Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Mar 15, 2025; 17(3): 99673
Published online Mar 15, 2025. doi: 10.4251/wjgo.v17.i3.99673
Pharmacological role of Herba Patriniae and Coix seed in colorectal cancer
Yu-Xin Zhang, Hui Wan, Guan-Yue Shan, Yi-Ying Liu, Wen-Na Shi, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
Jun-Ya Cheng, Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130061, Jilin Province, China
Hai-Jun Li, Institute of Liver Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
ORCID number: Guan-Yue Shan (0000-0002-3380-9418); Hai-Jun Li (0000-0002-2515-3076).
Author contributions: Zhang YX, Wan H, Shan GY and Shi WN collected the information; Cheng JY and Liu YY drew and modified the illustrations; Zhang YX wrote the paper; Li HJ revised the paper; all of the authors read and approved the final version of the manuscript to be published.
Supported by National Natural Science Foundation of China, No. 81970529; and The Natural Science Foundation of Jilin Province, No. 20230508074RC and No. YDZJ202401218ZYTS.
Conflict-of-interest statement: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Hai-Jun Li, MD, PhD, Associate Professor, Institute of Liver Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun 130061, Jilin Province, China. hjli2012@jlu.edu.cn
Received: July 27, 2024
Revised: October 31, 2024
Accepted: December 25, 2024
Published online: March 15, 2025
Processing time: 201 Days and 21.6 Hours

Abstract

Colorectal cancer (CRC) is the most prevalent cancer globally, and its traditional treatment modalities commonly encompass radiation therapy, chemotherapy, surgery and the administration of cytotoxic drugs. Currently, novel chemotherapy drugs that combine traditional Chinese medicine (TCM) with herbal extracts exhibit superior comprehensive benefits. Herein, we delved into an article authored by Wang et al, focusing specifically on the pharmacological effects of "Herba Patriniae and Coix seed (HC)" and their targeted mechanisms in combating CRC. From the perspective of TCM philosophy, damp-heat stagnation and toxicity are the cardinal pathogenic factors underlying CRC. HC, renowned for their abilities to antipyretic and enhance diuresis, have demonstrated promising efficacy in preliminary studies for the treatment of CRC. These findings offer potential insights in favor of fostering anti-cancer medications.

Key Words: Colorectal cancer; Herba Patriniae; Coix seed; Antitumor; Chinese medicine

Core Tip: Colorectal cancer (CRC) can be treated with novel chemotherapeutic agents combining traditional Chinese medicine (TCM) and herbal extracts in addition to traditional treatments including radiotherapy, chemotherapy, surgery and cytotoxic drugs. The TCMs Herba Patriniae and Coix seed can act on the signaling pathways of CRC, such as phosphoinositide 3 kinase/Akt/mammalian target of rapamycin, p53, transforming growth factor-β/SMAD, NFkappaB and Wnt/β-catenin, to show better therapeutic effect, and their future treatments and applications in CRC are very valuable and promising.



INTRODUCTION

Colorectal cancer (CRC) is the third most common cancer globally, with an incidence rate of 9.6%, and it ranks second in terms of mortality, affecting 9.3% of cancer-related deaths worldwide[1]. Among the conventional treatments, including surgery, radiotherapy, chemotherapy, immunotherapy and targeted therapies, there are still many significant impediments to the side effects of these methods[2].

In recent years, traditional Chinese medicine (TCM) has shown significant promise in the treatment of CRC. TCM not only helps alleviate clinical symptoms and boosts the immune response in patients but also plays a crucial role in preventing tumor recurrence and metastasis. A notable example is the combination of Herba Patriniae and Coix seed (HC), which are valued in TCM for their heat-clearing and phlegm-reducing properties. Research suggests that the active components in these herbs can inhibit the apoptosis of CRC cells by modulating the phosphoinositide 3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), P53, NFkappaB (NF-κB), mitogen activated protein kinase, and transforming growth factor-β (TGF-β)/SMAD pathways[3,4]. Additionally, they have been shown to reverse epithelial-mesenchymal transition (EMT) by inhibiting the Wnt/β-catenin signaling pathway, thus reducing the spread and migration of CRC cells. The objective of this article is to investigate the pharmacological effects of HC and their targeted mechanisms in CRC, drawing on an article by Wang et al[5] in the World Journal of Gastrointestinal Oncology. This study comprehensively reviews the scientific literature on the pathogenesis of CRC and the medicinal value of HC as well as its application in the treatment of colorectal cancer. Additionally, it elaborates on the major molecular mechanisms implicated in these processes. Finally, it discusses the future development directions and prospects of HC in the treatment of CRC.

PATHOGENESIS OF CRC

CRC arises from a complex interplay of genetic and environmental factors[6]. Genetically, the majority of CRC cases are sporadic, with approximately 80% lacking a clear familial predisposition[7]. Early detection and screening programs have been shown to be effective in preventing more than 90% of CRC cases[8]. In Western countries, it is suggested that up to 71% of CRC cases could be attributed to modifiable lifestyle factors[9]. While the incidence and mortality rates of CRC are showing signs of stabilization and decline in highly developed countries, this is largely attributed to extensive colonoscopy practices, national screening initiatives, and dietary changes[10]. Conversely, in many developing countries, the incidence of CRC is on the rise, reflecting the need for improved prevention strategies and healthcare infrastructure[11].

Environmental factors significantly contribute to the development of CRC, encompassing a range of elements such as obesity, height, diet, alcohol consumption, physical activity, and smoking[12]. Notably, dietary factors, including red meat, processed meat, animal-derived saturated fats and alcohol have been identified as influential in CRC development[13]. International Agency for Research on Cancer classifies processed meat consumption as a group 1 carcinogen[14]. Studies in rodent models have shown that heme iron in red meat can trigger lipid oxidation and the formation of 4-hydroxynonenal, which is associated with precancerous conditions[15]. In vitro, components such as animal fats, proteins from red meat, N-nitroso compounds, heterocyclic amines, and heme iron are implicated in CRC induction, with heme iron specifically enhancing cellular inflammation and permeability, leading to genetic damage[16,17]. Adherence to healthy dietary patterns, such as the Mediterranean, vegetarian, and semi-vegetarian diets, which include whole grains, full-fat dairy products, and calcium, has been shown to decrease CRC morbidity[18]. Ethanol, when consumed, is metabolized into acetaldehyde and subsequently oxidized into acetate, a known carcinogen that increases the risk of cancer. Moreover, ethanol's impact on the DNA methylation of the COLCA1 and COLCA2 genes is associated with an elevated risk of CRC[19]. Regular and sufficient physical activity has been demonstrated to reduce the incidence of CRC and improve survival rates among those diagnosed. Chen et al[20] utilized a polygenic risk score to analyze the relationship between physical activity and CRC risk, finding a consistent inverse association with CRC risk, regardless of genetic predisposition. Smoking is a primary factor in the etiology of early-stage CRC, and its long-term use is linked to the progression of advanced CRC[21]. Cigarette contains over 7000 harmful chemicals, including at least 70 known carcinogens that can affect nearly every biological system in the human body[22]. Toxic chemicals in cigarettes, such as nitrosamines, heterocyclic amines, benzene, and polycyclic aromatic hydrocarbons, have the potential to induce cancer in the colon and rectum, either through direct exposure or systemic circulation to the colorectal mucosa[23].

In terms of deeper pathogenesis, cancer stem cells (CSCs) constitute a small subset of cells within the tumor that share characteristics with embryonic stem cells (ESCs)[24]. The majority of CRCs are believed to arise from CSCs within the colonic epithelium, which accumulate genetic and epigenetic alterations over time. These cells possess the ability for self-renewal and differentiation, and they can suppress tumor suppressor genes while activating oncogenes. This leads to abnormal growth patterns and a slow rate of cellular turnover, making them resistant to therapies that target rapidly dividing cells[25]. The genesis of CSCs is attributed to two primary mechanisms: (1) First, the accumulation of oncogenic mutations in normal adult stem cells or ESCs can lead to a deregulation of cell proliferation, resulting in uncontrolled growth[26,27]; and (2) Second, differentiated cancer cells may revert to a stem-cell-like state through a process known as dedifferentiation. Biomarkers such as c-Myc, STAT3, aldehyde dehydrogenase, ABCG2, and CD133 are strongly associated with the presence of cancer. Furthermore, specific markers including CD44, LGR5, and EpCAM have been identified as particularly relevant in the context of CRC[28].

CRC can be classified into four distinct consensus molecular subtypes (CMS), which are delineated by their gene expression profiles. CMS1 is characterized by microsatellite instability (MSI) and immune system activation, CMS2 is considered the typical subtype, CMS3 is defined by its metabolic dysregulation, and CMS4 is distinguished by its mesenchymal features. The genetic and molecular pathways associated with each subtype are interrelated[29].

Tumors with MSI and those that are metabolic in nature are predominantly found in the right-sided colon. Specifically, CMS1 is associated with MSI, BRAF mutations, an increase in immune cell infiltration, and the upregulation of checkpoint inhibitors. In contrast, CMS2 is defined by chromosomal instability, the activation of the Wnt/MYC signaling pathway, and a decrease in immune cell presence. The CMS3 subtype is frequently associated with KRAS mutations, while CMS4 is marked by a mesenchymal phenotype, activation of the TGF-β pathway, and increased matrix deposition along with heightened immune cell infiltration[30].

NEW CRC TREATMENT DRUGS: "HERBA PATRINIAE AND COIX SEED"

Currently, the main approaches to treating CRC include surgery, radiotherapy, chemotherapy, immunotherapy and targeted therapy. Despite these methods, challenges such as persistent proliferation signaling, resistance to chemotherapy, postoperative complications, drug side effects, and high rates of metastasis and recurrence pose significant obstacles to effective CRC treatment[2]. These factors contribute to the high mortality rate associated with CRC.

Recent advancements in interdisciplinary treatment strategies have achieved notable breakthroughs over traditional methods. Specifically, molecular targeting and immunotherapy, which focus on tumor-associated proteins and the regulation of signaling pathways involved in tumor growth and progression, have shown promise in enhancing treatment efficacy and revolutionizing conventional treatment approaches[31].

TCM with its extensive history, has increasingly become a vital resource in the development of anti-CRC drugs[32]. TCM employs a multi-component approach, targeting multiple protein and signaling pathways, which can significantly mitigate the adverse effects of radiation and chemotherapy. This approach not only enhances immune function but also improves patients' overall quality of life, life expectancy, and reduces the incidence of tumor recurrence and metastasis[33]. Sijunzi Decoction, has been shown to effectively alleviate clinical symptoms, improve patients' quality of life, extend survival, and reduce the risk of tumor recurrence and metastasis[34].

The "Compendium of Materia Medica" recognizes Coicis Semen as a traditional Chinese medicinal material with both pharmacological value and culinary uses[3,35]. Historically esteemed in TCM, Coicis Semen is frequently combined with other herbs to treat various conditions. For instance, the 'Qian Jin Yao Fang' describes the Phragmites Stem Soup, a formulation that pairs Coicis Semen with Phragmites Stem and Winter Melon Seeds to address respiratory issues such as lung abscess, lobar pneumonia, and bronchitis. This blend aims to enhance urine excretion and regulate spleen function, thereby alleviating diarrhea. Furthermore, the 'Wen Bing Diao Bian' by Wu JT details the Sanzi Decoction, which includes Coicis Semen, Amomum Villosum Lour, and Almond among other ingredients, specifically formulated for damp-heat syndrome treatment[36].

Herba Patriniae, another traditional Chinese medicinal herb, is celebrated for its purifying and antioxidant properties and is used in treatments for diarrhea, acute hepatitis, ulcerative colitis, cancer, and other diseases[37]. First documented in the 'Shennong Herbal Classic', Herba Patriniae has been valued for its medicinal properties and applied to treat a range of conditions. Modern pharmacological studies have identified the plant's chemical constituents as having antioxidant, anti-inflammatory, and sedative effects[38].

In terms of modern clinical treatment, HC known for their diuretic, purgative, hemostatic, and anti-inflammatory properties[4,39]. This combination has demonstrated efficacy in improving experimental enteritis in mice by reducing the secretion of inflammatory cytokines and oxidative stress[40]. The Coicis Semen Polysaccharide component of HC is notable for its ability to activate the PI3K/Akt signaling pathway, which enhances the population of bacteria that produce short-chain fatty acids. This mechanism contributes to the lowering of blood sugar levels in mouse models of type 2 diabetes[41]. Coicis Semen has established itself as an anti-cancer agent, particularly effective against CRC[42]. It possesses the capacity to reduce the proliferation, invasion, and migration of various cancer cells. Furthermore, Coix has been shown to disrupt NF-κB signaling, which enhances the anti-tumor effects of gemcitabine in pancreatic cancer treatment[43]. Coicis Semen Oil (CSO) has also emerged as an agent with potential anti-cancer activity against triple-negative breast cancer by modulating the miR-205/S1PR1 axis[44]. The compound Kanglaite, extracted from Coicis Semen, has demonstrated clinical utility as a broad-spectrum anti-tumor agent[45].

Herba Patriniae, on its own, has shown promise in inducing apoptosis in CRC cells and suppressing tumor growth and angiogenesis, presenting itself as an innovative therapeutic option for CRC treatment[46].

MODE OF ACTION OF HERBA PATRINIAE AND COIX SEED IN CRC

The signal pathway related to CRC is a major direction of CRC treatment, and exploring the mechanism of the CRC-related signal pathway also helps to further clarify the drug targets for anti-CRC. EMT process is widely acknowledged to be involved in cancer invasion and metastasis, and is regulated by numerous transcription factors[47]. The process of EMT in cancer cells can be triggered by multiple intrinsic signaling pathways. These pathways include the PI3K/Akt/mTOR, TGF-β/SMAD and Wnt/β-catenin. We are mainly interested in HC in regulating the signaling pathways driven by PI3K/Akt/mTOR, p53, TGF-β/SMAD, NF-κB and Wnt/β-catenin (Figure 1).

Figure 1
Figure 1 A graphical illustration showcasing the immunomodulatory mechanisms of Herba Patriniae and Coix seed on signaling pathways implicated in the onset and progression of cancer. APC: Anaphase-promoting-complex; BAX: B cell lymphoma/leukemia-2-associated X protein; BCL-2: B cell lymphoma/leukemia-2; CK1: Casein kinase; DR5: Death receptor 5; DVL: Dishevelled; EGFR: Epidermal growth factor receptor; ERK: Extracellular signal-regulated protein kinases; IL: Interleukin; IKB: IkappaB; IKK: KappaB kinase; LRP: Lung resistance protein; MDM2: Murine double minute 2; MEK: Mitogen-activated protein kinase; MMPs: Matrix metalloproteinases; MTORC1: Mammalian target of rapamycin complex 1; MyD88: Myeloid differentiation factor 88; NF-κB: NFkappaB; PDK1: Phosphoinositide-dependent kinase 1; PI3K: Phosphoinositide 3 kinase; RAS: Rat sarcoma; RTKs: Receptor tyrosine kinases; TBK1: TANK-binding kinase 1; TGF: Transforming growth factor; TGFBR2: Transforming growth factor-beta1 receptor 2; TLR: Toll like receptor; TNF-α: Tumour necrosis factor-alpha; TRAF2: Tumour necrosis factor receptor associated factor 2; TRAIL: Tumor necrosis factor-related apoptosis-inducing ligand; TRADD: Tumour necrosis factor receptor-associated death domain; TSC1/2: Tuberous sclerosis complex 1/2.

The PI3K/Akt/mTOR signaling pathway is a crucial intracellular signaling mechanism, primarily functioning as a downstream effector of receptor tyrosine kinases and G protein-coupled receptors. It has been implicated in various cancer types, including CRC[48,49]. Research by Ni et al[50] has demonstrated that CSO can target the PI3K/Akt signaling pathway, leading to the arrest of HT-29 cells in the G2 phase and the induction of apoptosis. Similarly, Yang et al[51] reported that Coix lacryma oil influences mitochondrial function by modulating the PTEN/PI3K/Akt pathway, thereby promoting apoptosis in human pancreatic cancer cells. The aqueous extract of Herba Patriniae exert anticancer effects by activating kinase signals including p38, extracellular signal-regulated protein kinases 1 and 2, c-Jun N-terminal kinase, mitogen extracellular kinase 1 and 2, and mitogen-and stress-activated protein kinase 1 and 2[52]. An extract from Patrinia heterophylla, a species categorized under Herba Patriniae, has exhibited significant anti-CRC potential by markedly reducing the mRNA and protein expression levels of epidermal growth factor receptor, PI3K, and Akt in HCT116 cells[53].

The p53 protein plays a pivotal role in various cellular processes, including the regulation of cell cycle arrest, apoptosis, DNA repair, and senescence by binding to specific DNA response elements[54]. It is widely recognized as a crucial gene that protects against cancer. However, in the context of cancer, the p53 function is often compromised due to inactivation or mutation[55]. Coicis Semen Extract has been shown to induce apoptosis through multiple mechanisms, such as the inhibition of Bcl-2, upregulation of p53, and modulation of cleaved poly (adenosine diphosphate-ribose) polymerase, as well as cleaved caspase-3 and caspase-8[56]. Kang Lai Te (KLT) injection, an antitumor drug derived from Coix, primarily exerts its effects by inhibiting tumor cell proliferation. It achieves this by reducing cell mitosis through the blockade of the G2/M phase of the cell cycle and by activating pro-apoptotic factors, thereby promoting cell apoptosis[57]. Similarly, Herba Patriniae and its constituent isovitexin have demonstrated the ability to effectively combat CRC by inducing apoptosis and arresting the cell cycle through the activation of p53[52].

NF-κB family is a prevalent class of transcription factors that play a significant role in cellular processes. Activation of NF-κB is known to promote cell growth, decelerate cell death, and is implicated in disease progression, including genetic instability and the potential activation of resistance genes, which may contribute to the development of rectal cancer[58]. A substantial body of research has indicated that HC possess the ability to inhibit NF-κB function. For instance, Hu et al[59] discovered that Coixol, a component of Coix seed, can suppress the activation of the mitogen-activated protein kinase, NF-κB pathway, and the NOD-like receptor thermal protein domain associated protein 3 inflammasome. Similarly, Chen et al[60] reported that KLT, an active extract derived from Coix lacryma-jobi seeds, enhances the effectiveness of cisplatin in hepatocellular carcinoma treatment by inhibiting chemokine-like factor 1-mediated NF-κB signaling and modulating transporter-associated drug efflux. Furthermore, ginsenosides present in Herba Patriniae have been shown to exert anti-invasive effects on CRC by dampening the NF-κB signaling pathway and regulating the expression of EMT markers, such as E-cadherin and N-cadherin[61].

The TGF-β/SMAD signaling cascade is a critical regulatory mechanism for EMT in colon cancer. It impacts a broad spectrum of human diseases by modulating cellular processes such as proliferation, apoptosis, differentiation, lineage commitment, motility, and the maintenance of stem cell homeostasis[62]. The aqueous extract of Herba Patriniae mediates its therapeutic effects through the regulation of key signaling pathways, specifically the TGF-βR1-SMAD2/3-E-cadherin and FAK-RhoA-cofilin axes. This modulation influences the immune cell composition and cytokine levels within the tumor microenvironment, as well as the gut microbiota, which are essential factors in cancer progression[63].

The Wnt/β-catenin signaling pathway is a critical mediator in the progression of human CRC and is considered a significant target for therapeutic intervention[64]. This pathway involves a complex network of proteins, including glycogen synthase kinase 3β (GSK3β), casein kinase 1 (CK1), the tumor suppressor Axin, and adenomatous polyposis coli (APC), which together form a destruction complex. Axin acts as the structural framework of this complex, enabling the interaction of β-catenin with APC, CK1, and GSK3β[65]. Upon activation, β-catenin translocates to the nucleus, where it interacts with transcription factors to drive the transcription of essential target genes such as c-Myc, Cyclin D1, and matrix metalloproteinases[66]. Plant-derived medicinal foods, with Coix seed as a primary component, have shown a robust ability to inhibit tumor growth. This is achieved by disrupting the Wnt/β-catenin signaling cascade and reversing EMT process[67].

CONCLUSION

HC, widely recognized as plant-based medicines across many Asian countries, have attracted considerable interest due to their notable anticancer properties. A plethora of studies has attested to the efficacious tumor suppressive and immune regulatory capabilities of these botanical agents, which can counteract numerous oncogenic signal molecules and pathways. To date, the inhibitory impact of HC on signaling pathways associated with CRC has been highly encouraging, with the targeting of the PI3K/Akt/mTOR pathway showing particular promise and value. Despite these advances, research into their application in CRC cell lines and the disease's clinical context remains relatively limited.

Further investigation is warranted to delineate the precise mechanisms of action and the specific signaling pathways modulated by these compounds. Utilizing CRC cell lines as a screening platform can facilitate this evaluation. Based on these findings, HC along with their isolated compounds, opens up promising directions and offers valuable insights for the development of innovative anticancer therapeutics. This exploration can deepen our understanding of the pharmacological value and therapeutic potential of these natural agents in treating CRC. In summary, HC present themselves as candidates for novel combination chemotherapy agents in the management of CRC.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Janyakhantikul S S-Editor: Luo ML L-Editor: A P-Editor: Zhao YQ

References
1.  Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in RCA: 1262]  [Article Influence: 1262.0]  [Reference Citation Analysis (0)]
2.  Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022;12:31-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 664]  [Cited by in RCA: 4166]  [Article Influence: 1388.7]  [Reference Citation Analysis (0)]
3.  Zhou Q, Yu R, Liu T, Li Y, Zhong J, Zhang T, Liu Z, Hu Y. Coix Seed Diet Ameliorates Immune Function Disorders in Experimental Colitis Mice. Nutrients. 2021;14:123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
4.  Li J, Zhou F, Shang L, Liu N, Liu Y, Zhang M, Wang S, Yang S. Integrated network pharmacology and experimental verification to investigate the mechanisms of YYFZBJS against colorectal cancer via CDK1/PI3K/Akt signaling. Front Oncol. 2022;12:961653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
5.  Wang CL, Yang BW, Wang XY, Chen X, Li WD, Zhai HY, Wu Y, Cui MY, Wu JH, Meng QH, Zhang N. Targeting colorectal cancer with Herba Patriniae and Coix seed: Network pharmacology, molecular docking, and in vitro validation. World J Gastrointest Oncol. 2024;16:3539-3558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
6.  Modest DP, Pant S, Sartore-Bianchi A. Treatment sequencing in metastatic colorectal cancer. Eur J Cancer. 2019;109:70-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in RCA: 211]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
7.  Young PE, Womeldorph CM. Colonoscopy for colorectal cancer screening. J Cancer. 2013;4:217-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in RCA: 63]  [Article Influence: 5.3]  [Reference Citation Analysis (1)]
8.  Winawer SJ, Fletcher RH, Miller L, Godlee F, Stolar MH, Mulrow CD, Woolf SH, Glick SN, Ganiats TG, Bond JH, Rosen L, Zapka JG, Olsen SJ, Giardiello FM, Sisk JE, Van Antwerp R, Brown-Davis C, Marciniak DA, Mayer RJ. Colorectal cancer screening: clinical guidelines and rationale. Gastroenterology. 1997;112:594-642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1353]  [Cited by in RCA: 1225]  [Article Influence: 43.8]  [Reference Citation Analysis (0)]
9.  Shaw E, Farris MS, Stone CR, Derksen JWG, Johnson R, Hilsden RJ, Friedenreich CM, Brenner DR. Effects of physical activity on colorectal cancer risk among family history and body mass index subgroups: a systematic review and meta-analysis. BMC Cancer. 2018;18:71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in RCA: 43]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
10.  Clinton SK, Giovannucci EL, Hursting SD. The World Cancer Research Fund/American Institute for Cancer Research Third Expert Report on Diet, Nutrition, Physical Activity, and Cancer: Impact and Future Directions. J Nutr. 2020;150:663-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in RCA: 451]  [Article Influence: 90.2]  [Reference Citation Analysis (0)]
11.  Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009;22:191-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1215]  [Cited by in RCA: 1375]  [Article Influence: 98.2]  [Reference Citation Analysis (0)]
12.  World Cancer Research Fund  Diet, Nutrition, Physical Activity and Colorectal Cancer. 2018.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Platz EA, Willett WC, Colditz GA, Rimm EB, Spiegelman D, Giovannucci E. Proportion of colon cancer risk that might be preventable in a cohort of middle-aged US men. Cancer Causes Control. 2000;11:579-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in RCA: 193]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
14.  Diallo A, Deschasaux M, Latino-Martel P, Hercberg S, Galan P, Fassier P, Allès B, Guéraud F, Pierre FH, Touvier M. Red and processed meat intake and cancer risk: Results from the prospective NutriNet-Santé cohort study. Int J Cancer. 2018;142:230-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in RCA: 75]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
15.  Marnett LJ. Oxyradicals and DNA damage. Carcinogenesis. 2000;21:361-370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1282]  [Cited by in RCA: 1223]  [Article Influence: 48.9]  [Reference Citation Analysis (0)]
16.  Bastide N, Morois S, Cadeau C, Kangas S, Serafini M, Gusto G, Dossus L, Pierre FH, Clavel-Chapelon F, Boutron-Ruault MC. Heme Iron Intake, Dietary Antioxidant Capacity, and Risk of Colorectal Adenomas in a Large Cohort Study of French Women. Cancer Epidemiol Biomarkers Prev. 2016;25:640-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
17.  Martin OCB, Olier M, Ellero-Simatos S, Naud N, Dupuy J, Huc L, Taché S, Graillot V, Levêque M, Bézirard V, Héliès-Toussaint C, Estrada FBY, Tondereau V, Lippi Y, Naylies C, Peyriga L, Canlet C, Davila AM, Blachier F, Ferrier L, Boutet-Robinet E, Guéraud F, Théodorou V, Pierre FHF. Haem iron reshapes colonic luminal environment: impact on mucosal homeostasis and microbiome through aldehyde formation. Microbiome. 2019;7:72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
18.  Veettil SK, Wong TY, Loo YS, Playdon MC, Lai NM, Giovannucci EL, Chaiyakunapruk N. Role of Diet in Colorectal Cancer Incidence: Umbrella Review of Meta-analyses of Prospective Observational Studies. JAMA Netw Open. 2021;4:e2037341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in RCA: 124]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
19.  Zhou X, Wang L, Xiao J, Sun J, Yu L, Zhang H, Meng X, Yuan S, Timofeeva M, Law PJ, Houlston RS, Ding K, Dunlop MG, Theodoratou E, Li X. Alcohol consumption, DNA methylation and colorectal cancer risk: Results from pooled cohort studies and Mendelian randomization analysis. Int J Cancer. 2022;151:83-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in RCA: 33]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
20.  Chen X, Guo F, Chang-Claude J, Hoffmeister M, Brenner H. Physical activity, polygenic risk score, and colorectal cancer risk. Cancer Med. 2023;12:4655-4666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
21.  Botteri E, Iodice S, Bagnardi V, Raimondi S, Lowenfels AB, Maisonneuve P. Smoking and colorectal cancer: a meta-analysis. JAMA. 2008;300:2765-2778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 510]  [Cited by in RCA: 552]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
22.  Drope J, Schluger NW.   The Tobacco Atlas. 6th ed. Atlanta: American Cancer Society, 2018.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Cheng J, Chen Y, Wang X, Wang J, Yan Z, Gong G, Li G, Li C. Meta-analysis of prospective cohort studies of cigarette smoking and the incidence of colon and rectal cancers. Eur J Cancer Prev. 2015;24:6-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in RCA: 59]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
24.  Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther. 2024;9:170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
25.  Ricci-Vitiani L, Fabrizi E, Palio E, De Maria R. Colon cancer stem cells. J Mol Med (Berl). 2009;87:1097-1104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in RCA: 157]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
26.  Mondal K, Posa MK, Shenoy RP, Roychoudhury S. KRAS Mutation Subtypes and Their Association with Other Driver Mutations in Oncogenic Pathways. Cells. 2024;13:1221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Cao J, Zhao M, Liu J, Zhang X, Pei Y, Wang J, Yang X, Shen B, Zhang J. RACK1 Promotes Self-Renewal and Chemoresistance of Cancer Stem Cells in Human Hepatocellular Carcinoma through Stabilizing Nanog. Theranostics 20199:811-828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in RCA: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
28.  Munro MJ, Wickremesekera SK, Peng L, Tan ST, Itinteang T. Cancer stem cells in colorectal cancer: a review. J Clin Pathol. 2018;71:110-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in RCA: 203]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
29.  Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P, Bot BM, Morris JS, Simon IM, Gerster S, Fessler E, De Sousa E Melo F, Missiaglia E, Ramay H, Barras D, Homicsko K, Maru D, Manyam GC, Broom B, Boige V, Perez-Villamil B, Laderas T, Salazar R, Gray JW, Hanahan D, Tabernero J, Bernards R, Friend SH, Laurent-Puig P, Medema JP, Sadanandam A, Wessels L, Delorenzi M, Kopetz S, Vermeulen L, Tejpar S. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350-1356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3408]  [Cited by in RCA: 3361]  [Article Influence: 336.1]  [Reference Citation Analysis (0)]
30.  Timar J, Kashofer K. Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metastasis Rev. 2020;39:1029-1038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in RCA: 174]  [Article Influence: 34.8]  [Reference Citation Analysis (0)]
31.  Jensen M, Yilmaz M, Pedersen B. Everyday life with long-term chemotherapy induced peripheral neuropathy among patient in adjuvant treatment for colorectal cancer: A multi methods study. Ann Oncol. 2019;30:v843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
32.  Kong MY, Li LY, Lou YM, Chi HY, Wu JJ. Chinese herbal medicines for prevention and treatment of colorectal cancer: From molecular mechanisms to potential clinical applications. J Integr Med. 2020;18:369-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
33.  Wei X, Leng X, Li G, Wang R, Chi L, Sun D. Advances in research on the effectiveness and mechanism of Traditional Chinese Medicine formulas for colitis-associated colorectal cancer. Front Pharmacol. 2023;14:1120672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
34.  Wu B, Xuan ZR. Progress in research on applying Sijunzi Decoction in treating digestive malignant tumor. Chin J Integr Med. 2007;13:156-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
35.  Pan X, Shen Q, Zhang C, Zhang X, Li Y, Chang Z, Pang B. Coicis Semen for the treatment of malignant tumors of the female reproductive system: A review of traditional Chinese medicinal uses, phytochemistry, pharmacokinetics, and pharmacodynamics. Front Pharmacol 202314:1129874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
36.  Li H, Peng L, Yin F, Fang J, Cai L, Zhang C, Xiang Z, Zhao Y, Zhang S, Sheng H, Wang D, Zhang X, Liang Z. Research on Coix seed as a food and medicinal resource, it's chemical components and their pharmacological activities: A review. J Ethnopharmacol. 2024;319:117309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
37.  Gong L, Zou W, Zheng K, Shi B, Liu M. The Herba Patriniae (Caprifoliaceae): A review on traditional uses, phytochemistry, pharmacology and quality control. J Ethnopharmacol. 2021;265:113264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
38.  Zuo F, Wei ZC, Tang C, Wang WQ, Tong D, Meng XL, Zhang Y. [Medication rules for prescriptions containing Pterocephali Herba based on data mining]. Zhongguo Zhong Yao Za Zhi. 2017;42:3213-3218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
39.  39 Luo X, Li H, Jiang D, Meng J, Zhang F, Xu Q, Chen X, Liu C, Yang Y. Analysis of Fungi on Coix (Coix lacryma-jobi) Seed and the Effect of Its Aqueous Extract on the Growth of Aspergillus flavus. J Food Prot 201982:1775-1782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
40.  40 Sui H, Zhang L, Gu K, Chai N, Ji Q, Zhou L, Wang Y, Ren J, Yang L, Zhang B, Hu J, Li Q. YYFZBJS ameliorates colorectal cancer progression in Apc(Min/+) mice by remodeling gut microbiota and inhibiting regulatory T-cell generation. Cell Commun Signal 2020;18:113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 68]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
41.  Xia T, Liu CS, Hu YN, Luo ZY, Chen FL, Yuan LX, Tan XM. Coix seed polysaccharides alleviate type 2 diabetes mellitus via gut microbiota-derived short-chain fatty acids activation of IGF1/PI3K/AKT signaling. Food Res Int. 2021;150:110717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 64]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
42.  Kuo CC, Chen HH, Chiang W. Adlay ( yì yĭ; "soft-shelled job's tears"; the seeds of Coix lachryma-jobi L. var. ma-yuen Stapf) is a Potential Cancer Chemopreventive Agent toward Multistage Carcinogenesis Processes. J Tradit Complement Med. 2012;2:267-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in RCA: 36]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
43.  Qian Y, Yang B, Xiong Y, Gu M. Coix seed emulsion synergistically enhances the antitumor activity of gemcitabine in pancreatic cancer through abrogation of NF-κB signaling. Oncol Rep. 2016;36:1517-1525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
44.  Fang T, Jiang YX, Chen L, Huang L, Tian XH, Zhou YD, Nagle DG, Zhang DD. Coix Seed Oil Exerts an Anti-Triple-Negative Breast Cancer Effect by Disrupting miR-205/S1PR1 Axis. Front Pharmacol. 2020;11:529962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
45.  Hou D, Yang L, Xiong J, Xiong L. Efficacy and safety of Kanglaite injection for gastric cancer: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 2020;99:e21619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
46.  Liu L, Shen A, Chen Y, Wei L, Lin J, Sferra TJ, Hong Z, Peng J. Patrinia scabiosaefolia induces mitochondrial-dependent apoptosis in a mouse model of colorectal cancer. Oncol Rep. 2013;30:897-903.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
47.  Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. 2019;20:69-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1356]  [Cited by in RCA: 2331]  [Article Influence: 388.5]  [Reference Citation Analysis (0)]
48.  Duan S, Huang W, Liu X, Liu X, Chen N, Xu Q, Hu Y, Song W, Zhou J. IMPDH2 promotes colorectal cancer progression through activation of the PI3K/AKT/mTOR and PI3K/AKT/FOXO1 signaling pathways. J Exp Clin Cancer Res. 2018;37:304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in RCA: 115]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
49.  Moafian Z, Maghrouni A, Soltani A, Hashemy SI. Cross-talk between non-coding RNAs and PI3K/AKT/mTOR pathway in colorectal cancer. Mol Biol Rep. 2021;48:4797-4811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
50.  Ni C, Li B, Ding Y, Wu Y, Wang Q, Wang J, Cheng J. Anti-Cancer Properties of Coix Seed Oil against HT-29 Colon Cells through Regulation of the PI3K/AKT Signaling Pathway. Foods. 2021;10:2833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
51.  Yang J, Liu Y, Lu S, Sun X, Yin Y, Wang K, Liu S. Coix seed oil regulates mitochondrial functional damage to induce apoptosis of human pancreatic cancer cells via the PTEN/PI3K/AKT signaling pathway. Mol Biol Rep. 2022;49:5897-5909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Reference Citation Analysis (0)]
52.  Li J, Shang L, Zhou F, Wang S, Liu N, Zhou M, Lin Q, Zhang M, Cai Y, Chen G, Yang S. Herba Patriniae and its component Isovitexin show anti-colorectal cancer effects by inducing apoptosis and cell-cycle arrest via p53 activation. Biomed Pharmacother. 2023;168:115690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
53.  Yang H, Cheung MK, Yue GG, Leung PC, Wong CK, Lau CB. Integrated Network Pharmacology Analysis and In Vitro Validation Revealed the Potential Active Components and Underlying Mechanistic Pathways of Herba Patriniae in Colorectal Cancer. Molecules. 2021;26:6032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
54.  Hafner A, Bulyk ML, Jambhekar A, Lahav G. The multiple mechanisms that regulate p53 activity and cell fate. Nat Rev Mol Cell Biol. 2019;20:199-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 427]  [Cited by in RCA: 452]  [Article Influence: 75.3]  [Reference Citation Analysis (0)]
55.  Di Y, Jing X, Hu K, Wen X, Ye L, Zhang X, Qin J, Ye J, Lin R, Wang Z, He W. The c-MYC-WDR43 signalling axis promotes chemoresistance and tumour growth in colorectal cancer by inhibiting p53 activity. Drug Resist Updat. 2023;66:100909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
56.  Sabapathy K, Lane DP. Therapeutic targeting of p53: all mutants are equal, but some mutants are more equal than others. Nat Rev Clin Oncol. 2018;15:13-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in RCA: 226]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
57.  Lu Y, Li CS, Dong Q. Chinese herb related molecules of cancer-cell-apoptosis: a minireview of progress between Kanglaite injection and related genes. J Exp Clin Cancer Res. 2008;27:31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in RCA: 58]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
58.  Taniguchi K, Karin M. NF-κB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18:309-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1132]  [Cited by in RCA: 1783]  [Article Influence: 254.7]  [Reference Citation Analysis (0)]
59.  Hu Y, Zhou Q, Liu T, Liu Z. Coixol Suppresses NF-κB, MAPK Pathways and NLRP3 Inflammasome Activation in Lipopolysaccharide-Induced RAW 264.7 Cells. Molecules. 2020;25:894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
60.  Chen C, Ai QD, Wei YH. Kanglaite enhances the efficacy of cisplatin in suppression of hepatocellular carcinoma via inhibiting CKLF1 mediated NF-κB pathway and regulating transporter mediated drug efflux. J Ethnopharmacol. 2021;264:113388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
61.  Xia L, Zhang B, Yan Q, Ruan S. Effects of saponins of patrinia villosa against invasion and metastasis in colorectal cancer cell through NF-κB signaling pathway and EMT. Biochem Biophys Res Commun. 2018;503:2152-2159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
62.  Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol. 2016;12:325-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1512]  [Cited by in RCA: 2348]  [Article Influence: 260.9]  [Reference Citation Analysis (0)]
63.  Yang H, Yue GG, Yuen KK, Gao S, Leung PC, Wong CK, Lau CB. Mechanistic insights into the anti-tumor and anti-metastatic effects of Patrinia villosa aqueous extract in colon cancer via modulation of TGF-βR1-smad2/3-E-cadherin and FAK-RhoA-cofilin pathways. Phytomedicine. 2023;117:154900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
64.  Koveitypour Z, Panahi F, Vakilian M, Peymani M, Seyed Forootan F, Nasr Esfahani MH, Ghaedi K. Signaling pathways involved in colorectal cancer progression. Cell Biosci. 2019;9:97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in RCA: 223]  [Article Influence: 37.2]  [Reference Citation Analysis (0)]
65.  Disoma C, Zhou Y, Li S, Peng J, Xia Z. Wnt/β-catenin signaling in colorectal cancer: Is therapeutic targeting even possible? Biochimie. 2022;195:39-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
66.  Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, Zhou Z, Shu G, Yin G. Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther. 2022;7:3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in RCA: 899]  [Article Influence: 299.7]  [Reference Citation Analysis (0)]
67.  Chen X, Yue W, Tian L, Li N, Chen Y, Zhang L, Chen J. A plant-based medicinal food inhibits the growth of human gastric carcinoma by reversing epithelial-mesenchymal transition via the canonical Wnt/β-catenin signaling pathway. BMC Complement Med Ther. 2021;21:137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]