functions and roles in
cancer. Int. J. Mol. Sci. https ://doi.org/10.3390/ijms1 90409 74 (2018).
Content courtesy of Springer Nature, terms of use apply. Rights reserved

Vol.:(0123456789)
Scientic Reports | (2020) 10:18900 | 
www.nature.com/scientificreports/
62. Bahrami, A. et al. erapeutic potential of targeting Wnt/β-catenin pathway in treatment of colorectal cancer: rational and progress.
J. Cell Biochem. 118, 1979–1983. https ://doi.org/10.1002/jcb.25903 (2017).
63. Schrock, A. B. et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic
colorectal cancer. Ann. Oncol. O. J. Eur. Soc. Med. Oncol. https ://doi.org/10.1093/annon c/mdz13 4 (2019).
64. Cancer Genome Atlas, N. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337.
https ://doi.org/10.1038/natur e1125 2 (2012).
65. Becht, E. et al. Immune and stromal classication of colorectal cancer is associated with molecular subtypes and relevant for
precision immunotherapy. Clin. Cancer Res. O. J. Am. Assoc. Cancer Res. 22, 4057–4066. https ://doi.org/10.1158/1078-0432.
CCR-15-2879 (2016).
66. Calon, A. et al. Stromal gene expression denes poor-prognosis subtypes in colorectal cancer. Nat. Genet. 47, 320–329. https ://
doi.org/10.1038/ng.3225 (2015).
67. Tahara, T. et al. Colorectal carcinomas with CpG island methylator phenotype 1 frequently contain mutations in chromatin regula-
tors. Gastroenterology 146, 530-538.e535. https ://doi.org/10.1053/j.gastr o.2013.10.060 (2014).
68. Jones, P. A. & Baylin, S. B. e fundamental role of epigenetic events in cancer. Nat. Rev. Genet. 3, 415–428. https ://doi.org/10.1038/
nrg81 6 (2002).
69. Carter, C. A. et al. No patient le behind: the promise of immune priming with epigenetic agents. Oncoimmunology 6, e1315486.
https ://doi.org/10.1080/21624 02X.2017.13154 86 (2017).
70. B eauchemin, N. & Arabzadeh, A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression
and metastasis. Cancer Metastasis Rev. 32, 643–671. https ://doi.org/10.1007/s1055 5-013-9444-6 (2013).
71. Bacac, M. et al. A novel carcinoembryonic antigen T-cell bispecic antibody (CEA TCB) for the treatment of solid tumors. Clin.
Cancer Res. O. J. Am. Assoc. Cancer Res. 22, 3286–3297. https ://doi.org/10.1158/1078-0432.CCR-15-1696 (2016).
72. Quenet, et al. A UNICANCER phase III trial of hyperthermic intra-peritoneal chemotherapy (HIPEC) for colorectal peritoneal
carcinomatosis (PC): PRODIGE 7. J. Clin. Oncol. 36, LBA3503 (2018).
Author contributions
Study design: A.D.B., S.H., H.Y. Patient recruitment: S.H., H.Y. Molecular analysis: S.H., J.S., C.W., C.B., V.P.
Funding
e study was funded by a grant from the Good Hope Hospital Charity. ADB is funded by a Cancer Research
UK Advanced Clinician Scientist Award (C31641/A23923).
Competing interests
e authors declare no competing interests.
Additional information
Supplementary information is available for this paper at https ://doi.org/10.1038/s4159 8-020-75844 -6.
Correspondence and requests for materials should be addressed to A.D.B.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the articles Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http://creat iveco mmons .org/licen ses/by/4.0/.
© e Author(s) 2020
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... In spite of shortcomings in existing studies, and in agreement with findings in mCRC in general, PM-CRC patients with tumors that have mutations in the BRAF oncogene have been identified as a subgroup with less favorable prognosis than BRAF wild-type cases. A few transcriptomic studies have been performed on a limited number of PM-CRC cases (n = 4-52 cases) [3,6,7], focusing on differences in gene expression between subgroups of CMS4 tumors [6], between PM-CRC and primary cancers, and between responders and non-responders to CRS-HIPEC [7]. However, analyses to unravel molecular consequences of mutational subgroups on transcriptional changes have not been performed in PM-CRC, specifically. ...
... In spite of shortcomings in existing studies, and in agreement with findings in mCRC in general, PM-CRC patients with tumors that have mutations in the BRAF oncogene have been identified as a subgroup with less favorable prognosis than BRAF wild-type cases. A few transcriptomic studies have been performed on a limited number of PM-CRC cases (n = 4-52 cases) [3,6,7], focusing on differences in gene expression between subgroups of CMS4 tumors [6], between PM-CRC and primary cancers, and between responders and non-responders to CRS-HIPEC [7]. However, analyses to unravel molecular consequences of mutational subgroups on transcriptional changes have not been performed in PM-CRC, specifically. ...
Article
Full-text available
Background Patients with peritoneal metastasis from colorectal cancer (PM-CRC) have inferior prognosis and respond particularly poorly to chemotherapy. This study aims to identify the molecular explanation for the observed clinical behavior and suggest novel treatment strategies in PM-CRC. Methods Tumor samples (230) from a Norwegian national cohort undergoing surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) with mitomycin C (MMC) for PM-CRC were subjected to targeted DNA sequencing, and associations with clinical data were analyzed. mRNA sequencing was conducted on a subset of 30 samples to compare gene expression in tumors harboring BRAF or KRAS mutations and wild-type tumors. Results BRAF mutations were detected in 27% of the patients, and the BRAF-mutated subgroup had inferior overall survival compared to wild-type cases (median 16 vs 36 months, respectively, p < 0.001). BRAF mutations were associated with RNF43/RSPO aberrations and low expression of negative Wnt regulators (ligand-dependent Wnt activation). Furthermore, BRAF mutations were associated with gene expression changes in transport solute carrier proteins (specifically SLC7A6) and drug metabolism enzymes (CES1 and CYP3A4) that could influence the efficacy of MMC and irinotecan, respectively. BRAF-mutated tumors additionally exhibited increased expression of members of the novel butyrophilin subfamily of immune checkpoint molecules (BTN1A1 and BTNL9). Conclusions BRAF mutations were frequently detected and were associated with particularly poor survival in this cohort, possibly related to ligand-dependent Wnt activation and altered drug transport and metabolism that could confer resistance to MMC and irinotecan. Drugs that target ligand-dependent Wnt activation or the BTN immune checkpoints could represent two novel therapy approaches.
... In CRC, the CMS4 subtype is associated with upregulation of mesenchymal gene expression (Guinney et al, 2015). In four independent cohorts, CRC PM samples classified almost exclusively as CMS4 (71-100%) (Hallam et al, 2020;Narasimhan et al, 2020;Laoukili et al, 2022;Lenos et al, 2022). In addition, the majority of matched primary tumors was also classified as CMS4, and a higher incidence of PM was found in a cohort of primary CMS4 tumors Milky spots in the omentum contain various immune cells and dense capillary networks. ...
... Created with BioRender.com. (Hallam et al, 2020;Laoukili et al, 2022;Lenos et al, 2022). This suggests that the subtype of the primary tumor defines the ability to metastasize to the peritoneum. ...
Article
Full-text available
Peritoneal metastases are a common form of tumor cell dissemination in gastrointestinal malignancies. Peritoneal metastatic disease (PMD) is associated with severe morbidity and resistance to currently employed therapies. Given the distinct route of dissemination compared with distant organ metastases, and the unique microenvironment of the peritoneal cavity, specific tumor cell characteristics are needed for the development of PMD. In this review, we provide an overview of the known histopathological, genomic, and transcriptomic features of PMD. We find that cancers representing the mesenchymal subtype are strongly associated with PMD in various malignancies. Furthermore, we discuss the peritoneal niche in which the metastatic cancer cells reside, including the critical role of the peritoneal immune system. Altogether, we show that PMD should be regarded as a distinct disease entity, that requires tailored treatment strategies.
... In addition, APC alterations can contribute to the loss of cell adhesion and errors in cell cycle control or in DNA repair (28). Gene expression analysis has also shown that PM are associated with activation of the WNT/β-catenin pathway that can be activated because of other genomic alterations beyond APC (29). Therefore, WNT/β-catenin pathway may be more frequently activated in mCRC with PM. ...
Article
Full-text available
Peritoneal metastases (PM) are common in metastatic colorectal cancer (mCRC). We aimed to characterize patients with mCRC and PM from a clinical and molecular perspective using the American Association of Cancer Research Genomics Evidence Neoplasia Information Exchange (GENIE) Biopharma Collaborative (BPC) registry. Patients’ tumor samples underwent targeted next-generation sequencing. Clinical characteristics and treatment outcomes were collected retrospectively. Overall survival (OS) from advanced disease and progression-free survival (PFS) from start of cancer-directed drug regimen were estimated and adjusted for the left truncation bias. A total of 1,281 patients were analyzed, 244 (19%) had PM at time of advanced disease. PM were associated with female sex [OR: 1.67; 95% confidence interval (CI): 1.11–2.54; P = 0.014] and higher histologic grade (OR: 1.72; 95% CI: 1.08–2.71; P = 0.022), while rectal primary tumors were less frequent in patients with PM (OR: 0.51; 95% CI: 0.29–0.88; P < 0.001). APC occurred less frequently in patients with PM (N = 151, 64% vs. N = 788, 79%) while MED12 alterations occurred more frequently in patients with PM (N = 20, 10% vs. N = 32, 4%); differences in MED12 were not significant when restricting to oncogenic and likely oncogenic variants according to OncoKB. Patients with PM had worse OS (HR: 1.45; 95% CI: 1.16–1.81) after adjustment for independently significant clinical and genomic predictors. PFS from initiation of first-line treatment did not differ by presence of PM. In conclusion, PM were more frequent in females and right-sided primary tumors. Differences in frequencies of MED12 and APC alterations were identified between patients with and without PM. PM were associated with shorter OS but not with PFS from first-line treatment. Significance Utilizing the GENIE BPC registry, this study found that PM in patients with colorectal cancer occur more frequently in females and right-sided primary tumors and are associated with worse OS. In addition, we found a lower frequency of APC alterations and a higher frequency in MED12 alterations in patients with PM.
... [167] Interestingly, another transcriptome analysis revealed unique characteristics of isolated metastatic peritoneal carcinoma in comparison to primary colorectal cancer. These features include a higher prevalence of mutations in negative regulators of the Wnt/β-catenin signaling pathway, TET2 mutations, mismatch repair gene mutations, and an increased tumor mutational burden [174]. In summary, the exploration of molecular mechanisms and gene alterations associated with colorectal cancer metastasizing to the liver, lungs, and peritoneum underscores their criticality in advancing targeted therapeutic strategies and propelling precision medicine. ...
Article
Full-text available
Distant metastasis remains a leading cause of mortality among patients with colorectal cancer (CRC). Organotropism, referring to the propensity of metastasis to target specific organs, is a well-documented phenomenon in CRC, with the liver, lungs, and peritoneum being preferred sites. Prior to establishing premetastatic niches within host organs, CRC cells secrete substances that promote metastatic organotropism. Given the pivotal role of organotropism in CRC metastasis, a comprehensive understanding of its molecular underpinnings is crucial for biomarker-based diagnosis, innovative treatment development, and ultimately, improved patient outcomes. In this review, we focus on metabolic reprogramming, tumor-derived exosomes, the immune system, and cancer cell-organ interactions to outline the molecular mechanisms of CRC organotropic metastasis. Furthermore, we consider the prospect of targeting metastatic organotropism for CRC therapy.
... Our results indicated that TMB was higher in non-responders than that in responders, which is consistent with a previous finding of colorectal PM treated with CRS/HIPEC (48). Conversely, the correlation between high TMB and better responses to HIPEC was reported in gastric PM patients (49). ...
Article
Full-text available
Introduction Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are considered for patients with peritoneal metastasis (PM). However, patients selection that relies on conventional prognostic factors is not yet optimal. In this study, we performed whole exome sequencing (WES) to establish tumor molecular characteristics and expect to identify prognosis profiles for PM management. Methods In this study, blood and tumor samples were collected from patients with PM before HIPEC. Tumor molecular signatures were determined using WES. Patient cohort was divided into responders and non-responders according to 12-month progression-free survival (PFS). Genomic characteristics between the two cohorts were compared to study potential targets. Results In total, 15 patients with PM were enrolled in this study. Driver genes and enriched pathways were identified from WES results. AGAP5 mutation was found in all responders. This mutation was significantly associated with better OS (p = 0.00652). Conclusions We identified prognostic markers that might be useful to facilitate decision-making before CRS/HIPEC.
Article
Full-text available
Metastasized colorectal cancer (CRC) is associated with a poor prognosis and rapid disease progression. Besides hepatic metastasis, peritoneal carcinomatosis is the major cause of death in UICC (Union for International Cancer Control) stage IV CRC patients. Insights into differential site-specific reconstitution of tumour cells and the corresponding tumour microenvironment are still missing. Here, we analysed the transcriptome of single cells derived from murine multivisceral CRC and delineated the inter-metastatic cellular heterogeneity regarding tumour epithelium, stroma and immune cells. Interestingly, we found an intercellular site-specific network of cancer associated fibroblasts and tumour epithelium during peritoneal metastasis as well as an autologous feed-forward loop in cancer stem cells. We furthermore deciphered a metastatic dysfunctional adaptive immunity by a loss of B cell dependent antigen presentation and consecutive effector T cell exhaustion. Furthermore, we demonstrated major similarities of this murine metastatic CRC model with human disease and -based on the results of our analysis- provided an auspicious site-specific immune modulatory treatment approach for stage IV CRC by intraperitoneal checkpoint inhibition.
Article
Colorectal cancer peritoneal metastases (CRC-PM) are present in 5 to 15% of instances of CRC, and the overall survival (OS) of patients with CRC-PM is much lower than that of patients with other isolated metastatic locations. In recent years, the introduction of cytoreductive surgery (CRS) in conjunction with hyperthermic intraperitoneal chemotherapy has resulted in a significant improvement in CRC-PM patients' OS. Despite this, a significant proportion of CRS patients continue to suffer complications of grades III to V or even die during the perioperative period. Early diagnosis, optimization of patient selection criteria, and refining of individualized combination therapy are necessary for these patients. In this review, we evaluate studies examining the relationship between molecular status and CRS in CRC-PM. Our objective is to gain a comprehensive understanding of how the altered molecular status of CRC-PM impacts CRS, which could increase the likelihood of tailored therapy in the future.
Article
Full-text available
Peritoneal metastases are associated with a low response rate to immune checkpoint blockade (ICB) therapy. The numbers of peritoneal resident macrophages (PRMs) are reversely correlated with the response rate to ICB therapy. We have previously shown that TLR9 in fibroblastic reticular cells (FRCs) plays a critical role in regulating peritoneal immune cell recruitment. However, the role of TLR9 in FRCs in regulating PRMs is unclear. Here, we demonstrated that the class A TLR9 agonist, ODN1585, markedly enhanced the efficacy of anti-PD-1 therapy in mouse models of colorectal peritoneal metastases. ODN1585 injected i.p. reduced the numbers of Tim4+ PRMs and enhanced CD8+ T cell antitumor immunity. Mechanistically, treatment of ODN1585 suppressed the expression of genes required for retinoid metabolism in FRCs, and this was associated with reduced expression of the PRM lineage-defining transcription factor GATA6. Selective deletion of TLR9 in FRCs diminished the benefit of ODN1585 in anti-PD-1 therapy in reducing peritoneal metastases. The crosstalk between PRMs and FRCs may be utilized to develop new strategies to improve the efficacy of ICB therapy for peritoneal metastases.
Article
Full-text available
Background: Microsatellite instability (MSI) is a biomarker for response to immune checkpoint inhibitors (ICPIs). PD-1 inhibitors in metastatic colorectal carcinoma (mCRC) with MSI-high (MSI-H) have demonstrated a high disease control rate and favorable progression-free survival (PFS); however, reported response rates to pembrolizumab and nivolumab are variable and often <50%, suggesting that additional predictive biomarkers are needed. Methods: Clinicopathologic data were collected from patients with MSI-H mCRC confirmed by hybrid capture-based next-generation sequencing (NGS) treated with PD-1/L1 inhibitors at five institutes. Tumor mutational burden (TMB) was determined on 0.8-1.1 Mb of sequenced DNA and reported as mutations/Mb. Potential biomarkers of response and time to progression were analyzed by univariate and multivariate analyses. Once TMB was confirmed as a predictive biomarker, a larger dataset of 18 140 unique CRC patients was analyzed to define the relevance of the identified TMB cut-point. Results: A total of 22 patients were treated with PD-1/L1 inhibitors including 19 with pembrolizumab monotherapy. Among tested variables, TMB showed the strongest association with objective response (OR; P < 0.001) and PFS, by univariate (P < 0.001) and multivariate analysis (P < 0.01). Using log-rank statistics, the optimal predictive cut-point for TMB was estimated between 37 and 41 mutations/Mb. All 13 TMBhigh cases responded, while 6/9 TMBlow cases had progressive disease. The median PFS for TMBhigh has not been reached (median follow-up >18 months) while the median PFS for TMBlow was 2 months. A TMB of 37.4 mutations/Mb in a large MSI-H mCRC population (821/18, 140 cases; 4.5%) evaluated by NGS corresponded to the 35th percentile cut-point. Conclusions: TMB appears to be an important independent biomarker within MSI-H mCRC to stratify patients for likelihood of response to ICPIs. If validated in prospective studies, TMB may play an important role in guiding the sequencing and/or combinations of ICPIs in MSI-H mCRC.
Article
Full-text available
Background: Up to 15 per cent of colorectal cancers present with peritoneal metastases (CPM). Cytoreductive surgery and heated intraperitoneal chemotherapy (CRS + HIPEC) aims to achieve macroscopic tumour resection combined with HIPEC to destroy microscopic disease. CRS + HIPEC is a major operation with significant morbidity and effects on quality of life (QoL). Improving patient selection is crucial to maximize patient outcomes while minimizing morbidity and mortality. The aim of this study was to identify prognostic factors for patients with CPM undergoing CRS + HIPEC. Methods: A systematic search of MEDLINE, Embase and Cochrane Library electronic databases was performed using terms for colorectal cancer, peritoneal metastasis and CRS + HIPEC. Included studies focused on the impact of prognostic factors on overall survival following CRS + HIPEC in patients with CPM. Results: Twenty-four studies described 3128 patients. Obstruction or perforation of the primary tumour (hazard ratio (HR) 2·91, 95 per cent c.i. 1·5 to 5·65), extent of peritoneal metastasis as described by the Peritoneal Carcinomatosis Index (PCI) (per increase of 1 PCI point: HR 1·07, 1·02 to 1·12) and the completeness of cytoreduction (CC score above zero: HR 1·75, 1·18 to 2·59) were associated with reduced overall survival after CRS + HIPEC. Conclusion: Primary tumour obstruction or perforation, PCI score and CC score are valuable prognostic factors in the selection of patients with CPM for CRS + HIPEC.
Article
Full-text available
Colorectal peritoneal metastases (CPM) are associated with abbreviated survival and significantly impaired quality of life. In patients with CPM, radical multimodality treatment consisting of cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) has demonstrated oncological superiority over systemic chemotherapy alone. In highly selected patients undergoing CRS + HIPEC, overall survival of over 60% has been reported in some series. These are patients in whom the disease burden is limited and where the diagnosis is made at an early stage in the disease course. Early diagnosis and a deeper understanding of the biological mechanisms that regulate CPM are critical to refining patient selection for radical treatment, personalising therapeutic approaches, enhancing prognostication, and ultimately improving long-term survivorship. In the present study, we outline three broad themes which represent critical future research targets in CPM: (1) enhanced radiological strategies for early detection and staging; (2) identification and validation of translational biomarkers for diagnostic, prognostic, and therapeutic deployment; and (3) development of optimized approaches for surgical cytoreduction as well as more precise strategies for intraperitoneal drug selection and delivery. Herein, we provide a contemporary narrative review of the state of the art in these three areas. A systematic review in accordance with PRISMA guidelines was undertaken on all English language studies published between 2007 and 2017. In vitro and animal model studies were deemed eligible for inclusion in the sections pertaining to biomarkers and therapeutic optimisation, as these areas of research currently remain in the early stages of development. Acquired data were then divided into hierarchical thematic categories (imaging modalities, translational biomarkers (diagnostic/prognostic/therapeutic), and delivery techniques) and subcategories. An interactive sunburst figure is provided for intuitive interrogation of the CPM research landscape.
Article
Full-text available
Clinical evidence demonstrates that treatment with immune checkpoint inhibitor immunotherapy agents can have considerable benefit across multiple tumours. However, there is a need for the development of predictive biomarkers that identify patients who are most likely to respond to immunotherapy. Comprehensive characterisation of tumours using genomic, transcriptomic, and proteomic approaches continues to lead the way in advancing precision medicine. Genetic correlates of response to therapy have been known for some time, but recent clinical evidence has strengthened the significance of high tumour mutational burden (TMB) as a biomarker of response and hence a rational target for immunotherapy. Concordantly, immune checkpoint inhibitors have changed clinical practice for lung cancer and melanoma, which are tumour types with some of the highest mutational burdens. TMB is an implementable approach for molecular biology and/or pathology laboratories that provides a quantitative measure of the total number of mutations in tumour tissue of patients and can be assessed by whole genome, whole exome, or large targeted gene panel sequencing of biopsied material. Currently, TMB assessment is not standardised across research and clinical studies. As a biomarker that affects treatment decisions, it is essential to unify TMB assessment approaches to allow for reliable, comparable results across studies. When implementing TMB measurement assays, it is important to consider factors that may impact the method workflow, the results of the assay, and the interpretation of the data. Such factors include biopsy sample type, sample quality and quantity, genome coverage, sequencing platform, bioinformatic pipeline, and the definitions of the final threshold that determines high TMB. This review outlines the factors for adoption of TMB measurement into clinical practice, providing an understanding of TMB assay considerations throughout the sample journey, and suggests principles to effectively implement TMB assays in a clinical setting to aid and optimise treatment decisions.
Article
Full-text available
Background: Approximately 90% of colorectal cancer (CRC) deaths are caused by tumors ability to migrate into the adjacent tissues and metastase into distant organs. More than 40 genes have been causally linked to the development of CRC but no mutations have been associated with metastasis yet. To identify molecular basis of CRC metastasis we performed whole-exome and genome-scale transcriptome sequencing of 7 liver metastases along with their matched primary tumours and normal tissue. Multiple, spatially separated fragments of primary tumours were analyzed in each case. Uniformly malignant tissue specimen were selected with macrodissection, for three samples followed with laser microdissection. Results: > 100 sequencing coverage allowed for detection of genetic alterations in subpopulation of tumour cells. Mutations in KRAS, APC, POLE, and PTPRT, previously associated with CRC development, were detected in most patients. Several new associations were identified, including PLXND1, CELSR3, BAHD1 and PNPLA6. Conclusions: We confirm the essential role of inflammation in CRC progression but question the mechanism of matrix metalloproteinases activation described in other work. Comprehensive sequencing data made it possible to associate genome-scale mutation distribution with gene expression patterns. To our knowledge, this is the first work to report such link in CRC metastasis context.
Article
Full-text available
Purpose: Dysregulation of the Wnt pathway is a crucial step in the tumorigenesis of colorectal cancer (CRC). This study aimed to determine whether DNA methylation of Wnt pathway genes helps predict treatment response and survival in patients with metastatic or recurrent CRC. Materials and methods: We retrospectively collected primary tumor tissues from 194 patients with metastatic or recurrent CRC. Pyrosequencing was used to examine the methylation of 10 CpG island loci in DNA extracted from formalin-fixed paraffin-embedded specimens. To elucidate the predictive role of DNA methylation markers, Kaplan-Meier survival estimation and Cox regression were performed for progression-free survival and overall survival (OS). Results: The methylation frequencies of the 10 genes analyzed (p16, p14, MINT1, MINT2, MINT31, hMLH1, DKK3, WNT5A, AXIN2, and TFAP2E) were 47.9%, 10.8%, 21.1%, 16.0%, 20.6%, 0.5%, 53.1%, 32.0%, 2.6%, and 2.1%, respectively. We divided patients into three groups based on the number of methylated genes (group 1, no methylation n=38; group 2, 1-2 methylations n=92; group 3, 3 or more methylations n=64). Among patients treated with palliative chemotherapy (n=167), median OSs of groups 1, 2, and 3 were 39.1, 39.7, and 29.1 months, respectively (log rank p=0.013). After adjustment, number of methylations was identified as an independent poor prognostic factor (0-2 methylated vs. ≥3 methylated: hazard ratio, 1.72; 95% confidence interval, 1.16-2.56, p=0.007). Conclusion: This study suggests that methylation of Wnt pathway genes, in addition to known CpG island methylator phenotype markers, may help predict treatment outcome and survival in patients with CRC.
Article
Full-text available
The evolutionary features of clear-cell renal cell carcinoma (ccRCC) have not been systematically studied to date. We analyzed 1,206 primary tumor regions from 101 patients recruited into the multi-center prospective study, TRACERx Renal. We observe up to 30 driver events per tumor and show that subclonal diversification is associated with known prognostic parameters. By resolving the patterns of driver event ordering, co-occurrence, and mutual exclusivity at clone level, we show the deterministic nature of clonal evolution. ccRCC can be grouped into seven evolutionary subtypes, ranging from tumors characterized by early fixation of multiple mutational and copy number drivers and rapid metastases to highly branched tumors with >10 subclonal drivers and extensive parallel evolution associated with attenuated progression. We identify genetic diversity and chromosomal complexity as determinants of patient outcome. Our insights reconcile the variable clinical behavior of ccRCC and suggest evolutionary potential as a biomarker for both intervention and surveillance.
Article
Background: Adequate selection of patients with peritoneal metastasis (PM) for cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) remains critical for successful long-term outcomes. Factors reflecting tumor biology are currently poorly represented in the selection process. The prognostic relevance of RAS/RAF mutations in patients with PM remains unclear. Methods: Survival data of patients with colorectal PM operated in 6 European tertiary centers were retrospectively collected and predictive factors for survival identified by Cox regression analyses. A simple point-based risk score was developed to allow patient selection and outcome prediction. Results: Data of 524 patients with a median age of 59 years and a median peritoneal cancer index of 7 (interquartile range: 3-12) were collected. A complete resection was possible in 505 patients; overall morbidity and 90-day mortality were 50.9% and 2.1%, respectively. PCI [hazard ratio (HR): 1.08], N1 stage (HR: 2.15), N2 stage (HR: 2.57), G3 stage (HR: 1.80) as well as KRAS (HR: 1.46) and BRAF (HR: 3.97) mutations were found to significantly impair survival after CRS/HIPEC on multivariate analyses. Mutations of RAS/RAF impaired survival independently of targeted treatment against EGFR. Consequently, a simple point-based risk score termed BIOSCOPE (BIOlogical Score of COlorectal PEritoneal metastasis) based on PCI, N-, G-, and RAS/RAF status was developed, which showed good discrimination [development area under the curve (AUC) = 0.72, validation AUC = 0.70], calibration (P = 0.401) and allowed categorization of patients into 4 groups with strongly divergent survival outcomes. Conclusion: RAS/RAF mutations impair survival after CRS/HIPEC. The novel BIOSCOPE score reflects tumor biology, adequately stratifies long-term outcomes, and improves patient assessment and selection.
Article
LBA3503 Background: Promising results have been obtained during the last decade using cytoreductive surgery (CRS) plus HIPEC for selected patients with colorectal PC who are amenable to complete macroscopic resection. This is the first trial to evaluate the specific role of HIPEC, after CRS, for the treatment of PC of colorectal origin. Methods: Prodige 7 is a randomized phase III, multicenter trial. Patients with histologically proven and isolated PC, peritoneal cancer index (PCI) ≤25 were eligible. Randomization (1:1) was stratified by center, complete macroscopic resection (R0/1 vs R2), and neoadjuvant systemic chemotherapy. Patients were treated with CRS plus HIPEC with oxaliplatin or CRS alone, in association with systemic chemotherapy. The primary endpoint was the overall survival (OS). Secondary endpoints were relapse-free survival (RFS) and toxicity. 264 patients were required to show a gain in median OS from 30 to 48 months (HR = 0.625) with a two-sided α = 0,046 and 80% power. Results: 265 patients from 17 centers were included between February 2008 and January 2014: 132 in Arm without HIPEC and 133 in Arm with HIPEC. The median age was 60 years (range: 30-74). Baseline characteristics were well balanced. The overall post-operative mortality rate was 1.5% and was not different between the two arms. The morbidity rates did not differ statistically at 30 days. At 60 days, the grade 3-5 morbidity rate was significantly higher with HIPEC (24.1% vs. 13.6%, p= 0.030). After a median follow up of 63.8 months (95% CI: 58.9-69.8), the median OS was 41.2 months (95% CI 35.1-49.7) in the non-HIPEC Arm and 41.7 months (95% CI: 36.2-52.8) in the HIPEC Arm, HR = 1.00 (95% CI: 0.73-1.37) p = 0.995. The median RFS was 11.1 months (95% CI: 9-12.7) in non-HIPEC Arm and 13.1 months (95% CI: 12.1-15.7) in HIPEC Arm, HR = 0.90 (95% CI: 0.69-1.90) (p = 0.486), whilst the 1-year RFS rates were 46.1% in non-HIPEC Arm and 59 % in the HIPEC Arm. Conclusions: The therapeutic curative management of PC from colorectal cancer by CRS shows satisfactory survival results. While the addition of HIPEC with oxaliplatin does not influence the OS. Clinical trial information: NCT00769405.
Article
Background: It is well known that peritoneal carcinomatosis (PC) from colorectal cancer (CRC) is associated with a poor prognosis. However, data on the prognostic significance of modern chemotherapy containing bevacizumab, cetuximab or panitumumab are not available. Materials and Methods: This retrospective review concerned 526 patients with metastatic CRC who were classified into two groups according to the presence or absence of PC, and were treated with systemic chemotherapy, with or without bevacizumab or anti-EGFR antibodies. The genetic background, in particular KRAS, BRAF, and PIK3CA gene mutations, and overall survival (OS) were compared between the two groups. Results: The median OS values were 23.3 and 29.1 months for PC and non-PC patients, respectively (hazard ratio [HR]=1.20; p=0.17). Among all patients, tumor location, number of metastatic sites and BRAF mutation status were significant prognostic factors, whereas the presence of PC was not. In the PC group, chemotherapy with bevacizumab resulted in a significantly longer OS than forchemotherapy without bevacizumab (HR=0.38, p < 0.01), but this was not the case in the non-PC group (HR=0.80, p=0.10). Furthermore, the incidence of the BRAF V600E mutation was significantly higher in PC than in non-PC patients (27.7% versus 7.3%, p < 0.01). BRAF mutations displayed a strong correlation with shorter OS in non-PC (HR=2.26), but not PC patients (HR=1.04). Conclusions: Systemic chemotherapy, especially when combined with bevacizumab, improved survival in patients with PC from CRC as well as non-PC patients. While BRAF mutation demonstrated a high frequency in PC patients, but it was not associated with prognosis.