Next Article in Journal
Conjugation of Carbon Dots with β-Galactosidase Enzyme: Surface Chemistry and Use in Biosensing
Previous Article in Journal
Effects of High Pressure Modification on Conformation and Digestibility Properties of Oyster Protein
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Pien-Tze-Huang: A Review on Its Chemical Composition, Pharmacology, and Clinical Application

1
College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian, China
2
Engineering Innovation Center of Marine Biological Resource Development and Utilization, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, China
3
State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2019, 24(18), 3274; https://doi.org/10.3390/molecules24183274
Submission received: 12 August 2019 / Revised: 3 September 2019 / Accepted: 6 September 2019 / Published: 9 September 2019

Abstract

:
Pien-Tze-Huang (PTH) is a famous and commonly used traditional Chinese medicine formula in China. It was first formulated by a royal physician of the Ming Dynasty (around 1555 AD). Recently, PTH has attracted attention worldwide due to its beneficial effects against various diseases, especially cancer. This paper systematically reviewed the up-to-date information on its chemical composition, pharmacology, and clinical application. A range of chemical compounds, mainly ginsenosides and bile acids, have been identified and quantified from PTH. Pharmacological studies indicated that PTH has beneficial effects against various cancers, hepatopathy, and ischemic stroke. Furthermore, PTH has been used clinically to treat various diseases in China, such as colorectal cancer, liver cancer, and hepatitis. In summary, PTH is a potential agent with extensive therapeutic effects for the treatment of various diseases. However, the lack of information on the side effects and toxicity of PTH is a non-negligible issue, which needs to be seriously studied in the future.

1. Introduction

Recently, traditional Chinese medicine (TCM) with thousands of years of clinical practice has increasingly attracted interest in the treatment of various human diseases around the world. It not only provides an alternative and promising therapeutic strategy, but also contributes a precious drug discovery source. Pien-Tze-Huang (PTH) is a well-known and precious Chinese patent medicine in China and Southeast Asian countries. It was first formulated by a royal physician of the Ming Dynasty in 1555 AD. It is composed of four TCM ingredients, including Radix et Rhizoma Notoginseng (Sanqi in Chinese, 85%), Moschus (Shexiang in Chinese, 3%), Calculus Bovis (Niuhuang in Chinese, 5%), and Snake Gall (Shedan in Chinese, 7%), which together have functions of reducing fever, detoxifying, promoting blood circulation, reducing blood stasis and swelling, and relieving pain [1]. PTH is officially used to treat acute and chronic viral hepatitis, ulcer, deep-rooted boil, traumatic injuries, and various inflammatory diseases [1,2,3]. In addition, PTH has been commonly applied to treat cancer and stroke. Its therapeutic effects on hepatocellular carcinoma and colon cancer have been recognized in clinical trials [4,5]. PTH has been categorized as one of the national treasures in the catalogue of the National Protected Traditional Chinese Medicines in 1994 attributing to its long history and significant beneficial effect against various diseases.
Although many properties of PTH have been extensively studied, a systematic and comprehensive review on its chemical, experimental, and clinical properties has not been conducted. Therefore, the aim of this review is to summarize the up-to-date progress concerning the chemical composition, pharmacology, and clinical application of PTH, thus to better understand its therapeutic potential and support its clinical application and in-depth study in the future.

2. Chemical Composition

PTH consists of four natural TCM materials, including Radix et Rhizoma Notoginseng, Moschus, Calculus Bovis, and Snake Gall. Many studies have been conducted to determine the main bioactive compounds of these materials. The results showed that Radix et Rhizoma Notoginseng mainly contains protopanaxadiol-type (e.g., notoginsenoside R1 and ginsenosides Re, Rf, Rg1, and Rg2) and protopanaxatriol-type ginsenosides (e.g., ginsenosides Rb1, Rc, Rd, and Rg3), which are considered its main bioactive compounds [6]. Moschus mainly contains macrocyclic ketones (e.g., muscone) and steroids (e.g., androsterone), which possess extensive biological activities [7]. Calculus Bovis mainly contains bile acids and their glycine or taurine conjugates, such as cholic acid, deoxycholic acid, chenodeoxycholic acid, hyodeoxycholic acid, and ursodeoxycholic acid, which are supposed to be its main characteristic and bioactive compounds [8]. Snake Gall mainly contains taurine-conjugated bile acids, such as taurocholic acid, taurochenodeoxycholic acid, taurodeoxycholic acid, and glycodeoxycholic acid, but their contents are markedly different from those of Calculus Bovis, which is the main reason for their varied efficacies [9].
Given the complicated influences of collection, processing, production, and storage of raw materials and products, the chemical composition of PTH is somewhat different to the chemical composition of its four components. Therefore, several analytical methods, including thin layer chromatography scanning (TLCS), high-performance liquid chromatography (HPLC), HPLC coupled with quadrupole time-of-flight mass spectrometry (HPLC-QTOF-MS), and ultra-performance LC coupled with triple quadrupole MS (UPLC-QqQ-MS), have been applied to separate and identify the chemical composition of PTH in past decades [10,11,12,13]. For example, only ginsenoside Rg1 in PTH was determined by TLCS after complicated and time-consuming alumina column purification [14], and three saponin components (notoginsenoside R1, ginsenoside Rb1, and ginsenoside Rg1) in PTH were determined by HPLC [15]. In our recent studies, 27 compounds, including taurine, citric acid, malic acid, notoginsenoside R1, ginsenosides Re, Rg1, Rf, Rg2, Rh1, Rb1, Rd, Rg3, Rc, Rh2, Rh3, cholic acid, deoxycholic acid, chenodeoxycholic acid, ursodeoxycholic acid, hyodeoxycholic acid, taurocholic acid, tauroursodeoxycholic acid, taurochenodeoxycholic acid, taurohyodesoxycholic acid, glycocholic acid, glycodeoxycholic acid, and muscone, were definitely identified in PTH by HPLC-QTOF-MS (Figure 1) [10]. Among them, 21 compounds showed significant inhibitory activities on LPS-induced TNF-α production with IC50 values ranging from 30.82 μM to 147.24 μM in murine mononuclear macrophage RAW 264.7 cells [10]. Thus, they were chosen as quality biomarkers of PTH, and their contents in PTH were accurately quantified by UPLC-QqQ-MS [10,11]. Furthermore, we developed a UPLC-MS/MS method to investigate the pharmacokinetic behaviors of six active compounds of PTH, including ginsenosides Re, Rg1, Rb1, and Rd, notoginsenoside R1, and muscone, after oral administration to rats and partly revealed the clinical implication of PTH in several inflammation-related diseases [13].
These identified compounds in PTH have been reported to have beneficial effects against various diseases, which partly reflect the therapeutic function of this formula [16,17]. For example, ginsenoside Re, one of the protopanaxadiol-type ginsenosides in Radix et Rhizoma Notoginseng, has been reported to exhibit potential anti-oxidant, anti-inflammatory, anticarcinogenic, and neuroprotective effects, which was well tolerated up to a 375 mg/kg/d oral dosage level in rats [18,19,20,21]. As one of protopanaxatriol-type ginsenosides in this herb, ginsenoside Rb1 has been proved to possess remarkable anti-oxidant, anti-cancer, and neuroprotective effects [22,23,24]. In addition, muscone, a volatile bioactive compound in Moschus, has been shown to have significant anti-inflammatory and neuroprotective effects, which exhibited liver toxicity in mice at an oral dose of 100 mg/kg [25,26,27,28]. However, more studies are still needed to determine which compounds are responsible for its therapeutic effect against different diseases.

3. Pharmacology

3.1. Colorectal Cancer

Accumulated clinical and experimental studies have exhibited the anti-cancer properties of PTH against a broad spectrum of cancer cells, which multiply via signaling pathways. Colorectal cancer is a malignant tumor in the gastrointestinal tract, the incidence and mortality of which are both forefront worldwide [29]. In addition to its well-known anti-inflammatory activities, PTH has been shown to possess potent anti-proliferative effects against various human colorectal cancer cell lines with IC50s below 1 mg/mL [30,31,32,33]. The in vivo anti-cancer potential of PTH has been recognized in different xenograft animal models [31]. PTH not only overcomes multidrug resistance (MDR) in HCT-8/5-fluorouracil (5-FU) cells but also attenuates 5-FU-induced intestinal mucositis in CT-26 tumor-bearing xenograft mice [34,35,36]. Furthermore, it exhibits synergistic effects when combined with chemotherapy drugs, such as fluorouracil, oxaliplatin, and capecitabine, in clinical in China [37,38]. However, the molecular mechanism underlying its anti-cancer effect remains unclear.
The induction of cell cycle arrest and apoptosis is suggested to be the primary reason for the anti-proliferative effect of PTH. PTH inhibits the proliferation of Caco-2 and HCT-8 cells by inducing G1/S-phase arrest, which may be due to the inhibition of cyclin-dependent kinase (CDK)4, CDK6, Cyclin D1, and c-Myc [32,33]. PTH-induced cell cycle arrest is often accompanied by apoptosis. These two actions are at least partially mediated by inhibiting interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) pathway and its target genes, such as suppressor of cytokine signaling 3 (SOCS3) [31,39]. Moreover, the inhibition of the IL-6/STAT3 pathway has been suggested to account for the beneficial effects of PTH in the treatment of ulcerative colitis [40]. As one of the MiR-34 family, miR-34c exhibits anti-proliferative and pro-apoptotic functions. Wan et al. (2017) showed that PTH inhibits proliferation and induces apoptosis in HCT-8 cells partially by upregulating miR-34c-5p expression, which may provide a novel perspective for investigating its mechanisms.
Metastasis is a complicated process related to the spread of malignant cancer cells from a primary position to another position. In HCT-8 cells, PTH inhibits migration and invasion in a dose-dependent manner by decreasing the expressions of transforming growth factor β1 (TGF-β1), Smad2/3, Smad4, zinc finger E-box binding homeobox 1 (ZEB1), and ZEB2 and upregulating the expressions of miR-200a, miR-200b, and miR-200c [41]. In an orthotopic liver metastasis model of colorectal cancer, PTH exhibits beneficial effects against metastasis without significant toxicity. Furthermore, it inhibits the expressions of TGF-β, Smad2/3, p-Smad2/3, and Smad4 in tumor tissues, suggesting its inhibitive activities on the TGF-β/Smad pathway [42]. It also inhibits hypoxia-induced epithelial–mesenchymal transition (EMT) in HCT-8 cells through suppressing hypoxia-inducible factor 1 (HIF-1) pathway, which may be another important molecular mechanism [43]. A similar effect has also been found in HCT-8/5-FU cells [34]. Lymphangiogenesis, the production of new lymphatic vessels, plays a crucial role in tumor development, especially in cancer metastasis. Vascular endothelial growth factor-C (VEGF-C) is important in cancer metastasis as well. A recent study indicated that PTH reduces cell migration and VEGF-C expression in various colorectal cancer cell lines, and inhibits lymphangiogenesis by reducing cell migration and tube formation in a VEGF-C-stimulated human lymphatic endothelial cell model, indicated that inhibiting VEGF-C may be one of its potential molecular mechanisms [44].
Angiogenesis plays a critical role in tumor progress, which is one of the common therapeutic strategies in cancer treatment. An in vitro study showed that PTH inhibits migration and tube formation of human umbilical vein endothelial cells (HUVECs) in a dose-dependent manner without affecting their viability. Furthermore, PTH inhibits hypoxia-induced activation of HIF-1α and decreases VEGF-A and vascular endothelial growth factor receptor 2 (VEGFR2) expressions in both HT-29 and HUVECs [45]. In a colorectal cancer mouse xenograft model, PTH decreases tumor volume, tumor weight, microvessel density in tumor tissues; and the expressions of angiogenic factors, including iNOS, eNOS, VEGFR2, VEGF-A, bFGF, and basic fibroblast growth factor (bFGFR) [46]. However, more studies are needed to elucidate the anti-angiogenesis effect and molecular mechanisms of PTH.
Several studies demonstrated that PTH may be a potential multidrug resistance (MDR)-reversing drug that restores the sensitivity of MDR cancer cells [34,35,36]. PTH dose-dependently inhibits the proliferation of HCT-8/5-FU cells, increases the intercellular accumulation of Rhodamine-123, and downregulates ATP-binding cassette subfamily C member 2 (ABCC2) expressions in HCT-8/5-FU cells, indicating its ability to overcome MDR. Moreover, PTH inhibits MDR/EMT-enhanced migration and invasion capabilities and suppresses MDR-induced activation of TGF-β signaling in HCT-8/5-FU cells [34]. Another recent study also found that PTH inhibits proliferation and induces apoptosis of HCT-8/5-FU cells by regulating the expressions of CDK4, cyclin D1, B-cell lymphoma 2/Bcl-2-associated X protein (Bcl-2/Bax) ratio, caspase-9, and caspase-3, which may be mediated by modulating the miR-22/c-Myc signaling network [35]. Notably, PTH not only overcomes MDR but also attenuates chemotherapy drug-induced side effects. In CT-26 tumor-bearing xenograft mice, PTH improves 5-FU-induced diarrhea and intestinal pathological damages without significant effect on body weight loss, which is partly related to its anti-apoptotic effect in the intestinal crypt [36].
Cancer stem cells (CSCs) play a crucial role in tumor development and drug resistance, suggesting that targeting CSCs may be a promising approach for cancer treatment. In the side population (SP) isolated from HT-29 cells, PTH dose-dependently reduces the percentage of SP cells, inhibits their proliferation and sphere-forming capacity, and inhibits ABCB1 and ABCG2 levels, indicating that the inhibition of CSCs may be a potential mechanism of PTH [47]. A similar study by Qi et al. demonstrated that PTH dose-dependently inhibits proliferation and induces apoptosis and differentiation of SP cells isolated from SW480 cells. In addition, PTH treatment inhibits the mRNA and protein expressions of hairy and enhancer of split 1 (Hes1) and Notch1 of SP cells [48].

3.2. Liver Cancer

Previous experimental and clinical studies have exhibited promising therapeutic effects in the treatment of liver cancer. In human BEL-7402 hepatocellular carcinoma cell lines, PTH inhibits proliferation by inhibiting cyclin D1 and CDK4 expressions and induces apoptosis by decreasing Bcl-2/Bax ratio, which may be due to the upregulation of tumor suppressor miR-16 [49]. Similar anti-proliferative and anti-apoptotic effects have also been observed in human SMMC-7721 and HepG2 CSCs [50].

3.3. Osteosarcoma

Osteosarcoma is the most common form of primary bone cancer that usually occurs in teenagers or young adults. An in vitro study has shown that PTH dose-dependently inhibits the viability of MG-63 cells by inducing apoptosis, which may be mediated by downregulating p21, p53, Bcl-xl, Bcl-2, PI3K, p-Akt, and p-ERK1/2 levels and upregulating caspase-3, caspase-9, Bax, and cleaved-PARP levels [51]. In human osteosarcoma-bearing nude mice, PTH treatment alone or in combination with p27 gene recombinant adenovirus exhibited significant tumor growth inhibition effects (34.1% and 63.8%, respectively), suggesting that both treatments are beneficial for osteosarcoma treatment [52]. Moreover, PTH dose- and time-dependently inhibits the viability of U2OS/ADM cells through inducing G2/M-phase cell arrest and apoptosis, which may be mediated by the downregulation of Bcl-2, survivin, and P-gp and the upregulation of Bax [53].

3.4. Other Cancers

In addition to the abovementioned cancer types, PTH has been used in the treatment of other cancers, including neuroblastoma, ovarian cancer, and breast cancer, with similar mechanisms. The anti-proliferative activities of PTH in vitro are summarized in Table 1. A previous study showed that PTH can dose-dependently inhibit the survival of neuroblastoma SH-SY5Y cells without affecting normal fibroblast NIH-3T3 cells [54]. PTH also inhibits the viability of human ovarian cancer OVCAR-3 cell line by inducing G1/S-phase cell arrest without inducing apoptosis. This effect is at least partially mediated by inhibiting Akt–mammalian target of rapamycin (mTOR) pathway [55]. In human breast cancer MCF-7/ADR cells, PTH dose-dependently inhibits their proliferation, increases the intercellular accumulation of adriamycin, and reduces ABCG2 and ABCB1 expressions. Moreover, PTH inhibits EMT, migration, and invasion and suppresses the activation of TGF-β1 in MCF-7/adriamycin cells [56]. Further experimental and clinical studies are needed to clearly define these effects.

3.5. Hepatopathy

Accumulated evidence demonstrated that PTH can be potentially used in treating various hepatic diseases, such as viral hepatitis, alcoholic hepatitis, and fatty liver. Previous studies have shown that PTH protects against liver damage, decreases alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels in a mouse with acute hepatitis induced by carbon tetrachloride or galactosamine, and upregulates the mRNA expressions of anti-microbial protein 1 (AP1) and nuclear factor kappa-B (NF-κB) in hepatoma cells [2]. Moreover, PTH exhibits similar hepatoprotective effect in alcoholic and non-alcoholic fatty liver animal models. In alcoholic and high-fat diet-induced alcoholic fatty liver rats, PTH improves hepatic function defects, hepatic pathology, and impairment in lipid metabolism by inhibiting the homocysteine-related protein kinase RNA-like endoplasmic reticulum kinase (PERK)/eukaryotic translation initiation factor 2α (eIF2α) pathway [57]. In high-fat diet-induced non-alcoholic fatty liver rats, PTH improves liver function and lowers blood lipid, which may be related to regulating the farnesoid X receptor (FXR)/small heterodimer partner (SHP)/sterol regulatory element binding protein-1c (SREBP-1) pathway [58].

3.6. Ischemic Stroke

Stroke is one of the major causes of death and disability worldwide, and more than 80% of cases are categorized as ischemic stroke. Increasing evidence suggest that PTH is a potential protective and therapeutic agent for ischemic stroke. In spontaneously hypertensive rat (SHR) and stroke-prone SHR, PTH significantly reduces cell death in the hippocampus and cerebellum caused by chronic ischemia and hypertensive stroke possibly by inhibiting cell apoptosis or ROS/oxidative damage in the mitochondria [59]. Moreover, PTH can obviously make the stroke less serious and notably postpone death after its occurrence [59,60].
In our previous study, the transient middle cerebral artery occlusion (tMCAO) model was used to investigate the effect of PTH on cerebral ischemia/reperfusion injury. The results showed that PTH significantly improves neurological deficit infarct volume, and downregulates the levels of IL-1β, IL-6, and TNF-α in brain tissue of tMCAO rats. Furthermore, PTH decreases the cytosolic levels of cytochrome C, p53, Bax, cleaved caspase-3, and cleaved caspase-9 and increases the mitochondrial levels of cytochrome C and Bcl-xl and the phosphorylation levels of Akt and GSK-3β, indicating that inhibiting mitochondria-mediated neuronal apoptosis may be one of its important mechanisms [61]. Although much progress has been made in experimental studies, more clinical studies are needed to verify these effects. The pharmacological activities and mechanisms of PTH in vivo have been summarized in Table 2.
Notably, PTH was simultaneously used to treat cancer and stroke in different studies, whereas mechanistically, it has a pro-apoptotic effect in cancer and anti-apoptotic effect in stroke [31,61]. In general, the same drug or therapy can be used to treat different diseases and is considered to be one of the main characteristics of TCM. Besides, various results could be obtained due to different disease pathogenesis as well as diverse dosage regimens. However, the detailed molecular mechanisms are required for further study.

4. Clinical Application

4.1. Colorectal Cancer

In past decades, the clinical efficacy of PTH on colorectal cancer has been investigated in China. As early as 1993, PTH alone has been observed to improve clinical symptoms and life quality in a preliminary clinical observation containing 25 advanced colorectal cancer patients [5]. In another clinical observation containing 47 advanced colorectal cancer patients, PTH in combination with 5-FU, oxaliplatin, and leucovorin exhibited better short-term efficacy, reduced the toxicity and side effects of chemotherapy, and improved the life qualities of advanced colorectal cancer patients compared with single chemotherapy [37]. A similar synergistic effect of PTH combined with oxaliplatin and capecitabine was observed in another clinical observation containing 68 advanced colorectal cancer patients [38]. However, more restrictive clinical trials are needed to further confirm its efficacy in colorectal cancer treatment.

4.2. Liver Cancer

Several clinical studies on liver cancer have been conducted in the past decades. In a 1994 clinical observation containing 42 patients with moderate or advanced liver cancer, PTH combined with conventional therapies achieved lower exacerbation rate and improved clinical symptoms and life quality compared with single chemotherapy [62]. In another randomized, double-blind, placebo-controlled clinical trial that enrolled 207 patients with primary liver cancer, PTH in combination with interventional chemotherapy exerted significant improvements in reducing tumor size, improving life quality, relieving pain, and reducing the toxicity and side effects of chemotherapy compared with single interventional chemotherapy [63]. Intriguingly, PTH can improve weakened immunity inhibited by transcatheter arterial chemoembolization in 40 patients with primary liver cancer [64].

4.3. Hepatitis

A recent clinical observation demonstrated that PTH can improve clinical symptoms and signs, alleviate digestive tract symptoms, protect liver function, and decrease the levels of HBV-DNA and HbeAg in 47 patients with chronic hepatitis B, with an efficacy similar to or better than polyene phosphatidylcholine [65].

4.4. Other Diseases

PTH has also been applied in treating other diseases, including ulcers and phlebitis. Two clinical observations have verified that PTH can accelerate the healing of recurrent aphthous ulcer and decrease the risk of recurrence without an increase in obvious adverse reactions [66,67]. Moreover, in a clinical study containing 31 patients with phlebitis induced by amiodarone, PTH exhibited better clinical efficacy than magnesium sulfate [68].
Except for a well-controlled clinical study by Zhao et al. [63], most of above studies were preliminary clinical observation. Furthermore, most of studies were focused on the combination of PTH and routine therapy but not the single PTH. Clinical application of PTH on various diseases has been summarized in Table 3.

5. Conclusions and Prospects

This review mainly summarizes the up-to-date progress concerning the chemical composition, pharmacology, and clinical application of PTH. The findings indicate that PTH is a safe and effective therapeutic agent against various diseases. In the past decades, researchers have focused on the pharmacological activities of PTH, particularly on its anti-cancer effects. Despite the great progress in the study of PTH, several points still need to be noted for better understanding and utilization of this traditional formula. First, although PTH has been increasingly used to treat various diseases in China and Southeast Asian countries, especially cancer, only few relevant randomized, double-blind, placebo-control, and multi-center clinical trials exist. Therefore, more restrictive clinical trials are needed to evaluate the safety and efficacy of PTH alone or in combination with routine therapy. Second, in spite of no side effects or toxicity reported in the peer reviewed papers, we believe that the potential side effects or toxicity of PTH still need to be seriously studied in future research, and more attention should be paid to its misuse, overdose, long-term administration, or unreasonable drug combination. Third, more pharmacokinetic studies are needed to illuminate the absorption, distribution, metabolism, and excretion of PTH, which can better guide its clinical application. Fourth, the specific compounds responsible for the beneficial effects of PTH on different diseases, as well as the action of mechanism, still remain to be elucidated. Fifth, the quality control of PTH is still important for guaranteeing the safety and efficacy of its preparation, though some analysis methods have made significant contributions. Future studies should focus on the collection, processing, production, and storage of its raw materials and finished products. Finally, most of the research on PTH are from China currently, and it needs more and more abroad scientists’ participation. Collectively, finding of presenting studies highlight the therapeutic potential of PTH in various diseases, especially cancer.

Author Contributions

L.H. and Y.Z. shared the same amount of work on writing this manuscript. X.Z., X.C., J.L., and M.H. wrote and edited this manuscript. Y.W. provided the professional guidance. M.H. provided the financial support for this manuscript.

Funding

This work is supported by the National Natural Science Foundation of China (81673561, 81973437), the Natural Science Foundation of Fujian Province (2017J01838), the Research Fund of University of Macau (MYRG2018-00165-ICMS and CPG2019-00006-ICMS) and The Science and Technology Development Fund, Macao SAR (File no. 176/2017/A3).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Commission, C.P. Pharmacopoeia of the People’s Republic of China; Chinese Medical Science and Technology Press: Beijing, China, 2015; pp. 573–575. [Google Scholar]
  2. Lee, K.K.; Kwong, W.H.; Chau, F.T.; Yew, D.T.; Chan, W.Y. Pien Tze Huang protects the liver against carbon tetrachloride-induced damage. Pharm. Toxicol. 2002, 91, 185–192. [Google Scholar] [CrossRef] [PubMed]
  3. Chan, W.Y.; Chau, F.T.; Lee, K.K.; Kwong, W.H.; Yew, D.T. Substitution for natural musk in Pien Tze Huang does not affect its hepatoprotective activities. Hum. Exp. Toxicol. 2004, 23, 35–47. [Google Scholar] [CrossRef] [PubMed]
  4. Xu, Y.Y.; Yu, E.X. Clinical analysis of the effect of Pien Tze Huang in treatment of 42 patients with moderate or advanced liver cancer. Shanghai J. Tradit. Chin. Med. 1994, 12, 4–5. [Google Scholar]
  5. Gu, Z.X. Preliminary Therapeutical observation of advanced colon cancer. Chin. Tradit. Pat. Med. 1993, 15, 23. [Google Scholar]
  6. Lai, C.J.; Tan, T.; Zeng, S.L.; Qi, L.W.; Liu, X.G.; Dong, X.; Li, P.; Liu, E.H. An integrated high resolution mass spectrometric data acquisition method for rapid screening of saponins in Panax notoginseng (Sanqi). J. Pharm. Biomed. Anal. 2015, 109, 184–191. [Google Scholar] [CrossRef] [PubMed]
  7. Oh, S.R.; Lee, J.P.; Chang, S.Y.; Shin, D.H.; Ahn, K.S.; Min, B.S.; Lee, H.K. Androstane alkaloids from musk of Moschus moschiferus. Chem. Pharm. Bull. 2002, 50, 663–664. [Google Scholar] [CrossRef] [PubMed]
  8. Kong, W.J.; Xing, X.Y.; Xiao, X.H.; Wang, J.B.; Zhao, Y.L.; Yang, M.H. Multi-component analysis of bile acids in natural Calculus bovis and its substitutes by ultrasound-assisted solid-liquid extraction and UPLC-ELSD. Analyst 2012, 137, 5845–5853. [Google Scholar] [CrossRef] [PubMed]
  9. Qiao, X.; Ye, M.; Pan, D.L.; Miao, W.J.; Xiang, C.; Han, J.; Guo, D.A. Differentiation of various traditional Chinese medicines derived from animal bile and gallstone: Simultaneous determination of bile acids by liquid chromatography coupled with triple quadrupole mass spectrometry. J. Chromatography. A 2011, 1218, 107–117. [Google Scholar] [CrossRef] [PubMed]
  10. Huang, M.; Xu, W.; Zhang, Y.; Liu, J.; Zhang, X.; Lin, J.; Peng, J.; Lu, J. Identification and quantification of the anti-inflammatory constituents in Pian-Tze-Huang by liquid chromatography combined with quadrupole time-of-flight and triple quadrupole mass spectrometry. J. Chromatogr. Banalyt. Technol. Biomed. Life Sci. 2016, 1027, 27–39. [Google Scholar] [CrossRef]
  11. Huang, M.; Zhao, H.; Xu, W.; Chu, K.; Hong, Z.; Peng, J.; Chen, L. Rapid simultaneous determination of twelve major components in Pien Tze Huang by ultra-performance liquid chromatography coupled with triple quadrupole mass spectrometry. J. Sep. Sci. 2013, 36, 3866–3873. [Google Scholar] [CrossRef]
  12. Li, S.X.; Mu, Y.; Zheng, F.Y. Influence of gastrointestinal digestion and edible plant combination on oral bioavailability of triterpene saponins, using a biomimetic digestion and absorption system and determination by HPLC. J. Agric. Food Chem. 2013, 61, 10599–10603. [Google Scholar] [CrossRef] [PubMed]
  13. Xu, W.; Zhang, Y.; Zhou, C.; Tai, Y.; Zhang, X.; Liu, J.; Sha, M.; Huang, M.; Zhu, Y.; Peng, J.; et al. Simultaneous quantification six active compounds in rat plasma by UPLC-MS/MS and its application to a pharmacokinetic study of Pien-Tze-Huang. J. Chromatogr. Banalyt. Technol. Biomed. Life Sci. 2017, 1061–1062, 314–321. [Google Scholar] [CrossRef] [PubMed]
  14. Pang, X.; Xie, M. The determination of ginsenosides Rg1 in Pien Tze Huang by thin-layer chromatography. Chin. Herb. Med. 1997, 28, 536–537. [Google Scholar]
  15. Lai, Y.J.; Chen, J.P.; You, Y.J. Determination of Three Compounds in Pien Tze Huang by HPLC. Chin. Pharm. J. 2008, 43, 1906–1907. [Google Scholar]
  16. Zhang, J.; Ding, L.; Wang, B.; Ren, G.; Sun, A.; Deng, C.; Wei, X.; Mani, S.; Wang, Z.; Dou, W. Notoginsenoside R1 attenuates experimental inflammatory bowel disease via pregnane X receptor activation. J. Pharmacol. Exp. Ther. 2015, 352, 315–324. [Google Scholar] [CrossRef] [PubMed]
  17. Ning, C.; Gao, X.; Wang, C.; Huo, X.; Liu, Z.; Sun, H.; Yang, X.; Sun, P.; Ma, X.; Meng, Q.; et al. Hepatoprotective effect of ginsenoside Rg1 from Panax ginseng on carbon tetrachloride-induced acute liver injury by activating Nrf2 signaling pathway in mice. Environ. Toxicol. 2018, 33, 1050–1060. [Google Scholar] [CrossRef]
  18. Paul, S.; Shin, H.S.; Kang, S.C. Inhibition of inflammations and macrophage activation by ginsenoside-Re isolated from Korean ginseng (Panax ginseng C.A. Meyer). Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2012, 50, 1354–1361. [Google Scholar] [CrossRef] [PubMed]
  19. Jang, H.J.; Han, I.H.; Kim, Y.J.; Yamabe, N.; Lee, D.; Hwang, G.S.; Oh, M.; Choi, K.C.; Kim, S.N.; Ham, J.; et al. Anticarcinogenic effects of products of heat-processed ginsenoside Re, a major constituent of ginseng berry, on human gastric cancer cells. J. Agric. Food Chem. 2014, 62, 2830–2836. [Google Scholar] [CrossRef]
  20. Lim, K.H.; Lim, D.J.; Kim, J.H. Ginsenoside-Re ameliorates ischemia and reperfusion injury in the heart: A hemodynamics approach. J. Ginseng Res. 2013, 37, 283–292. [Google Scholar] [CrossRef]
  21. Liu, S.; Cheng, Y.; Rao, M.; Tang, M.; Dong, Z. Muscone Induces CYP1A2 and CYP3A4 Enzyme Expression in L02 Human Liver Cells and CYP1A2 and CYP3A11 Enzyme Expression in Kunming Mice. Pharmacology 2017, 99, 205–215. [Google Scholar] [CrossRef]
  22. Fernandez-Moriano, C.; Gonzalez-Burgos, E.; Iglesias, I.; Lozano, R.; Gomez-Serranillos, M.P. Evaluation of the adaptogenic potential exerted by ginsenosides Rb1 and Rg1 against oxidative stress-mediated neurotoxicity in an in vitro neuronal model. PLoS ONE 2017, 12, e0182933. [Google Scholar] [CrossRef]
  23. Lahiani, M.H.; Eassa, S.; Parnell, C.; Nima, Z.; Ghosh, A.; Biris, A.S.; Khodakovskaya, M.V. Carbon nanotubes as carriers of Panax ginseng metabolites and enhancers of ginsenosides Rb1 and Rg1 anti-cancer activity. Nanotechnology 2017, 28, 015101. [Google Scholar] [CrossRef]
  24. Radad, K.; Gille, G.; Moldzio, R.; Saito, H.; Ishige, K.; Rausch, W.D. Ginsenosides Rb1 and Rg1 effects on survival and neurite growth of MPP+-affected mesencephalic dopaminergic cells. J. Neural Transm. (Vienna Austria 1996) 2004, 111, 37–45. [Google Scholar] [CrossRef]
  25. Du, Y.; Gu, X.; Meng, H.; Aa, N.; Liu, S.; Peng, C.; Ge, Y.; Yang, Z. Muscone improves cardiac function in mice after myocardial infarction by alleviating cardiac macrophage-mediated chronic inflammation through inhibition of NF-kappaB and NLRP3 inflammasome. Am. J. Transl. Res. 2018, 10, 4235–4246. [Google Scholar]
  26. Shen, A.; Hong, F.; Liu, L.; Lin, J.; Wei, L.; Cai, Q.; Hong, Z.; Peng, J. Pien Tze Huang inhibits the proliferation of human colon carcinoma cells by arresting G1/S cell cycle progression. Oncol. Lett. 2012, 4, 767–770. [Google Scholar] [CrossRef]
  27. Liang, Q.Q.; Zhang, M.; Zhou, Q.; Shi, Q.; Wang, Y.J. Muscone protects vertebral end-plate degeneration by antiinflammatory property. Clin. Orthop. Relat. Res. 2010, 468, 1600–1610. [Google Scholar] [CrossRef]
  28. Lu, D.; Liu, J.; Zhao, W.; Li, P. Chronic toxicity of ginsenoside Re on Sprague-Dawley rats. J. Ethnopharmacol. 2012, 144, 656–663. [Google Scholar] [CrossRef]
  29. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  30. Lin, J.M.; Wei, L.H.; Chen, Y.Q.; Liu, X.X.; Hong, Z.F.; Sferra, T.J.; Peng, J. Pien Tze Huang induced apoptosis in human colon cancer HT-29 cells is associated with regulation of the Bcl-2 family and activation of caspase 3. Chin. J. Integr. Med. 2011, 17, 685–690. [Google Scholar] [CrossRef]
  31. Zhuang, Q.; Hong, F.; Shen, A.; Zheng, L.; Zeng, J.; Lin, W.; Chen, Y.; Sferra, T.J.; Hong, Z.; Peng, J. Pien Tze Huang inhibits tumor cell proliferation and promotes apoptosis via suppressing the STAT3 pathway in a colorectal cancer mouse model. Int. J. Oncol. 2012, 40, 1569–1574. [Google Scholar] [CrossRef]
  32. Shen, A.; Chen, Y.; Hong, F.; Lin, J.; Wei, L.; Hong, Z.; Sferra, T.J.; Peng, J. Pien Tze Huang suppresses IL-6-inducible STAT3 activation in human colon carcinoma cells through induction of SOCS3. Oncol. Rep. 2012, 28, 2125–2130. [Google Scholar] [CrossRef] [Green Version]
  33. Wan, Y.; Shen, A.; Qi, F.; Chu, J.; Cai, Q.; Sferra, T.J.; Peng, J.; Chen, Y. Pien Tze Huang inhibits the proliferation of colorectal cancer cells by increasing the expression of miR-34c-5p. Exp. Ther. Med. 2017, 14, 3901–3907. [Google Scholar] [CrossRef] [Green Version]
  34. Shen, A.; Chen, H.; Chen, Y.; Lin, J.; Lin, W.; Liu, L.; Sferra, T.J.; Peng, J. Pien Tze Huang Overcomes Multidrug Resistance and Epithelial-Mesenchymal Transition in Human Colorectal Carcinoma Cells via Suppression of TGF-beta Pathway. Evid.-Based Complement. Altern. Med. 2014, 2014, 679436. [Google Scholar] [CrossRef]
  35. Chen, Z.; Shen, A.; Liu, L.; Chen, Y.; Chu, J.; Cai, Q.; Qi, F.; Sferra, T.J.; Peng, J. Pien Tze Huang induces apoptosis and inhibits proliferation of 5-fluorouracil-resistant colorectal carcinoma cells via increasing miR-22 expression. Exp. Ther. Med. 2017, 14, 3533–3540. [Google Scholar] [CrossRef]
  36. Fu, C.; Chu, J.; Shen, A.; Liu, L.; Chen, H.; Lin, J.; Sferra, T.J.; Chen, Y.; Peng, J. Pien Tze Huang alleviates 5-fluorouracil-induced intestinal mucositis in CT-26 tumor-bearing mice. Exp. Ther. Med. 2017, 14, 2291–2297. [Google Scholar] [CrossRef] [Green Version]
  37. Lin, M.H.; Zhu, D.Z. Clinical observation of Pien Tze Huang combined with chemotherapy in the treatment of advacnced colon cancer. Fujian J. TCM Fubruary 2012, 43, 8–9. [Google Scholar]
  38. Huang, Z.; Zi, Y.; Lu, H. Clinical observation of Pien Tze Huang combined with XELOX chemotherapy regimen in the treatment of advacnced colon cancer. Fujian J. TCM Fubruary 2014, 45, 30–31. [Google Scholar]
  39. Shen, A.L.; Hong, F.; Liu, L.Y.; Lin, J.M.; Zhuang, Q.C.; Hong, Z.F.; Peng, J. Effects of Pien Tze Huang on angiogenesis in vivo and in vitro. Chin. J. Integr. Med. 2012, 18, 431–436. [Google Scholar] [CrossRef]
  40. Li, L.; Shen, A.; Chu, J.; Sferra, T.J.; Sankararaman, S.; Ke, X.; Chen, Y.; Peng, J. Pien Tze Huang ameliorates DSS-induced colonic inflammation in a mouse colitis model through inhibition of the IL-6/STAT3 pathway. Mol. Med. Rep. 2018, 1113–1119. [Google Scholar] [CrossRef]
  41. Shen, A.; Lin, W.; Chen, Y.; Liu, L.; Chen, H.; Zhuang, Q.; Lin, J.; Sferra, T.J.; Peng, J. Pien Tze Huang inhibits metastasis of human colorectal carcinoma cells via modulation of TGF-beta1/ZEB/miR-200 signaling network. Int. J. Oncol. 2015, 46, 685–690. [Google Scholar] [CrossRef]
  42. 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-beta signaling in an orthotopic model of colorectal cancer. Oncol. Rep. 2015, 33, 1922–1928. [Google Scholar] [CrossRef]
  43. Chen, H.; Shen, A.; Zhang, Y.; Chen, Y.; Lin, J.; Lin, W.; Sferra, T.; Peng, J. Pien Tze Huang inhibits hypoxia-induced epithelial-mesenchymal transition in human colon carcinoma cells through suppression of the HIF-1 pathway. Exp. Ther. Med. 2014, 7, 1237–1242. [Google Scholar] [CrossRef]
  44. Lin, J.; Feng, J.; Jin, Y.; Yan, Z.; Lai, Z.; Peng, J. Pien Tze Huang suppresses VEGF-C-mediated lymphangiogenesis in colorectal cancer. Oncol. Rep. 2016, 36, 3568–3576. [Google Scholar] [CrossRef] [Green Version]
  45. Chen, H.; Feng, J.; Zhang, Y.; Shen, A.; Chen, Y.; Lin, J.; Lin, W.; Sferra, T.J.; Peng, J. Pien Tze Huang Inhibits Hypoxia-Induced Angiogenesis via HIF-1 alpha /VEGF-A Pathway in Colorectal Cancer. Evid.-Based Complement. Altern. Med. 2015, 2015, 454279. [Google Scholar] [CrossRef]
  46. Shen, A.; Lin, J.; Chen, Y.; Lin, W.; Liu, L.; Hong, Z.; Sferra, T.J.; Peng, J. Pien Tze Huang inhibits tumor angiogenesis in a mouse model of colorectal cancer via suppression of multiple cellular pathways. Oncol. Rep. 2013, 30, 1701–1706. [Google Scholar] [CrossRef] [Green Version]
  47. Wei, L.; Chen, P.; Chen, Y.; Shen, A.; Chen, H.; Lin, W.; Hong, Z.; Sferra, T.J.; Peng, J. Pien Tze Huang suppresses the stem-like side population in colorectal cancer cells. Mol. Med. Rep. 2014, 9, 261–266. [Google Scholar] [CrossRef]
  48. Qi, F.; Wei, L.; Shen, A.; Chen, Y.; Lin, J.; Chu, J.; Cai, Q.; Pan, J.; Peng, J. Pien Tze Huang inhibits the proliferation, and induces the apoptosis and differentiation of colorectal cancer stem cells via suppression of the Notch1 pathway. Oncol. Rep. 2016, 35, 511–517. [Google Scholar] [CrossRef]
  49. Wei, L.H.; Qi, F.; Peng, J.; Yan, M.L.; Qiu, F.N. Effects of pien tze huang on proliferation and apoptosis of liver cancer stem cells. Fujian J. TCM 2017, 48, 27–30. [Google Scholar]
  50. Guo, P.L.; Liu, J.H.; Yu, B.Y. Study on antitumor effect of Pien Tze Huang in vitro. Drug Eval. Res. 2011, 34, 171–173. [Google Scholar]
  51. Fu, Y.; Zhang, L.; Hong, Z.; Zheng, H.; Li, N.; Gao, H.; Chen, B.; Zhao, Y. Methanolic Extract of Pien Tze Huang Induces Apoptosis Signaling in Human Osteosarcoma MG63 Cells via Multiple Pathways. Molecules 2016, 21, 283. [Google Scholar] [CrossRef]
  52. Ren, S.S.; Yuan, F.; Liu, Y.H.; Zhou, L.T.; Li, J. Effect of p27 gene combined with Pientzehuang on tumor growth in osteosarcoma-bearing nude mice. Chin. J. Integr. Med. 2015, 21, 830–836. [Google Scholar] [CrossRef]
  53. Zhang, Y.; Wang, Q.H.; Niu, S.S.; Liu, J.N.; Zhang, L. Pien Tze Huang induces apoptosis in multi-drugresistant U2OS/ADM cells via downregulation of Bcl2, survivin and P-gp and upregulation of Bax. Oncology reports 2014, 31, 763–770. [Google Scholar] [CrossRef]
  54. Lu, L.; Wai, M.S.; Yew, D.T.; Mak, Y.T. Pien Tze Huang, a composite Chinese traditional herbal extract, affects survival of neuroblastoma cells. Int. J. Neurosci. 2009, 119, 255–262. [Google Scholar] [CrossRef]
  55. He, F.; Wu, H.N.; Cai, M.Y.; Li, C.P.; Zhang, X.; Wan, Q.; Tang, S.B.; Cheng, J.D. Inhibition of ovarian cancer cell proliferation by Pien Tze Huang via the AKT-mTOR pathway. Oncol. Lett. 2014, 7, 2047–2052. [Google Scholar] [CrossRef] [Green Version]
  56. Chen, X.; Qi, F.; Shen, A.L.; Chu, J.F.; Sferra, T.J.; Chen, Y.Q.; Peng, J. Pien Tze Huang Overcomes Doxorubicin Resistance and Inhibits Epithelial-Mesenchymal Transition in MCF-7/ADR Cells. Chin. J. Integr. Med. 2018. [Google Scholar] [CrossRef]
  57. Yang, Y.; Chen, Z.; Deng, L.; Yu, J.; Wang, K.; Zhang, X.; Ji, G.; Li, F. Pien Tze Huang ameliorates liver injury by inhibiting the PERK/eIF2alpha signaling pathway in alcohol and high-fat diet rats. Acta Histochem. 2018, 120, 578–585. [Google Scholar] [CrossRef]
  58. Deng, L.Y.; Li, F.H.; Ji, G.; Cao, Y.C.; Zhang, X. Effect Study of Pien Tze Huang on nonalcoholic fatty liver disease in rats. Chin. J. Exp. Tradit. Med. Formulae 2015, 21, 124–128. [Google Scholar]
  59. Zhang, L.; Lam, W.; Lü, L.; Wang, C.; Wong, Y.; Lam, L.; Tang, H.; Wai, M.; Wang, M.; Kwong, W.; et al. Protective effects and potential mechanisms of Pien Tze Huang on cerebral chronic ischemia and hypertensive stroke. Chin. Med. 2010, 5. [Google Scholar] [CrossRef]
  60. Zhang, L.; Lam, W.P.; Lu, L.; J Wang, Y.X.J.; W Wong, Y.; H Lam, L.; C Tang, H.; S Wai, M.; T Mak, Y.; Wang, M.; et al. How Would Composite Traditional Chinese Medicine Protect the Brain—An Example of the Composite Formula “Pien Tze Huang”. Curr. Med. Chem. 2011, 18, 3590–3594. [Google Scholar] [CrossRef]
  61. Zhang, X.; Zhang, Y.; Tang, S.; Yu, L.; Zhao, Y.; Ren, Q.; Huang, X.; Xu, W.; Huang, M.; Peng, J. Pien-Tze-Huang protects cerebral ischemic injury by inhibiting neuronal apoptosis in acute ischemic stroke rats. J. Ethnopharmacol. 2018, 219, 117–125. [Google Scholar] [CrossRef]
  62. Xu, Y.Y.; Yu, E.X. Clinical analysis of 42 cases of advanced liver cancer treated with Pien Tze Huang. Shanghai J. Tradit. Chin. Med. 1994, 4–5. [Google Scholar]
  63. Zhao, S.L.; Pan, J. A clinical trial of combined use of Pien Tze Huang and chemotherapy in the treatment of primary liver cancer. Med. World 2006, 49–51. [Google Scholar]
  64. Meng, L.P.; Gu, X.F. Clinical observation of the changes of immune indexes of primary liver cancer treated with Pien Tze Huang and TACE. Jiu Jiang Med. J. 2008, 1, 31–33. [Google Scholar]
  65. Zhong, R.; Duan, Z.P.; Chen, Y.; Zhen, S.J.; Kong, M.; Xu, M.M. Clinical study of Pien tze huang in treatment of chronic hepatitis B. Chin. J. Gastroenterol. Hepatol. 2017, 26, 1290–1294. [Google Scholar]
  66. He, Y.L.; Chen, H.M.; Zhou, H.A. Clinical study of Pianzaihuang in the treatment of recurrent aphthous ulcer. Chin J. Clin. Pharm. 2015, 31, 780–783. [Google Scholar]
  67. Lin, G.Q.; Chen, Q.Q.; Zeng, G.Q.; Chen, W.L. Clinical observation of Pianzaihuang in the treatment of recurrent aphthous ulcer. Shenzheng J. Integr. Med. 2017, 27, 21–24. [Google Scholar]
  68. Lian, C.Y.; Liu, S.F.; Chen, N.Q. Clinical observation of Pien Tze Huang in the treatment of phlebitis induced by amiodarone. Today Nurse 2012, 3, 94–96. [Google Scholar]
Figure 1. Chemical structures of 27 compounds identified from Pien-Tze-Huang (PTH).
Figure 1. Chemical structures of 27 compounds identified from Pien-Tze-Huang (PTH).
Molecules 24 03274 g001
Table 1. The anti-proliferative activities of Pien-Tze-Huang (PTH) in vitro.
Table 1. The anti-proliferative activities of Pien-Tze-Huang (PTH) in vitro.
Cancer TypesCell LinesIC50Reference
Colorectal cancerHT-290.65 mg/mL/24 h[30]
Caco-2>1 mg/mL/24 h[26]
CT-260.75 mg/mL/24 h[42]
HCT-80.25-0.5 mg/mL/6 h[43]
0.25-0.5 mg/mL/24 h[41]
HCT-8/5-Fu>0.25 mg/mL/48 h[34]
SW480 SP0.75 mg/mL/48 h[48]
HCT-1160.75 mg/mL/24 h[44]
0.5-0.75 mg/mL/48 h[44]
SW620<0.75 mg/mL/24 h[44]
>0.5 mg/mL/48 h[44]
NeuroblastomaSH-SY5Y<400 μg/mL/24 h[54]
NIH-3T3>400 μg/mL/24 h[54]
OsteosarcomaU2OS1.2 mg/mL/48 h[53]
U2OS/ADM>1.2 mg/mL/24 h[53]
<1.2 mg/mL/48 h[53]
0.8-1.2 mg/mL/72 h[53]
MG-63500-750 μg/mL/24 h[51]
Ovarian cancerOVCAR-3>1000 mg/mL/24 h[55]
Liver cancerBEL-7402>0.75 mg/mL/48 h[49]
0.5-0.57 mg/mL/72 h[49]
Breast cancerMCF-7/ADM>0.75 mg/mL/48 h[56]
Note: IC50, half maximal inhibitory concentration; 5-Fu, fluorouracil; SP, side population; ADM, adriamycin.
Table 2. The pharmacological activities and mechanisms of PTH in vivo.
Table 2. The pharmacological activities and mechanisms of PTH in vivo.
Disease TypeAnimal ModelMethodEffectsMechanismsReference
Colorectal cancerHT-29 tumor-bearing xenograft mice234 mg/kg/d, 5 days a week for 16 daysreduced tumor volume and tumor weight without apparent adverse effect, inhibited tumor cell proliferation, promoted apoptosis↑Bax, Bax/Bcl-2;
↓Cyclin D1, CDK4, Bcl-2, p-STAT3
[31]
CT-26 tumor-bearing xenograft mice250 mg/kg/d for 4 daysalleviated the severity of 5-FU-induced diarrhea and morphological intestinal damages, inhibited cell apoptosis in the intestinal crypt↑Bax, Bax/Bcl-2;
↓Bcl-2,
[36]
HT-29 tumor-bearing xenograft mice234 mg/kg/d, 5 days a week for 16 dayssuppressed tumor volume and weight, inhibited tumor angiogenesis↓p-STAT3, p-ERK, p-Akt, p-JNK, p-p38, VEGF-A, VEGFR2, bFGF, bFGFR, iNOS, eNOS[46]
CT-26 cells liver metastasis animal model in athymic male nude mice234 mg/kg/d for 14 daysinhibited tumor liver metastasis without apparent toxicity, inhibited EMT↑E-cadherin;
↓N-cadherin, TGF-β, p-Smad2/3, Smad4
[42]
OsteosarcomaSaos-2 tumor-bearing xenograft mice0.234 mg/g twice daily for 6 weeks combined with p27 gene once for 3 daysinhibited tumor growth↑p27[52]
Hepatic diseasesacute hepatitis mouse induced by carbon tetrachloride 0.5 g/kg three times over 36 hameliorated hepatic pathology↑AP1, NF-κB;
↓ALT
[2]
alcohol and high-fat diet rats0.5, 1, and 2g/kg/d for 3 weeksameliorated the defects in hepatic function, hepatic pathology and the impairment in lipid metabolism↓GRP78, GRP9, p-eIF2a[57]
nonalcoholic fatty liver rats0. 5, 1.0, 2.0 g/kg/d for 4 weeksimproved liver function, lowered blood lipid↑SREBP-1c mRNA expression;
↓FXR, SHP mRNA expression
[58]
Ischemic strokeSHR rats and stroke prone SHR rats18 mg/kg/d for 3 monthsreduced cell death in hippocampus and cerebellum by chronic ischemia and hypertensive stroke↓QCR2, cleaved caspase-3[59]
MCAO rats180 mg/kg/d for 4 daysreduced cerebral infarct volume, improved neurological deficit, attenuated inflammatory response, inhibited neuronal apoptosis↑NeuN, cytosolic Cyt C, Bax, P53,cleaved caspase-3;
↓IL-1β, IL-6, TNF-α, Bcl-xl, p-AKT, p-GSK-3β, caspase-9, mitochondrial Cyt C
[61]
Note:↑, upregulation; ↓, downregulation; CRC, colorectal cancer; Bcl-2, B cell lymphoma/lewkmia-2; Bax, Bcl-2 associated X protein; CDK, cyclin-dependent kinase; p-STAT3, phosphor-signal transducer and activator of transcription 3; p-ERK, phosphor-endoplasmic reticulum kinase; p-JNK, phosphor-c-Jun N-terminal kinase; VEGF-A, vascular endothelial growth factor-A; VEGFR2, vascular endothelial growth factor receptor 2; bFGF, basic fibroblast growth factor; bFGFR, basic fibroblast growth factor receptor; iNOS, inducible nitric oxide synthase; eNOS, endothelial nitric oxide synthase; TGF-β1, transforming growth factor-β1; Smad, drosophila mothers against decapentaplegic protein; AP1, anti-microbial protein 1; NF-κB, nuclear factor kappa-B; ALT, alanine aminotransferase; GRP, glucose-regulated protein; p-eIF2α, phosphor-eukaryotic translation initiation factor 2α; SREBP-1c, sterol regulatory element binding protein-1c; FXR, farnesoid X receptor; SHP, small heterodimer partner; SHR, spontaneously hypertensive rat; QCR2, Cytochrome b-c1 complex subunit 2; caspase-3, cysteine aspartic protease 3; MCAO, middle cerebral artery occlusion; NeuN, nuclear antigen neuron; IL-1β, interleukin-1β; IL-6: interleukin-6; TNF-α, tumor necrosis factor α; p-GSK-3β, phosphor-glycogen synthase kinase 3β; caspase-9, cysteine aspartic protease 9; Cyt C, cytochrome C.
Table 3. Clinical application of PTH on various diseases.
Table 3. Clinical application of PTH on various diseases.
Disease TypeDose and Course of TreatmentCombined MedicationPTH/Control (n)EfficacySide EffectsReference
Advanced colorectal cancer1.8 g/d, p.o., 28 days-25/0improved clinical symptoms and life qualityN/A[5]
1.2 g/d, p.o., 24 weeks5-FU (300 mg/m2/d, i.v., 2 days), oxaliplatin (85 mg/m2/d, i.v., 2 days), leucovorin (200 mg/m2/d, i.v., 1 day)24/23had better short-term efficacy, reduced the toxicity and side effects of chemotherapy, and improved life qualitiesN/A[38]
1.2 g/d, p.o., 24 weeksoxaliplatin (130 mg/m2/d, i.v., 1 day) and capecitabine (1000 mg/m2/d, p.o.)34/34improved life quality, reduced the toxicity and side effects of chemotherapyN/A[37]
Moderate or advanced liver cancer1.0 g/d, p.o.,30 daysroutine radiotherapy or chemotherapy42/0achieved lower exacerbation rate, improved clinical symptoms and life quality compared with single chemotherapyN/A[62]
Primary liver cancer1.8 g/d, p.o.,56 daysroutine interventional chemotherapy102/105reduced tumor size, improved life quality, relieved pain, reduced the toxicity and side effects of chemotherapyN/A[63]
1.8 g/d, p.o.,56 daystranscatheter arterial chemoembolization20/20increased NK, CD3, CD4 and CD4/CD8 levelsN/A[64]
Chronic hepatitis B1.8 g/d, p.o., 3 monthspolyenephosphatidylcholine (1368 mg/d, p.o.)47/47improved digestive tract symptoms, protected liver functionN/A[65]
Recurrent aphthous ulcer1.5 g/d, p.o.Niuhuang Jiedu Pills (2.43 g/d, p.o.)31/35accelerated the healing of recurrent aphthous ulcer, decreased the risk of recurrenceN/A[66]
2.13 g/d, p.o.Niuhuang Jiedu Pills (2.13 g/d, p.o.)37/37accelerated the healing of recurrent aphthous ulcer, decreased the risk of recurrenceN/A[67]
Phlebitis induced by amiodarone0.75 g/d, external treatment, 4 days50% magnesium sulfate31/31improved redness, swelling, heat, painN/A[68]
Note: PTH, the PTH-treated group; Control, the control group; NK, natural killer cell; CD, cluster of differentiation; N/A, not available.

Share and Cite

MDPI and ACS Style

Huang, L.; Zhang, Y.; Zhang, X.; Chen, X.; Wang, Y.; Lu, J.; Huang, M. Therapeutic Potential of Pien-Tze-Huang: A Review on Its Chemical Composition, Pharmacology, and Clinical Application. Molecules 2019, 24, 3274. https://doi.org/10.3390/molecules24183274

AMA Style

Huang L, Zhang Y, Zhang X, Chen X, Wang Y, Lu J, Huang M. Therapeutic Potential of Pien-Tze-Huang: A Review on Its Chemical Composition, Pharmacology, and Clinical Application. Molecules. 2019; 24(18):3274. https://doi.org/10.3390/molecules24183274

Chicago/Turabian Style

Huang, Lili, Yiping Zhang, Xiaoqin Zhang, Xiuping Chen, Yitao Wang, Jinjian Lu, and Mingqing Huang. 2019. "Therapeutic Potential of Pien-Tze-Huang: A Review on Its Chemical Composition, Pharmacology, and Clinical Application" Molecules 24, no. 18: 3274. https://doi.org/10.3390/molecules24183274

Article Metrics

Back to TopTop