Consensus molecular subtype 4 (CMS4)-targeted therapy in primary colon cancer: A proof-of-concept study

Niek A Peters, Alexander Constantinides, Inge Ubink, Joyce van Kuik, Haiko J Bloemendal, Joyce M van Dodewaard, Menno A Brink, Thijs P Schwartz, Martijn P J K Lolkema, Miangela M Lacle, Leon M Moons, Joost Geesing, Wilhelmina M U van Grevenstein, Jeanine M L Roodhart, Miriam Koopman, Sjoerd G Elias, Inne H M Borel Rinkes, Onno Kranenburg, Niek A Peters, Alexander Constantinides, Inge Ubink, Joyce van Kuik, Haiko J Bloemendal, Joyce M van Dodewaard, Menno A Brink, Thijs P Schwartz, Martijn P J K Lolkema, Miangela M Lacle, Leon M Moons, Joost Geesing, Wilhelmina M U van Grevenstein, Jeanine M L Roodhart, Miriam Koopman, Sjoerd G Elias, Inne H M Borel Rinkes, Onno Kranenburg

Abstract

Background: Mesenchymal Consensus Molecular Subtype 4 (CMS4) colon cancer is associated with poor prognosis and therapy resistance. In this proof-of-concept study, we assessed whether a rationally chosen drug could mitigate the distinguishing molecular features of primary CMS4 colon cancer.

Methods: In the ImPACCT trial, informed consent was obtained for molecular subtyping at initial diagnosis of colon cancer using a validated RT-qPCR CMS4-test on three biopsies per tumor (Phase-1, n=69 patients), and for neoadjuvant CMS4-targeting therapy with imatinib (Phase-2, n=5). Pre- and post-treatment tumor biopsies were analyzed by RNA-sequencing and immunohistochemistry. Imatinib-induced gene expression changes were associated with molecular subtypes and survival in an independent cohort of 3232 primary colon cancer.

Results: The CMS4-test classified 52/172 biopsies as CMS4 (30%). Five patients consented to imatinib treatment prior to surgery, yielding 15 pre- and 15 post-treatment samples for molecular analysis. Imatinib treatment caused significant suppression of mesenchymal genes and upregulation of genes encoding epithelial junctions. The gene expression changes induced by imatinib were associated with improved survival and a shift from CMS4 to CMS2.

Conclusion: Imatinib may have value as a CMS-switching drug in primary colon cancer and induces a gene expression program that is associated with improved survival.

Keywords: ImPACCT; colorectal cancer; consensus molecular subtype 4; imatinib; platelet-derived growth factor receptor (PDGFR).

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Peters, Constantinides, Ubink, van Kuik, Bloemendal, van Dodewaard, Brink, Schwartz, Lolkema, Lacle, Moons, Geesing, van Grevenstein, Roodhart, Koopman, Elias, Borel Rinkes and Kranenburg.

Figures

Figure 1
Figure 1
ImPACCT study flowchart. Individuals scheduled for a colonoscopy procedure were approached to obtain informed consent for acquisition of 5 additional biopsies for CMS4 testing in case suspect lesions were found, and for approval to approach them again in case the tumor was diagnosed as CMS4. Patients with CMS4 CRC were approached to obtain informed consent for the second part of the study (imatinib treatment), and were screened for eligibility. Five patients received imatinib treatment for 14 days prior to surgery. Pre-treatment diagnostic biopsies and post-treatment biopsies from the resected primary tumors were used for CMS classification and additional molecular analyses.CMS, consensus molecular subtype; RT-qPCR, real-time quantitative polymerase chain reaction; IC, informed consent; CRC, colorectal cancer.
Figure 2
Figure 2
. CMS4 assessment on diagnostic biopsies and patient selection. Diagnostic biopsies (3 per tumor) were processed for RNA isolation and subsequent CMS4 testing, using a previously designed and validated RT-qPCR test (9). The heatmap shows CMS4 probabilities per-biopsy (top) and the weighed mean probabilities per tumor, to account for intra-tumor CMS4 heterogeneity. If the weighed mean probability was higher than 50%, tumors were classified as CMS4 (n=24) and the patients were approached for the second part of the study. The cohort contained 3 histologically confirmed adenomas (right sub-panel).
Figure 3
Figure 3
Imatinib treatment of primary CMS4 CRC results in a mesenchymal-to-epithelial phenotype shift. (A) Bar graph summarizing CMS classification of tumor tissue samples PRE and POST imatinib treatment, measured by the RT-qPCR test and the CMS random forest (RF) classifier applied to RNA sequencing data. (B) XY-plot showing the correlation between CMS4 probabilities of pre-treatment diagnostic biopsies as measured by the RT-qPCR test and the RF classifier. ρm denotes the marginal Pearson correlation coefficient for clustered data (34) with two-sided p-value. (C) Dot-plots showing expression (mean z-scores) of a signature comprised of the 4 genes in the CMS4 test (PDGFRA, PDGFRB, PDGFC, KIT) and the CMS4 probabilities generated by the RF classifier, in tissue samples PRE and POST imatinib treatment. P values were generated using ANOVA and a linear mixed model. (D) Dot plots showing 2log expression levels of PDGFRA, PDGFRB, ZEB1, and CD36 in tissue samples PRE and POST imatinib treatment. P values were generated using a two-sided Student’s t-test. (E) Dot plots Graphs showing 2log expression values of epithelial junction genes (CDH1, JUP, and CTNNA) and expression of signatures for Adherens Junctions, Desmosomes, and genes upregulated in epithelial cell clusters versus single cells in tissue samples PRE and POST imatinib treatment. P values were generated using ANOVA and a linear mixed on pre- vs post-treatment biopsies. (F) XY-plot showing the (negative) correlation between CDH1 expression and CMS4 probabilities (RF) in tissue samples PRE and POST imatinib treatment. ρm denotes the marginal Pearson correlation coefficient for clustered data with two-sided p-value. (G) Dot plot showing ZEB1 expression in tissue samples PRE and POST imatinib treatment in individual patients with color-coded CMS classification. The black lines indicate the change in mean ZEB1 expression following imatinib treatment.
Figure 4
Figure 4
Imatinib treatment of primary CMS4 CRC causes increased expression of proliferation-associated genes. (A) Tukey box and violin plots showing expression of the proliferation marker MKI67 and signatures reflecting cell cycle activity (KEGG), WNT target genes (31), and MYC target genes (39) in CMS1–4 in the CMS–3232 cohort. Statistically significant differences were identified using one–way analysis of variance (ANOVA) with subsequent post–hoc pairwise comparisons using t–tests with pooled SD using Bonferroni multiple comparison p–value adjustment. (B) XY–plot demonstrating the (negative) correlation between CMS4–identfying genes in the RF classifier and the KEGG pathway signature genes reflecting cell cycle activity. R denotes the Pearson correlation coefficient with two–sided p–value in the CMS–3232 cohort. CMS1–4 are color–coded. (C) As in (A) but in the ImPACCT cohort. Statistically significant differences were identified using two–sided ANOVA and a linear mixed model.
Figure 5
Figure 5
Imatinib treatment of primary CMS4 CRC induces a phenotype that is associated with better prognosis. (A) Principle component analysis based on expression of all genes. PRE and POST imatinib samples are color–coded. (B) Bar plot showing the significant up– and down–regulated cancer hallmark signatures (40) (n = 10/50) between pre– and post–imatinib biopsies ranked according to significance (min–log10 p–values). (C) Heatmap showing expression of the 10 significantly upregulated hallmark signatures and a compendium of immune signatures (29) in PRE and POST imatinib treatment samples. (D) Differential gene expression analysis (ANOVA FDR p ≤ 0.001) identified 680 differentially expressed genes of which 228 were up– and 452 were down–regulated after imatinib therapy. The 228 imatinib–induced genes were then used to cluster the CMS3232 cohort (1) into LOW and HIGH expression subgroups using the k–means algorithm. (E) Heatmap showing expression of imatinib–induced genes in the LOW and HIGH expression subgroups. (F) Stacked barplot showing the CMS distribution in subgroups of tumors expressing LOW and HIGH levels of imatinib–induced genes. (G) Kaplan Meier curves showing overall (left) and relapse–free (right) survival in subgroups of stage II–III tumors in the CMS3232 cohort (1) expressing LOW and HIGH levels of imatinib–induced genes. A two–sided log–rank test was applied to assess the significance of the survival differences between the two groups.
Figure 6
Figure 6
Imatinib inhibits ribosomal protein S6 phosphorylation and causes transcriptional activation of the mTORC1 pathway. Expression levels of (A) mTORC1 TOP target mRNAs (33), (B) ribosomal protein S6 (RPS6), (C) the Hallmark mTORC1 signature, and the individual mTORC1 components (D)RPTOR, (E)MLST8, (F)DEPTOR, (G)AKT1S1, and (H)MTOR. Statistically significant expression differences were identified using ANOVA and a linear mixed model. (I) Immunohistochemistry (IHC) for the detection of phosphorylated ribosomal protein S6 (pS6) on PRE–treatment (upper row) and POST–treatment (lower row) biopsies. Representative images of the stained sections are shown. Scale bar, 50 μm (J) QuPath software (41) was used to quantify the pS6 IHC signal in the epithelial compartment in pre– and post–imatinib biopsies. Values were then plotted in Tukey boxplots and the significance of the observed staining difference was assessed using a two–sided paired Student’s t–test.

References

    1. Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, et al. . The consensus molecular subtypes of colorectal cancer. Nat Med (2015) 21(11):1350–6. doi: 10.1038/nm.3967
    1. Trinh A, Trumpi K, De Sousa EMF, Wang X, de Jong JH, Fessler E, et al. . Practical and robust identification of molecular subtypes in colorectal cancer by immunohistochemistry. Clin Cancer Res an Off J Am Assoc Cancer Res (2017) 23(2):387–98. doi: 10.1158/1078-0432.CCR-16-0680
    1. Song N, Pogue-Geile KL, Gavin PG, Yothers G, Kim SR, Johnson NL, et al. . Clinical outcome from oxaliplatin treatment in stage Ii/Iii colon cancer according to intrinsic subtypes: Secondary analysis of nsabp c-07/Nrg oncology randomized clinical trial. JAMA Oncol (2016) 2(9):1162–9. doi: 10.1001/jamaoncol.2016.2314
    1. Ten Hoorn S, de Back TR, Sommeijer DW, Vermeulen L. Clinical value of consensus molecular subtypes in colorectal cancer: A systematic review and meta-analysis. J Natl Cancer Institute (2021) 114(4):503–16. doi: 10.1093/jnci/djab106
    1. Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, et al. . Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet (2015) 47(4):320–9. doi: 10.1038/ng.3225
    1. Isella C, Terrasi A, Bellomo SE, Petti C, Galatola G, Muratore A, et al. . Stromal contribution to the colorectal cancer transcriptome. Nat Genet (2015) 47(4):312–9. doi: 10.1038/ng.3224
    1. Steller EJ, Raats DA, Koster J, Rutten B, Govaert KM, Emmink BL, et al. . Pdgfrb promotes liver metastasis formation of mesenchymal-like colorectal tumor cells. Neoplasia (2013) 15(2):204–17. doi: 10.1593/neo.121726
    1. Fatrai S, van Schelven SJ, Ubink I, Govaert KM, Raats D, Koster J, et al. . Maintenance of clonogenic kit(+) human colon tumor cells requires secretion of stem cell factor by differentiated tumor cells. Gastroenterology (2015) 149(3):692–704. doi: 10.1053/j.gastro.2015.05.003
    1. Ubink I, Elias SG, Moelans CB, Lacle MM, van Grevenstein WMU, van Diest PJ, et al. . A novel diagnostic tool for selecting patients with mesenchymal-type colon cancer reveals intratumor subtype heterogeneity. J Natl Cancer Inst (2017) 109(8):1–8. doi: 10.1093/jnci/djw303
    1. Heldin CH. Targeting the pdgf signaling pathway in the treatment of non-malignant diseases. J Neuroimmune Pharmacol (2014) 9(2):69–79. doi: 10.1007/s11481-013-9484-2
    1. Kitadai Y, Sasaki T, Kuwai T, Nakamura T, Bucana CD, Fidler IJ. Targeting the expression of platelet-derived growth factor receptor by reactive stroma inhibits growth and metastasis of human colon carcinoma. AmJPathol (2006) 169(6):2054–65. doi: 10.2353/ajpath.2006.060653[doi
    1. Xing S, Wang C, Tang H, Guo J, Liu X, Yi F, et al. . Down-regulation of pdgfrbeta suppresses invasion and migration in osteosarcoma cells by influencing epithelial-mesenchymal transition. FEBS Open Bio (2020) 10(9):1748–57. doi: 10.1002/2211-5463.12915
    1. Hou H, Jia D, Yan W, Zhang X, Wang C, Li Y, et al. . Kit/Pdgfra/Kdr amplification defines a novel molecular subtype of adenoid cystic carcinoma patients who may benefit from treatment with tyrosine kinase inhibitors. Trans Cancer Res (2020) 9(8):4703–14. doi: 10.21037/tcr-20-637
    1. Nehoff H, Parayath NN, McConnell MJ, Taurin S, Greish K. A combination of tyrosine kinase inhibitors, crizotinib and dasatinib for the treatment of glioblastoma multiforme. Oncotarget (2015) 6(35):37948–64. doi: 10.18632/oncotarget.5698
    1. Dunne PD, McArt DG, Bradley CA, O'Reilly PG, Barrett HL, Cummins R, et al. . Challenging the cancer molecular stratification dogma: Intratumoral heterogeneity undermines consensus molecular subtypes and potential diagnostic value in colorectal cancer. Clin Cancer Res an Off J Am Assoc Cancer Res (2016) 22(16):4095–104. doi: 10.1158/1078-0432.CCR-16-0032
    1. Marisa L, Blum Y, Taieb J, Ayadi M, Pilati C, Le Malicot K, et al. . Intratumor cms heterogeneity impacts patient prognosis in localized colon cancer. Clin Cancer Res an Off J Am Assoc Cancer Res (2021) 27(17):4768–80. doi: 10.1158/1078-0432.CCR-21-0529
    1. Trumpi K, Ubink I, Trinh A, Djafarihamedani M, Jongen JM, Govaert KM, et al. . Neoadjuvant chemotherapy affects molecular classification of colorectal tumors. Oncogenesis (2017) 6(7):e357. doi: 10.1038/oncsis.2017.48
    1. Schlicker A, Ellappalayam A, Beumer IJ, Snel MHJ, Mittempergher L, Diosdado B, et al. . Investigating the concordance in molecular subtypes of primary colorectal tumors and their matched synchronous liver metastasis. Int J Cancer (2020) 147(8):2303–15. doi: 10.1002/ijc.33003
    1. Eide PW, Moosavi SH, Eilertsen IA, Brunsell TH, Langerud J, Berg KCG, et al. . Metastatic heterogeneity of the consensus molecular subtypes of colorectal cancer. NPJ Genom Med (2021) 6(1):59. doi: 10.1038/s41525-021-00223-7
    1. Ubink I, Bloemendal HJ, Elias SG, Brink MA, Schwartz MP, Holierhoek YCW, et al. . Imatinib treatment of poor prognosis mesenchymal-type primary colon cancer: A proof-of-Concept study in the preoperative window period (Impacct). BMC Cancer (2017) 17(1):282. doi: 10.1186/s12885-017-3264-y
    1. Chen EC, Karl TA, Kalisky T, Gupta SK, O'Brien CA, Longacre TA, et al. . Kit signaling promotes growth of colon xenograft tumors in mice and is up-regulated in a subset of human colon cancers. Gastroenterology (2015) 149(3):705–17 e2. doi: 10.1053/j.gastro.2015.05.042
    1. Hoehler T, von Wichert G, Schimanski C, Kanzler S, Moehler MH, Hinke A, et al. . Phase I/Ii trial of capecitabine and oxaliplatin in combination with bevacizumab and imatinib in patients with metastatic colorectal cancer: Aio krk 0205. Br J Cancer (2013) 109(6):1408–13. doi: 10.1038/bjc.2013.409
    1. Michael M, Zalcberg J, Gibbs P, Lipton L, Gouillou M, Jefford M, et al. . A phase I trial of imatinib in combination with Mfolfox6-bevacizumab in patients with advanced colorectal cancer. Cancer ChemotherPharmacol (2013) 71(2):321–30. doi: 10.1007/s00280-012-2009-5[doi
    1. Marisa L, de Reynies A, Duval A, Selves J, Gaub MP, Vescovo L, et al. . Gene expression classification of colon cancer into molecular subtypes: Characterization, validation, and prognostic value. PloS Med (2013) 10(5):e1001453. doi: 10.1371/journal.pmed.1001453
    1. Cancer Genome Atlas N. Comprehensive molecular characterization of human colon and rectal cancer. Nature (2012) 487(7407):330–7. doi: 10.1038/nature11252
    1. Davis MI, Hunt JP, Herrgard S, Ciceri P, Wodicka LM, Pallares G, et al. . Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol (2011) 29(11):1046–51. doi: 10.1038/nbt.1990
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for rna-seq data with Deseq2. Genome Biol (2014) 15(12):550. doi: 10.1186/s13059-014-0550-8
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. . Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U.S.A. (2005) 102(43):15545–50. doi: 10.1073/pnas.0506580102
    1. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. . Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity (2013) 39(4):782–95. doi: 10.1016/j.immuni.2013.10.003
    1. Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, et al. . Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell (2014) 158(5):1110–22. doi: 10.1016/j.cell.2014.07.013
    1. Van der Flier LG, Sabates-Bellver J, Oving I, Haegebarth A, De Palo M, Anti M, et al. . The intestinal Wnt/Tcf signature. Gastroenterology (2007) 132(2):628–32. doi: 10.1053/j.gastro.2006.08.039
    1. Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. Kegg: Integrating viruses and cellular organisms. Nucleic Acids Res (2021) 49(D1):D545–D51. doi: 10.1093/nar/gkaa970
    1. Fonseca BD, Smith EM, Yelle N, Alain T, Bushell M, Pause A. The ever-evolving role of mtor in translation. Semin Cell Dev Biol (2014) 36:102–12. doi: 10.1016/j.semcdb.2014.09.014
    1. Lorenz DJ, Datta S, Harkema SJ. Marginal association measures for clustered data. Stat Med (2011) 30(27):3181–91. doi: 10.1002/sim.4368
    1. Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S, Tabernero J. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer (2017) 17(2):79–92. doi: 10.1038/nrc.2016.126
    1. Anjomshoaa A, Nasri S, Humar B, McCall JL, Chatterjee A, Yoon HS, et al. . Slow proliferation as a biological feature of colorectal cancer metastasis. Br J Cancer (2009) 101(5):822–8. doi: 10.1038/sj.bjc.6605229
    1. Melo DSE, Colak S, Buikhuisen J, Koster J, Cameron K, de Jong JH, et al. . Methylation of cancer-Stem-Cell-Associated wnt target genes predicts poor prognosis in colorectal cancer patients. Cell Stem Cell (2011) 9(5):476–85. doi: 10.1016/j.stem.2011.10.008
    1. Emmink BL, van Houdt WJ, Vries RG, Hoogwater FJ, Govaert KM, Verheem A, et al. . Differentiated human colorectal cancer cells protect tumor-initiating cells from irinotecan. Gastroenterology (2011) 141(1):269–78. doi: 10.1053/j.gastro.2011.03.052
    1. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The molecular signatures database (Msigdb) hallmark gene set collection. Cell Syst (2015) 1(6):417–25. doi: 10.1016/j.cels.2015.12.004
    1. Thomas LW, Esposito C, Stephen JM, Costa ASH, Frezza C, Blacker TS, et al. . Chchd4 regulates tumour proliferation and emt-related phenotypes, through respiratory chain-mediated metabolism. Cancer Metab (2019) 7:7. doi: 10.1186/s40170-019-0200-4
    1. Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y, McArt DG, Dunne PD, et al. . Qupath: Open source software for digital pathology image analysis. Sci Rep (2017) 7(1):16878. doi: 10.1038/s41598-017-17204-5
    1. Magnuson B, Ekim B, Fingar DC. Regulation and function of ribosomal protein S6 kinase (S6k) within mtor signalling networks. Biochem J (2012) 441(1):1–21. doi: 10.1042/BJ20110892
    1. Faller WJ, Jackson TJ, Knight JR, Ridgway RA, Jamieson T, Karim SA, et al. . Mtorc1-mediated translational elongation limits intestinal tumour initiation and growth. Nature (2015) 517(7535):497–500. doi: 10.1038/nature13896
    1. Evdokimova V, Tognon C, Ng T, Sorensen PH. Reduced proliferation and enhanced migration: Two sides of the same coin? molecular mechanisms of metastatic progression by yb-1. Cell Cycle (2009) 8(18):2901–6. doi: 10.4161/cc.8.18.9537
    1. Shiwarski DJ, Shao C, Bill A, Kim J, Xiao D, Bertrand CA, et al. . To "Grow" or "Go": Tmem16a expression as a switch between tumor growth and metastasis in scchn. Clin Cancer Res an Off J Am Assoc Cancer Res (2014) 20(17):4673–88. doi: 10.1158/1078-0432.CCR-14-0363
    1. Cargnello M, Tcherkezian J, Roux PP. The expanding role of mtor in cancer cell growth and proliferation. Mutagenesis (2015) 30(2):169–76. doi: 10.1093/mutage/geu045
    1. Li J, Dang Y, Gao J, Li Y, Zou J, Shen L. Pi3k/Akt/Mtor pathway is activated after imatinib secondary resistance in gastrointestinal stromal tumors (Gists). Med Oncol (2015) 32(4):111. doi: 10.1007/s12032-015-0554-6
    1. Burchert A, Wang Y, Cai D, von Bubnoff N, Paschka P, Muller-Brusselbach S, et al. . Compensatory Pi3-Kinase/Akt/Mtor activation regulates imatinib resistance development. Leukemia (2005) 19(10):1774–82. doi: 10.1038/sj.leu.2403898
    1. Singh P, Kumar V, Gupta SK, Kumari G, Verma M. Combating tki resistance in cml by inhibiting the Pi3k/Akt/Mtor pathway in combination with tkis: A review. Med Oncol (2021) 38(1):10. doi: 10.1007/s12032-021-01462-5
    1. Schoffski P, Reichardt P, Blay JY, Dumez H, Morgan JA, Ray-Coquard I, et al. . A phase I-ii study of everolimus (Rad001) in combination with imatinib in patients with imatinib-resistant gastrointestinal stromal tumors. Ann Oncol Off J Eur Soc Med Oncol / ESMO (2010) 21(10):1990–8. doi: 10.1093/annonc/mdq076
    1. Piskol R, Huw L, Sergin I, Kljin C, Modrusan Z, Kim D, et al. . A clinically applicable gene-expression classifier reveals intrinsic and extrinsic contributions to consensus molecular subtypes in primary and metastatic colon cancer. Clin Cancer Res an Off J Am Assoc Cancer Res (2019) 25(14):4431–42. doi: 10.1158/1078-0432.CCR-18-3032
    1. Sirinukunwattana K, Domingo E, Richman SD, Redmond KL, Blake A, Verrill C, et al. . Image-based consensus molecular subtype (Imcms) classification of colorectal cancer using deep learning. Gut (2021) 70(3):544–54. doi: 10.1136/gutjnl-2019-319866
    1. Strating E, Wassenaar E, Verhagen M, Rauwerdink P, van Schelven S, de Hingh I. Strating eea. fibroblast activation protein identifies consensus molecular subtype 4 in colorectal cancer and allows its detection by 68ga-fapi pet imaging. Br J Cancer (2022) 127(1):145–55. doi: 10.1038/s41416-022-01748-z

Source: PubMed

3
Abonnere