Next Article in Journal
Novel Multiparametric Magnetic Resonance Imaging-Based Deep Learning and Clinical Parameter Integration for the Prediction of Long-Term Biochemical Recurrence-Free Survival in Prostate Cancer after Radical Prostatectomy
Next Article in Special Issue
Controlled Plasma Membrane Delivery of FGFR1 and Modulation of Signaling by a Novel Regulated Anterograde RTK Transport Pathway
Previous Article in Journal
Externally Applied Electromagnetic Fields and Hyperthermia Irreversibly Damage Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review of Herbal Resources Inducing Anti-Liver Metastasis Effects in Gastrointestinal Tumors via Modulation of Tumor Microenvironments in Animal Models

1
Liver and Immunology Research Center, Collage of Korean Medicine, Daejeon University, Daejeon 35235, Republic of Korea
2
East-West Cancer Center, Cheonan Korean Medicine Hospital, Daejeon University, Cheonan 31099, Republic of Korea
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(13), 3415; https://doi.org/10.3390/cancers15133415
Submission received: 22 April 2023 / Revised: 22 June 2023 / Accepted: 27 June 2023 / Published: 29 June 2023

Abstract

:

Simple Summary

Based on the importance of the tumor microenvironment and liver environment in liver metastases, we conducted a systematic review to discover herbal candidates inhibiting the liver metastasis of tumors originating from the digestive system. We produced a list of potent herbs such as Curcuma longa Linn and single compounds such as curcumin and tanshinone II-A, along with their underlying mechanisms. The list of herbal agents and their mechanisms produced in this study is helpful for the development of anti-liver metastasis drugs in the future via modulation of liver environments.

Abstract

Liver metastases remain a major obstacle for the management of all types of tumors arising from digestive organs, and the tumor microenvironment has been regarded as an important factor in metastasis. To discover herbal candidates inhibiting the liver metastasis of tumors originating from the digestive system via the modulation of the tumor microenvironment and liver environment, we searched three representative public databases and conducted a systematic review. A total of 21 studies that employed experimental models for pancreatic (9), colon (8), and stomach cancers (4) were selected. The herbal agents included single-herb extracts (5), single compounds (12), and multiherbal decoctions (4). Curcuma longa Linn was most frequently studied for its anti-colon–liver metastatic effects, and its possible mechanisms involved the modulation of tumor microenvironment components such as vascular endothelial cells and immunity in both tumor tissues and circulating cells. The list of herbal agents and their mechanisms produced in this study is helpful for the development of anti-liver metastasis drugs in the future.

1. Introduction

Tumors in the digestive system, including the stomach, colon, rectum, and pancreas, accounted for approximately one-fifth of both new cases of cancer and cancer-related deaths in 2020 worldwide [1]. The 5-year survival rate of these gastrointestinal (GI) cancers has recently significantly increased; however, metastasis to secondary organs is a crucial event shortening the survival period [2,3]. Due to the presence of the portal vein, which brings a substantial blood flow, the liver is the most predominant site of metastasis in patients with malignancies of GI organs and occurs in almost 50~80% of tumors of the digestive tract at the late stage [4,5]. The liver is a central organ that maintains body homeostasis; therefore, liver metastasis reduces the survival rate of cancer patients. For example, patients with liver metastasis survive for only approximately one-fifth of the period of patients without liver metastasis in both colorectal and gastric cancer [6,7].
Despite advances in tumor treatments such as surgical techniques, radiotherapy, targeted therapy, and immunology therapy, the survival rates of digestive-system cancer patients with distant metastasis remain poor compared with other cancer types [8]. On the other hand, researchers have discovered that tumor metastasis is affected not only by metastasis-favoring genetic alterations of cancer cells but also by changes in the tumor microenvironment (TME) [9,10]. The TME is the location in which cancer cells communicate with adjacent stromal cells, making tumors more prone to progression and metastatic behavior [11,12]. Contrary to the genetic aberrations predominantly responsible for tumor progression, the TME plays a key role in modulating the aggressiveness, motility, dissemination, and colonization of cancer cells in distal tissues/organs [13,14]. For example, the invasion of colorectal cancer cells into liver tissue can be accelerated under metastasis-favorable microenvironments, including hepatic steatosis, inflammation, or fibrotic circumstances [15,16].
There are experiments showing the potential of targeting the TME to control tumor growth as well as the metastatic risk [17,18]. As pharmaceutical resources, many herbal plants such as Panax ginseng C.A. Mayer and Coptis chinensis F. have shown anti-tumor or beneficial effects in preclinical and clinical studies [19,20]. Some researchers have also reported the anti-metastatic properties of herbal remedies, especially in cancers of the GI tract, along with the effects on the TME [21,22]. These findings indicate the potential of herbal resources to inhibit the liver metastasis of GI cancers, especially given their effects on the TME.
To help develop herb-derived agents to protect against liver metastasis, we systematically searched for studies of herbal candidates inhibiting the liver metastasis of digestive organ tumors by modulating the TME.

2. Materials and Methods

2.1. Literature Search

In this study, we systematically surveyed herbal candidates that showed effects on the liver metastasis of GI-system tumors via the modulation of the TME. Relevant published studies were identified by searching PubMed, Embase, and Cochrane from each database’s inception to December 2021. To search for GI-system tumor metastasis-related articles, we searched for ‘neoplasm metastasis’ OR ‘tumor metastasis’ OR ‘cancer metastasis’ OR ‘metastasis’ OR ‘metast* in the title, abstract, or keywords. In addition, we used ‘herbal medicine’ OR ‘natural’ OR ‘plant’ OR ‘phytogenic’ OR ‘botanical’ OR ‘herb*’ OR ‘phyto*’ OR ‘botanic*’. In order to not to miss results, we manually searched for all cancer metastasis-related articles instead of for any specific organ name or abbreviations. Both free-text searches and MeSH-term searches for titles, abstracts, and keywords were employed. The database searches were supplemented with additional studies from the references of all retrieved relevant studies. The search was restricted to publications in the English language for the collection of the qualified data.

2.2. Review and Selection of Relevant Articles

At the time as title and abstract screening, we used the following inclusion and exclusion criteria to select relevant articles. We included human or animal-based studies that focused on the anti-liver metastatic effects of herbal resources, mostly medicinal plants, herbal decoctions, herb-derived compounds, and animal-derived resources. We excluded the following: (i) studies that were not human-derived or animal-based; (ii) studies that included resource(s) already approved for cancer treatment in the National Comprehensive Cancer Network (NCCN) guidelines, such as paclitaxel and irinotecan; (iii) diagnostic method studies; (iv) review articles; (v) studies of metastases in organs other than the liver, such as lung metastases; (vi) retracted articles; and (vii) studies described in other languages. In the full-text screening, we also excluded (i) studies in which the metastatic site was not the liver and (ii) studies in which the described mechanisms were not related to the TME and were only related to the cytotoxic effect of targeting cancer cells (Figure 1).

2.3. Analysis of Major Mechanisms Related to the Effects on Metastasis by Targeting the TME

We carefully read the text of the articles and extracted the anti-metastatic mechanisms given by the authors, such as modulation of the extracellular matrix, angiogenesis, endothelial cells, or immunity. In addition, we performed a search for each herbal candidate’s effects on liver environments using hepatopathology terms such as fibrosis, inflammation, injury, and steatosis, which are well-known factors promoting liver metastasis.

3. Results

3.1. Characteristics of the Selected Studies

Ultimately, 21 studies were included in our analysis. Regarding the origin organs of the cancers, the pancreas was the most frequent (9 studies), followed by the colon (8 studies, including 1 xenograft), the stomach (4 studies), and 1 primary tumor developed using a Helicobacter felis infection [3]. A total of 11 studies (57.1%) performed an orthotopic implantation of cell lines in the pancreas, colon, or stomach, while others performed splenic, subcutaneous, intraperitoneal, intravenous, or intrahepatic injections. A total of 12 studies employed an oral administration of herbal candidates, while 7 studies employed intraperitoneal and 2 studies employed intravenous injections (Table 1).
Overall, 15 studies (71.4%) evaluated the efficacy of drugs by comparing the number of metastatic nodules in the liver with those in the control group. Similarly, comparing the size of the liver metastatic area or luciferase activity was also used in some studies. Seven studies compared the effect of their herbal candidates with that of other approved anti-cancer drugs, including FOLFOX (5-FU, fluorouracil, and oxaliplatin), 5-FU, cisplatin, gemcitabine, mithramycin, or capecitabine (Table 1).

3.2. Characteristics of the Herbal Candidates

Among the 21 studies, 11 studies researched the effects of 9 herb-derived single compounds, 4 studies researched the effects of 3 single-herb extracts, 4 studies researched the effects of 3 multiherbal decoctions, and 2 studies used an animal-derived resource (1 single compound and 1 water extract). Also included were 3 multiherbal decoctions; Qing Yi Hua Ji (consisting of 7 herbs) [4,5], Juzen-taiho-to (10 herbs) [6], and Pien Tze Huang (consisting of 1 herb and 3 materials from animals) [7]. Regarding medicinal plant-derived resources, Curcuma longa L. (C. longa, including curcumin) was studied in 3 papers [3,8,9], while emodin from Rheum palmatum L. [10,11], Venenum bufonis (including cinobufacini) [12,13,14], and honokiol from Magnolia acuminate L. [14,15] were assessed in 2 studies (Table 2).

3.3. Anti-Liver Metastatic Effects on the TME

Regarding the effects on preventing liver metastasis by modulating the TME, 3 studies investigated the TME using metastasized hepatic tissue [7,11,12], while others used the organ/tissues in which the tumor developed (16 studies) or blood (2 studies) (Table 2). In the enrolled studies, the main mechanisms of the herbal candidates could be categorized as effects on cell adhesion or trafficking molecules, angiogenesis, the extracellular matrix, immunity, and inflammatory components.
Eight single compounds, including curcumin, emodin, cinobufacini, and honokiol, increased E-cadherin with/without suppression of the N-cadherin expression in transplanted tumor organs/tissues and/or cells in the circulation [11,13,15,16,17,18,19,20]. The acetate extract of Celastrus orbiculatus T. and Pien tze huang also had similar effects [7,21]. Water or ethanol extracts of Curcuma longa L., Venenum bufonis, and Qing Yi Hua Ji regulated angiogenesis via the key cytokine vascular endothelial growth factor (VEGF) in metastasized liver or rectum tissues and cancer cells transplanted into the pancreas [4,8,9,12]. Five agents, including emodin and tanshinone II-A, showed anti-liver metastatic effects via the modulation of matrix metalloproteinases (MMPs) with/without effects on the tissue inhibitors of metalloproteinases (TIMPs) [10,12,13,16,19,22]. Two compounds (honokiol and antartina) and two decoctions (Qing Yi Hua Ji and Juzen-taiho-to) activated tumor immune components, including macrophages and/or dendritic cells [5,6,14,23], while some agents (emodin, honokiol, antartina, Qing Yi Hua Ji, and Pien tze huang) showed anti-inflammatory effects via the suppression of proinflammatory molecules such as interleukin-6 (IL-6) or nuclear factor-κB (NF-κB) [23] in transplanted tumor tissues, including the colon, pancreas, and peritoneal cavity [5,7,10,18,21]. Further information is summarized in Table 2 and Figure 2.

3.4. Effect of Herbal Candidates on the Liver Environment

We additionally investigated the pharmacological effects of these herbal resources on the hepatic microenvironment in 3 liver metastasis-favoring pathologic conditions: hepatic steatosis, inflammation, and fibrosis. As shown in Table 2, 9 of the 15 candidates previously reported beneficial effects against at least one of the three pathologic conditions. Four compounds (curcumin, emodin, honokiol, and tanshinone II-A) and two decoctions (Juzen-taiho-to and Pien tze huang) previously showed beneficial effects on three pathologic states [26,27,28,29,37,38,41,42]. Deoxyelephantopin showed positive effects against liver inflammation [40], while extracts of both Annona muricata L. and Celastrus orbiculatus T. showed effects on liver steatosis and inflammation [30,33,34,39]. Further information is summarized in Table 2 and Figure 2.

4. Discussion

Medicinal herbs are attractive sources for anti-tumor drug development, and several agents such as paclitaxel, vinblastine, and vincristine are successfully being used owing to their anti-cancer activities [35]. Recently, these natural resources have gained attention not only for cancer prevention and treatment but also for reductions in cancer recurrence and metastasis [36,43]. Given the clinical impact of cancer metastasis, this study systematically reviewed herbal resources with anti-metastatic properties, especially those with effects on the liver metastasis of GI-tract tumors via modulation of the TME.
A total of 21 studies investigated pharmaceutical effects on the liver metastasis of tumors from 3 organs, including the pancreas, colon, and stomach, using 15 types of resources. Four phytochemicals (emodin, honokiol, deoxyelephantopin, and NGD16) and two herbal extracts (Elephantopus scaber L. and Annona muricata L.), including one decoction (Qing Yi Hua Ji), showed anti-liver metastatic effects in four orthotopic implantations and four splenic injection models [5,10,12,14,17,20]. Liver metastasis is a common feature of pancreatic cancer; more than 50% of patients are diagnosed with liver metastases, which is an important poor prognostic factor [44]. However, one clinical study showed that administration with Chinese herbal medicine (CHM) significantly increased the 1-year survival rate in patients with liver-metastasized pancreatic cancer compared with patients who did not receive CHM (21.9% versus 4.8%) [45]. Among the herbal candidates mentioned above, emodin is a main compound derived from Rheum palmatum L. that has been traditionally used as a representative herb to treat inflammation-related diseases, including GI-system tumors [46]. Many studies have shown the direct anti-cancer activity of emodin in not only pancreatic tumors but also breast cancer cells [47,48]. From our data, emodin modulated the TME in both tumor-bearing pancreas tissue (suppressing NF-κB and MMP-9) and metastasis-bearing hepatic tissue (increasing E-cadherin) in pancreas and splenic injection models, respectively [10,11].
Regarding gastric and colorectal tumors, the wide and early application of endoscopic screening has led to a reduction in metastasis; however, the death rates due to these malignancies are still very high, ranking second and third, respectively, after lung cancer, especially in many Asian countries [49]. Liver metastasis has the poorest prognostic factor, and it occurs during diagnosis, treatment, or surveillance in approximately 50% of both gastric and colorectal cancer patients [50,51]. Bigelovin, tanshinone II-A, antartina, cinobufacini, curcumin, honokiol, and peperomin E showed anti-metastatic effects via the modulation of MMP-2/9, TIMPs, and N-cadherin under various experimental models of transplanted colon/gastric tumor tissues and cytolytic T cells in blood [3,13,15,16,18,19,23]. The TME-associated activities of these compounds indicate their potential. For example, tanshinone II-A was previously shown to prevent the metastasis of colon tumor cells by the modulation of migration and angiogenesis [52,53]. The ethanol extract of C. longa L. and its main compound curcumin have also shown effects on colon/stomach liver metastasis by inhibiting angiogenesis and CXCL12/CXCR4 expression [3,8,9]. C. longa L. and curcumin are well-known inhibitors of angiogenesis in a wide variety of tumor cells [54]. The CXCL12/CXCR4 axis plays a crucial role in tumor–stromal cell interactions, leading to progression-related behaviors such as angiogenesis, migration, and metastasis [55].
We reviewed herb-derived agents with actions on the TME. The TME, consisting of cellular and structural components, plays a vital role in the complex and multistep metastatic cascade; thus, the TME has become a target of anti-cancer and anti-metastatic agents [56]. The TME encompasses the surrounding stromal cells, including tumor-infiltrating immune cells, cancer-associated fibroblasts (CAFs), adipose cells, blood endothelial cells, and pericytes as well as the extracellular matrix (ECM), the basement membrane (BM), and cancer-cell secreted growth factors and cytokines such as IL-6 and TGF-β [57]. The herb-derived candidates were found to modulate many of these TME components, mainly inside transplanted tumor tissues, as described above. In our data, two studies (antatina and Juzen-taiho-to) revealed the activation of immune cells such monocytes or cytolytic cells in the bloodstream [6,23] and three studies (emodin, Venenum bufonis, and Qing Yi Hua Ji) showed effects on the hepatic TME of metastases from pancreatic cancer [4,11,12]. Cancer cells encounter immune cells, platelets, and multiple cytokines in the blood microenvironment and need to escape from immune surveillance to successfully metastasize into secondary regions [58]. Juzen-Taiho-To showed an effect on the liver metastasis of colon cancer via the regulation of blood immune cells [6].
On the other hand, the state of the liver itself in addition to the primary tumor (stomach, pancreas, or colorectal tumors) also notably affects the end step of metastasis. Alterations in the liver microenvironment such as inflammation, steatosis, and fibrotic changes accelerate liver metastasis [59]. We previously found that the alcohol consumption-induced acceleration of colon–liver metastasis is related to the upregulation of intercellular adhesion molecule 1 (ICAM1) and mild inflammation in hepatic tissue [60]. Furthermore, many studies have found asymptomatic dormant cancer cells in the liver that disseminated from the primary lesion, likely the stomach or colon; under some conditions, the hepatic microenvironment could reactivate cancer cells to proliferate to generate metastasis [61]. One group showed the prevention of colorectal liver metastasis via the administration of interleukin-12 (IL-12), carried in chitosan into the portal vein [62]. IL-12 is one of the most effective inducers of anti-tumor immunity via enhancing the cellular immunity of natural killer cytolytic T cells and interferon (IFN)-γ production [63]. In one cohort study of patients who underwent radical surgery for colon cancer and subsequent routine radiotherapy and chemotherapy, the administration of herbal decoctions showed significant anti-liver metastatic effects, with 26.2 ± 4.5 versus 14.2 ± 4.2 months for the average time to relapse and metastasis, respectively [64].
In our study, 9 candidates presented protective or therapeutic effects in pathologically altered hepatic environmental conditions such as inflammation, steatosis, or fibrosis (Table 2). Based on our data, curcumin, emodin, and honokiol are the most promising candidates for inhibiting liver metastasis. These compounds are known to exert pharmacological activities in the TME and liver environment. For example, curcumin inhibits tumor-promoting M2 macrophages [65] and attenuates hepatic inflammation, steatosis, and fibrosis by controlling Kupffer cells and hepatic stellate cells [66]. Both emodin and honokiol have also shown effects on hepatosteatosis, inflammation, and fibrosis in preclinical studies [26,27], along with other effects on TME components such as tumor-specific immune cells [32,67].
The data have some limitations, including the use of only animal studies (no clinical data exist yet) and a relatively small number of articles. It is worth noting that our data collection was limited to English language publications for the collection of the qualified data.

5. Conclusions

In this study, we reviewed the recent research findings on herb-derived resources with effects on liver metastasis. Our results provide a basis for scientists working to develop drugs to prevent metastasis via modulation of the TME, especially for GI tumor–liver metastasis.

Author Contributions

S.-K.K. was responsible for the literature search, data collection, and drafted the manuscript. N.-H.L. contributed to the discussion and full revision process. C.-G.S. was responsible for supervision of the study and contributed to writing the manuscript. All authors read and approved the final report. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a National Research Foundation of Korea (NRF) grant, funded by the Ministry of Science, ICT & Future Planning (NRF-2018R1A6A1A03025221, NRF 2019R1A2C2088201).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare that there are no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Lv, Y.; Patel, N.; Zhang, H.-J. The progress of non-alcoholic fatty liver disease as the risk of liver metastasis in colorectal cancer. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 1169–1180. [Google Scholar] [CrossRef]
  3. Gu, X.; Zhang, Q.; Zhang, W.; Zhu, L. Curcumin inhibits liver metastasis of gastric cancer through reducing circulating tumor cells. Aging 2019, 11, 1501. [Google Scholar] [CrossRef]
  4. Yin, J.-H.; Shi, W.-D.; Zhu, X.-Y.; Chen, Z.; Liu, L.-M. Qingyihuaji formula inhibits progress of liver metastases from advanced pancreatic cancer xenograft by targeting to decrease expression of Cyr61 and VEGF. Integr. Cancer Ther. 2012, 11, 37–47. [Google Scholar] [CrossRef] [Green Version]
  5. Zhang, J.; Wang, P.; Ouyang, H.; Yin, J.; Liu, A.; Ma, C.; Liu, L. Targeting cancer-related inflammation: Chinese herbal medicine inhibits epithelial-to-mesenchymal transition in pancreatic cancer. PLoS ONE 2013, 8, e70334. [Google Scholar] [CrossRef] [Green Version]
  6. Ohnishi, Y.; Fujii, H.; Hayakawa, Y.; Sakukawa, R.; Yamaura, T.; Sakamoto, T.; Tsukada, K.; Fujimaki, M.; Nunome, S.; Komatsu, Y. Oral administration of a Kampo (Japanese herbal) medicine Juzen-taiho-to inhibits liver metastasis of Colon 26-L5 carcinoma cells. Jpn. J. Cancer Res. 1998, 89, 206–213. [Google Scholar] [CrossRef]
  7. Lin, W.; Zhuang, Q.; Zheng, L.; Cao, Z.; Shen, A.; Li, Q.; Fu, C.; Feng, J.; Peng, J. Pien Tze Huang inhibits liver metastasis by targeting TGF-β signaling in an orthotopic model of colorectal cancer. Oncol. Rep. 2015, 33, 1922–1928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Li, M.; Yue, G.G.-L.; Luo, L.; Tsui, S.K.-W.; Fung, K.-P.; Ng, S.S.-M.; Lau, C.B.-S. Turmeric Is therapeutic in vivo on patient-derived colorectal cancer xenografts: Inhibition of growth, metastasis, and tumor recurrence. Front. Oncol. 2021, 10, 574827. [Google Scholar] [CrossRef]
  9. Li, M.; Yue, G.G.-L.; Tsui, S.K.-W.; Fung, K.-P.; Bik-San Lau, C. Turmeric extract, with absorbable curcumin, has potent anti-metastatic effect in vitro and in vivo. Phytomedicine 2018, 46, 131–141. [Google Scholar] [CrossRef]
  10. Liu, A.; Chen, H.; Wei, W.; Ye, S.; Liao, W.; Gong, J.; Jiang, Z.; Wang, L.; Lin, S. Antiproliferative and antimetastatic effects of emodin on human pancreatic cancer. Oncol. Rep. 2011, 26, 81–89. [Google Scholar]
  11. Li, N.; Wang, C.; Zhang, P.; You, S. Emodin inhibits pancreatic cancer EMT and invasion by up-regulating microRNA-1271. Mol. Med. Rep. 2018, 18, 3366–3374. [Google Scholar] [CrossRef]
  12. Yin, J.-H.; Zhu, X.-Y.; Shi, W.-D.; Liu, L.-M. Huachansu injection inhibits metastasis of pancreatic cancer in mice model of human tumor xenograft. BMC Complement. Altern. Med. 2014, 14, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wang, J.; Cai, H.; Liu, Q.; Xia, Y.; Xing, L.; Zuo, Q.; Zhang, Y.; Chen, C.; Xu, K.; Yin, P. Cinobufacini inhibits colon cancer invasion and metastasis via suppressing Wnt/β-catenin signaling pathway and EMT. Am. J. Chin. Med. 2020, 48, 703–718. [Google Scholar] [CrossRef] [PubMed]
  14. Averett, C.; Bhardwaj, A.; Arora, S.; Srivastava, S.K.; Khan, M.A.; Ahmad, A.; Singh, S.; Carter, J.E.; Khushman, M.; Singh, A.P. Honokiol suppresses pancreatic tumor growth, metastasis and desmoplasia by interfering with tumor–stromal cross-talk. Carcinogenesis 2016, 37, 1052–1061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Pan, H.-C.; Lai, D.-W.; Lan, K.-H.; Shen, C.-C.; Wu, S.-M.; Chiu, C.-S.; Wang, K.-B.; Sheu, M.-L. Honokiol thwarts gastric tumor growth and peritoneal dissemination by inhibiting Tpl2 in an orthotopic model. Carcinogenesis 2013, 34, 2568–2579. [Google Scholar] [CrossRef] [Green Version]
  16. Shan, Y.-F.; Shen, X.; Xie, Y.-K.; Chen, J.-C.; Shi, H.-Q.; Yu, Z.-P.; Song, Q.-T.; Zhou, M.-T.; Zhang, Q.-Y. Inhibitory effects of tanshinone II-A on invasion and metastasis of human colon carcinoma cells. Acta Pharmacol. Sin. 2009, 30, 1537–1542. [Google Scholar] [CrossRef] [Green Version]
  17. Katoch, A.; Suklabaidya, S.; Chakraborty, S.; Nayak, D.; Rasool, R.U.; Sharma, D.; Mukherjee, D.; Faheem, M.M.; Kumar, A.; Sharma, P.R. Dual role of Par-4 in abrogation of EMT and switching on mesenchymal to epithelial transition (MET) in metastatic pancreatic cancer cells. Mol. Carcinog. 2018, 57, 1102–1115. [Google Scholar] [CrossRef]
  18. Li, M.; Yue, G.G.-L.; Song, L.-H.; Huang, M.-B.; Lee, J.K.-M.; Tsui, S.K.-W.; Fung, K.-P.; Tan, N.-H.; Bik-San Lau, C. Natural small molecule bigelovin suppresses orthotopic colorectal tumor growth and inhibits colorectal cancer metastasis via IL6/STAT3 pathway. Biochem. Pharmacol. 2018, 150, 191–201. [Google Scholar] [CrossRef]
  19. Wanga, X.-Z.; Gu, J.-L.; Gao, M.; Bian, Y.; Liang, J.-Y.; Wen, H.-M.; Wu, H. Peperomin E induces promoter hypomethylation of metastatic-suppressor genes and attenuates metastasis in poorly differentiated gastric cancer. Cell. Physiol. Biochem. 2018, 50, 2341–2364. [Google Scholar] [CrossRef]
  20. Ji, D.; Zhong, X.; Huang, P.; Kang, P.; Leng, K.; Zheng, W.; Wang, Z.; Xu, Y.; Cui, Y. Deoxyelephantopin induces apoptosis via oxidative stress and enhances gemcitabine sensitivity in vitro and in vivo through targeting the NF-κB signaling pathway in pancreatic cancer. Aging 2020, 12, 11116. [Google Scholar] [CrossRef]
  21. Zhu, Y.; Liu, Y.; Qian, Y.; Dai, X.; Yang, L.; Chen, J.; Guo, S.; Hisamitsu, T. Antimetastatic Effects of Celastrus orbiculatus on Human Gastric Adenocarcinoma by Inhibiting Epithelial–Mesenchymal Transition and NF-κB/Snail Signaling Pathway. Integr. Cancer Ther. 2015, 14, 271–281. [Google Scholar] [CrossRef] [Green Version]
  22. Torres, M.P.; Rachagani, S.; Purohit, V.; Pandey, P.; Joshi, S.; Moore, E.D.; Johansson, S.L.; Singh, P.K.; Ganti, A.K.; Batra, S.K. Graviola: A novel promising natural-derived drug that inhibits tumorigenicity and metastasis of pancreatic cancer cells in vitro and in vivo through altering cell metabolism. Cancer Lett. 2012, 323, 29–40. [Google Scholar] [CrossRef] [Green Version]
  23. Malvicini, M.; Gutierrez-Moraga, A.; Rodriguez, M.M.; Gomez-Bustillo, S.; Salazar, L.; Sunkel, C.; Nozal, L.; Salgado, A.; Hidalgo, M.; Lopez-Casas, P.P. A tricin derivative from Deschampsia antarctica Desv. inhibits colorectal carcinoma growth and liver metastasis through the induction of a specific immune response. Mol. Cancer Ther. 2018, 17, 966–976. [Google Scholar] [CrossRef] [Green Version]
  24. Jia, X.; Iwanowycz, S.; Wang, J.; Saaoud, F.; Yu, F.; Wang, Y.; Hu, J.; Chatterjee, S.; Wang, Q.; Fan, D. Emodin attenuates systemic and liver inflammation in hyperlipidemic mice administrated with lipopolysaccharides. Exp. Biol. Med. 2014, 239, 1025–1035. [Google Scholar] [CrossRef] [Green Version]
  25. Zhao, X.-A.; Chen, G.; Liu, Y.; Wu, H.; Chen, J.; Xiong, Y.; Tian, C.; Jia, B.; Wang, G.; Xia, J. Emodin alleviates liver fibrosis of mice by reducing infiltration of Gr1hi monocytes. Evid.-Based Complement. Altern. Med. 2018, 2018, 5738101. [Google Scholar] [CrossRef] [Green Version]
  26. Kataoka, S.; Umemura, A.; Okuda, K.; Taketani, H.; Seko, Y.; Nishikawa, T.; Yamaguchi, K.; Moriguchi, M.; Kanbara, Y.; Arbiser, J.L. Honokiol acts as a potent anti-fibrotic agent in the liver through Inhibition of TGF-β1/SMAD signaling and autophagy in hepatic stellate cells. Int. J. Mol. Sci. 2021, 22, 13354. [Google Scholar] [CrossRef] [PubMed]
  27. Zhai, T.; Xu, W.; Liu, Y.; Qian, K.; Xiong, Y.; Chen, Y. Honokiol alleviates methionine-choline deficient diet-induced hepatic steatosis and oxidative stress in C57BL/6 mice by regulating CFLAR-JNK pathway. Oxidative Med. Cell. Longev. 2020, 2020, 2313641. [Google Scholar] [CrossRef] [PubMed]
  28. Li, X.-X.; Lu, X.-Y.; Zhang, S.-J.; Chiu, A.P.; Lo, L.H.; Largaespada, D.A.; Chen, Q.-B.; Keng, V.W. Sodium tanshinone IIA sulfonate ameliorates hepatic steatosis by inhibiting lipogenesis and inflammation. Biomed. Pharmacother. 2019, 111, 68–75. [Google Scholar] [CrossRef]
  29. Shi, M.-J.; Yan, X.-L.; Dong, B.-S.; Yang, W.-N.; Su, S.-B.; Zhang, H. A network pharmacology approach to investigating the mechanism of Tanshinone IIA for the treatment of liver fibrosis. J. Ethnopharmacol. 2020, 253, 112689. [Google Scholar] [CrossRef] [PubMed]
  30. Kim, G.T.; Cho, K.H.; Sharma, A.; Devi, S.; Park, T.S. Annona muricata leaf extract attenuates hepatic lipogenesis and adipogenesis. Food Funct. 2021, 12, 4621–4629. [Google Scholar] [CrossRef]
  31. Takahashi, Y.; Soejima, Y.; Kumagai, A.; Watanabe, M.; Uozaki, H.; Fukusato, T. Inhibitory effects of Japanese herbal medicines sho-saiko-to and juzen-taiho-to on nonalcoholic steatohepatitis in mice. PLoS ONE 2014, 9, e87279. [Google Scholar] [CrossRef]
  32. Jiraviriyakul, A.; Songjang, W.; Kaewthet, P.; Tanawatkitichai, P.; Bayan, P.; Pongcharoen, S. Honokiol-enhanced cytotoxic T lymphocyte activity against cholangiocarcinoma cells mediated by dendritic cells pulsed with damage-associated molecular patterns. World J. Gastroenterol. 2019, 25, 3941. [Google Scholar] [CrossRef]
  33. Lu, Y.; Zheng, W.; Lin, S.; Guo, F.; Zhu, Y.; Wei, Y.; Liu, X.; Jin, S.; Jin, L.; Li, Y. Identification of an Oleanane-Type Triterpene Hedragonic Acid as a Novel Farnesoid X Receptor Ligand with Liver Protective Effects and Anti-inflammatory Activity. Mol. Pharm. 2018, 93, 63–72. [Google Scholar] [CrossRef]
  34. Zhang, Y.; Si, Y.; Zhai, L.; Yang, N.; Yao, S.; Sang, H.; Zu, D.; Qin, S.; Wang, J. Celastrus orbiculatus Thunb. ameliorates high-fat diet-induced non-alcoholic fatty liver disease in guinea pigs. Die Pharm.-Int. J. Pharm. Sci. 2013, 68, 850–854. [Google Scholar]
  35. Gezici, S.; Sekeroglu, N. Current Perspectives in the Application of Medicinal Plants Against Cancer: Novel Therapeutic Agents. Anticancer Agents Med. Chem. 2019, 19, 101–111. [Google Scholar] [CrossRef]
  36. Upreti, S.; Pandey, S.C.; Bisht, I.; Samant, M. Evaluation of the target-specific therapeutic potential of herbal compounds for the treatment of cancer. Mol. Divers. 2022, 26, 1823–1835. [Google Scholar] [CrossRef] [PubMed]
  37. Zheng, H.; Wang, X.; Zhang, Y.; Chen, L.; Hua, L.; Xu, W. Pien-Tze-Huang ameliorates hepatic fibrosis via suppressing NF-κB pathway and promoting HSC apoptosis. J. Ethnopharmacol. 2019, 244, 111856. [Google Scholar] [CrossRef] [PubMed]
  38. Zhu, Z.; Zhou, W.; Yang, Y.; Wang, K.; Li, F.; Dang, Y. Quantitative profiling of oxylipin reveals the mechanism of Pien-Tze-Huang on alcoholic liver disease. Evid.-Based Complement. Altern. Med. 2021, 2021, 9931542. [Google Scholar] [CrossRef]
  39. Abdul Wahab, S.M.; Jantan, I.; Haque, M.A.; Arshad, L. Exploring the leaves of Annona muricata L. as a source of potential anti-inflammatory and anticancer agents. Front. Pharmacol. 2018, 9, 661. [Google Scholar] [CrossRef]
  40. Huang, C.-C.; Lin, K.-J.; Cheng, Y.-W.; Hsu, C.-A.; Yang, S.-S.; Shyur, L.-F. Hepatoprotective effect and mechanistic insights of deoxyelephantopin, a phyto-sesquiterpene lactone, against fulminant hepatitis. J. Nutr. Biochem. 2013, 24, 516–530. [Google Scholar] [CrossRef]
  41. Saadati, S.; Sadeghi, A.; Mansour, A.; Yari, Z.; Poustchi, H.; Hedayati, M.; Hatami, B.; Hekmatdoost, A. Curcumin and inflammation in non-alcoholic fatty liver disease: A randomized, placebo controlled clinical trial. BMC Gastroenterol. 2019, 19, 1–6. [Google Scholar] [CrossRef]
  42. Kong, D.; Zhang, Z.; Chen, L.; Huang, W.; Zhang, F.; Wang, L.; Wang, Y.; Cao, P.; Zheng, S. Curcumin blunts epithelial-mesenchymal transition of hepatocytes to alleviate hepatic fibrosis through regulating oxidative stress and autophagy. Redox Biol. 2020, 36, 101600. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, S.; Chen, W.; Wang, Y.; Wu, J.; Xu, L.; Yu, Y.; Tian, J.; Xu, R.; Fang, Z.; Jiang, L.; et al. Chinese Herbal Prescription Fu-Zheng-Qu-Xie Prevents Recurrence and Metastasis of Postoperative Early-Stage Lung Adenocarcinoma: A Prospective Cohort Study Followed with Potential Mechanism Exploration. Oxid Med. Cell Longev. 2021, 2021, 6673828. [Google Scholar] [CrossRef]
  44. Smeenk, H.G.; Incrocci, L.; Kazemier, G.; van Dekken, H.; Tran, K.T.; Jeekel, J.; van Eijck, C.H. Adjuvant 5-FU-based chemoradiotherapy for patients undergoing R-1/R-2 resections for pancreatic cancer. Dig. Surg. 2005, 22, 321–328. [Google Scholar] [CrossRef]
  45. Ouyang, H.; Wang, P.; Meng, Z.; Chen, Z.; Yu, E.; Jin, H.; Chang, D.Z.; Liao, Z.; Cohen, L.; Liu, L. Multimodality treatment of pancreatic cancer with liver metastases using chemotherapy, radiation therapy, and/or Chinese herbal medicine. Pancreas 2011, 40, 120–125. [Google Scholar] [CrossRef] [Green Version]
  46. Zhang, Q.; Liu, J.; Li, R.; Zhao, R.; Zhang, M.; Wei, S.; Ran, D.; Jin, W.; Wu, C. A Network Pharmacology Approach to Investigate the Anticancer Mechanism and Potential Active Ingredients of Rheum palmatum L. Against Lung Cancer via Induction of Apoptosis. Front. Pharm. 2020, 11, 528308. [Google Scholar] [CrossRef] [PubMed]
  47. Tong, H.; Huang, Z.; Chen, H.; Zhou, B.; Liao, Y.; Wang, Z. Emodin Reverses Gemcitabine Resistance of Pancreatic Cancer Cell Lines Through Inhibition of IKKbeta/NF-kappaB Signaling Pathway. Onco Targets 2020, 13, 9839–9848. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, N.; Wang, J.; Sheng, A.; Huang, S.; Tang, Y.; Ma, S.; Hong, G. Emodin Inhibits the proliferation of MCF-7 human breast cancer cells through activation of aryl hydrocarbon receptor (AhR). Front. Pharmacol. 2021, 11, 622046. [Google Scholar] [CrossRef] [PubMed]
  49. Matsuda, T. Expectations for and challenges in population-based endoscopic gastric and colorectal cancer screening. Dig. Endosc. 2022, 34, 15–19. [Google Scholar] [CrossRef]
  50. Page, A.J.; Cosgrove, D.C.; Herman, J.M.; Pawlik, T.M. Advances in understanding of colorectal liver metastasis and implications for the clinic. Expert Rev. Gastroenterol. Hepatol. 2015, 9, 245–259. [Google Scholar] [CrossRef]
  51. Yu, P.; Zhang, Y.; Ye, Z.; Chen, X.; Huang, L.; Du, Y.; Cheng, X. Treatment of synchronous liver metastases from gastric cancer: A single-center study. Cancer Manag. Res. 2020, 12, 7905. [Google Scholar] [CrossRef]
  52. Zhou, L.-H.; Hu, Q.; Sui, H.; Ci, S.-J.; Wang, Y.; Liu, X.; Liu, N.-N.; Yin, P.-H.; Qin, J.-M.; Li, Q. Tanshinone II-a inhibits angiogenesis through down regulation of COX-2 in human colorectal cancer. Asian Pac. J. Cancer Prev. 2012, 13, 4453–4458. [Google Scholar] [CrossRef] [Green Version]
  53. Xue, J.; Jin, X.; Wan, X.; Yin, X.; Fang, M.; Liu, T.; Zhao, S. Effects and mechanism of tanshinone II A in proliferation, apoptosis, and migration of human colon cancer cells. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 4793. [Google Scholar] [CrossRef] [PubMed]
  54. Shakeri, A.; Ward, N.; Panahi, Y.; Sahebkar, A. Anti-angiogenic activity of curcumin in cancer therapy: A narrative review. Curr. Vasc. Pharmacol. 2019, 17, 262–269. [Google Scholar] [CrossRef] [PubMed]
  55. Guo, F.; Wang, Y.; Liu, J.; Mok, S.; Xue, F.; Zhang, W. CXCL12/CXCR4: A symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene 2016, 35, 816–826. [Google Scholar] [CrossRef] [PubMed]
  56. Neophytou, C.M.; Panagi, M.; Stylianopoulos, T.; Papageorgis, P. The role of tumor microenvironment in cancer metastasis: Molecular mechanisms and therapeutic opportunities. Cancers 2021, 13, 2053. [Google Scholar] [CrossRef]
  57. Sato, A.; Rahman, N.I.A.; Shimizu, A.; Ogita, H. Cell-to-cell contact-mediated regulation of tumor behavior in the tumor microenvironment. Cancer Sci. 2021, 112, 4005–4012. [Google Scholar] [CrossRef]
  58. Heeke, S.; Mograbi, B.; Alix-Panabières, C.; Hofman, P. Never travel alone: The crosstalk of circulating tumor cells and the blood microenvironment. Cells 2019, 8, 714. [Google Scholar] [CrossRef] [Green Version]
  59. Williamson, T.; Sultanpuram, N.; Sendi, H. The role of liver microenvironment in hepatic metastasis. Clin. Transl. Med. 2019, 8, 1–7. [Google Scholar] [CrossRef]
  60. Im, H.-J.; Kim, H.-G.; Lee, J.-S.; Kim, H.-S.; Cho, J.-H.; Jo, I.-J.; Park, S.-J.; Son, C.-G. A preclinical model of chronic alcohol consumption reveals increased metastatic seeding of colon cancer cells in the liver. Cancer Res. 2016, 76, 1698–1704. [Google Scholar] [CrossRef] [Green Version]
  61. Akkoc, Y.; Gozuacik, D. Autophagy and hepatic tumor microenvironment associated dormancy. J. Gastrointest. Cancer 2021, 52, 1277–1293. [Google Scholar] [CrossRef] [PubMed]
  62. Xu, Q.; Guo, L.; Gu, X.; Zhang, B.; Hu, X.; Zhang, J.; Chen, J.; Wang, Y.; Chen, C.; Gao, B. Prevention of colorectal cancer liver metastasis by exploiting liver immunity via chitosan-TPP/nanoparticles formulated with IL-12. Biomaterials 2012, 33, 3909–3918. [Google Scholar] [CrossRef]
  63. Xu, M.; Mizoguchi, I.; Morishima, N.; Chiba, Y.; Mizuguchi, J.; Yoshimoto, T. Regulation of antitumor immune responses by the IL-12 family cytokines, IL-12, IL-23, and IL-27. Clin. Dev. Immunol. 2010, 2010, 832454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Luo, L.; Yang, Y.; Li, P. Cohort study on fuzheng capsule and quxie capsule in reducing relapse and metastasis of cancer in patients with stage II and III colorectal carcinoma after operation. Zhongguo Zhong Xi Yi Jie He Za Zhi Zhongguo Zhongxiyi Jiehe Zazhi = Chin. J. Integr. Tradit. West. Med. 2006, 26, 677–680. [Google Scholar]
  65. Mukherjee, S.; Baidoo, J.N.; Sampat, S.; Mancuso, A.; David, L.; Cohen, L.S.; Zhou, S.; Banerjee, P. Liposomal TriCurin, a synergistic combination of curcumin, epicatechin gallate and resveratrol, repolarizes tumor-associated microglia/macrophages, and eliminates glioblastoma (GBM) and GBM stem cells. Molecules 2018, 23, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. She, L.; Xu, D.; Wang, Z.; Zhang, Y.; Wei, Q.; Aa, J.; Wang, G.; Liu, B.; Xie, Y. Curcumin inhibits hepatic stellate cell activation via suppression of succinate-associated HIF-1α induction. Mol. Cell. Endocrinol. 2018, 476, 129–138. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, W.; Li, H.; Bu, H.; Chen, H.; Tong, H.; Liu, D.; Guo, H.; Lin, S.-Z. Emodin inhibits the differentiation and maturation of dendritic cells and increases the production of regulatory T cells. Int. J. Mol. Med. 2012, 29, 159–164. [Google Scholar]
Figure 1. Flow diagram of selection strategy and article review.
Figure 1. Flow diagram of selection strategy and article review.
Cancers 15 03415 g001
Figure 2. Summary of anti-liver metastatic TMEs’ modulation by herbal candidates. ECM: extracellular matrix; DET: deoxyelephantopin; NGD16: 1,1′-β-D-glucopyranosyl-3,3′-bis (5-bromoindolyl)-octyl methane.
Figure 2. Summary of anti-liver metastatic TMEs’ modulation by herbal candidates. ECM: extracellular matrix; DET: deoxyelephantopin; NGD16: 1,1′-β-D-glucopyranosyl-3,3′-bis (5-bromoindolyl)-octyl methane.
Cancers 15 03415 g002
Table 1. Summary of the characteristics of the included studies.
Table 1. Summary of the characteristics of the included studies.
ClassificationTotal Number of Studies/Reference
Final number of included studies21
Original plant (only single compound or extract)12
Medication
    Single compound12/[3,10,11,13,14,15,16,17,18,19,20,23]
    Single-herb extract5/[8,9,12,21,22]
    Herbal decoction4/[4,5,6,7]
Organ of origin
    Pancreas9/[4,5,10,11,12,14,17,20,22]
    Colon8/[6,7,8,9,13,16,18,23]
    Stomach4/[3,15,19,21]
Experimental design
  Lesion implantation area
    Pancreatic region4/[10,14,17,22]
    Colon region4/[7,9,13,18]
    Stomach region3/[3,15,19]
    Spleen injection5/[4,5,11,12,15,20]
    Subcutaneous injection1/[8]
    Intraperitoneal injection1/[21]
  Intrahepatic injection1/[23]
    Intravenous injection (tail vein)2/[6,16]
  Administration method
    Oral12/[4,5,6,7,8,9,10,11,16,19,21,22]
    Intraperitoneal injection7/[12,13,14,15,17,20,23]
    Intravenous injection2/[3,18]
Positive control
  FOLFOX (5-FU, fluorouracil, and oxaliplatin)2/[8,9]
  5-FU1/[23]
  Cisplatin1/[6]
  Gemcitabine1/[20]
  Mithramycin1/[19]
  Capecitabine1/[21]
  None14/[3,4,5,7,10,11,12,13,14,15,16,17,18,22]
Efficacy assessment tool
  Number of liver metastasis nodules15/[3,4,6,7,10,11,12,13,14,15,16,17,20,21,22]
  Size of liver metastatic area5/[5,8,9,18,23]
  Luciferase activity in liver1/[19]
Table 2. List of natural products having preventative effects against liver metastasis of digestive organ cancers.
Table 2. List of natural products having preventative effects against liver metastasis of digestive organ cancers.
Original PlantOrigin of Cancer (Method of Transplantation) and Drug Formulation (Route of Administration)Main Mechanisms Related to the Tumor Microenvironment (Site of Tissue Taken for Assay)Liver Environment Factors
Curcuma longa L.Xenograft, colon ca. (sc), and ethanol ex (po) [8] Regulates EMT and angiogenesis at the original site (rectum)Steatosis, inflammation, and fibrosis [24,25]
Colon 26-M01 (oi) and ethanol ex (po) [9,18] Regulates immune components and the vascular endothelium at the original site (rectum)
Primary gastric cancer cell (sc) and curcumin (iv) [3]Inhibits CXCL12/CXCR4 expression at the original site (limb)
Rheum palmatum L.Pancreas SW1990 (oi) and emodin (po) [10]Suppresses NF-κB and MMP-9 protein expression at the original site (pancreas)Steatosis, inflammation, and fibrosis [26,27]
Pancreas SW1990 (si) and emodin (po) [11]Increases E-cadherin expression via miRNA-1271 at the metastasis site (liver)
Venenum bufonisPancreas SW1990 (si) and water ex (ip) [12]Regulates MMP-2/9 and VEGF expression at the metastasis site (liver)
Colon HCT116 (oi) and cinobufacini (ip) [13]Regulates MMP-2/9 and increases E-cadherin expression at the original site (colon)
Magnolia acuminate L.Pancreas MIA PaCa-2 (oi) and honokiol (ip) [14]Interferes with tumor–stromal crosstalk at the original site (pancreas)Steatosis, inflammation, and fibrosis [28,29]
Stomach MKN45 (oi) and honokiol (ip) [15]Increases E-cadherin and suppresses TGF-β expression at the original site (stomach)
Elephantopus scaber L.Pancreas BxPC-3 (si) and deoxyelephantopin (ip) [20]Increases E-cadherin and suppresses NF-κB expression at the original site (pancreas)Inflammation [30]
Inula helianthus-aquaticaColon HCT116 (oi) and bigelovin (iv) [18]Decreases IL-6 and suppresses N-cadherin expression at the original site (colon)
Salvia miltiorrhiza B.Colon SW480 (iv) and tanshinone II-A (po) [16]Regulates MMP-2/9 and TIMP-1/2 expression at the original site (colon)Steatosis, inflammation, and fibrosis [31,32]
Deschampsia Antarctica D.Colon CT-26 (ih) and antartina (ip) [23]Activates immunity, mainly via dendritic cells and CD8 T cells (blood)
Peperomia dindygulensis M.Stomach NCI-N87-luc (oi) and peperomin E (po) [19]Activates E-cadherin and TIMP-3 at the original site (stomach)
Cruciferous vegetablesPancreas UN-KC-6141 (oi) and NGD16 (ip) [17]Increases E-cadherin expression at the original site in a Par4-dependent manner (pancreas)
Annona muricata L. Pancreas CD18/HPAF (oi) and ethanol ex (po) [22]Suppresses MMP-9 and MUC4 expression at the original site (pancreas)Steatosis and inflammation [33,34]
Celastrus orbiculatus T.Stomach SGC-7901 (ip) and acetate ex (po) [21]Decreases NF-κB, increases E-cadherin, and suppresses N-cadherin expression at the original site (peritoneal)Steatosis and inflammation [35,36]
Qing Yi Hua JiPancreas SW1990HM (si) and decoction (po) [5]Decreases IL-6 expression and increases tumor-associated macrophages at the original site (pancreas)
Pancreas SW1990HM (si) and decoction (po) [4] Suppresses VEGF releases at the metastasis site (liver)
Juzen-taiho-toColon 26-L5 (pv) and decoction (po) [6]Regulates macrophages and/or T cells (blood)Steatosis, inflammation, and fibrosis [37,38]
PientzehuangColon CT-26 (oi) and decoction (po) [7]Decreases TGF-β, increases E-cadherin, and suppresses N-cadherin expression at the original site (colon)Steatosis, inflammation, and fibrosis [39,40]
Ex: extract; DMSO: dimethyl sulfoxide; po: per os (by mouth); sc: subcutaneous injection; oi: orthotopic implantation; iv: intravenous injection; ip: intraperitoneal injection; si: splenic injection; pv: portal vein; ih: intrahepatic inoculation; Wnt: wingless-related integration site; Src: SRC proto-oncogene tyrosine-protein kinase; KRAS: gene Kirsten rat sarcoma viral oncogene homolog; EGFR: epidermal growth factor receptor; SDF-1: stromal cell-derived factor-1; CXCR4: C-X-C chemokine receptor type 4; NF-κB: nuclear factor-κB; miRNA: microRNA; VEGF: vascular endothelial growth factor; SHH: sonic hedgehog; Tpl2: tumor progression locus 2; IL-6: interleukin-6; STAT3: signal transducer and activator of transcription 3; ALK2: activin A receptor type II-like kinase 2; Smad4: mothers against decapentaplegic homolog 4; TGF-β: transforming growth factor-β; NGD16: 1,1′-β-D-glucopyranosyl-3,3′-bis(5-bromoindolyl)-octyl methane: TAM: tumor-associated macrophage; EMT: epithelial mesenchymal transition; MMP: matrix metalloproteinase; TIMP: tissue inhibitor of metalloproteinase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, S.-K.; Lee, N.-H.; Son, C.-G. A Review of Herbal Resources Inducing Anti-Liver Metastasis Effects in Gastrointestinal Tumors via Modulation of Tumor Microenvironments in Animal Models. Cancers 2023, 15, 3415. https://doi.org/10.3390/cancers15133415

AMA Style

Kim S-K, Lee N-H, Son C-G. A Review of Herbal Resources Inducing Anti-Liver Metastasis Effects in Gastrointestinal Tumors via Modulation of Tumor Microenvironments in Animal Models. Cancers. 2023; 15(13):3415. https://doi.org/10.3390/cancers15133415

Chicago/Turabian Style

Kim, Sul-Ki, Nam-Hun Lee, and Chang-Gue Son. 2023. "A Review of Herbal Resources Inducing Anti-Liver Metastasis Effects in Gastrointestinal Tumors via Modulation of Tumor Microenvironments in Animal Models" Cancers 15, no. 13: 3415. https://doi.org/10.3390/cancers15133415

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop