Molecular portraits of colorectal cancer morphological regions

  1. Eva Budinská
  2. Martina Hrivňáková
  3. Tina Catela Ivkovic
  4. Marie Madrzyk
  5. Rudolf Nenutil
  6. Beatrix Bencsiková
  7. Dagmar Al Tukmachi
  8. Michaela Ručková
  9. Lenka Zdražilová Dubská
  10. Ondřej Slabý
  11. Josef Feit
  12. Mihnea-Paul Dragomir
  13. Petra Borilova Linhartova
  14. Sabine Tejpar
  15. Vlad Popovici  Is a corresponding author
  1. RECETOX, Faculty of Science, Masarykova Univerzita, Czech Republic
  2. Central European Institute of Technology, Masarykova Univerzita, Czech Republic
  3. Masaryk Memorial Cancer Institute, Czech Republic
  4. Faculty of Medicine, Masarykova Univerzita, Czech Republic
  5. Central European Institute of Technology, Department of Biology, Faculty of Medicine, Masarykova Univerzita, Czech Republic
  6. Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masarykova Univerzita, Czech Republic
  7. Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany
  8. Berlin Institute of Health, Germany
  9. German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Germany
  10. Faculty of Medicine, Digestive Oncology Unit, Katholieke Universiteit Leuven, Belgium
6 figures, 1 table and 14 additional files

Figures

Morphological patterns and their distribution in the dataset.

(A) The six CRC morphological patterns of interest (morphotypes). Left: example of an original annotation used for macrodissection and RNA extraction. Note that the original annotations in the image are not identical to the ones used in the main text. Here, A-SE stands for serrated (SE) in the text, B-DE for desmoplastic (DE) in the text, C-MUC for mucinous (MU) in the text, and D-ST for solid/trabecular (TB) in the text, respectively. Also, N indicates a tumor-adjacent normal epithelial region and S a supportive stroma region, respectively. Right: examples of morphotypes – complex tubular (CT), desmoplastic (DE), mucinous (MU), papillary (PP), serrated (SE), and solid/trabecular (TB). (B) Morphotype distribution per case (unique tumor) and intersections thereof: some cases had several morphotypes profiled.

Figure 2 with 3 supplements
CRC morphotypes: in silico decomposition of the cellular admixture.

(A) Boxplots of the tumor purity (epithelial content – ESTIMATE method) in each tumor morphotype and the two non-tumor regions, ordered by increasing median values. (B) Signatures specific to colon crypt compartments and major cell types estimated from gene expression data in terms of normalized enrichment scores (NES): only statistically significant scores are shown. (C) Immune cell fractions (and unassigned fractions) inferred from gene expression data using quanTIseq method. (D) Types of cancer-associated fibroblasts (CAFs) as estimated from gene expression using the signatures from Khaliq et al., 2022; Kieffer et al., 2020.

Figure 2—figure supplement 1
Epithelial signatures from Pelka et al., 2021.

Only statistically significant scores (NES) are shown.

Figure 2—figure supplement 2
Immune signatures from Pelka et al., 2021.

Only statistically significant scores (NES) are shown.

Figure 2—figure supplement 3
Stromal signatures from Pelka et al., 2021.

Only statistically significant scores (NES) are shown.

Figure 3 with 2 supplements
Top differentially expressed genes and hallmark pathways.

(A) GSEA scores for hallmark pathways in the six morphotypes and two non-tumoral regions. Only pathways with statistically significant scores are shown. (B) Principal component analysis of hallmark pathways: the median profiles of the six morphotypes (CT: complex tubular, DE: desmoplastic, MU: mucinous, PP: papillary, SE: serrated, and TB: solid/trabecular) and the two non-tumoral regions (NR: tumor-adjacent normal and ST: supportive stroma) are projected onto the space defined by first two principal components (74% of the total variance). The top pathways contributing to the principal axes are shown as well. See also Figure 3—figure supplement 1. (C) Heatmap of top 5 up- and down-regulated genes for each of the six morphotypes.

Figure 3—figure supplement 1
Principal component analysis of hallmark pathways GSEA scores: loadings for the first two principal components, i.e., contribution of pathways to the first two axes.
Figure 3—figure supplement 2
Hallmark pathways differential activation between pairs of morphotypes.

Here we compare the results from GSEA applied to differentially expressed genes between pairs of morphotypes originating from all cases to results of GSEA applied to differentially expressed genes between pairs of morphotypes originating from the same section (tumor; i.e., matched pairs of morphotypes). All results are shown, including the statistically not significant ones. First, four columns correspond to pairs of morphotypes from all cases, while the last four, to matched pairs of morphotypes.

Figure 4 with 1 supplement
Intra-tumoral heterogeneity and the morphotypes (for all core samples, including those unassigned by the classifiers).

Only cases with at least two distinct morphotypes present are shown. (A) Left: CMS assignment for tumors represented by multiple regions. Right: CMS assignment per morphotype (and two non-tumoral patterns). (B) Left: iCMS assignment for tumors represented by multiple regions. Right: iCMS assignment per morphotype (and two non-tumoral patterns). (C) Differences between paired signatures: morphotypes vs whole tumor (each signature was normalized to [0,1] prior to computing the differences). Only four (morphotype, whole tumor) pairs were represented enough in the data. (D) Boxplots for the ten (normalized) signatures across morphotypes. The ‘Eschrich’ and ‘Jorissen’ signatures vary significantly (Kruskal-Wallis’s test) across morphotypes. For equivalent plots for all samples, including non-core, see Figure 4—figure supplement 1.

Figure 4—figure supplement 1
Molecular subtypes and morphotypes in all samples, including non-core samples.

Note that the sets of samples in A and C are the same as in Figure 4, as only core samples also had at least two distinct morphological regions.

Figure 5 with 2 supplements
Intra-tumoral heterogeneity case study.

For the same case, different CMS labels are assigned to regions and whole tumor profile. The hallmark pathways show various levels of activation (as computed by GSVA) within same section. The relative change in prognostic scores indicate potential underestimation of risk for some signatures, while others appear to be stable across tumor. See also Figure 5—figure supplements 1 and 2. Note that in the pathology section image, the original annotations were preserved, and they are not identical to the ones used in the main text. Here, MUC stands for mucinous (MU) in the text. Also, N indicates a tumor-adjacent normal epithelial region and S a supportive stroma region, respectively.

Figure 5—figure supplement 1
Intra-tumoral heterogeneity additional case study.
Figure 5—figure supplement 2
Intra-tumoral heterogeneity additional case study.
Figure 6 with 1 supplement
Normalized enrichment scores from GSEA for selected resistance signatures (from C2 section of MSigDB).

Only significant scores are shown.

Figure 6—figure supplement 1
Resistance scores (GSVA) per patient and morphotype for cases where the whole–tumor prediction is contradicted by some regional score.

Tables

Table 1
Results of comparison of each morphotype (and the two non-tumoral regions) with the average profile.

The table shows the top 20 up- and down- regulated genes and significantly activated hallmark pathways and processes (as result of GSEA). The genes not significant after p-value adjustment (at FDR = 0.15) have their symbols greyed. See also Supplementary files 5–6.

MorphTop 20 up-regulated genes (compared to mean)Top 20 down-regulated genes (compared to mean)Hallmark pathways with high scoreActive processes (based on the active hallmark pathways)
MUARF4, MUC2, SULF1, FNDC1, LOXL1, LGALS1, ANTXR1, BGN, COL12A1, PALLD, MEG8, DKK3, ACVR1, GPX8, CALD1, FBN1, MLLT11, CSRP2, TUSC3, GREM1TIMD4, PRELID3BP3, EREG, KDM4A, CCDC175, TDP2, CHMP1B2P, ACE2, NLRP7, UGT2A3, SLC26A3, A1CF, TSPAN6, CLDN10, TMIGD1, BMP5, MS4A12, FAM3B, CLCA4, MEP1AEMT, TNF a signaling via NFKB, Complement, IL2 STAT5 signaling, hypoxia, inflammatory response, KRAS signaling, UV response, myogenesis, coagulation, apical junction, allograft rejection, IL6 JAK STAT3 signaling, interferon gamma response, apoptosis, TGF-beta signaling, angiogenesis, hedgehog signaling, estrogen response early, NOTCH signaling, WNT beta catenin signaling, cholesterol homeostasisInflammation, neoangiogenesis, increased metastatic potential, apoptosis, development
DEOLFML2B, INHBA, LUM, SULF1, PTPN14, PRDM6, SPOCK1, RDX, EDNRA, COL12A1, CTHRC1, PRRX1, LGALS1, COPZ2, COL10A1, TNFAIP6, IGFL1P1, ST6GAL2, FAP, BGNSLC17A4, ANPEP, DEFA5, RAP1GAP, MRAP2, ADH1C, TRIQK, REG1A, SLC4A4, UGT2B15, REG4, SEMA6A, L1TD1, MS4A12, SI, SPINK4, CLCA4, MUC2, CLCA1, CA1EMT, TNF a signaling via NFKB, Complement, IL2 STAT5 signaling, hypoxia, inflammatory response, KRAS signaling, UV response, myogenesis, coagulation, apical junction, apoptosis, TGF-beta signaling, angiogenesis, hedgehog signaling, estrogen response earlyInflammation, neoangiogenesis, increased metastatic potential, apoptosis
PPPTPRD, KNDC1, MIMT1, UPK3B, MPZ, MMP15, CYP4F12, SNORD4A, SNAR-C3, TMTC4, LRCOL1, GATA5, SNAR-E, EPHA7, IPO4, SNAR-I, CASC21, NUTF2, SNAR-B2, RPL31P50IGKV3-11, IGHV4-39, ANPEP, OR4F8P, HEPACAM2, ADAM28, CPS1, TMIGD1, NPY6R, ITLN1, SI, ADH1C, CAV1, MMP2, FDCSP, CLU, REG1A, RSPO3, PAX8-AS1, PALMDMYC targets V1, MYC targets V2, E2F targets, KRAS signaling DOWN, WNT beta catenin signaling,
SEPPAN-P2RY11, TUBB4BP7, JADE3, PFDN6, CLDN2, YAF2, BOLL, SLAMF9, SLC12A2, CCDC175, GRIN2B, TUBB3P2, GAPDHP71, RPS2P25, MAT1A, NOX1, SNORD12C, SMAD6, MECOM, EXTL2IGKV2D-29, MYLK, TAGLN, CNTNAP3P2, GLI3, CPXM2, NR3C1, CNN1, PECAM1, COLEC12, IGKV4-1, IGKV2D-30, DPYD, CLU, TSHZ2, ADH1B, IL10RA, PDE7B, ABCA8, CDC42SE2MYC targets V1, MYC targets V2, E2F targets, G2M checkpoint,
CTTMEM97, RPL13, CLDN1, TFDP1, CKS2, CDCA7, TPX2, ANLN, RAD54B, KRT18, HSPH1, CCT6A, PLK1, TMEM97P2, CSE1L, MIPEP, SNORA71D, SNORA71C, PTTG1, PLBD1CR2, OGN, SNORD114-21, SLC30A10, CLCA4, SNORD114-12, DCLK1, FAT4, CPA3, ADH1B, SLC26A2, SNORD114-20, SFRP1, ZG16, FGF7, SNORD113-1, ABCA8, B4GALNT2, MS4A12, CA1MYC targets V1, MYC targets V2, E2F targets, G2M checkpoint, MTORC1 signaling, unfolded protein response, Glycolysis, oxidative phosphorylation, fatty acid metabolism, protein secretionProliferation, Catabolism, oxidative stress, cell cycle disruption
TBCKAP2, HSP90AA1, PPP3CA, REEP4, MSH6, TOP2A, HSPE1, PPP2R5C, TBCA, VRK2, NIFK, TXNL4A, MNAT1, ERI1, XPO1, VTRNA1-2, ANP32A, ARF6, RNF2, EIF4A1P7FLJ22763, TMEM236, NPY6R, IGKV3D-20, IGKV2D-30, OLFM4, SELENBP1, LRRC19, CDHR1, IGHA1, SNORD123, SLC26A3, CXCL14, SLC3A1, SEMA5A, MS4A12, IGHA2, CLCA4, NXPE4, NXPE1MYC targets V1, MYC targets V2, E2F targets, G2M checkpoint, MTORC1 signaling, unfolded protein response, Glycolysis, oxidative phosphorylation, fatty acid metabolism, protein secretion, cholesterol homeostasis,Inflammation, catabolism, apoptosis, oxidative stress, proliferation, cell cycle disruption
NRPIGR, SLC26A3, ADH1B, NXPE1, IGHA2, CLCA1, JCHAIN, IGHA1, FCGBP, IGK, NXPE4, SLC9A2, MUC2, NR3C2, TMEM236, MS4A12, FABP1, IGLC3, IGKV1D-39, LRRC19TACSTD2, FAM83D, ASPN, CXCL11, CTHRC1, SLC39A6, IFNE, SULF1, HSPH1, ELFN1-AS1, THBS2, CLDN1, SIM2, SLC22A3, SPARC, FN1, AHNAK2, COL11A1, SPP1, INHBAHeme metabolism, bile acid metabolism, xenobiotic metabolism, fatty acid metabolism
STSFRP2, ADH1B, EMCN, STEAP4, ADAMTS1, ABI3BP, SPARCL1, DCN, PTGDS, PALMD, NOVA1, SLIT3, OGN, SERPINF1, RSPO3, CPA3, FBLN5, C3, EFEMP1, PBX3FRK, AADACP1, CKS2, HOOK1, CLDN1, ANLN, S100P, UGT8, MACC1, EXPH5, CYP3A5, OCIAD2, SLC12A2, GK, EVADR, TMC5, REG4, TFF1, TCN1, CXCL8EMT, TNF a signaling via NFKB, Complement, IL2 STAT5 signaling, hypoxia, inflammatory response, KRAS signaling, UV response, myogenesis, coagulation, apical junction, allograft rejection, IL6 JAK STAT3 signaling, interferon gamma responseInflammation, neoangiogenesis, increased metastatic potential

Additional files

Supplementary file 1

Main clinical parameters of the study cohort.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp1-v1.xlsx
Supplementary file 2

Distribution of main clinical parameters per morphotype (and tumor-adjacent normal and supportive stroma).

https://cdn.elifesciences.org/articles/86655/elife-86655-supp2-v1.xlsx
Supplementary file 3

Table of gene expression signatures.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp3-v1.xlsx
Supplementary file 4

Table of GSEA scores (NES) for “other” signatures (see also Supplementary file 3 for signatures).

https://cdn.elifesciences.org/articles/86655/elife-86655-supp4-v1.xlsx
Supplementary file 5

List of differentially expressed genes (limma tables) per morphotype in contrast with pooled profile.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp5-v1.xlsx
Supplementary file 6

GSEA results for genes in Supplementary file 5, for whole MSigDB collection.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp6-v1.xlsx
Supplementary file 7

List of differentially expressed genes (limma tables) per morphotype in contrast with all other five morphotypes.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp7-v1.xlsx
Supplementary file 8

GSEA results for genes in Supplementary file 7, for whole MSigDB collection.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp8-v1.xlsx
Supplementary file 9

List of differentially expressed genes (limma tables) per matched pairs of morphotypes.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp9-v1.xlsx
Supplementary file 10

GSEA results for genes in Supplementary file 9, for whole MSigDB collection.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp10-v1.xlsx
Supplementary file 11

List of differentially expressed genes (limma tables) for pairs of morphotypes.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp11-v1.xlsx
Supplementary file 12

GSEA results for genes in Supplementary file 11, for whole MSigDB collection.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp12-v1.xlsx
Supplementary file 13

List of prognostic signatures tested.

https://cdn.elifesciences.org/articles/86655/elife-86655-supp13-v1.xlsx
MDAR checklist
https://cdn.elifesciences.org/articles/86655/elife-86655-mdarchecklist1-v1.docx

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Eva Budinská
  2. Martina Hrivňáková
  3. Tina Catela Ivkovic
  4. Marie Madrzyk
  5. Rudolf Nenutil
  6. Beatrix Bencsiková
  7. Dagmar Al Tukmachi
  8. Michaela Ručková
  9. Lenka Zdražilová Dubská
  10. Ondřej Slabý
  11. Josef Feit
  12. Mihnea-Paul Dragomir
  13. Petra Borilova Linhartova
  14. Sabine Tejpar
  15. Vlad Popovici
(2023)
Molecular portraits of colorectal cancer morphological regions
eLife 12:RP86655.
https://doi.org/10.7554/eLife.86655.3