Colorectal Cancer: Pathogenesis and Targeted Therapy

Jingyuan Li , Jiashu Pan , Lisheng Wang , Guang Ji , Yanqi Dang

MedComm ›› 2025, Vol. 6 ›› Issue (3) : e70127

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (3) : e70127 DOI: 10.1002/mco2.70127
REVIEW

Colorectal Cancer: Pathogenesis and Targeted Therapy

Author information +
History +
PDF

Abstract

Colorectal cancer (CRC) ranks among the most prevalent malignant neoplasms globally. A growing body of evidence underscores the pivotal roles of genetic alterations and dysregulated epigenetic modifications in the pathogenesis of CRC. In recent years, the reprogramming of tumor cell metabolism has been increasingly acknowledged as a hallmark of cancer. Substantial evidence suggests a crosstalk between tumor cell metabolic reprogramming and epigenetic modifications, highlighting a complex interplay between metabolism and the epigenetic genome that warrants further investigation. Biomarkers associated with the pathogenesis and metabolic characteristics of CRC hold significant clinical implications. Nevertheless, elucidating the genetic, epigenetic, and metabolic landscapes of CRC continues to pose considerable challenges. Here, we attempt to summarize the key genes driving the onset and progression of CRC and the related epigenetic regulators, clarify the roles of gene expression and signaling pathways in tumor metabolism regulation, and explore the potential crosstalk between epigenetic events and tumor metabolic reprogramming, providing a comprehensive mechanistic explanation for the malignant progression of CRC. Finally, by integrating reliable targets from genetics, epigenetics, and metabolic processes that hold promise for translation into clinical practice, we aim to offer more strategies to overcome the bottlenecks in CRC treatment.

Keywords

colorectal cancer / genetics / epigenetics / tumor metabolism / molecular mechanisms / targeted therapy

Cite this article

Download citation ▾
Jingyuan Li, Jiashu Pan, Lisheng Wang, Guang Ji, Yanqi Dang. Colorectal Cancer: Pathogenesis and Targeted Therapy. MedComm, 2025, 6(3): e70127 DOI:10.1002/mco2.70127

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

H. Sung, J. Ferlay, R. L. Siegel, et al., “Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries,” CA: A Cancer Journal for Clinicians 71, no. 3 (2021): 209–249.

[2]

E. Morgan, M. Arnold, A. Gini, et al., “Global Burden of Colorectal Cancer in 2020 and 2040: Incidence and Mortality Estimates From GLOBOCAN,” Gut 72, no. 2 (2023): 338–344.

[3]

A. Howren, E. C. Sayre, J. M. Loree, et al., “Trends in the Incidence of Young-Onset Colorectal Cancer With a Focus on Years Approaching Screening Age: A Population-Based Longitudinal Study,” JNCI: Journal of the National Cancer Institute 113, no. 7 (2021): 863–868.

[4]

B. T. Dickinson, J. Kisiel, D. A. Ahlquist, and W. M. Grady, “Molecular Markers for Colorectal Cancer Screening,” Gut 64, no. 9 (2015): 1485–1494.

[5]

V. Lee, R. Parkinson, M. Zahurak, et al., “A Phase II Study of Guadecitabine Combined With Irinotecan vs Regorafenib or TAS-102 in Irinotecan-Refractory Metastatic Colorectal Cancer Patients,” International Journal of Cancer 154, no. 10 (2024): 1794–1801.

[6]

Y. Okugawa, W. M. Grady, and A. Goel, “Epigenetic Alterations in Colorectal Cancer: Emerging Biomarkers,” Gastroenterology 149, no. 5 (2015): 1204–1225. e12.

[7]

Y. Kanai and S. Hirohashi, “Alterations of DNA Methylation Associated with Abnormalities of DNA Methyltransferases in Human Cancers During Transition from a Precancerous to a Malignant State,” Carcinogenesis 28, no. 12 (2007): 2434–2442.

[8]

J. Gil, A. Ramírez-Torres, and S. Encarnación-Guevara, “Lysine Acetylation and Cancer: A Proteomics Perspective,” Journal of Proteomics 150 (2017): 297–309.

[9]

S. La Vecchia and C. Sebastián, “Metabolic Pathways Regulating Colorectal Cancer Initiation and Progression,” Seminars in Cell & Developmental Biology 98 (2020): 63–70.

[10]

M. S. Pino and D. C. Chung, “The Chromosomal Instability Pathway in Colon Cancer,” Gastroenterology 138, no. 6 (2010): 2059–2072.

[11]

E. R. Fearon and B. Vogelstein, “A Genetic Model for Colorectal Tumorigenesis,” Cell 61, no. 5 (1990): 759–767.

[12]

Comprehensive Molecular Characterization of Human Colon and Rectal Cancer. Nature 2012; 487(7407): 330–337

[13]

Z. A. Zhong, M. N. Michalski, P. D. Stevens, E. A. Sall, and B. O. Williams, “Regulation of Wnt Receptor Activity: Implications for Therapeutic Development in Colon Cancer,” Journal of Biological Chemistry 296 (2021): 100782.

[14]

G. Zhu, L. Pei, H. Xia, Q. Tang, and F. Bi, “Role of Oncogenic KRAS in the Prognosis, Diagnosis and Treatment of Colorectal Cancer,” Molecular cancer 20, no. 1 (2021): 143.

[15]

I. López, L. PO, P. Tucci, F. Alvarez-Valín, R. AC, and M. Marín, “Different Mutation Profiles Associated to P53 Accumulation in Colorectal Cancer,” Gene 499, no. 1 (2012): 81–87.

[16]

B. Iacopetta, A. Russo, V. Bazan, et al., “Functional Categories of TP53 Mutation in Colorectal Cancer: Results of an International Collaborative Study,” Annals of Oncology 17, no. 5 (2006): 842–847.

[17]

M. Baretti and D. T. Le, “DNA mismatch repair in cancer,” Pharmacology & Therapeutics 189 (2018): 45–62.

[18]

R. Parsons, L. L. Myeroff, B. Liu, et al., “Microsatellite instability and mutations of the transforming growth factor beta type II receptor gene in colorectal cancer,” Cancer Research 55, no. 23 (1995): 5548–5550.

[19]

Y. Shimada, Y. Tajima, M. Nagahashi, et al., “Clinical Significance of BRAF Non-V600E Mutations in Colorectal Cancer: A Retrospective Study of Two Institutions,” Journal of Surgical Research 232 (2018): 72–81.

[20]

L. H. Nguyen, A. Goel, and D. C. Chung, “Pathways of Colorectal Carcinogenesis,” Gastroenterology 158, no. 2 (2020): 291–302.

[21]

T. Kambara, L. A. Simms, V. L. Whitehall, et al., “BRAF mutation is associated With DNA methylation in serrated polyps and cancers of the colorectum,” Gut 53, no. 8 (2004): 1137–1144.

[22]

C. Gallois, P. Laurent-Puig, and J. Taieb, “Methylator phenotype in colorectal cancer: A prognostic factor or not?,” Critical Reviews in Oncology/Hematology 99 (2016): 74–80.

[23]

W. C. Fernando, M. S. Miranda, D. L. Worthley, et al., “The CIMP Phenotype in BRAF Mutant Serrated Polyps From a Prospective Colonoscopy Patient Cohort,” Gastroenterology Research and Practice 2014 (2014): 374926.

[24]

L. Shen, M. Toyota, Y. Kondo, et al., “Integrated genetic and epigenetic analysis identifies three different subclasses of colon cancer,” PNAS 104, no. 47 (2007): 18654–18659.

[25]

D. W. Rosenberg, S. Yang, D. C. Pleau, et al., “Mutations in BRAF and KRAS differentially distinguish serrated versus non-serrated hyperplastic aberrant crypt foci in humans,” Cancer Research 67, no. 8 (2007): 3551–3554.

[26]

T. Jess, M. Gamborg, P. Matzen, P. Munkholm, and T. I. Sørensen, “Increased risk of intestinal cancer in Crohn’s disease: A meta-analysis of population-based cohort studies,” American Journal of Gastroenterology 100, no. 12 (2005): 2724–2729.

[27]

S. H. Itzkowitz, Y. X. Inflammation, and I. V. cancer, “Colorectal cancer in inflammatory bowel disease: The role of inflammation,” American journal of physiology Gastrointestinal and liver physiology 287, no. 1 (2004): G7–G17.

[28]

S. C. Shah and S. H. Itzkowitz, “Colorectal Cancer in Inflammatory Bowel Disease: Mechanisms and Management,” Gastroenterology 162, no. 3 (2022): 715–730. e3.

[29]

L. B. Meira, J. M. Bugni, S. L. Green, et al., “DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice,” Journal of Clinical Investigation 118, no. 7 (2008): 2516–2525.

[30]

S. I. Grivennikov, “Inflammation and colorectal cancer: Colitis-associated neoplasia,” Seminars in Immunopathology 35, no. 2 (2013): 229–244.

[31]

R. L. Siegel, K. D. Miller, and A. Jemal, “Cancer statistics, 2020,” CA: A Cancer Journal for Clinicians 70, no. 1 (2020): 7–30.

[32]

V. Greger, E. Passarge, W. Höpping, et al., “Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma,” Human Genetics 83, no. 2 (1989): 155–158.

[33]

Y. Baba, T. Yagi, H. Sawayama, et al., “Long Interspersed Element-1 Methylation Level as a Prognostic Biomarker in Gastrointestinal Cancers,” Digestion 97, no. 1 (2018): 26–30.

[34]

A. N. Gargalionis, C. Piperi, C. Adamopoulos, and A. G. Papavassiliou, “Histone modifications as a pathogenic mechanism of colorectal tumorigenesis,” International Journal of Biochemistry & Cell Biology 44, no. 8 (2012): 1276–1289.

[35]

A. F Vega-Benedetti, E. Loi, L. Moi, et al., “Colorectal Cancer Early Detection in Stool Samples Tracing CpG Islands Methylation Alterations Affecting Gene Expression,” International Journal of Molecular Sciences 21, no. 12 (2020): 4494.

[36]

D. Müller and B. Győrffy, “DNA methylation-based diagnostic, prognostic, and predictive biomarkers in colorectal cancer,” Biochimica et Biophysica Acta - Reviews on Cancer 1877, no. 3 (2022): 188722.

[37]

D. Yang, M. Thangaraju, K. Greeneltch, et al., “Repression of IFN regulatory factor 8 by DNA methylation is a molecular determinant of apoptotic resistance and metastatic phenotype in metastatic tumor cells,” Cancer Research 67, no. 7 (2007): 3301–3309.

[38]

S. Tapial, S. Olmedillas-López, D. Rueda, et al., “Cimp-Positive Status is More Representative in Multiple Colorectal Cancers Than in Unique Primary Colorectal Cancers,” Scientific Reports 9, no. 1 (2019): 10516.

[39]

E. Nazemalhosseini Mojarad, P. J. Kuppen, H. A. Aghdaei, and M. R. Zali, “The CpG island methylator phenotype (CIMP) in colorectal cancer,” Gastroenterology and Hepatology from Bed to Bench 6, no. 3 (2013): 120–128.

[40]

V. Amodio, S. Lamba, R. Chilà, et al., “Genetic and pharmacological modulation of DNA mismatch repair heterogeneous tumors promotes immune surveillance,” Cancer Cell 41, no. 1 (2023): 196–209. e5.

[41]

R. D. Kornberg and Y. Lorch, “Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome,” Cell 98, no. 3 (1999): 285–294.

[42]

M. R. Marunde, H. A. Fuchs, J. M. Burg, et al., “Nucleosome conformation dictates the histone code,” Elife 13 (2024): e78866.

[43]

K. Murray, “The occurrence of epsilon-n-methyl lysine in histones,” Biochemistry 3 (1964): 10–15.

[44]

J. C. Black, C. Van Rechem, and J. R. Whetstine, “Histone lysine methylation dynamics: Establishment, regulation, and biological impact,” Molecular Cell 48, no. 4 (2012): 491–507.

[45]

L. Dong, J. Zhu, A. Deng, et al., “Relationship Between histone demethylase LSD family and development and prognosis of gastric cancer,” Frontiers in Immunology 14 (2023): 1170773.

[46]

S. M. Kooistra and K. Helin, “Molecular mechanisms and potential functions of histone demethylases,” Nature Reviews Molecular Cell Biology 13, no. 5 (2012): 297–311.

[47]

T. Huang, C. Lin, L. L. Zhong, et al., “Targeting histone methylation for colorectal cancer,” Therapeutic Advances in Gastroenterology 10, no. 1 (2017): 114–131.

[48]

M. Di Donato, E. Di Zazzo, A. Salvati, et al., “RIZ2 at the crossroad of the EGF/EGFR signaling in colorectal cancer,” Journal of translational medicine 21, no. 1 (2023): 736.

[49]

F. Meng, X. Liu, C. Lin, et al., “SMYD2 suppresses APC2 expression to activate the Wnt/β-catenin pathway and promotes epithelial-mesenchymal transition in colorectal cancer,” American Journal of Cancer Research 10, no. 3 (2020): 997–1011.

[50]

Y. Zhang, L. Zhou, Y. Xu, et al., “Targeting SMYD2 inhibits angiogenesis and increases the efficiency of apatinib by suppressing EGFL7 in colorectal cancer,” Angiogenesis 26, no. 1 (2023): 1–18.

[51]

S. J. Samyesudhas, L. Roy, and K. D. Cowden Dahl, “Differential expression of ARID3B in normal adult tissue and carcinomas,” Gene 543, no. 1 (2014): 174–180.

[52]

X. Zhang, X. Qiu, W. Zhao, et al., “Over-Expression of ARID3B Suppresses Tumor Progression and Predicts Better Prognosis in Patients With Gastric Cancer,” Cancer Control 30 (2023): 10732748231169403.

[53]

P. Dausinas, K. Pulakanti, S. Rao, J. M. Cole, R. Dahl, and K. D. Cowden Dahl, “ARID3A and ARID3B induce stem promoting pathways in ovarian cancer cells,” Gene 738 (2020): 144458.

[54]

K. Kobayashi, T. Era, A. Takebe, L. M. Jakt, and S. Nishikawa, “ARID3B induces malignant transformation of mouse embryonic fibroblasts and is strongly associated With malignant neuroblastoma,” Cancer Research 66, no. 17 (2006): 8331–8336.

[55]

T. T. Liao, C. C. Lin, J. K. Jiang, S. H. Yang, H. W. Teng, and M. H. Yang, “Harnessing stemness and PD-L1 expression by AT-rich interaction domain-containing protein 3B in colorectal cancer,” Theranostics 10, no. 14 (2020): 6095–6112.

[56]

S. Carvalho, M. Freitas, L. Antunes, et al., “Prognostic value of histone marks H3K27me3 and H3K9me3 and modifying enzymes EZH2, SETDB1 and LSD-1 in colorectal cancer,” Journal of Cancer Research and Clinical Oncology 144, no. 11 (2018): 2127–2137.

[57]

V. G. Allfrey, R. Faulkner, and A. E. Mirsky, “Acetylation And Methylation of Histones and Their Possible Role in The Regulation of RNA Synthesis,” PNAS 51, no. 5 (1964): 786–794.

[58]

E. Verdin and M. Ott, “50 years of protein acetylation: From gene regulation to epigenetics, metabolism and Beyond,” Nature Reviews Molecular Cell Biology 16, no. 4 (2015): 258–264.

[59]

K. Struhl, “Histone acetylation and transcriptional regulatory mechanisms,” Genes & Development 12, no. 5 (1998): 599–606.

[60]

M. Pérez-Salvia and M. Esteller, “Bromodomain inhibitors and cancer therapy: From structures to applications,” Epigenetics 12, no. 5 (2017): 323–339.

[61]

F. Fuks, W. A. Burgers, A. Brehm, L. Hughes-Davies, and T. Kouzarides, “DNA methyltransferase Dnmt1 associates With histone deacetylase activity,” Nature Genetics 24, no. 1 (2000): 88–91.

[62]

K. Taniue, T. Hayashi, Y. Kamoshida, et al., “UHRF1-KAT7-mediated regulation of TUSC3 expression via histone methylation/acetylation is critical for the proliferation of colon cancer cells,” Oncogene 39, no. 5 (2020): 1018–1030.

[63]

N. Garmpis, C. Damaskos, A. Garmpi, et al., “Histone Deacetylases and their Inhibitors in Colorectal Cancer Therapy: Current Evidence and Future Considerations,” Current Medicinal Chemistry 29, no. 17 (2022): 2979–2994.

[64]

J. Karczmarski, T. Rubel, A. Paziewska, et al., “Histone H3 lysine 27 acetylation is altered in colon cancer,” Clinical Proteomics 11, no. 1 (2014): 24.

[65]

H. Ashktorab, K. Belgrave, F. Hosseinkhah, et al., “Global histone H4 acetylation and HDAC2 expression in colon adenoma and carcinoma,” Digestive Diseases and Sciences 54, no. 10 (2009): 2109–2117.

[66]

M. F. Fraga, E. Ballestar, A. Villar-Garea, et al., “Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer,” Nature Genetics 37, no. 4 (2005): 391–400.

[67]

H. Zhang, J. Chi, J. Hu, et al., “Intracellular AGR2 transduces PGE2 stimuli to promote epithelial-mesenchymal transition and metastasis of colorectal cancer,” Cancer Letters 518 (2021): 180–195.

[68]

H. Tamagawa, T. Oshima, M. Shiozawa, et al., “The global histone modification pattern correlates With overall survival in metachronous liver metastasis of colorectal cancer,” Oncology Reports 27, no. 3 (2012): 637–642.

[69]

Z. Shen, B. Wang, J. Luo, et al., “Global-scale profiling of differential expressed lysine acetylated proteins in colorectal cancer tumors and paired liver metastases,” Journal of Proteomics 142 (2016): 24–32.

[70]

A. Benard, I. J Goossens-Beumer, A. Q. van Hoesel, et al., “Nuclear expression of histone deacetylases and their histone modifications predicts clinical outcome in colorectal cancer,” Histopathology 66, no. 2 (2015): 270–282.

[71]

B. N. Singh, G. Zhang, Y. L. Hwa, J. Li, S. C. Dowdy, and S. W. Jiang, “Nonhistone protein acetylation as cancer therapy targets,” Expert Review of Anticancer Therapy 10, no. 6 (2010): 935–954.

[72]

A. J. Wilson, D. S. Byun, N. Popova, et al., “Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer,” Journal of Biological Chemistry 281, no. 19 (2006): 13548–13558.

[73]

K. Kanda, J. Sakamoto, Y. Matsumoto, et al., “Nardilysin controls intestinal tumorigenesis Through HDAC1/p53-dependent transcriptional regulation,” JCI Insight 3, no. 8 (2018): e91316.

[74]

B. Wang, Y. Ye, X. Yang, et al., “SIRT2-dependent IDH1 deacetylation inhibits colorectal cancer and liver metastases,” Embo Reports 21, no. 4 (2020): e48183.

[75]

H. Jiang, L. Wu, J. Chen, et al., “Sulfiredoxin Promotes Colorectal Cancer Cell Invasion and Metastasis Through a Novel Mechanism of Enhancing EGFR Signaling,” Molecular Cancer Research 13, no. 12 (2015): 1554–1566.

[76]

R. C. Friedman, K. K. Farh, C. B. Burge, and D. P. Bartel, “Most mammalian mRNAs are conserved targets of microRNAs,” Genome Research 19, no. 1 (2009): 92–105.

[77]

D. Zapletal, K. Kubicek, P. Svoboda, and R. Stefl, “Dicer structure and function: Conserved and evolving features,” Embo Reports 24, no. 7 (2023): e57215.

[78]

T. Treiber, N. Treiber, and G. Meister, “Regulation of microRNA biogenesis and its crosstalk With other cellular pathways,” Nature Reviews Molecular Cell Biology 20, no. 1 (2019): 5–20.

[79]

Y. W. Cheng, C. J. Chou, and P. M. Yang, “Ten-eleven translocation 1 (TET1) gene is a potential target of miR-21-5p in human colorectal cancer,” Surgical Oncology 27, no. 1 (2018): 76–81.

[80]

M. Toyota, H. Suzuki, Y. Sasaki, et al., “Epigenetic silencing of microRNA-34b/c and B-cell translocation gene 4 is associated With CpG island methylation in colorectal cancer,” Cancer Research 68, no. 11 (2008): 4123–4132.

[81]

R. Kogo, T. Shimamura, K. Mimori, et al., “Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated With poor prognosis in colorectal cancers,” Cancer Research 71, no. 20 (2011): 6320–6326.

[82]

D. Hanahan and R. A. Weinberg, “Hallmarks of cancer: The next generation,” Cell 144, no. 5 (2011): 646–674.

[83]

B. Faubert, A. Solmonson, and R. J. DeBerardinis, “Metabolic reprogramming and cancer progression,” Science 368, no. 6487 (2020): eaaw5473.

[84]

O. Warburg, F. Wind, and E. Negelein, “The metabolism of tumors in the body,” Journal of General Physiology 8, no. 6 (1927): 519–530.

[85]

J. Zheng, “Energy metabolism of cancer: Glycolysis versus oxidative phosphorylation (Review),” Oncology Letters 4, no. 6 (2012): 1151–1157.

[86]

M. Jiang, S. Liu, J. Lin, et al., “A pan-cancer analysis of molecular characteristics and oncogenic role of hexokinase family genes in human tumors,” Life Sciences 264 (2021): 118669.

[87]

F. Li, X. Wan, Z. Li, and L. Zhou, “High glucose inhibits autophagy and promotes the proliferation and metastasis of colorectal cancer Through the PI3K/AKT/mTOR pathway,” Cancer Medicine 13, no. 11 (2024): e7382.

[88]

S. Mizuno, R. Seishima, J. Yamasaki, et al., “Angiopoietin-Like 4 promotes glucose metabolism by regulating glucose transporter expression in colorectal cancer,” Journal of Cancer Research and Clinical Oncology 148, no. 6 (2022): 1351–1361.

[89]

N. Rahmani-Kukia, M. Zamani, and P. Mokaram, “ERMP1 Facilitates The Malignant Characteristics of Colorectal Cancer Cells Through Modulating PI3K/AKT/β-Catenin Pathway and Localization of GRP78,” Cell Journal 25, no. 7 (2023): 470–482.

[90]

W. B. Long, X. Pu, Y. Tang, et al., “Arginine ADP-ribosyltransferase 1 Regulates Glycolysis in Colorectal Cancer via the PI3K/AKT/HIF1α Pathway,” Current Medical Science 42, no. 4 (2022): 733–741.

[91]

C. Li, Q. Chen, Y. Zhou, et al., “S100A2 promotes glycolysis and proliferation via GLUT1 regulation in colorectal cancer,” Faseb Journal 34, no. 10 (2020): 13333–13344.

[92]

M. Talieri, D. K. Alexopoulou, A. Scorilas, et al., “Expression analysis and clinical evaluation of kallikrein-related peptidase 10 (KLK10) in colorectal cancer,” Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine 32, no. 4 (2011): 737–744.

[93]

H. Wei, C. Dong, and Z. Shen, “Kallikrein-related peptidase (KLK10) cessation blunts colorectal cancer cell growth and glucose metabolism by regulating the PI3K/Akt/mTOR pathway,” Neoplasma 67, no. 4 (2020): 889–897.

[94]

F. Dupuy, S. Tabariès, S. Andrzejewski, et al., “PDK1-Dependent Metabolic Reprogramming Dictates Metastatic Potential in Breast Cancer,” Cell Metabolism 22, no. 4 (2015): 577–589.

[95]

S. Dong, S. Liang, Z. Cheng, et al., “ROS/PI3K/Akt and Wnt/β-catenin signalings activate HIF-1α-induced metabolic reprogramming to impart 5-fluorouracil resistance in colorectal cancer,” Journal of Experimental & Clinical Cancer Research 41, no. 1 (2022): 15.

[96]

P. H. Cha, J. H. Hwang, D. K. Kwak, E. Koh, K. S. Kim, and K. Y. Choi, “APC loss induces Warburg effect via increased PKM2 transcription in colorectal cancer,” British Journal of Cancer 124, no. 3 (2021): 634–644.

[97]

A. J. Valvezan, F. Zhang, J. A. Diehl, and P. S. Klein, “Adenomatous polyposis coli (APC) regulates multiple signaling pathways by enhancing glycogen synthase kinase-3 (GSK-3) activity,” Journal of Biological Chemistry 287, no. 6 (2012): 3823–3832.

[98]

I. T. Sandoval, R. G. Delacruz, B. N. Miller, et al., “A metabolic switch controls intestinal differentiation downstream of Adenomatous polyposis coli (APC),” Elife 6 (2017): e22706.

[99]

R. Boidot, F. Végran, A. Meulle, et al., “Regulation of monocarboxylate transporter MCT1 expression by p53 mediates inward and outward lactate fluxes in tumors,” Cancer Research 72, no. 4 (2012): 939–948.

[100]

J. Drury, P. G. Rychahou, D. He, et al., “Inhibition of Fatty Acid Synthase Upregulates Expression of CD36 to Sustain Proliferation of Colorectal Cancer Cells,” Frontiers in Oncology 10 (2020): 1185.

[101]

A. M. Niculae, M. Dobre, V. Herlea, et al., “Lipid Handling Protein Gene Expression in Colorectal Cancer: CD36 and Targeting miRNAs,” Life (Basel) 12, no. 12 (2022): 2127.

[102]

K. Kawaguchi, S. Senga, C. Kubota, Y. Kawamura, Y. Ke, and H. Fujii, “High expression of Fatty Acid-Binding Protein 5 promotes cell growth and metastatic potential of colorectal cancer cells,” FEBS Open Bio 6, no. 3 (2016): 190–199.

[103]

Y. Y. Zaytseva, J. W. Harris, M. I. Mitov, et al., “Increased expression of fatty acid synthase provides a survival advantage to colorectal cancer cells via upregulation of cellular respiration,” Oncotarget 6, no. 22 (2015): 18891–18904.

[104]

J. Wen, X. Min, M. Shen, et al., “ACLY facilitates colon cancer cell metastasis by CTNNB1,” Journal of Experimental & Clinical Cancer Research 38, no. 1 (2019): 401.

[105]

C. Li, L. Zhang, Z. Qiu, W. Deng, and W. Wang, “Key Molecules of Fatty Acid Metabolism in Gastric Cancer,” Biomolecules 12, no. 5 (2022): 706.

[106]

N. Zaidi, I. Royaux, J. V. Swinnen, and K. Smans, “ATP citrate lyase knockdown induces growth arrest and apoptosis Through different cell-and environment-dependent mechanisms,” Molecular Cancer Therapeutics 11, no. 9 (2012): 1925–1935.

[107]

E. Piccinin, M. Cariello, and A. Moschetta, “Lipid metabolism in colon cancer: Role of Liver X Receptor (LXR) and Stearoyl-CoA Desaturase 1 (SCD1),” Molecular Aspects of Medicine 78 (2021): 100933.

[108]

D. R. Plas and C. B. Thompson, “Akt-dependent transformation: There is more to growth Than just surviving,” Oncogene 24, no. 50 (2005): 7435–7442.

[109]

Y. A. Wen, X. Xiong, Y. Y. Zaytseva, et al., “Downregulation of SREBP inhibits tumor growth and initiation by altering cellular metabolism in colon cancer,” Cell Death & Disease 9, no. 3 (2018): 265.

[110]

Z. Qiu, W. Deng, Y. Hong, et al., “Biological Behavior and Lipid Metabolism of Colon Cancer Cells are Regulated by a Combination of Sterol Regulatory Element-Binding Protein 1 and ATP Citrate Lyase,” OncoTargets and Therapy 14 (2021): 1531–1542.

[111]

J. Quan, C. Cheng, Y. Tan, et al., “Acyl-CoA synthetase long-chain 3-mediated fatty acid oxidation is required for TGFβ1-induced epithelial-mesenchymal transition and metastasis of colorectal carcinoma,” International Journal of Biological Sciences 18, no. 6 (2022): 2484–2496.

[112]

R. V. Farese and T. C. Walther, “Lipid droplets finally get a little R-E-S-P-E-C-T,” Cell 139, no. 5 (2009): 855–860.

[113]

L. Tirinato, C. Liberale, S. Di Franco, et al., “Lipid droplets: A new player in colorectal cancer stem cells unveiled by spectroscopic imaging,” Stem Cells 33, no. 1 (2015): 35–44.

[114]

S. S. Mayengbam, A. Singh, A. D. Pillai, and M. K. Bhat, “Influence of cholesterol on cancer progression and therapy,” Translational Oncology 14, no. 6 (2021): 101043.

[115]

C. Wang, P. Li, J. Xuan, et al., “Cholesterol Enhances Colorectal Cancer Progression via ROS Elevation and MAPK Signaling Pathway Activation,” Cellular Physiology and Biochemistry 42, no. 2 (2017): 729–742.

[116]

W. Dai, W. Xiang, L. Han, et al., “PTPRO represses colorectal cancer tumorigenesis and progression by reprogramming fatty acid metabolism,” Cancer Communications (Lond) 42, no. 9 (2022): 848–867.

[117]

P. Gao, C. Zhou, L. Zhao, G. Zhang, and Y. Zhang, “Tissue amino acid profile could be used to differentiate advanced adenoma From colorectal cancer,” Journal of Pharmaceutical and Biomedical Analysis 118 (2016): 349–355.

[118]

A. M. Hosios, V. C. Hecht, L. V. Danai, et al., “Amino Acids Rather Than Glucose Account for the Majority of Cell Mass in Proliferating Mammalian Cells,” Developmental Cell 36, no. 5 (2016): 540–549.

[119]

T. Q. Tran, E. A. Hanse, A. N. Habowski, et al., “α-Ketoglutarate attenuates Wnt signaling and drives differentiation in colorectal cancer,” Nature Cancer 1, no. 3 (2020): 345–358.

[120]

A. K. Najumudeen, F. Ceteci, S. K. Fey, et al., “The amino acid transporter SLC7A5 is required for efficient growth of KRAS-mutant colorectal cancer,” Nature Genetics 53, no. 1 (2021): 16–26.

[121]

C. C. Wong, J. Xu, X. Bian, et al., “In Colorectal Cancer Cells With Mutant KRAS, SLC25A22-Mediated Glutaminolysis Reduces DNA Demethylation to Increase WNT Signaling, Stemness, and Drug Resistance,” Gastroenterology 159, no. 6 (2020): 2163–2180. e6.

[122]

S. S. Hu, Y. Han, T. Y. Tan, et al., “SLC25A21 downregulation promotes KRAS-mutant colorectal cancer progression by increasing glutamine anaplerosis,” JCI Insight 8, no. 21 (2023): e167874.

[123]

N. Venkateswaran and C.-S. M. Kynurenine, “an oncometabolite in colon cancer,” Cell Stress 4, no. 1 (2020): 24–26.

[124]

L. Sun, H. Zhang, and P. Gao, “Metabolic reprogramming and epigenetic modifications on the path to cancer,” Protein Cell 13, no. 12 (2022): 877–919.

[125]

T. Ge, X. Gu, R. Jia, et al., “Crosstalk Between metabolic reprogramming and epigenetics in cancer: Updates on mechanisms and therapeutic opportunities,” Cancer Commun (Lond) 42, no. 11 (2022): 1049–1082.

[126]

C. Choudhary, B. T. Weinert, Y. Nishida, E. Verdin, and M. Mann, “The growing landscape of lysine acetylation links metabolism and cell signalling,” Nature Reviews Molecular Cell Biology 15, no. 8 (2014): 536–550.

[127]

S. Trefely, C. D. Lovell, N. W. Snyder, and K. E. Wellen, “Compartmentalised acyl-CoA metabolism and roles in chromatin regulation,” Molecular Metabolis 38 (2020): 100941.

[128]

E. V. Prochownik and H. Wang, “The Metabolic Fates of Pyruvate in Normal and Neoplastic Cells,” Cells 10, no. 4 (2021): 762.

[129]

J. B. Felix, A. R. Cox, and S. M. Hartig, “Acetyl-CoA and Metabolite Fluxes Regulate White Adipose Tissue Expansion,” Trends in Endocrinology and Metabolism 32, no. 5 (2021): 320–332.

[130]

Y. Xu and W. Wan, “Acetylation in the regulation of autophagy,” Autophagy 19, no. 2 (2023): 379–387.

[131]

J. V. Lee, A. Carrer, S. Shah, et al., “Akt-dependent metabolic reprogramming regulates tumor cell histone acetylation,” Cell Metabolism 20, no. 2 (2014): 306–319.

[132]

E. McDonnell, S. B. Crown, D. B. Fox, et al., “Lipids Reprogram Metabolism to Become a Major Carbon Source for Histone Acetylation,” Cell Reports 17, no. 6 (2016): 1463–1472.

[133]

R. A. Henry, Y. M. Kuo, V. Bhattacharjee, T. J. Yen, and A. J. Andrews, “Changing the selectivity of p300 by acetyl-CoA modulation of histone acetylation,” ACS Chemical Biology 10, no. 1 (2015): 146–156.

[134]

M. Lu, W. W. Zhu, X. Wang, et al., “ACOT12-Dependent Alteration of Acetyl-CoA Drives Hepatocellular Carcinoma Metastasis by Epigenetic Induction of Epithelial-Mesenchymal Transition,” Cell Metabolism 29, no. 4 (2019): 886–900. e5.

[135]

P. Icard, Z. Wu, L. Fournel, A. Coquerel, H. Lincet, and M. Alifano, “ATP citrate lyase: A central metabolic enzyme in cancer,” Cancer Letters 471 (2020): 125–134.

[136]

T. Golias, M. Kery, S. Radenkovic, and I. Papandreou, “Microenvironmental control of glucose metabolism in tumors by regulation of pyruvate dehydrogenase,” International Journal of Cancer 144, no. 4 (2019): 674–686.

[137]

Y. A. Wen, X. Xing, J. W. Harris, et al., “Adipocytes activate mitochondrial fatty acid oxidation and autophagy to promote tumor growth in colon cancer,” Cell Death & Disease 8, no. 2 (2017): e2593.

[138]

A. Carracedo, L. C. Cantley, and P. P. Pandolfi, “Cancer metabolism: Fatty acid oxidation in the limelight,” Nature Reviews Cancer 13, no. 4 (2013): 227–232.

[139]

X. Li, W. Yu, X. Qian, et al., “Nucleus-Translocated ACSS2 Promotes Gene Transcription for Lysosomal Biogenesis and Autophagy,” Molecular Cell 66, no. 5 (2017): 684–697. e9.

[140]

K. Liu, F. Li, Q. Sun, et al., “p53 β-hydroxybutyrylation attenuates p53 activity,” Cell Death & Disease 10, no. 3 (2019): 243.

[141]

L. M. Rodrigues, S. Uribe-Lewis, B. Madhu, D. J. Honess, M. Stubbs, and J. R. Griffiths, “The action of β-hydroxybutyrate on the growth, metabolism and global histone H3 acetylation of spontaneous mouse mammary tumours: Evidence of a β-hydroxybutyrate paradox,” Cancer & Metabolism 5 (2017): 4.

[142]

U. E Martinez-Outschoorn, M. Prisco, A. Ertel, et al., “Ketones and lactate increase cancer cell “stemness,” driving recurrence, metastasis and poor clinical outcome in breast cancer: Achieving personalized medicine via Metabolo-Genomics,” Cell Cycle 10, no. 8 (2011): 1271–1286.

[143]

K. J. Menzies, H. Zhang, E. Katsyuba, and J. Auwerx, “Protein acetylation in metabolism—metabolites and cofactors,” Nature Reviews Endocrinology 12, no. 1 (2016): 43–60.

[144]

M. Zhou, D. Wang, X. Li, et al., “Farnesoid-X receptor as a therapeutic target for inflammatory bowel disease and colorectal cancer,” Frontiers in Pharmacology 13 (2022): 1016836.

[145]

S. Pathak, S. Pandanaboyana, I. Daniels, N. Smart, and K. R. Prasad, “Obesity and colorectal liver metastases: Mechanisms and management,” Surgical Oncology 25, no. 3 (2016): 246–251.

[146]

S. Ocvirk, A. S. Wilson, C. N. Appolonia, T. K. Thomas, and S. J. D. O’Keefe, “Fiber, Fat, and Colorectal Cancer: New Insight Into Modifiable Dietary Risk Factors,” Current Gastroenterology Reports 21, no. 11 (2019): 62.

[147]

J. Yang, H. Wei, Y. Zhou, et al., “High-Fat Diet Promotes Colorectal Tumorigenesis Through Modulating Gut Microbiota and Metabolites,” Gastroenterology 162, no. 1 (2022): 135–149. e2.

[148]

D. Chen, J. Bruno, E. Easlon, et al., “Tissue-specific regulation of SIRT1 by calorie restriction,” Genes & Development 22, no. 13 (2008): 1753–1757.

[149]

B. Sosnowska, M. Mazidi, P. Penson, A. Gluba-Brzózka, J. Rysz, and M. Banach, “The sirtuin family members SIRT1, SIRT3 and SIRT6: Their role in vascular biology and atherogenesis,” Atherosclerosis 265 (2017): 275–282.

[150]

J. G. Ryall, S. Dell’Orso, A. Derfoul, et al., “The NAD(+)-dependent SIRT1 deacetylase translates a metabolic switch Into regulatory epigenetics in skeletal muscle stem cells,” Cell Stem Cell 16, no. 2 (2015): 171–183.

[151]

W. Kim, A. Deik, C. Gonzalez, et al., “Polyunsaturated Fatty Acid Desaturation Is a Mechanism for Glycolytic NAD(+) Recycling,” Cell Metabolism 29, no. 4 (2019): 856–870. e7.

[152]

C. C. Wong, Y. Qian, and J. Yu, “Interplay Between epigenetics and metabolism in oncogenesis: Mechanisms and therapeutic approaches,” Oncogene 36, no. 24 (2017): 3359–3374.

[153]

N. Shiraki, Y. Shiraki, T. Tsuyama, et al., “Methionine metabolism regulates maintenance and differentiation of human pluripotent stem cells,” Cell Metabolism, no. 5 (2014): 780–794.

[154]

S. J. Mentch, M. Mehrmohamadi, L. Huang, et al., “Histone Methylation Dynamics and Gene Regulation Occur Through the Sensing of One-Carbon Metabolism,” Cell Metabolism 22, no. 5 (2015): 861–873.

[155]

M. A. Caudill, J. C. Wang, S. Melnyk, et al., “Intracellular S-adenosylhomocysteine concentrations predict global DNA hypomethylation in tissues of methyl-deficient cystathionine beta-synthase heterozygous mice,” Journal of Nutrition 131, no. 11 (2001): 2811–2818.

[156]

K. Ito, S. Ikeda, N. Kojima, et al., “Correlation Between the expression of methionine adenosyltransferase and the stages of human colorectal carcinoma,” Surgery Today 30, no. 8 (2000): 706–710.

[157]

J. Wang, Z. H. Zhu, H. B. Yang, et al., “Cullin 3 targets methionine adenosyltransferase IIα for ubiquitylation-mediated degradation and regulates colorectal cancer cell proliferation,” Febs Journal 283, no. 13 (2016): 2390–2402.

[158]

X. Xie, H. Liu, Y. Wang, et al., “Nicotinamide N-methyltransferase enhances resistance to 5-fluorouracil in colorectal cancer cells Through inhibition of the ASK1-p38 MAPK pathway,” Oncotarget 7, no. 29 (2016): 45837–45848.

[159]

H. Xie, L. Wei, Q. Wang, S. Tang, and J. Gan, “Elevated serum homocysteine levels associated With poor recurrence-free and overall survival in patients With colorectal cancer,” Scientific Reports 14, no. 1 (2024): 10057.

[160]

B. W. Carey, L. W. Finley, J. R. Cross, C. D. Allis, and C. B. Thompson, “Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells,” Nature 518, no. 7539 (2015): 413–416.

[161]

R. Chowdhury, K. K. Yeoh, Y. M. Tian, et al., “The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases,” Embo Reports 12, no. 5 (2011): 463–469.

[162]

C. Gerecke, F. Schumacher, A. Berndzen, T. Homann, and B. Kleuser, “Vitamin C in combination With inhibition of mutant IDH1 synergistically activates TET enzymes and epigenetically modulates gene silencing in colon cancer cells,” Epigenetics 15, no. 3 (2020): 307–322.

[163]

I. Zucchi, S. Astigiano, G. Bertalot, et al., “Distinct populations of tumor-initiating cells derived From a tumor generated by rat mammary cancer stem cells,” PNAS 105, no. 44 (2008): 16940–16945.

[164]

T. K. Ha, N. G. Her, M. G. Lee, et al., “Caveolin-1 increases aerobic glycolysis in colorectal cancers by stimulating HMGA1-mediated GLUT3 transcription,” Cancer Research 72, no. 16 (2012): 4097–4109.

[165]

C. Shen, B. Xuan, T. Yan, et al., “m(6)A-dependent glycolysis enhances colorectal cancer progression,” Molecular Cancer 19, no. 1 (2020): 72.

[166]

K. Zhang, T. Zhang, Y. Yang, et al., “N(6)-methyladenosine-mediated LDHA induction potentiates chemoresistance of colorectal cancer cells Through metabolic reprogramming,” Theranostics 12, no. 10 (2022): 4802–4817.

[167]

W. Wei, Z. Y. Zhang, B. Shi, et al., “METTL16 promotes glycolytic metabolism reprogramming and colorectal cancer progression,” Journal of Experimental & Clinical Cancer Research 42, no. 1 (2023): 151.

[168]

B. W. Katona, Y. Liu, A. Ma, J. Jin, and X. Hua, “EZH2 inhibition enhances the efficacy of an EGFR inhibitor in suppressing colon cancer cells,” Cancer Biology & Therapy 15, no. 12 (2014): 1677–1687.

[169]

C. W. Chou, M. S. Wu, W. C. Huang, and C. C. Chen, “HDAC inhibition decreases the expression of EGFR in colorectal cancer cells,” PLoS ONE 6, no. 3 (2011): e18087.

[170]

T. TeSlaa, M. Ralser, J. Fan, and J. D. Rabinowitz, “The pentose phosphate pathway in health and disease,” Nature Metabolism 5, no. 8 (2023): 1275–1289.

[171]

B. Chen, Y. Hong, R. Gui, et al., “N6-methyladenosine modification of circ_0003215 suppresses the pentose phosphate pathway and malignancy of colorectal cancer Through the miR-663b/DLG4/G6PD axis,” Cell Death & Disease 13, no. 9 (2022): 804.

[172]

H. Yan, R. Talty, A. Jain, et al., “Discovery of decreased ferroptosis in male colorectal cancer patients With KRAS mutations,” Redox Biology 62 (2023): 102699.

[173]

L. A. Broadfield, A. A. Pane, A. Talebi, J. V. Swinnen, and S. M. Fendt, “Lipid metabolism in cancer: New perspectives and emerging mechanisms,” Developmental Cell 56, no. 10 (2021): 1363–1393.

[174]

E. Currie, A. Schulze, R. Zechner, and T. C. Walther, “Cellular fatty acid metabolism and cancer,” Cell Metabolism 18, no. 2 (2013): 153–161.

[175]

H. Hou, D. Chen, K. Zhang, et al., “Gut microbiota-derived short-chain fatty acids and colorectal cancer: Ready for clinical translation?,” Cancer Letters 526 (2022): 225–235.

[176]

Y. Yoshii, A. Waki, T. Furukawa, et al., “Tumor uptake of radiolabeled acetate reflects the expression of cytosolic acetyl-CoA synthetase: Implications for the mechanism of acetate PET,” Nuclear Medicine and Biology 36, no. 7 (2009): 771–777.

[177]

C. Zhang, X. Y. Wang, P. Zhang, et al., “Cancer-derived exosomal HSPC111 promotes colorectal cancer liver metastasis by reprogramming lipid metabolism in cancer-associated fibroblasts,” Cell Death & Disease 13, no. 1 (2022): 57.

[178]

H. N. Abramson, “The lipogenesis pathway as a cancer target,” Journal of Medicinal Chemistry 54, no. 16 (2011): 5615–5638.

[179]

H. Chen, C. Xie, Q. Chen, and S. Zhuang, “HDAC11, an emerging therapeutic target for metabolic disorders,” Frontiers in Endocrinology (Lausanne) 13 (2022): 989305.

[180]

N. Mitro, C. Godio, E. De Fabiani, et al., “Insights in the regulation of cholesterol 7alpha-hydroxylase gene reveal a target for modulating bile acid synthesis,” Hepatology 46, no. 3 (2007): 885–897.

[181]

Y. Zhang, S. S. Bharathi, M. J. Rardin, et al., “SIRT3 and SIRT5 regulate the enzyme activity and cardiolipin binding of very long-chain acyl-CoA dehydrogenase,” PLoS ONE 10, no. 3 (2015): e0122297.

[182]

M. D. Hirschey, T. Shimazu, E. Goetzman, et al., “SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation,” Nature 464, no. 7285 (2010): 121–125.

[183]

Z. Wei, J. Xia, J. Li, et al., “SIRT1 promotes glucolipid metabolic conversion to facilitate tumor development in colorectal carcinoma,” International Journal of Biological Sciences 19, no. 6 (2023): 1925–1940.

[184]

W. C. Hsieh, B. M. Sutter, H. Ruess, S. D. Barnes, V. S. Malladi, and B. P. Tu, “Glucose starvation induces a switch in the histone acetylome for activation of gluconeogenic and fat metabolism genes,” Molecular Cell 82, no. 1 (2022): 60–74. e5.

[185]

C. Corbet, A. Pinto, R. Martherus, J. P. Santiago de Jesus, F. Polet, and O. Feron, “Acidosis Drives the Reprogramming of Fatty Acid Metabolism in Cancer Cells Through Changes in Mitochondrial and Histone Acetylation,” Cell Metabolism 24, no. 2 (2016): 311–323.

[186]

T. Bouras, M. Fu, A. A. Sauve, et al., “SIRT1 deacetylation and repression of p300 involves lysine residues 1020/1024 Within the cell cycle regulatory domain 1,” Journal of Biological Chemistry 280, no. 11 (2005): 10264–10276.

[187]

Q. J. Zhang, Z. Wang, H. Z. Chen, et al., “Endothelium-specific overexpression of class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein E-deficient mice,” Cardiovascular Research 80, no. 2 (2008): 191–199.

[188]

Y. Majeed, N. Halabi, A. Y. Madani, et al., “SIRT1 promotes lipid metabolism and mitochondrial biogenesis in adipocytes and coordinates adipogenesis by targeting key enzymatic pathways,” Scientific Reports 11, no. 1 (2021): 8177.

[189]

X. Li, S. Zhang, G. Blander, J. G. Tse, M. Krieger, and L. Guarente, “SIRT1 deacetylates and positively regulates the nuclear receptor LXR,” Molecular Cell 28, no. 1 (2007): 91–106.

[190]

W. C. Hallows, S. Lee, and J. M. Denu, “Sirtuins deacetylate and activate mammalian acetyl-CoA synthetases,” PNAS 103, no. 27 (2006): 10230–10235.

[191]

J. Li, P. Song, L. Zhu, et al., “Synthetic lethality of glutaminolysis inhibition, autophagy inactivation and asparagine depletion in colon cancer,” Oncotarget 8, no. 26 (2017): 42664–42672.

[192]

Y. Han, Y. Pu, X. Liu, et al., “YTHDF1 regulates GID8-mediated glutamine metabolism to promote colorectal cancer progression in m6A-dependent manner,” Cancer Letters 601 (2024): 217186.

[193]

Y. Liu, C. E. Tu, X. Guo, et al., “Tumor-suppressive function of EZH2 is Through inhibiting glutaminase,” Cell Death & Disease 12, no. 11 (2021): 975.

[194]

J. Li, P. Song, T. Jiang, et al., “Heat Shock Factor 1 Epigenetically Stimulates Glutaminase-1-Dependent mTOR Activation to Promote Colorectal Carcinogenesis,” Molecular Therapy 26, no. 7 (2018): 1828–1839.

[195]

Y. Ohhara, N. Fukuda, S. Takeuchi, et al., “Role of targeted therapy in metastatic colorectal cancer,” World Journal of Gastrointestinal Oncology 8, no. 9 (2016): 642–655.

[196]

M. Yan, R. Yang, Q. Li, et al., “Anti-angiogenic and antitumor effects of anlotinib combined With bevacizumab for colorectal cancer,” Translational Oncology 41 (2024): 101887.

[197]

C. Cremolini, F. Loupakis, G. Masi, et al., “FOLFOXIRI or FOLFOXIRI plus bevacizumab as first-line treatment of metastatic colorectal cancer: A propensity score-adjusted analysis From two randomized clinical trials,” Annals of Oncology 27, no. 5 (2016): 843–849.

[198]

H. S. Hochster, P. Catalano, M. Weitz, et al., “Combining antivascular endothelial growth factor and anti-epidermal growth factor receptor antibodies: Randomized phase II study of irinotecan and cetuximab With/Without ramucirumab in second-line colorectal cancer (ECOG-ACRIN E7208),” JNCI: Journal of the National Cancer Institute 116, no. 9 (2024): 1487–1494.

[199]

N. S. González, P. V. Marchese, I. Baraibar, et al., “Epidermal growth factor receptor antagonists in colorectal cancer: Emerging strategies for precision therapy,” Expert Opinion on Investigational Drugs 33, no. 6 (2024): 613–625.

[200]

B. You and E. X. Chen, “Anti-EGFR monoclonal antibodies for treatment of colorectal cancers: Development of cetuximab and panitumumab,” Journal of Clinical Pharmacology 52, no. 2 (2012): 128–155.

[201]

E. Van Cutsem, C. H. Köhne, E. Hitre, et al., “Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer,” New England Journal of Medicine 360, no. 14 (2009): 1408–1417.

[202]

A. Carrato, A. Abad, B. Massuti, et al., “First-line panitumumab plus FOLFOX4 or FOLFIRI in colorectal cancer With multiple or unresectable liver metastases: A randomised, phase II trial (PLANET-TTD),” European Journal of Cancer 81 (2017): 191–202.

[203]

C. S. Karapetis, S. Khambata-Ford, D. J. Jonker, et al., “K-ras mutations and benefit From cetuximab in advanced colorectal cancer,” New England Journal of Medicine 359, no. 17 (2008): 1757–1765.

[204]

F. Di Nicolantonio, M. Martini, F. Molinari, et al., “Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer,” Journal of Clinical Oncology 26, no. 35 (2008): 5705–5712.

[205]

S. Siena, M. Di Bartolomeo, K. Raghav, et al., “Trastuzumab deruxtecan (DS-8201) in patients With HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): A multicentre, open-label, phase 2 trial,” The Lancet Oncology 22, no. 6 (2021): 779–789.

[206]

A. Sartore-Bianchi, L. Trusolino, C. Martino, et al., “Dual-targeted therapy With trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): A proof-of-concept, multicentre, open-label, phase 2 trial,” The Lancet Oncology 17, no. 6 (2016): 738–746.

[207]

F. Meric-Bernstam, H. Hurwitz, K. P. S. Raghav, et al., “Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): An updated report From a multicentre, open-label, phase 2a, multiple basket study,” The Lancet Oncology 20, no. 4 (2019): 518–530.

[208]

K. A. Klute, M. Rothe, E. Garrett-Mayer, et al., “Cobimetinib Plus Vemurafenib in Patients With Colorectal Cancer With BRAF Mutations: Results From the Targeted Agent and Profiling Utilization Registry (TAPUR) Study,” JCO Precision Oncology 6 (2022): e2200191.

[209]

E. Elez, S. Kopetz, J. Tabernero, et al., “SEAMARK: Phase II study of first-line encorafenib and cetuximab plus pembrolizumab for MSI-H/dMMR BRAFV600E-mutant mCRC,” Future Oncology 20, no. 11 (2024): 653–663.

[210]

S. Flis, A. Gnyszka, and K. Flis, “DNA methyltransferase inhibitors improve the effect of chemotherapeutic agents in SW48 and HT-29 colorectal cancer cells,” PLoS ONE 9, no. 3 (2014): e92305.

[211]

M. J. Overman, V. Morris, H. Moinova, et al., “Phase I/II study of azacitidine and capecitabine/oxaliplatin (CAPOX) in refractory CIMP-high metastatic colorectal cancer: Evaluation of circulating methylated vimentin,” Oncotarget 7, no. 41 (2016): 67495–67506.

[212]

N. S. Azad, A. El-Khoueiry, J. Yin, et al., “Combination epigenetic therapy in metastatic colorectal cancer (mCRC) With subcutaneous 5-azacitidine and entinostat: A phase 2 consortium/stand up 2 cancer study,” Oncotarget 8, no. 21 (2017): 35326–35338.

[213]

M. Buchwald, O. H. Krämer, and T. Heinzel, “HDACi–targets Beyond chromatin,” Cancer Letters 280, no. 2 (2009): 160–167.

[214]

S. Abou Najem, G. Khawaja, M. H. Hodroj, and S. Rizk, “Synergistic Effect of Epigenetic Inhibitors Decitabine and Suberoylanilide Hydroxamic Acid on Colorectal Cancer In vitro,” Current Molecular Pharmacology 12, no. 4 (2019): 281–300.

[215]

S. Tanaka, M. Hosokawa, A. Tatsumi, S. Asaumi, R. Imai, and K. I. Ogawara, “Improvement of resistance to oxaliplatin by vorinostat in human colorectal cancer cells Through inhibition of Nrf2 nuclear translocation,” Biochemical and Biophysical Research Communications 607 (2022): 9–14.

[216]

W. Fazzone, P. M. Wilson, M. J. Labonte, H. J. Lenz, and R. D. Ladner, “Histone deacetylase inhibitors suppress thymidylate synthase gene expression and synergize With the fluoropyrimidines in colon cancer cells,” International Journal of Cancer 125, no. 2 (2009): 463–473.

[217]

E. J. Lee, B. B. Lee, S. J. Kim, Y. D. Park, J. Park, and D. H. Kim, “Histone deacetylase inhibitor scriptaid induces cell cycle arrest and epigenetic change in colon cancer cells,” International Journal of Oncology 33, no. 4 (2008): 767–776.

[218]

Y. Shi, Y. Fu, X. Zhang, et al., “Romidepsin (FK228) regulates the expression of the immune checkpoint ligand PD-L1 and suppresses cellular immune functions in colon cancer,” Cancer Immunology, Immunotherapy 70, no. 1 (2021): 61–73.

[219]

J. W. Han, S. H. Ahn, S. H. Park, et al., “Apicidin, a histone deacetylase inhibitor, inhibits proliferation of tumor cells via induction of p21WAF1/Cip1 and gelsolin,” Cancer Research 60, no. 21 (2000): 6068–6074.

[220]

L. Liu, B. Chen, S. Qin, et al., “A novel histone deacetylase inhibitor Chidamide induces apoptosis of human colon cancer cells,” Biochemical and Biophysical Research Communications 392, no. 2 (2010): 190–195.

[221]

L. Liu, S. Qiu, Y. Liu, et al., “Chidamide and 5-flurouracil show a synergistic antitumor effect on human colon cancer xenografts in nude mice,” Neoplasma 63, no. 2 (2016): 193–200.

[222]

A. Saito, T. Yamashita, Y. Mariko, et al., “A synthetic inhibitor of histone deacetylase, MS-27-275, With marked in vivo antitumor activity Against human tumors,” PNAS 96, no. 8 (1999): 4592–4597.

[223]

A. J. Langlands, T. D. Carroll, Y. Chen, and I. Näthke, “Chir99021 and Valproic acid reduce the proliferative advantage of Apc mutant cells,” Cell Death & Disease 9, no. 3 (2018): 255.

[224]

J. R. Davie, “Inhibition of histone deacetylase activity by butyrate,” Journal of Nutrition 133, no. 7 Suppl (2003): 2485s–2493s.

[225]

R. M. Stilling, M. van de Wouw, G. Clarke, C. Stanton, T. G. Dinan, and J. F. Cryan, “The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis?,” Neurochemistry International 99 (2016): 110–132.

[226]

T. E. Wood, S. Dalili, C. D. Simpson, et al., “A novel inhibitor of glucose uptake sensitizes cells to FAS-induced cell death,” Molecular Cancer Therapeutics 7, no. 11 (2008): 3546–3555.

[227]

D. Kraus, J. Reckenbeil, N. Veit, et al., “Targeting glucose transport and the NAD pathway in tumor cells With STF-31: A re-evaluation,” Cellular Oncology (Dordr) 41, no. 5 (2018): 485–494.

[228]

X. Lei, K. Li, Y. Liu, et al., “Co-delivery nanocarriers targeting folate receptor and encapsulating 2-deoxyglucose and α-tocopheryl succinate enhance anti-tumor effect in vivo,” International Journal of Nanomedicine 12 (2017): 5701–5715.

[229]

W. Priebe, R. Zielinski, I. Fokt, et al., “EXTH-07. DESIGN AND EVALUATION OF WP1122, AN INHIBITOR OF GLYCOLYSIS WITH INCREASED CNS UPTAKE,” Neuro-oncol 20, no. suppl_6 (2018): vi86–vi86.

[230]

Y. Jia, Z. Ma, X. Liu, et al., “Metformin prevents DMH-induced colorectal cancer in diabetic rats by reversing the warburg effect,” Cancer Medicine 4, no. 11 (2015): 1730–1741.

[231]

J. Tong, G. Xie, J. He, J. Li, F. Pan, and H. Liang, “Synergistic antitumor effect of dichloroacetate in combination With 5-fluorouracil in colorectal cancer,” Journal of Biomedicine and Biotechnology 2011 (2011): 740564.

[232]

Y. Liang, D. Zhu, L. Zhu, et al., “Dichloroacetate Overcomes Oxaliplatin Chemoresistance in Colorectal Cancer Through the miR-543/PTEN/Akt/mTOR Pathway,” Journal of Cancer 10, no. 24 (2019): 6037–6047.

[233]

B. C. Xing, C. Wang, F. J. Ji, and X. B. Zhang, “Synergistically suppressive effects on colorectal cancer cells by combination of mTOR inhibitor and glycolysis inhibitor, Oxamate,” International Journal of Clinical and Experimental Pathology 11, no. 9 (2018): 4439–4445.

[234]

D. Krauß, O. Fari, and M. Sibilia, “Lipid Metabolism Interplay in CRC-An Update,” Metabolites 12, no. 3 (2022): 213.

[235]

A. Bleve, B. Durante, A. Sica, and F. M. Consonni, “Lipid Metabolism and Cancer Immunotherapy: Immunosuppressive Myeloid Cells at the Crossroad,” International Journal of Molecular Sciences 21, no. 16 (2020): 5854.

[236]

Y. Zaytseva, “Lipid Metabolism as a Targetable Metabolic Vulnerability in Colorectal Cancer,” Cancers (Basel) 13, no. 2 (2021): 301.

[237]

P. Mozolewska, K. Duzowska, A. Pakiet, A. Mika, and T. ŚledziŃski, “Inhibitors of Fatty Acid Synthesis and Oxidation as Potential Anticancer Agents in Colorectal Cancer Treatment,” Anticancer Research 40, no. 9 (2020): 4843–4856.

[238]

T. Lu, L. Sun, Z. Wang, Y. Zhang, Z. He, and C. Xu, “Fatty acid synthase enhances colorectal cancer cell proliferation and metastasis via regulating AMPK/mTOR pathway,” OncoTargets and Therapy 12 (2019): 3339–3347.

[239]

C. W. Fhu and A. Ali, “Fatty Acid Synthase: An Emerging Target in Cancer,” Molecules (Basel, Switzerland) 25, no. 17 (2020): 3935.

[240]

P. L. Huang, S. N. Zhu, S. L. Lu, Z. S. Dai, and Y. L. Jin, “Inhibitor of fatty acid synthase induced apoptosis in human colonic cancer cells,” World Journal of Gastroenterology 6, no. 2 (2000): 295–297.

[241]

S. Murata, K. Yanagisawa, K. Fukunaga, et al., “Fatty acid synthase inhibitor cerulenin suppresses liver metastasis of colon cancer in mice,” Cancer Science 101, no. 8 (2010): 1861–1865.

[242]

L. Chang, P. Wu, R. Senthilkumar, et al., “Loss of fatty acid synthase suppresses the malignant phenotype of colorectal cancer cells by Down-regulating energy metabolism and mTOR signaling pathway,” Journal of Cancer Research and Clinical Oncology 142, no. 1 (2016): 59–72.

[243]

J. Wils, “Adjuvant treatment of colon cancer: Past, present and future,” Journal of Chemotherapy 19, no. 2 (2007): 115–122.

[244]

R. Shiragami, S. Murata, C. Kosugi, et al., “Enhanced antitumor activity of cerulenin combined With oxaliplatin in human colon cancer cells,” International Journal of Oncology 43, no. 2 (2013): 431–438.

[245]

A. R. Rendina and D. Cheng, “Characterization of the inactivation of rat fatty acid synthase by C75: Inhibition of partial reactions and protection by substrates,” Biochemical Journal 388, no. Pt 3 (2005): 895–903.

[246]

Y. Tu, J. N. Thupari, E. K. Kim, et al., “C75 alters central and peripheral gene expression to reduce food intake and increase energy expenditure,” Endocrinology 146, no. 1 (2005): 486–493.

[247]

T. M. Loftus, D. E. Jaworsky, G. L. Frehywot, et al., “Reduced food intake and body weight in mice treated With fatty acid synthase inhibitors,” Science 288, no. 5475 (2000): 2379–2381.

[248]

H. Orita, J. Coulter, E. Tully, et al., “High levels of fatty acid synthase expression in esophageal cancers represent a potential target for therapy,” Cancer Biology & Therapy 10, no. 6 (2010): 549–554.

[249]

H. Y. Chuang, Y. F. Chang, and J. J. Hwang, “Antitumor effect of orlistat, a fatty acid synthase inhibitor, is via activation of caspase-3 on human colorectal carcinoma-bearing animal,” Biomedicine & Pharmacotherapy 65, no. 4 (2011): 286–292.

[250]

A. Czumaj, J. Zabielska, A. Pakiet, et al., “In Vivo Effectiveness of Orlistat in the Suppression of Human Colorectal Cancer Cell Proliferation,” Anticancer Research 39, no. 7 (2019): 3815–3822.

[251]

G. Falchook, J. Infante, H. T. Arkenau, et al., “First-in-human study of the safety, pharmacokinetics, and pharmacodynamics of first-in-class fatty acid synthase inhibitor TVB-2640 alone and With a taxane in advanced tumors,” EClinicalMedicine 34 (2021): 100797.

[252]

R. Ventura, K. Mordec, J. Waszczuk, et al., “Inhibition of de novo Palmitate Synthesis by Fatty Acid Synthase Induces Apoptosis in Tumor Cells by Remodeling Cell Membranes, Inhibiting Signaling Pathways, and Reprogramming Gene Expression,” EBioMedicine 2, no. 8 (2015): 808–824.

[253]

D. Y. Lim, Y. Jeong, A. L. Tyner, and J. H. Park, “Induction of cell cycle arrest and apoptosis in HT-29 human colon cancer cells by the dietary compound luteolin,” American Journal of Physiology Gastrointestinal and Liver Physiology 292, no. 1 (2007): G66–75.

[254]

Y. Schneider, F. Vincent, B. Duranton, et al., “Anti-proliferative effect of resveratrol, a natural component of grapes and wine, on human colonic cancer cells,” Cancer Letters 158, no. 1 (2000): 85–91.

[255]

L. Tessitore, A. Davit, I. Sarotto, and G. Caderni, “Resveratrol depresses the growth of colorectal aberrant crypt foci by affecting bax and p21(CIP) expression,” Carcinogenesis 21, no. 8 (2000): 1619–1622.

[256]

J. Vanamala, L. Reddivari, S. Radhakrishnan, and C. Tarver, “Resveratrol suppresses IGF-1 induced human colon cancer cell proliferation and elevates apoptosis via suppression of IGF-1R/Wnt and activation of p53 signaling pathways,” BMC Cancer 10 (2010): 238.

[257]

O. Bilen and C. M. Ballantyne, “Bempedoic Acid (ETC-1002): An Investigational Inhibitor of ATP Citrate Lyase,” Current Atherosclerosis Reports 18, no. 10 (2016): 61.

[258]

C. Qiao, W. Huang, J. Chen, et al., “IGF1-mediated HOXA13 overexpression promotes colorectal cancer metastasis Through upregulating ACLY and IGF1R,” Cell Death & Disease 12, no. 6 (2021): 564.

[259]

S. Nishizawa, H. Sumi, Y. Satoh, et al., “In vitro and in vivo antitumor activities of T-3764518, a novel and orally available small molecule stearoyl-CoA desaturase 1 inhibitor,” European Journal of Pharmacology 807 (2017): 21–31.

[260]

S. Wang, Y. Zhang, X. Yang, et al., “Betulinic acid arrests cell cycle at G2/M phase by up-regulating metallothionein 1G inhibiting proliferation of colon cancer cells,” Heliyon 10, no. 1 (2024): e23833.

[261]

L. Potze, S. di Franco, J. H. Kessler, G. Stassi, and J. P. Medema, “Betulinic Acid Kills Colon Cancer Stem Cells,” Current Stem Cell Research & Therapy 11, no. 5 (2016): 427–433.

[262]

I. R. Schlaepfer and M. Joshi, “CPT1A-mediated Fat Oxidation, Mechanisms, and Therapeutic Potential,” Endocrinology 161, no. 2 (2020): bqz046.

[263]

F. Hossain, A. A Al-Khami, D. Wyczechowska, et al., “Inhibition of Fatty Acid Oxidation Modulates Immunosuppressive Functions of Myeloid-Derived Suppressor Cells and Enhances Cancer Therapies,” Cancer Immunology Research 3, no. 11 (2015): 1236–1247.

[264]

E. Hernlund, L. S. Ihrlund, O. Khan, et al., “Potentiation of chemotherapeutic drugs by energy metabolism inhibitors 2-deoxyglucose and etomoxir,” International Journal of Cancer 123, no. 2 (2008): 476–483.

[265]

B. Dhakal, Y. Tomita, P. Drew, et al., “Perhexiline: Old Drug, New Tricks? A Summary of Its Anti-Cancer Effects,” Molecules (Basel, Switzerland) 28, no. 8 (2023): 3624.

[266]

Y. Wang, J. H. Lu, F. Wang, et al., “Inhibition of fatty acid catabolism augments the efficacy of oxaliplatin-based chemotherapy in gastrointestinal cancers,” Cancer Letters 473 (2020): 74–89.

[267]

S. Y. Jun, A. J. Brown, N. K. Chua, et al., “Reduction of Squalene Epoxidase by Cholesterol Accumulation Accelerates Colorectal Cancer Progression and Metastasis,” Gastroenterology 160, no. 4 (2021): 1194–1207. e28.

[268]

Y. Xiao, Q. Liu, N. Peng, et al., “Lovastatin Inhibits RhoA to Suppress Canonical Wnt/β-Catenin Signaling and Alternative Wnt-YAP/TAZ Signaling in Colon Cancer,” Cell Transplantation 31 (2022): 9636897221075749.

[269]

L. L. Kodach, R. J. Jacobs, P. W. Voorneveld, et al., “Statins augment the chemosensitivity of colorectal cancer cells inducing epigenetic reprogramming and reducing colorectal cancer cell ‘stemness’ via the bone morphogenetic protein pathway,” Gut 60, no. 11 (2011): 1544–1553.

[270]

R. C. Willis and J. E. Seegmiller, “The inhibition by 6-diazo-5-oxo-l-norleucine of glutamine catabolism of the cultured human lymphoblast,” Journal of Cellular Physiology 93, no. 3 (1977): 375–382.

[271]

M. I. Gross, S. D. Demo, J. B. Dennison, et al., “Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer,” Molecular Cancer Therapeutics 13, no. 4 (2014): 890–901.

[272]

Y. Li, S. Wu, Y. Zhao, et al., “Neutrophil extracellular traps induced by chemotherapy inhibit tumor growth in murine models of colorectal cancer,” Journal of Clinical Investigation 134, no. 5 (2024): e175031.

[273]

A. K. Holt, A. K. Najumudeen, T. J. Collard, et al., “Aspirin reprogrammes colorectal cancer cell metabolism and sensitises to glutaminase inhibition,” Cancer & Metabolism 11, no. 1 (2023): 18.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

183

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/