Letter to the Editor Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Dec 28, 2024; 30(48): 5221-5224
Published online Dec 28, 2024. doi: 10.3748/wjg.v30.i48.5221
Calculus bovis hijacks the tumor microenvironment in liver cancer cells in a multifaceted approach: A falling row of dominoes
Said G Farhat, Department of Internal Medicine, Division of Gastroenterology, Saint Georges Hospital University Medical Center, Beirut 3187, Beyrouth, Lebanon
Said G Farhat, Department of Gastroenterology, Dr. Sulaiman Al habib, Dubai 505005, Dubai, United Arab Emirates
Karam Karam, Department of Gastroenterology, University of Balamand, Beirut 3187, Beyrouth, Lebanon
ORCID number: Said G Farhat (0000-0002-8071-4681); Karam Karam (0009-0001-1914-320X).
Co-first authors: Said G Farhat and Karam Karam.
Author contributions: Farhat SG and Karam K contributed to conceptualization, data curation, drafting and writing original draft.
Conflict-of-interest statement: The authors declare that they have no conflict of interest.
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: Said G Farhat, MD, Chief Doctor, Department of Internal Medicine, Division of Gastroenterology, Saint Georges Hospital University Medical Center, Rmeil Street, Ashrafieh, Beirut 3187, Beyrouth, Lebanon. saidfarhat@hotmail.com
Received: October 3, 2024
Revised: October 25, 2024
Accepted: November 13, 2024
Published online: December 28, 2024
Processing time: 56 Days and 22.8 Hours

Abstract

Calculus bovis (C. bovis) is widely used in traditional Chinese medicine due to its anti-tumor effects. C. bovis shifts liver cancer tumor microenvironment towards regression by hindering tumor-associated macrophages polarization. Huang et al have demonstrated in their study that C. bovis inhibits M2-tumour-associated macrophages (TAM) polarization by halting the Wnt/β-catenin pathway. The mechanism of action by which C. bovis exerts its anti-tumor effects is multifaceted and includes network pharmacology, transcriptomics and molecular docking. In vitro assays demonstrated that C. bovis-containing serum inhibited M2-TAMs polarization in human hepatocellular carcinomas cells. C. bovis was found to have 22 active components of which 11 were detected in the bloodstream. The anti-neoplastic activity of C. bovis lies in suppressing M2-TAM polarization by modulation of the Wnt/B-catenin pathway. In vitro and in vivo experiments have shown that C. bovis suppresses M2-TAM polarization and halts the Wnt signaling pathway. The inhibitory effect of C. bovis on M2-TAM was reversed by SKL2001, a Wnt agonist, which highlights C. bovis’s selectivity and specificity. C. bovis inhibits M2-TAM polarization by modulating the Wnt/ β-catenin pathway, thus impeding liver cancer growth. Owing to the “cross-talk” between transforming growth factor-β (TGF-β) signaling pathways, this paper highlights the potential significance of C. bovis in controlling the tumor microenvironment not only through hindering the polarization of M2-TAMs via the Wnt signaling pathway, but also by downregulating TGF-β. Therefore, C. bovis serves as an igniter to fuel a cascade of signaling events that culminates in the regression of the tumor microenvironment by compromising oncogenesis and angiogenesis. TGF-β is also known for its pro-fibrotic properties. Therefore, C. bovis may play a pivotal role in treating liver fibrosis by downregulating TGF-β, thus hindering oncogenesis, angiogenesis and liver fibrosis. Hence, the “domino effect”.

Key Words: Calculus bovis; Wnt/β-catenin signaling pathway; M2-tumor-associated macrophages; Liver cancer; Transforming growth factor-β; Angiogenesis; Liver fibrosis

Core Tip: Calculus bovis (C. bovis) is an herb used in traditional Chinese medicine known for its various anti-inflammatory and anti-tumorigenic effects. C. bovis influences the tumor microenvironment by targeting immune-related pathways. Transcriptome sequencing revealed that C. bovis plays a crucial role in the regulation of M2-tumor-associated macrophages polarization and halting the Wnt/β-catenin pathway. This study provides a solid and promising evidence concerning potential drug therapy in the treatment of liver cancer. In this paper, we shed light on the inevitable “cross talk” between the Wnt and transforming growth factor-β signaling pathways, thus connecting angiogenesis, oncogenesis and liver fibrosis. C. bovis is deemed a priming molecule that sets the stage for a series of related and inter-connected events, such as angiogenesis, hepatocarcinogenesis and hepatic fibrosis. This process brings about the “domino effect” whereby one event uncovers another related event as a falling row of dominoes.



TO THE EDITOR

Liver cancer ranks 6th in prevalence and 4th in cancer-related mortality worldwide[1]. Liver cancer is malignant, rapidly progressive and is associated with poor prognosis with a 5-year survival rate of 3%[2]. Surgical resection, liver transplantation, interventional modalities, local ablation and targeted immunotherapies are the main treatment modalities for liver cancer[3]. These therapeutic options have their limitations as the survival rate remains low in patients undergoing treatment for liver cancer due to liver cancer cells resistance to treatment, tumor relapse, organ toxicity and metastasis[3]. This fact obviates the need for the advent of novel pharmacotherapies by gaining insights into the molecular pathogenesis of liver cancer. Traditional Chinese medicine is deemed to have multi-component and multi-target abilities as it influences Tumor microenvironment (TME) in liver cancer, thus offering a new perspective for the treatment of liver cancer[4]. Huang et al[5] have successfully elucidated the mechanism of action by which Calculus bovis(C. bovis) induces TME regression at the molecular level[5]. TME harbors a myriad of cells whereby macrophages play a pivotal role in controlling the microenvironment. Tumor-associated macrophages (TAMs), derived from circulating monocytes, can adopt either the M1 or M2 phenotypes. M1-TAM is a tumor-regressing phenotype, whereas M2-TAM is a tumor-promoting phenotype. Different cytokines and growth factors dictate the phenotypic switch of macrophages to an M1 or M2 phenotypes[5]. For instance, M1-TAMs are anti-tumor macrophages regulated by interferon-gamma and tumor necrosis factor-α, whereas M2-TAMs are tumorigenic macrophages regulated by nuclear factor kappa-B, interleukin-6/signal transducers and activators of transcription-3 and Wnt/β-catenin pathways[6]. Furthermore, the molecular pathways that control M2-TAM polarization foster proliferation, migration, invasion and angiogenesis[7]. Thus, reversing TAM polarization and halting the Wnt/β-catenin signaling molecular pathway provide promising venues for the treatment of liver cancer by shifting TME into regression.

Insights into the mechanism of action of C. bovis at the molecular level

C. bovis is a valuable herb used in traditional Chinese medicine due to its anti-tumor effects in various models[8]. The influence of C. bovis on TME remains elusive and an area of active research due to the heterogeneity of TME. Understanding the heterogeneity of TME lies in deciphering its composition and constituents. The study of Huang et al[5] opens new therapeutic venues as it revealed that C. bovis exerts its anti-neoplastic effect on M2-TAM polarization via the Wnt/β-catenin pathway. TAMs, key players in TME, exhibit phenotypic plasticity as they can either adopt a tumor-promoting phenotype (M2-TAM) or a tumor-regressing phenotype (M1-TAM). In light of this, C. bovis hinders M2-polarized TAM differentiation by modulating the Wnt/β-catenin molecular pathway[5]. In vitro and in vivo assays have demonstrated that C. bovis effectuates its anti-cancer activity through its active constituents, cellular targets and signaling pathways[5]. C. bovis plays a regulatory role in macrophage phenotypic plasticity and Wnt/β-catenin pathway within TME[5]. This study lays a foundation for developing C. bovis-derived anti-neoplastic therapeutic modalities for liver cancer. C. bovis mitigates the proliferative and migratory properties of liver cancer cells by hindering M2-TAM polarization via the Wnt signaling pathway. Modulating TME by shifting it into regression constitutes a new perspective for the treatment of liver cancer. The immunomodulatory and anti-inflammatory effects of C. bovis are well-documented in medical literature[9]. The mechanism of action of C. bovis involves the Wnt/β-catenin signaling pathway that is always involved in the pathogenesis of liver cancer[10]. C. bovis impedes tumor progression by altering M2-TAM polarization via Wnt pathway.

Cross-talk between signaling pathways, liver fibrosis, angiogenesis and future directions

Serum analysis revealed 11 bioactive compounds for C. bovis. Bilirubin is one of the bioactive compounds of C. bovis that is known for its anti-oxidant, anticancer and anti-inflammatory effects[11]. Another bioactive compounds derived from C. bovis are acid esters and bile acid-like components known for their enterohepatic circulation and anti-tumor effects[12]. Hence, C. bovis has a hepatoprotective profile in addition to its anti-tumorigenic profile. For instance, the Wnt pathway drives the upregulation of transforming growth factor-β (TGF-β), which connects inflammatory, fibrotic, angiogenic and oncogenic processes. Enhancing our understanding about the pharmacology of C. bovis can offer promising treatment options for liver fibrosis and liver cancer. Therefore, C. bovis’s regulatory effect on TGF-β needs to be evaluated in future studies to elucidate a potential therapeutic effect of C. bovis in liver fibrosis. TGF-β signaling induces cell plasticity in liver fibrosis and hepatocarcinogenesis[13]. TGF-β is considered a pro-fibrotic mediator through the activation of quiescent hepatic stellate cell (HSC) to a myofibroblast (MFB) phenotype[14]. MFBs potentiate extracellular matrix (ECM) accumulation, which drives fibrogenesis. Furthermore, TGF-β stimulates an epithelial-to-mesenchymal transition in hepatocytes, thus aggravating fibrogenesis[15]. TGF-β also has a pro-tumorigenic effect by potentiating the pro-migratory and invasive potential of hepatic tumor cells. TGF-β enhances tumor cell plasticity by conferring properties of migratory tumor initiating cells[16]. Thus, TGF-β is both pro-fibrotic and pro-tumorigenic. TGF-β signaling pathway “cross-talks” with other signaling pathways, such as the Wnt signaling pathway. C. bovis suppresses M2-TAM polarization by halting the Wnt pathway, which downregulates TGF-β. Thus, C. bovis is deemed to harbor not just anti-tumor properties, but also anti-fibrotic potential, which lays a new foundation in treating both liver fibrosis and hepatocarcinogenesis.

Kathuria and Singla[17] posited that the study of Huang et al[5] includes areas that require further investigations, such as C. bovis’s potential effect on angiogenesis[17]. Kathuria and Singla[17] stated that future studies are needed to understand the effect of C. bovis on angiogenesis and to gain deeper insights into the mechanism by which C. bovis potentially exerts its anti-angiogenic effect[17]. For instance, TME harbors a myriad of cells, such as HSC. TME is also an environment of constant ECM remodeling and altered vasculature. It has been well-documented that TGF-β promotes angiogenesis. By virtue of paracrine signaling from endothelial cells to mesenchymal cells, TGF-β drives vascular smooth muscle cell and pericyte differentiation during blood vessel coverage by smooth muscle cells[18]. Therefore, TGF-β regulates angiogenic process. C. bovis downregulates TGF-β and thus vasculature is altered and angiogenesis ceases. Hence, C. bovis indirectly halts angiogenesis by suppressing the Wnt signaling pathway, which in turn downregulates TGF-β and its pro-angiogenic effects. It becomes clear that C. bovis mitigates TME in a multifaceted pattern. C. bovis suppresses the polarization of M2-TAM via the Wnt signaling pathway which downregulates TFG-β. Lower expression of TGF-β hinders angiogenesis and engenders TME regression. In other words, the “cross-talk” between TGF-β signaling pathways brings about the “Domino effect” whereby one event, in this case C. bovis’s suppression of M2-TAM polarization via regulation of the Wnt pathway, sets off a series of related events, in this case downregulation of TGF-β and halting its resultant pro-fibrotic and pro-angiogenic potentials. This falling row of “dominoes” culminates into TEM regression.

CONCLUSION

This study uncovered the molecular pathway behind C. bovis’s anti-tumor effects in liver cancer cells. C. bovis suppresses the polarization of M2-TAM by halting the Wnt/β-catenin signaling pathway. The Wnt pathway upregulates TGF-β, a growth factor involved in liver fibrosis, angiogenesis and oncogenesis. A suppressed Wnt signaling pathway engenders the downregulation of TGF-β. This process counteracts the pro-tumorigenic, pro-angiogenic and pro-fibrotic effects of TGF-β. Thus, C. bovis deems not only anti-tumorigenic, but also anti-fibrotic and anti-angiogenic. This lays a new foundation regarding the potential therapeutic effects of C. bovis in patients with liver fibrosis. Owing to the “cross-talk” between the Wnt signaling pathway and TGF-β signaling pathway, future studies should be generated to further elucidate the anti-tumor effects of C. bovis at the molecular level and evaluating its potential therapeutic benefits in liver fibrosis.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Lebanon

Peer-review report’s classification

Scientific Quality: Grade C

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Bedouhene S S-Editor: Fan M L-Editor: A P-Editor: Zheng XM

References
1.  Peng J, Lü M, Peng Y, Tang X. Global incidence of primary liver cancer by etiology among children, adolescents, and young adults. J Hepatol. 2023;79:e92-e94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
2.  Laface C, Fedele P, Maselli FM, Ambrogio F, Foti C, Molinari P, Ammendola M, Lioce M, Ranieri G. Targeted Therapy for Hepatocellular Carcinoma: Old and New Opportunities. Cancers (Basel). 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 25]  [Reference Citation Analysis (0)]
3.  Muñoz-Martínez S, Iserte G, Sanduzzi-Zamparelli M, Llarch N, Reig M. Current pharmacological treatment of hepatocellular carcinoma. Curr Opin Pharmacol. 2021;60:141-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
4.  Shi J, Zhu L, Tang BY, Yang WQ, Xi SY, Zhang CL, Li PF, Wang YJ, Guo KH, Huang JR, Huang CR, Yu ZX, Yu BK, Zhang CF, Zhang YM. Regulatory effect of Yinchenhao decoction on bile acid metabolism to improve the inflammatory microenvironment of hepatocellular carcinoma in mice. J Nat Med. 2024;78:633-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Huang Z, Meng FY, Lu LZ, Guo QQ, Lv CJ, Tan NH, Deng Z, Chen JY, Zhang ZS, Zou B, Long HP, Zhou Q, Tian S, Mei S, Tian XF. Calculus bovis inhibits M2 tumor-associated macrophage polarization via Wnt/β-catenin pathway modulation to suppress liver cancer. World J Gastroenterol. 2024;30:3511-3533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 2]  [Reference Citation Analysis (5)]
6.  Yang Y, Ye YC, Chen Y, Zhao JL, Gao CC, Han H, Liu WC, Qin HY. Crosstalk between hepatic tumor cells and macrophages via Wnt/β-catenin signaling promotes M2-like macrophage polarization and reinforces tumor malignant behaviors. Cell Death Dis. 2018;9:793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 205]  [Article Influence: 34.2]  [Reference Citation Analysis (0)]
7.  Cronan MR, Beerman RW, Rosenberg AF, Saelens JW, Johnson MG, Oehlers SH, Sisk DM, Jurcic Smith KL, Medvitz NA, Miller SE, Trinh LA, Fraser SE, Madden JF, Turner J, Stout JE, Lee S, Tobin DM. Macrophage Epithelial Reprogramming Underlies Mycobacterial Granuloma Formation and Promotes Infection. Immunity. 2016;45:861-876.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 154]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
8.  Naramore S, Virojanapa A, Bell M, Jhaveri PN. Bezoar in a Pediatric Oncology Patient Treated with Coca-Cola. Case Rep Gastroenterol. 2015;9:227-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
9.  Miao TG, Nan YM. [Hepatocellular carcinoma immune microenvironment]. Zhonghua Gan Zang Bing Za Zhi. 2022;30:923-930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
10.  Wang W, Smits R, Hao H, He C. Wnt/β-Catenin Signaling in Liver Cancers. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 240]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
11.  Yu ZJ, Xu Y, Peng W, Liu YJ, Zhang JM, Li JS, Sun T, Wang P. Calculus bovis: A review of the traditional usages, origin, chemistry, pharmacological activities and toxicology. J Ethnopharmacol. 2020;254:112649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
12.  Sen B, Aggarwal S, Nath R, Sehgal R, Singh R, Agrawal K, Shashidhara AN, Rastogi A, Bajpai M, Pamecha V, Trehanpati N, Ramakrishna G. Secretome of senescent hepatoma cells modulate immune cell fate by macrophage polarization and neutrophil extracellular traps formation. Med Oncol. 2022;39:134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
13.  Kim KK, Sheppard D, Chapman HA. TGF-β1 Signaling and Tissue Fibrosis. Cold Spring Harb Perspect Biol. 2018;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 447]  [Cited by in F6Publishing: 434]  [Article Influence: 72.3]  [Reference Citation Analysis (0)]
14.  Dooley S, ten Dijke P. TGF-β in progression of liver disease. Cell Tissue Res. 2012;347:245-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 468]  [Cited by in F6Publishing: 546]  [Article Influence: 42.0]  [Reference Citation Analysis (1)]
15.  Dewidar B, Soukupova J, Fabregat I, Dooley S. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis: Updated. Curr Pathobiol Rep. 2015;3:291-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 6]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
16.  Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G, Fabregat I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 227]  [Article Influence: 37.8]  [Reference Citation Analysis (0)]
17.  Kathuria I, Singla B. Anti-tumor efficacy of Calculus bovis: Suppressing liver cancer by targeting tumor-associated macrophages. World J Gastroenterol. 2024;30:4249-4253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
18.  Guerrero PA, Mccarty JH.   TGF-β Activation and Signaling in Angiogenesis. In: Simionescu D, Simionescu A, editors. Physiologic and Pathologic Angiogenesis - Signaling Mechanisms and Targeted Therapy. London: IntechOpen, 2017.  [PubMed]  [DOI]  [Cited in This Article: ]