Category: Moms

Cancer-fighting effects of herbal extracts

Cancer-fighting effects of herbal extracts

Park J-S, Yeom Cancer-fighying, Park W-S, Joo Recovery for couples, Rho Effect, Kim DH, Increase cognitive endurance xetracts. Individual therapies Find detailed information and research into some of the many different complementary and alternative therapies used by people with cancer. Register your interest. These studies are not mesothelioma-focused but may still interest patients. Lu, Y. Navari, R. Lung Cancer 68, —

Cancer-fighting effects of herbal extracts -

However, the anti-cancer targets of these pharmacodynamic compounds are still not clear, and this is the major obstacle for the application and development of Chinese herbal medicine. This review in Chinese herbal medicine and cancer focuses on summarizing experimental results and conclusions from English literatures reported since gov using the following keywords: Cancer, Tumor, Neoplasm, Chinese herbs, Chinese medicine, Herbal medicine.

To provide new insights into the critical path ahead, the pharmacological effects, novel mechanism of action, relevant clinical studies, innovative applications in combined therapy, and immunomodulation of the popular compounds originated from Chinese herbal medicine were reviewed systemically.

Different natural products derived from Chinese herbal medicine, including curcumin, EGCG, berberine, artemisinins, ginsenosides, ursolic acid UA , silibinin, emodin, triptolide, cucurbitacins, tanshinones, ordonin, shikonin, gambogic acid GA , artesunate, wogonin, β-elemene, and cepharanthine, were identified with emerging anti-cancer activities, such as anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as autophagy regulation, multidrug resistance reversal, immunity balance, and chemotherapy improvement in vitro and in vivo.

These compounds are considered popular with over supported publications and are selected to be discussed in more details.

Figure 1 shows the word cloud of these compounds. In this review, the advantages and drawbacks of representative Chinese herbal medicine-derived compounds in different types of cancers were also highlighted and summarized.

The anti-cancer compounds from Chinese herbal medicine CHM. Curcumin Fig. with many biological activities, but it has poor water solubility and stability [ 11 ]. Clinical evidence and extensive studies showed that curcumin has various pharmacology effects, including anti-cancer, anti-inflammatory, and anti-oxidative activities [ 12 , 13 , 14 ].

Curcumin and its analogues are shown to be emerging as effective agents for the treatment of several malignant diseases such as cancer. Numerous studies have shown that curcumin and its preparations can inhibit tumors in almost all parts of the body, including head and neck, ovarian, skin and gastric cancers [ 15 , 16 , 17 , 18 , 19 , 20 ].

Curcumin is shown to exhibit many anti-cancer effects through the inhibition of cell proliferation, promotion of cell apoptosis, prevention of tumor angiogenesis and metastasis, and the induction of autophagy [ 21 , 22 , 23 , 24 , 25 ].

Curcumin inhibits cell growth, induces cell cycle arrest and apoptosis in esophageal squamous cell carcinoma EC1, EC, KYSE, TE13 cells through STAT3 activation [ 12 ]. It also induces oxidative stress, which disrupts the mitochondrial membrane potential and causes the release of cytochrome c, thus inducing apoptosis [ 26 ].

Besides, curcumin is shown to induce autophagy [ 8 , 21 , 27 , 28 , 29 , 30 ]. Moreover, curcumin can ameliorate Warburg effect in human non-small cell lung cancer NSCLC H, breast cancer MCF-7, cervical cancer HeLa and prostate cancer PC-3 cells through pyruvate kinase M2 down-regulation, a key regulator of Warburg effect [ 18 ].

In addition, tumor metastasis has always been a frustrating problem for anti-cancer therapy, and curcumin also exhibits anti-metastasis effects [ 31 , 32 , 33 , 34 , 35 ]. By pulmonary administration of curcumin in mice, it overcomes the problem of its low bioavailability, and inhibits lung metastasis of melanoma [ 35 ].

The main target molecules and signaling involved in the pharmacological processes include reactive oxygen species ROS , matrix metalloproteinases MMPs , nuclear factor kappa-light-chain-enhancer of activated B cells NF-κB , signal transducer and activator of transcription and cell cycle-related proteins [ 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 ].

In addition, mammalian target of rapamycin mTOR plays a vital role in curcumin-induced autophagy and apoptosis [ 30 , 48 , 49 , 50 ]. Curcumin can also exert immunomodulatory effects against cancer cells.

Theracurmin, a highly bioavailable form of curcumin, decreases pro-inflammatory cytokine secretion from activated T cells, and enhances T cell-induced cytotoxicity in human esophageal adenocarcinoma OE33 and OE19 cells, so it increases the sensitivity of the cells to T cell-induced cytotoxicity [ 51 ].

The natural killing NK cells can directly kill cancer cells, and curcumin can enhance the cytotoxicity effect of NK cells when NK cells are co-cultured with human breast cancer MDA-MB cells, which is highly associated with signal transducer and activator of transcription 4 STAT4 and signal transducer and activator of transcription 5 STAT5 activation [ 52 ].

Besides, myeloid-derived suppressor cells MDSCs are immune-suppressive cells which are found in most cancer patients. Curcumin decreases interleukin IL -6 levels in the tumor tissues and serum of Lewis lung carcinoma LLC -bearing mice to impair the growth of MDSCs, so targeting MDSCs is important for the treatment of lung cancer [ 13 ].

In order to overcome the solubility issues of curcumin and facilitate its intracellular delivery, a curcumin-loaded nanoparticle, curcumin-PLGA-NP, is synthesized.

It has a tenfold increase in water solubility compared to curcumin, and shows threefold increased anti-cancer activities in human breast cancer MDA-MB and NSCLC A cells [ 56 ].

Another curcumin-capped nanoparticle exhibits promising anti-oxidative and selective anti-cancer activities in human colorectal cancer HT and SW cells [ 57 ]. Moreover, a curcumin analog, WZ35, has high chemical stability, and higher efficacy in anti-cancer effects compared to curcumin in human gastric cancer SGC cells and SGC xenograft mice [ 20 ].

Another analog, B63, induces cell death and reduces tumor growth through ROS and caspase-independent paraptosis in human gastric cancer SGC, BGC and SNU cells, 5-fluorouracil-resistant gastric cancer cells, and SGC xenograft mice [ 58 ]. Curcumin can be used with other chemotherapeutic agents to achieve synergistic effects, reduce adverse effects and enhance sensitivity.

Tamoxifen and curcumin are packed into a diblocknanopolymer, and this nanopolymer reduces the toxicity of tamoxifen in normal cells and exhibits better anti-proliferative and pro-apoptotic effects in human breast cancer tamoxifen-sensitive and -resistant MCF-7 cells [ 59 ].

Triptolide has strong liver and kidney toxicities, and when combined with curcumin, they exert synergistic anti-cancer effects in ovarian cancer, as well as reduce the side effects of triptolide [ 60 ].

In addition, adriamycin, sildenafil, 5-fluorouracil, irinotecan, doxorubicin, paclitaxel, sorafenib, Kruppel-like factor 4, emodin, docosahexaene acid and apigenin are shown to exhibit synergistic effects with curcumin [ 61 , 62 , 63 , 64 , 65 , 66 , 67 , 68 , 69 , 70 , 71 ].

Similarly, copper supplementation significantly enhances the anti-tumor effects of curcumin in several oral cancer cells [ 72 ], while epigallocatechingallic acid ester EGCG increases the ability of curcumin to inhibit cell growth and induce apoptosis in human uterine leiomyosarcoma SKN cells [ 73 ].

Clinical trials can confirm or reveal the effects, adverse reactions and pharmacokinetics of the drugs. As the bioavailability of curcumin is very poor, many curcumin preparations are synthesized and tested in clinical trials [ 74 , 75 , 76 ].

A phase I study was conducted to investigate the safety and pharmacokinetics of theracurmin in pancreatic and biliary tract cancer patients who failed with standard chemotherapy [ 76 ]. They administered theracurmin every day with standard gemcitabine-based chemotherapy.

No new adverse effects and no increase in the incidence of adverse effects were observed among these patients. This study has provided additional evidence for a high response rate and better tolerability with the use of curcumin during cancer therapy [ 77 ].

EGCG, also known as epigallocatechingallate Fig. Epidemiological studies have indicated that consumption of green tea has potential impact of reducing the risk of many chronic diseases, such as cardiovascular diseases and cancer [ 78 , 79 ].

EGCG possesses various biological effects including anti-obesity and anti-hyperuricemia, anti-oxidative, anti-viral, anti-bacterial, anti-infective, anti-angiogenic, anti-inflammatory and anti-cancer activities [ 80 , 81 , 82 , 83 , 84 ]. It is reported to present anti-cancer effects in variety of cancer cells, including lung, colorectal, prostate, stomach, liver, cervical, breast, leukemia, gastric, bladder cancers [ 85 , 86 , 87 , 88 , 89 , 90 ].

Among its anti-cancer activities, EGCG exhibits multiple pharmacological actions, including the suppression of cell growth, proliferation, metastasis and angiogenesis, induction of apoptosis, and enhancement of anti-cancer immunity [ 85 , 86 , 91 , 92 , 93 , 94 ].

EGCG can inhibit cell proliferation through multiple ways in many types of cancer cells. It inhibits cell proliferation in human bladder cancer SW, breast cancer MDA-MB and NSCLC A cells, and inhibits tumor growth in gastric cancer SGC xenograft mice [ 89 , 94 , 95 ].

It also induces apoptosis in human oral cancer KB, head and neck cancer FaDu, NSCLC A, and breast cancer MCF-7 cells [ 96 , 97 ]. Besides, EGCG induces autophagy, and inhibition of autophagy can enhance EGCG-induced cell death in human mesothelimoa ACC-meso, Y-meso, EHMES, EHMES-1 and MSTOH, and primary effusion lymphoma BCBL-1 and BC-1 cells [ 98 , 99 ].

In contrast, it induces cell death via apoptosis and autophagy in oral squamous cell carcinoma SCC-4 cells [ 84 ], so autophagy plays a dual role in EGCG-induced cell death. It can also suppress metastasis in human melanoma SK-MEL-5, SK-MEL, A and G, NSCLC CL, A and H cells, and lung metastasis mice [ 85 , 93 , ].

In addition, EGCG suppresses tumor angiogenesis in human NSCLC A cells and A xenograft mice [ ]. EGCG mediates apoptosis which involves pro- and anti-apoptotic proteins in various cancer cells. It up-regulates pro-apoptotic proteins such as Bclassociated X protein Bax , and down-regulates anti-apoptotic proteins including B-cell lymphoma 2 Bcl-2 , B-cell lymphoma-extra large Bcl-xL and survivin [ 97 , , , ].

ER stress also plays an important role in EGCG-induced cell death. EGCG inhibits endoplasmic reticulum ER stress-induced protein kinase R-like endoplasmic reticulum kinase PERK and eukaryotic translation-initiation factor 2α eIF2α phosphorylation [ ].

Besides, poly ADP-ribose polymerase PARP 16 is shown to activate ER stress markers, PERK and inositol-requiring enzyme 1α IRE1α [ ]. ER stress-induced apoptosis, PERK and eIF2α phosphorylation by EGCG are suppressed in PARPdeficient hepatocellular carcinoma QGY cells, so EGCG mediates apoptosis through ER stress, which is dependent on PARP16 [ ].

In addition to anti-cancer effects, EGCG shows a significant inhibitory effect on interferon-γ IFN-γ -induced indoleamine 2,3-dioxygenase IDO expression, an enzyme that guides cancer to regulate immune response, in human colorectal cancer SW cells [ ], so this suggests that EGCG might be useful for chemoprevention and colorectal cancer treatment, and could be a potential agent for anti-tumor immunotherapy.

EGCG is also found to be a potential immune checkpoint inhibitor, which down-regulates IFN-γ-induced B7 homolog 1 B7-H1 levels, an immunoglobulin-like immune suppressive molecule, in human NSCLC A cells [ ]. Although EGCG has numerous biological activities through different pathways, its efficacy demonstrated in in vivo studies is not always consistent with the results of in vitro studies.

This can be due to its low oil solubility, metabolic instability and poor bioavailability [ ]. Therefore, EGCG analogs and EGCG-loaded nanoparticles by modifying EGCG are developed, and they have been reported to enhance anti-cancer effects [ , , ].

Besides, EGCG-DHA docosahexaenoic ester, a lipophilic derivative of EGCG, shows improved anti-oxidative effects compared to EGCG, and suppresses colon carcinogenesis in mice [ , ]. These EGCG nanoparticles can improve the targeting ability and efficacy of EGCG, which greatly promote the clinical application and development of EGCG analogs.

EGCG antagonizes toxicity induced by anti-cancer chemotherapeutic agents, and sensitizes chemo-resistant cancer cells. It also exerts synergistic effects with anti-cancer agents in various cancer cells, such as cisplatin, oxaliplatin, temozolomide, resveratrol, doxorubicin, vardenafil, curcumin, erlotinib [ , , , , , , , ].

EGCG can enhance the sensitivity of cisplatin through copper transporter 1 CTR1 up-regulation, which results in the accumulation of cellular cisplatin and cisplatin—DNA adducts in human ovarian cancer SKOV3 and OVCAR3 cells, and the combination of EGCG and cisplatin suppresses tumor growth in OVCAR3 xenograft mice [ ].

The combined low concentration of EGCG and curcumin remarkably inhibits cell and tumor growth in human NSCLC A and NCI-H cells, and A xenograft mice through cell cycle arrest [ ].

To evaluate the tolerance, safety, pharmacokinetics and efficacy of EGCG in humans, clinical trials have been or are currently being conducted for cancer treatment.

During a phase I clinical trial for the treatment of radiation dermatitis, patients with breast cancer received adjuvant radiotherapy and EGCG solution. It was found that the maximum dose μM of EGCG was well tolerated and the maximum tolerated dose was undetermined [ ].

It was concluded that EGCG was effective for treating radiation dermatitis. Moreover, a phase II clinical trial was conducted to investigate the benefits of EGCG as a treatment for acute radiation-induced esophagitis ARIE for patients with stage III lung cancer.

The oral administration of EGCG was shown to be effective and phase III clinical trial to study the potential effects of EGCG to ARIE treatment was anticipated [ ]. Berberine Fig. Berberine has diverse pharmacological effects and is normally used for the treatment of gastroenteritis [ , ].

It exhibits significant anti-cancer effects in a wide spectrum of cancers including ovarian, breast, esophageal, and thyroid cancers, leukemia, multiple myeloma, nasopharyngeal carcinoma, and neuroblastoma, through inducing cell cycle arrest and apoptosis, inhibiting metastasis and angiogenesis [ , , , , , , , , ].

Berberine can induce cell cycle arrest in various cancer cells [ , , ]. However, berberine induces G1 phase arrest in human estrogen receptor positive breast cancer MCF-7 cells but not in estrogen receptor negative MDA-MB cells [ ]. Besides, it inhibits cell proliferation by inducing apoptosis in human colorectal cancer HCT-8 cells [ ].

In pnull leukemia EU-4 cells, berberine induces pindependent and X-linked inhibitor of apoptosis protein XIAP -mediated apoptosis, which is associated with mouse double minute 2 homolog MDM2 and proteasomal degradation [ ].

Mitochondrial-mediated apoptosis with Bcllike protein 11 Bim up-regulation and Forkhead box O FoxO nuclear retention is vital in berberine-induced apoptosis [ ]. In contrast, berberine induces protective autophagy in human malignant pleural mesothelioma NCI-H cells, and inhibition of autophagy promotes berberine-induced apoptosis [ ].

Therefore, autophagy plays a dual role in berberine-induced apoptosis. Furthermore, berberine also inhibits tumor migration and invasion [ , ]. It up-regulates plasminogen activator inhibitor-1 PAI-1 , a tumor suppressor that down-regulates urokinase-type plasminogen activator uPA and antagonizes uPA receptor to suppress metastasis in human hepatocellular carcinoma Bel and SMMC cells [ ].

Berberine interacts with diverse molecular targets as it binds to nucleic acids via specific deoxyribonucleic acid DNA sequences [ ].

Berberine also induces mitochondrial-mediated apoptosis through the loss of mitochondrial membrane potential, cytochrome c release, caspase and PARP activation, up-regulation of pro-apoptotic Bcl-2 family proteins, and down-regulation of anti-apoptotic Bcl-2 family proteins [ , , , ].

It can also activate apoptosis-inducing factor to induce ROS-mediated cell death in pancreatic, breast, and colon cancers [ , , ]. Immunotherapy has made great progress to cancer treatment over the past few years.

Toll-like receptors TLRs can activate innate immune responses for host defense [ ]. Berberine inhibits proto-oncogene tyrosine kinase Src activation and TLR4-mediated chemotaxis in lipopolysaccharide LPS -induced macrophages [ ].

Besides, IDO1 inhibitors are promising candidates for cancer immunotherapy [ ]. Berberine and its derivatives are shown to exhibit anti-cancer activity through cell killing by NK cells via IDO1 [ ].

IL-8 is associated with metastasis, and berberine decreases IL-8 levels to inhibit cell growth and invasion in triple-negative breast cancer cells [ ].

Berberine has low oral bioavailability as well as poor intestinal absorption [ ]. As it has pronounced anti-microbial activity against gut microbiota, high dosage can translates into adverse events [ ]. This limits the clinical use of berberine, and different approaches have been applied to improve the bioavailability of berberine.

d -α-Tocopheryl polyethylene glycol succinate enhances the intestinal absorption of berberine by inhibiting P-gp activity in rats [ ]. A self-microemulsifying drug delivery system is developed to improve the bioavailability of berberine, the bioavailability is increased by 2.

Ber8, a 9-alkylated derivative of berberine, has better cytotoxicity and cellular uptake than berberine, and further inhibits cell proliferation and induces cell cycle arrest in different cell lines, including SiHa, HL, and A cells [ ]. The combination of berberine and chemo- or radio-therapies provides synergistic anti-cancer effects [ , ].

Taxol combined with berberine significantly slows down cell growth in human epidermal growth factor receptor 2 HER2 -overexpressed breast cancer cells [ ], while the combined administration of berberine and caffeine enhances cell death through apoptosis and necroptosis in human ovarian cancer OVCAR3 cells [ ].

The combination therapy of berberine and niraparib, a PARP inhibitor, markedly enhances apoptosis and inhibits tumor growth in ovarian cancer A xenograft mice [ ].

Therefore, combination of berberine with other therapies is a promising treatment for the alternative cancer therapy. Previous pre-clinical research and animal studies have demonstrated the anti-tumor action of berberine hydrochloride.

The people with a history of colorectal cancer might be at higher risk for adenomas, thus they are particularly suitable for the study of the chemopreventive effects of berberine hydrochloride in adenomas. A randomized, double-blind, placebo-controlled trial was designed to determine whether the daily intake of mg of berberine hydrochloride could decrease the occurrence of new colorectal adenomas in patients with a history of colorectal cancer, and it is currently ongoing.

Another phase II clinical trial of berberine and gefitinib is also ongoing in patients with advanced NSCLC and activating EGFR mutations. Artemisinin Fig. Since the Nobel Prize in Physiology or Medicine conferred to Chinese scientist, Youyou Tu, artemisinin drew attention to worldwide [ ].

Beside from their well-established anti-malarial effects, artemisinin and its derivatives ARTs , including dihydroartemisinin DHA , artesunate, artemether and arteether, are also found to exhibit potent anti-cancer activities in many studies [ , , , , , ].

DHA and artesunate are the most studied ART derivatives for cancer treatment, and artesunate will be discussed in a separate section. The anti-cancer effects of ARTs are demonstrated in a broad spectrum of cancer cells including lung, liver, pancreatic, colorectal, esophageal, breast, ovarian, cervical, head and neck, and prostate cancers [ , , , , , , , , ].

The anti-cancer activities of ARTs include induction of apoptosis and cell cycle arrest, inhibition of cell proliferation and growth, metastasis and angiogenesis [ , , , , ].

ART inhibits cell proliferation, migration and invasion, and induces apoptosis in human breast cancer MCF-7 cells [ , ], while DHA suppresses cell growth through cell cycle arrest and apoptosis in human hepatocellular carcinoma HepG2 cells and HepG2 xenograft mice [ ]. Similarly, ART induces apoptosis in murine mastocytome P cells and hamster kidney adenocarcinoma BSR cells, and inhibits tumor growth in P xenograft mice [ ].

Moreover, autophagy plays a vital role in ART-mediated anti-cancer activities [ , , , , , ]. DHA can induce autophagy-dependent cell death in human cervical cancer HeLa cells, cholangiocarcinoma KKU, KKU and KKU, and tongue squamous cell carcinoma Cal cells [ , , ], while ART induces autophagy-mediated cell cycle arrest in human ovarian cancer SKOV3 cells [ ].

DHA is also shown to induce autophagy by suppressing NF-κB activation in several cancer cells including RPMI , NB4, HCT, and HeLa cells [ ]. Furthermore, ART and DHA can also inhibit metastasis in various cancer cells such as non-small-cell lung carcinoma NSCLC , ovarian and lung cancer cells [ , , ].

Apart from apoptosis and metastasis, the inhibition of angiogenesis is also a crucial approach in cancer treatment. ART inhibits angiogenesis through mitogen-activated protein kinase MAPK activation in osteosarcoma [ ], whilst DHA exerts strong anti-angiogenic effect by repressing extracellular signal—regulated kinase ERK and NF-κB pathways in human umbilical vein endothelial cells HUVECs and pancreatic cancer, respectively [ , ].

In the past decades, studies have been focused on studying the anti-cancer mechanisms of ARTs, but there are contentions. ARTs inhibit cancer cell proliferation mainly by the induction of apoptosis through mitochondrial-dependent pathways [ , , ].

ART mediates the release of cytochrome c and caspase-9 cleavage, leading to increased apoptosis in human breast cancer MCF-7 cells [ ]. DHA induces apoptosis through Bcl-2 down-regulation in human cervical cancer HeLa and Caski cells [ ], and via Bim-dependent intrinsic pathway in human hepatocellular carcinoma HepG2 and Huh7 cells [ ].

Interestingly, ART is demonstrated to be an inhibitor of anti-cancer target, histone deacetylases HDAC [ ]. In addition, another mechanism of killing tumor cells by ARTs is iron-dependent cell death called ferroptosis, a new form of cell death, so ferroptosis becomes an attractive strategy for cancer treatment [ , ].

DHA can enhance the anti-tumor cytolytic activity of γδ T cells against human pancreatic cancer SW, BxPC-3 and Panc-1 cells [ ], and ART also potentiates the cytotoxicity of NK cells to mediate anti-tumor activity [ ].

Similarly, ART inhibits tumor growth through T cell activation and T reg suppression in breast cancer 4T1 xenograft mice [ ]. Therefore, this provides a novel strategy for treating pancreatic cancer with immunotherapy.

ART has poor water solubility and bioavailability. In order to solve this issue, ART is encapsulated into micelles by nanoprecipitation to form ART-loaded micelles [ ].

The ART-loaded micelles enhance the drug exposure time and accumulation in breast cancer 4T1 xenograft mice, and shows specific toxicity in human and murine breast cancer MCF-7 and 4T1 cells. A mitochondrial-targeting analog of ART is also synthesized to specifically target mitochondria for enhancing the inhibition of cell proliferation in various cancer cells including HCT, MDA-MB, HeLa and SKBR3 cells [ ].

Moreover, dimmers of ART are also synthesized by polyamine linkers, and they further inhibit cell proliferation in human breast cancer MCF-7 cells and angiogenesis in HUVECs [ ]. Many studies show the synergistic effects of ARTs with other compounds or therapeutic approaches.

The combined treatment of ART and resveratrol markedly inhibits cell proliferation and migration, and enhances apoptosis and ROS production in human cervical cancer HeLa and hepatocellular carcinoma HepG2 cells [ ].

Similarly, the use of combined DHA and gemcitabine exhibits strong synergistic effects on the loss of mitochondrial membrane potential and induction of apoptosis in human NSCLC A cells [ ].

DHA also reinforces the anti-cancer activity of chemotherapeutic agent, cisplatin, in cisplatin-resistant ovarian cancer cells [ ]. Studies also demonstrate the enhancement of sensitivity by DHA in photodynamic therapy in esophageal cancer [ , ].

Therefore, this suggests that ARTs could be potential anti-cancer agents. The salivary DHA concentration was proportionally correlated with the plasma DHA concentration, so saliva is a good use for monitoring DHA levels in the body. An artemisinin analog, Artenimol-R, was shown to improve clinical symptoms and tolerability in patients with advanced cervical cancer [ ].

Ginsenosides Fig. Chen, Panax ginseng and Cinnamomum cassia Presl. Ginsenosides mainly exert anti-cancer effects in colorectal, breast, liver and lung cancers, through inhibiting cell proliferation and migration, angiogenesis, and reversing drug resistance [ 7 , , , , , , , , ].

Ginsenoside Rg3, ginsenoside Rh2, and compound K are the primary bioactive compounds among ginsenosides for cancer prevention. Ginsenoside Rg3 inhibits cell viability and induces cell apoptosis in human ovarian cancer HO cells [ ], hepatocellular carcinoma Hep, HepG2 and SMMC, breast cancer MCF-7, MDA-MB, MDA-MB and BT, and NSCLC A, H23 and Lewis lung carcinoma cells [ , , , , , , ].

It induces cell cycle arrest at G1 phase in human melanoma A, and multiple myeloma U, RPMI and SKO cells [ , ], and inhibits cell migration in human colorectal cancer LoVo, SW and HCT cells [ ].

Ginsenoside Rg3 can also modulate the tumor environment through inhibiting angiogenesis and enhancing anti-tumor immune responses [ ]. Moreover, ginsenoside Rh2 exhibits anti-tumor activity in human NSCLC H cells and H xenograft mice, through the induction of ROS-mediated ER-stress-dependent apoptosis [ ].

It also suppresses cell proliferation and migration, and induces cell cycle arrest in human hepatocellular carcinoma HepG2 and Hep3B cells, and inhibits tumor growth in HepG2 xenograft mice [ ].

Compound K, an intestinal bacterial metabolite of ginsenosides, also induces cell cycle arrest and apoptosis in human colorectal cancer HCT cells, and suppresses tumor growth in HCT xenograft mice [ ].

It also efficiently inhibits cell proliferation and induces apoptosis through mitochondrial-related pathways in human hepatocellular carcinoma MHCCH cells [ ]. Furthermore, 20 S -ginsenoside Rg3 induces autophagy to mediate cell migration and invasion in human ovarian cancer SKOV3 cells [ ].

In contrast, it sensitizes NSCLC cells to icotinib and hepatocellular carcinoma cells to doxorubicin through the inhibition of autophagy [ , ]. Besides, ginsenoside Rh2 inhibits cell growth partially through the coordination of autophagy and β-cateninin signaling in human heptocellular carcinoma HepG2 and Huh7 cells [ ].

Therefore, autophagy plays a dual role in cancer via different signaling routes. Moreover, 20 S -ginsenoside Rh2 is shown to bind to recombinant and intracellular annexin A2 directly, and this inhibits the interaction between annexin A2 and NF-κB p50 subunit, which decreases NF-κB activation [ ].

NF-κB is important in cell survival, and 20 S -ginsenoside Rh2 can inhibit cell survival through NF-κB pathway. Furthermore, p53 also plays a vital role in ginsenoside-induced anti-cancer activities [ , , ].

For the promotion of immunity, ginsenoside Rg3 can enhance lymphocyte proliferation and T helper type 1 cell Th1 -related cytokine secretion including IL-2 and IFN-γ in hepatacellular carcinoma Hbearing mice, and inhibit tumor growth partly through the induction this cellular immunity [ ].

Ginsenoside Rg3 can also down-regulate the levels of B7-H1 and B7 homolog 3 B7-H3 , immunoglobulin-like immune suppressive molecules, to modulate tumor microenvironment and enhance anti-tumor immunity, and these molecules are negatively associated with overall survival in colorectal cancer patients [ ].

In addition, ginsenoside Rh2 can also enhance anti-tumor immunity in melanoma mice by promoting T cell infiltration in the tumor and cytotoxicity in spleen lymphocytes [ ]. The combination of ginsenosides with other chemotherapeutic agents provides significant advantages for cancer treatment.

Ginsenoside Rg3 alone demonstrates modest anti-angiogenic effects, and displays additive anti-angiogenic effects in B6 glioblastoma rats when combined with temozolomide [ ].

When it is combined with paclitaxel, it enhances cytotoxicity and apoptosis through NF-κB inhibition in human triple-negative breast cancer MDA-MB, MDA-MB and BT cells [ ]. Ginsenosides have a long history of use as traditional medicine to treat many diseases in China.

Relatively few clinical studies have been performed in humans eventhough ginseng products are widely recognized to have therapeutic effects when used alone or in combination with other chemotherapeutic agents.

Therefore, clinical studies are needed to confirm the safety of such uses. A phase II clinical trial is conducting to assess the safety and efficacy of ginsenoside Rg3 in combination with first-line chemotherapy in advanced gastric cancer.

Patients with advanced NSCLC and epidermal growth factor receptor-tyrosine kinase inhibitor EGFR-TKI mutation were recruited in a study that investigated the safety and efficacy of the combined therapy, ginsenoside Rg3 and EGFR-TKI.

It was shown that this therapy increased progression-free survival, overall survival and objective response rate compared to EGFR-TKI alone [ ]. In another study, the safety and efficacy of combined ginsenoside Rg3 and transcatheter arterial chemoembolization TACE were studied in patients with advanced hepatocellular carcinoma.

The results showed that this therapy ameliorated TACE-induced adverse effects and prolonged the overall survival compared to the use of TACE alone [ ].

As an ursane-type pentacyclic triterpenic acid, UA Fig. cranberry , Arctostaphylos uva - ursi L. Spreng bearberry , Rhododendron hymenanthes Makino , Eriobotrya japonica, Rosemarinus officinalis, Calluna vulgaris, Eugenia jambolana and Ocimum sanctum, as well as in the wax-like protective coatings of fruits such as pears, apples and prunes [ ].

UA has numerous biochemical and pharmacological effects including anti-inflammatory, anti-oxidative, anti-proliferative, anti-atherosclerotic, anti-leukemic, anti-viral, and anti-diabetic effects [ , , , , , , ].

It also exerts anti-cancer activities in ovarian, breast, gastric, prostate, lung, liver, bladder, pancreatic, and colorectal cancers [ , , , , , , , , ]. UA can be used as a potential therapeutic agent for the treatment of various cancers [ , , , , , , , , ].

It induces apoptosis through both extrinsic death receptor and mitochondrial death pathways in human breast cancer MDA-MB cells [ ], and inhibits cell proliferation and induces pro-apoptosis in human breast cancer MCF-7 cells by FoxM1 inhibition [ ].

UA also inhibits cell and tumor growth through suppressing NF-κB and STAT3 pathways in human prostate cancer DU and LNCaP cells, and DU xenograft mice [ ], and induces apoptosis in human prostate cancer PC-3 cells [ ].

Similarly, UA induces apoptosis and inhibits cell proliferation in human colorectal cancer HCT, HCT, HT and Caco-2 cells [ , ]. UA is also shown to induce autophagy to mediate cell death in murine cervical cancer TC-1 cells [ ], and promote cytotoxic autophagy and apoptosis in human breast cancer MCF-7, MD-MB and SKBR3 cells [ ].

It also inhibits cell growth by inducing autophagy and apoptosis in human breast cancer cells T47D, MCF-7 and MD-MB cells [ ]. In contrast, UA induces autophagy, but the inhibition of autophagy enhances UA-induced apoptosis in human oral cancer Ca and SCC, and prostate cancer PC-3 cells [ , ].

Therefore, autophagy plays a dual role in UA-induced apoptosis via different signaling pathways. In addition, UA inhibits tumor angiogenesis through mitochondrial-dependent pathway in Ehrlich ascites carcinoma xenograft mice [ ].

UA is demonstrated to have apoptosis-promoting and anti-proliferative capacities via modulating the expressions of mitochondrial-related proteins such as Bax, Bcl-2, cytochrome c and caspase-9 [ , ]. It can also induce oxidative stress and disruption of mitochondrial membrane permeability to mediate apoptosis in human osteosarcoma MG63 and cervical cancer HeLa cells [ , ].

In addition, p53 pathway also contributes to the anti-cancer effects of UA. UA induces apoptosis and cell arrest through pmediated p53 activation in human colorectal cancer SW and breast cancer MCF-7 cells [ , ], and this p53 activation is through inhibiting negative regulators of p53, MDM2 and T-LAK cell-originated protein kinase TOPK [ ].

Studies have reported the cancer immunomodulatory activities of UA [ , ]. UA down-regulates NF-κB to inhibit cell growth and suppress inflammatory cytokine levels including TNF-α, IL-6, IL-1β, IL and IFN-γ in human breast cancer T47D, MCF-7 and MDA-MB cells [ ].

It also modulates the tumor environment by modulating cytokine production such as TNF-α and IL in ascites Ehrlich tumor [ ]. UA is insoluble in water, with poor pharmacokinetic properties including poor oral bioavailability, low dissolution and weak membrane permeability [ ].

Some new drug delivery technologies have been developed to overcome these problems including the uses of liposomes [ , , , , ], solid dispersions [ ], niossomal gels [ ], and nanoliposomes [ ]. Liposome is the most commonly used drug delivery system.

A chitosan-coated UA liposome is synthesized with tumor targeting and drug controlled release properties, and has fewer side effects [ ].

It enhances the inhibition of cell proliferation and tumor growth in human cervical cancer HeLa cells and U14 xenograft mice. Besides, a pH-sensitive pro-drug delivery system is also synthesized, and this pro-drug enhances cellular uptake and bioavailability of UA [ ].

It further inhibits cell proliferation, cell cycle arrest and induces apoptosis in human hepatocellular carcinoma HepG2 cells. UA can also be used in combination with other drugs. The combined treatment of zoledronic acid and UA enhances the induction of apoptosis and inhibition of cell proliferation through oxidative stress and autophagy in human osteosarcoma U2OS and MG63 cells [ ], whilst the combination of UA and curcumin inhibits tumor growth compared to UA alone in skin cancer mice [ ].

A human clinical study was conducted to investigate the toxicity and pharmacokinetics of UA-liposomes UAL including dose-limiting toxicity and maximum tolerated dose in healthy adult volunteers and patients with advanced solid tumors [ ].

Silibinin Fig. Gaertn, is commonly exploited for the treatment hepatic diseases in China, Germany and Japan. Previous studies have reported that silibinin exerts remarkable effects in numerous cancers such as renal, hepatocellular and pancreatic carcinoma, bladder, breast, colorectal, ovarian, lung, salivary gland, prostate and gastric cancers, through the induction of apoptosis, inhibition of tumor growth, metastasis and angiogenesis [ , , , , , , , , , , ].

Silibinin suppresses epidermal growth factor-induced cell adhesion, migration and oncogenic transformation through blocking STAT3 phosphorylation in triple negative breast cancer cells [ ]. It strongly suppresses cell proliferation and induces apoptosis in human pancreatic cancer AsPC-1, BxPC-3 and Panc-1 cells, and induces cell cycle arrest at G1 phase in AsPC-1 cells [ ].

It can also induce apoptosis via non-steroidal anti-inflammatory drug-activated gene-1 NAG-1 up-regulation in human colorectal cancer HT cells [ ], and induces mitochondrial dysfunction to mediate apoptosis in human breast cancer MCF-7 and MDA-MB cells [ ].

Moreover, silibinin induces autophagic cell death via ROS-dependent mitochondrial dysfunction in human breast cancer MCF-7 cells [ ]. In contrast, it induces autophagy to exert protective effect against apoptosis in human epidermoid carcinoma A, glioblastoma A and SR, and breast cancer MCF-7 cells [ , , ], and autophagy inhibition enhances silibinin-induced apoptosis in human prostate cancer PC-3 cells [ ].

Silibinin also induces autophagy to inhibit metastasis in human renal carcinoma ACHN and O cells, and salivary gland adenoid cystic carcinoma cells [ ].

Therefore, autophagy plays a dual role in silibinin-induced anti-cancer effects. In addition, silibinin inhibits angiogenesis in human prostate cancer PCa, LNCaP and 22Rv1 cells [ ]. Silibinin exhibits anti-cancer activities mainly due to the cell cycle arrest [ , , , , ].

It induces G1 phase arrest in human pancreatic cancer SW and AsPC-1, and breast cancer MCF-7 and MCFA cells [ , , ], whilst it causes G2 phase arrest in human cervical cancer HeLa, and gastric cancer MGC and SGC cells [ , ].

It also decreases the expressions of CDKs such as CDK1, CDK2, CDK4 and CDK6 that are involved in G1 and G2 progression [ , ]. In addition, silibinin induces apoptosis and inhibits proliferation through the suppression of NF-κB activation [ , , , ].

On the other hand, silibinin is shown to induce apoptosis through the promotion of mitochondrial dysfunction, including increased cytochrome c and Bcl-2 levels, the loss of mitochondrial membrane potential, and decreased adenosine triphosphate ATP levels [ , , , ].

Silibinin has immunomodulatory effects in cancer and immunity. The MDSCs are associated with immunosuppression in cancer, and silibinin increases the survival rate in breast cancer 4T1 xenograft mice, and reduces the population of MDSCs in their blood and tumor [ ].

There was also a reduction in macrophage infiltration and neutrophil population in silibinin-treated prostate cancer TRAMPC1 xenograft mice [ ].

These studies suggest a role of immunity in its anti-tumor effects. Silibinin has poor water solubility and bioavailability, so it limits its efficacy in anti-cancer activities [ ]. Advanced technologies such as nanoprecipitation technique are used to solve this issue [ , , , , ].

Silbinin is encapsulated in Eudragit ® E nanoparticles in the presence of polyvinyl alcohol, and these nanoparticles enhance apoptosis and cytotoxicity in human oral cancer KB cells [ ]. The silibinin-loaded magnetic nanoparticles further inhibit cell proliferation in human NSCLC A cells [ ], while silibinin-loaded chitosan nanoparticles enhances cytotoxicity compared to silibinin alone in human prostate cancer DU cells [ ].

The combination of silibinin and other drugs are used in cancer treatment to enhance the efficacy of anti-cancer effects [ , , , ]. The combination of curcumin and silibinin enhances the inhibition of cell growth and reduction in telomerase gene expression compared to silibinin alone in human breast cancer T47D cells [ ].

The mixture of luteolin and silibinin also shows synergistic effects on the attenuation of cell migration and invasion, and induction of apoptosis in human glioblastoma LN18 and SNB19 cells [ ]. Silibinin and paclitaxel combination enhances apoptosis and up-regulates tumour suppressor genes, p53 and p21, in human ovarian cancer SKOV3 cells [ ].

Silibinin has been widely used as anti-cancer drug in vitro and in vivo, and its combination with other therapies is a promising treatment for cancer, so clinical trials are needed to confirm its safety and efficacy in humans, and to develop as an anti-cancer drug.

Emodin Fig. It exhibits remarkable biological effects such as anti-inflammation, anti-oxidant, prevention of intrahepatic fat accumulation and DNA damage [ , , , , , , ]. Many studies have shown that emodin can attenuate numerous cancers including nasopharyngeal, gall bladder, lung, liver, colorectal, oral, ovarian, bladder, prostate, breast, stomach and pancreatic cancers, through the inhibition of cell proliferation and growth, metastasis, angiogenesis, and induction of apoptosis [ , , , , , , , , , , , , ].

Emodin suppresses ATP-induced cell proliferation and migration through inhibiting NF-κB activation in human NSCLC A cells [ ], and induces apoptosis through cell cycle arrest and ROS production in human hepatocellular carcinoma HepaRG cells [ ]. It also induces autophagy to mediate apoptosis through ROS production in human colorectal cancer HCT cells [ ].

Moreover, emodin can inhibit tumor growth and metastasis in triple negative breast cancer cells, and human colorectal cancer HCT cells [ , ], whilst it suppresses cell migration and invasion through microRNA up-regulation in human pancreatic cancer SW cells [ ].

In addition, emodin can also inhibit angiogenesis in thyroid and pancreatic cancers [ , , ]. Emodin exerts anti-cancer effects through various mechanisms. Besides, mitochondria and ER stress also play an important role in mediating emodin-induced anti-cancer effects [ , , , ].

Emodin induces apoptosis through the loss of mitochondrial membrane potential, modulation of Bcl-2 family proteins, and caspase activation in human colorectal cancer CoCa cells and hepatocellular carcinoma HepaRG cells [ , ]. ER stress is activated in emodin-treated human osteosarcoma U2OS cells, and emodin-induced apoptosis is suppressed by ER stress inhibition with 4-phenylbutyrate 4-PBA in human NSCLC A and H cells [ , ].

Emodin has immunomodulatory effects in cancer and immunity. It inhibits cell growth and metastasis through blocking the tumor-promoting feed forward loop between macrophages and breast cancer cells [ ].

It also down-regulates CXCR4 to suppress C—X—C motif chemokine 12 CXCL -induced cell migration and invasion in hepatocellular carcinoma HepG2 and HepG3 cells [ ].

In addition, emodin inhibits the differentiation of maturation of DCs [ ], and can modulate macrophage polarization to restore macrophage homeostasis [ ]. Aloe-emodin is a derivate of emodin, which exhibits superior bioactivities in some cancers.

It can inhibit cell proliferation through caspase-3 and caspase-9 activation in human oral squamous cell carcinoma SCC cells [ ], and induce apoptosis in human cervical cancer HeLa and SiHa cells, which is associated with glucose metabolism [ ].

Another derivative of emodin, rhein, can also induce apoptosis in human pancreatic cancer Panc-1 cells, and inhibit tumor growth in pancreatic cancer xenograft mice [ ]. The combination of emodin and other chemotherapies is widely used for cancer treatment.

Emodin can promote the anti-tumor effects of gemcitabine in pancreatic cancer [ , , ]. It enhances apoptosis in human pancreatic cancer SW cells, and further inhibits tumor growth in SW xenograft mice, through suppressing NF-κB pathway [ , ].

The combination of emodin and curcumin can also enhance the inhibition of cell proliferation, survival, and invasion in human breast cancer MDA-MB, MDA-MB and A1 cells [ 64 ].

Moreover, emodin enhances cisplatin-induced cytotoxicity through ROS production and multi-drug resistance-associated protein 1 MRP1 down-regulation in human bladder cancer T24 and J82 cells [ ]. Emodin has been shown to have remarkable anti-cancer effects in vitro and in vivo, and its combination with other therapies is very effective in treating cancer, therefore it is important to evaluate the safety and efficacy of emodin as an anti-cancer drug as the next step.

Triptolide Fig. For cancer therapy, it has been used to treat breast, lung, bladder, liver, colorectal, pancreatic, ovarian, stomach, prostate, cervical, and oral cancers, melanoma, myeloma, leukemia, neuroblastoma, osteosarcoma, lymphoma, renal, nasopharyngeal, and endometrial carcinoma, through apoptosis, cell cycle arrest, inhibition of cell proliferation, metastasis and angiogenesis [ , , , , , , , , , , , , , , , , , , , ].

Various effects have been disclosed as key contributions to the anti-cancer effects of triptolide. Triptolide is shown to exhibit pro-apoptosis effects in various cancers [ , , , , ]. Moreover, triptolide induces autophagy to induce apoptosis and inhibit angiogenesis in human osteosarcoma MG63 cells, and breast cancer MCF-7 cells [ , ].

Therefore, autophagy plays a dual role in triptolide-induced anti-cancer effects. In addition, triptolide is able to inhibit cell migration and invasion in human prostate cancer PC-3 and DU cells, and in tongue squamous cell carcinoma SAS cells co-inoculated with human monocytes U cells [ , ].

Furthermore, triptolide also possesses anti-angiogenic effect by inhibiting VEGFA expression in human breast cancer MDA-MB and HsT cells, and through COX-2 and VEGF down-regulation in human pancreatic cancer Panc-1 cells [ , ]. Triptolide is a natural substance, which exerts its anti-cancer effects through multiple targets.

Triptolide is shown to induce mitochondrial-mediated apoptosis in various cancer cells, through decreased mitochondrial membrane potential, Bax and cytochrome c accumulation, PARP and caspase-3 activation, decreased ATP levels, and Bcl-2 down-regulation [ , , , , ].

Moreover, ERK is also shown to be important in mediating triptolide-induced anti-cancer activities. Triptolide induces apoptosis through ERK activation in human breast cancer MDA-MB and MCF-7 cells [ , ], and ERK activation leads to caspase activation, Bax up-regulation and Bcl-xL down-regulation [ ].

On the other hand, it can also inhibit metastasis through ERK down-regulation in esophageal squamous cell cancer KYSE and KYSE cells, and murine melanoma B16F10 cells [ , ].

Interestingly, ERα is shown to be a potential binding protein of triptolide and its analogues [ ]. In addition, triptolide-induced metastasis is shown to be through MMP-2 and MMP-9 down-regulation in human neuroblastoma SH-SY5Y cells, via decreased MMP-3 and MMP-9 expressions in T-cell lymphoblastic lymphoma cells, and through MMP-2, MMP-7 and MMP-9 down-regulation in human prostate cancer PC-3 and DU cells [ , , ].

Indeed, immunology has been frequently validated to be associated with cancer. The derivatives of triptolide are always needed to improve its ant-cancer therapy. Triptolide derivative, MRx, shows positive effects on anti-proliferation and anti-metastasis through Wnt inhibition in human NSCLC H and A cells, and H xenograft mice [ ].

Minnelide, a water-soluble pro-drug of triptolide, can inhibit tumor growth in pancreatic cancer MIA PaCa-2 xenograft mice.

Meanwhile, the combination of minnelide and oxaliplatin further inhibits tumor growth [ ]. Moreover, triptolide is poorly soluble in water and exhibits hepatotoxicity and nephrotoxicity, selective delivery is an effective strategy for further application in cancer treatment.

Triptolide loaded onto a peptide fragment TPS-PF-A — is specifically targeted to the kidney and with less toxicity [ ]. Some modified triptolide-loaded liposomes are reported to contribute a targeted delivery with lower toxicity and better efficacy in lung cancer treatment [ ].

Similarly, triptolide-loaded exosomes enhances apoptosis in human ovarian cancer SKOV3 cells [ ]. Triptolide has some side effects in various organs because of excessive dosage, so researchers have been looking for alternative triptolide therapies, and combination therapy has become a hot spot.

Triptolide plus ionizing radiation synergistically enhances apoptosis and anti-angiogenic effects through NF-κB p65 down-regulation in human nasopharyngeal carcinoma cells and xenograft mice, which provides a new chemotherapy to advanced nasopharyngeal malignancy [ ].

The combined therapy of triptolide and 5-fluorouracil further promotes apoptosis and inhibits tumor growth through down-regulating vimentin in human pancreatic cancer AsPC-1 cells and AsPC-1 xenograft mice [ ].

Besides, low concentration of triptolide potentiates cisplatin-induced apoptosis in human lung cancer HTB, A and CRL and CRL cells [ ], and triptolide with cisplatin synergistically enhances apoptosis and induces cell cycle arrest in human bladder cancer cisplatin-resistant cells [ ].

Triptolide has wide-spectrum activities in pre-clinical studies, but it has strong side effects and water insolubility, so it is not used in clinical studies. However, some of its derivatives and analogs have been used in clinical studies to test the safety and efficacy on anti-cancer effects [ , , , ].

Omtriptolide, a derivative of triptolide, is highly water soluble, and a phase I clinical trial was conducted in Europe with patients who had refractory and relapsed acute leukemia [ ]. Another phase I clinical trial was completed in patients with refractory gastrointestinal malignancies to study the dose escalation and pharmacokinectics of minnelide, a pro-drug of triptolide [ ].

The doses used were 0. LLDT-8, another triptolide derivative, has anti-cancer and immunosuppressive effects, and is going to proceed into phase II clinical trial to test its anti-cancer effects in China [ , ].

Moreover, minnelide is currently under phase II clinical trial to test anti-cancer effects in patients with advanced pancreatic cancer [ ]. Cucurbitacins Fig. Cucurbitacins A—T are twelve main curcurbitacins belonging to this family. Cucurbitacins have multiple therapeutic effects such as anti-inflammation, anti-proliferation, anti-angiogenesis, and anti-cancer [ , , , , ].

Besides, cucurbitacins have also been elucidated as a potential candidate for various cancer therapies, including oral cell carcinoma, breast, ovarian, prostate, lung, gastric, bladder, and thyroid cancers, neuroastoma, hepatoma, and osteosarcoma [ , , , , , , , , , , , , ]. Most of cucurbitacins have been reported with various anti-cancer activities, such as pro-apoptosis, anti-angiogenesis, autophagy induction, and inhibition of metastasis [ , , , , ].

Cucurbitacin B is the most abundant source of cucurbitacins which can explain why it receives more attention from researchers than other cucurbitacins do.

It suppresses cell proliferation and enhances apoptosis in human NSCLC A cells, colorectal cancer SW and Caco-2 cells [ , ], and induces G1 phase cell cycle arrest in human colorectal cancer SW and Caco-2, and gastric cancer MKN45 cells [ , ].

Moreover, cucurbitacins B, E and I are shown to induce autophagy, however inhibition of autophagy can enhance cucurbitacin-induced apoptosis [ , , ].

They also inhibit cell migration and invasion in human breast cancer MDA-MB and SKBR3, NSCLC HBrM3 and PC9-BrM3, and colorectal cancer COLO cells [ , , , ], as well as angiogenesis in HUVECs [ , ]. Various targets have been demonstrated to be responsible for the anti-cancer effects of cucurbitacins.

STAT3 signaling is a very common target for cancer treatment. Besides, cucurbitacin E induces cell cycle arrest through cyclins B1 and D1 down-regulation [ , ], while cucurbitacin D inhibits cyclin B expression [ ].

Moreover, mitochondria and ER stress also play an important role in cucurbitacin-induced anti-cancer effects. Cucurbitacins mediate apoptosis through mitochondrial-related pathway, which is characterized by the loss of the mitochondrial membrane potential, Bcl-2 down-regulation, Bax up-regulation, cytochrome c release, that eventually leads to caspase activation [ , ].

Cucurbitacin I induces cell death through ER stress, by up-regulating ER stress markers such as IRE1α and PERK in human ovarian cancer SKOV3 cells and pancreatic cancer Panc-1 cells [ ]. Cancer immunotherapy also plays a vital role in cucurbitacin treatment.

Cucurbitacins may influence the production of cytokines and transcription factors that suppress the immune system, and these mechanisms may help to prevent the development of cancer.

Cucurbitacin B is able to promote DC differentiation and anti-tumor immunity in patients with lung cancer [ ]. Although cucurbitacin B has very effective anti-tumor effects, it is shown to exhibit high toxicity, which restricts its clinical application on cancer therapy.

Therefore, studies have been focused on tackling this side effect, and some cucurbitacin B derivatives have been synthesized to screen for effective cancer therapy with safety and tolerability. Compound 10b, one of the derivatives of cucurbitacin B, shows more potent anti-cancer activity than cucurbitacin B [ ].

The in vivo acute toxicity study also shows that compound 10b has better tolerability and safety than cucurbitacin B. In addition, some other strategies have been applied to accelerate the clinical use of cucurbitacin B.

The collagen peptide-modified nanomicelles with cucurbitacin B were synthesized to enhance the oral availability of cucurbitacin B, and these nanomicelles show a higher bioavailability and better tumor inhibition [ ].

For a better cancer therapy, some combinations between cucurbitacins and other drugs have been employed. Low doses of cucurbitacin B or methotrexate cannot inhibit tumor growth in osteosarcoma xenograft mice, however when combined together, they synergistically inhibit tumor growth [ ].

Recently, cucurbitacin B is suggested to be a potential candidate when it is applied with withanone, this combination can enhance cytotoxicity in human NSCLC A cells, and inhibit tumor growth and metastasis in A xenograft mice [ ]. Cucurbitacin I is also shown to be a STAT3 inhibitor to mediate cell survival and proliferation, and when it is combined with irinotecan, and they further inhibit cell proliferation in human colorectal cancer SW and LST cells [ ].

The derivatives of cucurbitacins, cucurbitacin B-nanomicelles, and the combination therapies show promising treatment for cancer in vitro and in vivo, so clinical trials are needed to confirm their safety and efficacy in cancer treatment.

Tanshinone Fig. Tanshinone IIA is the primary bioactive constituent of tanshinones [ ], which has various pharmacological effects, including anti-inflammatory, anti-cancer and anti-atherosclerotic activities, and cardiovascular protection [ , , , ].

Tanshinone exhibits anti-cancer activities in stomach, prostate, lung, breast, and colon cancers, through inducing cell cycle arrest, apoptosis, autophagy, and inhibiting cell migration [ , , , , , , , , ].

Tanshinone IIA suppresses cell proliferation and apoptosis in numerous cancer cells, including human breast cancer BT, MDA-MB, SKBR3, BT, MCF-7 and MD-MB [ , , ], and gastric cancer MKN45 and SGC cells [ ].

It also induces cell cycle arrest at G1 phase in human breast cancer BT cells [ ], and inhibits cell migration in human gastric cancer SGC cells [ ], and cell migration and invasion in cervix carcinoma stemness-likes cells [ ].

Tanshinone I and cryptotanshinone are two other major bioactive compounds, which also induce cytotoxicity against cancer cells. In addition, tanshinones I and IIA and cryptotanshinone also inhibit tumor angiogenesis in endothelial and cancer cells [ , , , , ].

Moreover, tanshinone IIA is shown to exhibit anti-cancer activities through the interplay between autophagy and apoptosis in human prostate cancer PC-3 cells, mesothelioma H28 and H cells [ , ].

It inhibits epithelial—mesenchymal transition by modulating STAT3-chemokine C—C motif ligand 2 CCL2 pathway in human bladder cancer , BFTC and T24 cells [ ], and suppresses cell proliferation and migration via forkhead box protein M1 FoxM1 down-regulation in human gastric cancer SGC cells [ ].

On the other hand, tanshinone I induces apoptosis via Bcl-2 down-regulation in human gastric cancer BGC and SGC cells [ ], while cryptotanshinone induces apoptosis through mitochondrial-, cyclin- and caspase-dependent pathways in human NSCLC A and NCI-H cells [ ], as well as via ER stress in human hepatocellular carcinoma HepG2 and breast cancer MCF-7 cells [ ].

Tanshinone IIA is also shown to exhibit immunomdulatory effects in cancer [ ]. Furthermore, cryptotanshinone becomes a new promising anti-tumor immunotherapeutic agent [ ]. It induces mouse DC maturation and stimulates IL-1β, TNF-α, ILp70 secretion in DCs, and enhances T cell infiltration and Th1 polarization in Lewis-bearing tumor tissues [ ].

Tanshinone IIA has poor bioavailability, so a mixed micelle system is developed to form a tanshinone-encapsulated micelle [ ]. This micelle has higher cytotoxicity and pro-apoptotic effects in human hepatocellular carcinoma HepG2 cells compared to tanshinone IIA alone.

The tanshinone IIA-loaded nanoparticles improve the bioavailability tanshinone IIA and enhance its leukemic activity in human leukemia NB4 cells [ ], while the nanoparticles containing tanshinone IIA and α-mangostin show increased cytotoxicity in human prostate cancer PC-3 and DU cells [ ].

Tanshinone IIA is shown to enhance chemosensitivity and its efficacy when combined with other therapeutic agents. Tanshinone IIA can be an effective adjunctive agent in cancer, and it enhances the chemosensitivity to 5-fluorouracil therapy in human colorectal cancer HCT and COLO cells through NF-κB inhibition [ ].

The combination of tanshinone IIA with doxorubicin does not only enhance the chemosensitivity of doxorubicin, but also reduces the toxic side effects of doxorubicin in human breast cancer MCF-7 cells [ ]. In addition, tanshinone IIA and cryptotanshinone synergistically enhance apoptosis in human leukemia K cells [ ].

The anti-cancer effects of Tanshinone IIA have been demonstrated in various cancers in vitro and in vivo, and it can enhance chemosensitivity and its efficacy is very effective when combined with other therapeutic agents. Up to now, the clinical trials of Tanshinone IIA are completed only for the treatment of other diseases [ ], so well-designed clinical trials should be done to further confirm its safety and efficacy in cancer treatment.

Oridonin Fig. Hara, which is also the main active constituent of Rabdosia rubescens Hemsl. Hara [ ]. A complication shows that numbers of international agencies and associated groups use RUCAM for HILI cases Table 4 ; Teschke and Eickhoff, Compared with other causality tools, RUCAM has many advantages and is a wonderful tool to establish assess causality in HILI cases quantitatively.

RUCAM represents a standardized and effective diagnostic approach for hepatotoxicity, which uses scores of key items to express the course of HILI. In case reports, the scores give final grade of causality for each suspected HILI patients range from 14 to -3 , which are highly probable, probable, possible, unlikely and excluded.

RUCAM achieve the requirements that doctors have a great degree of confidence when they diagnose their patients who are suspected HILI, the results can be readily available in a few minutes Danan and Teschke, TABLE 4.

Selective complication of agencies applying the RUCAM scale for causality assessment in suspected HILI. Use the updated RUCAM scale to have a clinical evaluation, followed by optional expert discussions based on RUCAM scores.

This structured approach would help to improve the transparency of case data Benichou et al. Nephrotoxicity refers to side effects of kidney damage. It is a broad term that includes all side effects associated with filtration, reabsorption and excretion.

Chemotherapy-induced nephrotoxicity is one of the main factors limiting the time and dose of chemotherapy in cancer patients. Nephrotoxicity is especially severe when chemotherapy is combined with radiotherapy.

Acute renal injury and hypomagnesemia are two common manifestations of nephrotoxicity. As for now, investigations about using natural products to decrease the nephrotoxicity induced by chemotherapy are mainly performed on animal experiments, and clinical trials are expected in the future.

Bu-zhong-yi-qi decoction BZYQD, also called Zhong-Yi-Qi-Tangang, Bojungikki-tang, and Hochu-ekki-to is a famous Chinese medicine prescription, which is extracted from eight kinds of Chinese herbal medicines and is widely used in Asia to improve digestibility.

Animal experiments showed that 5-fu could lead to severe renal injury. BZYQD almost completely changed the renal function related indexes and antioxidant enzyme activity affected by 5-fu Xiong et al.

Honey and royal jelly are daily health foods, and a clinical study has shown that acute renal damage caused by platinum chemotherapeutic drugs can be protected by the use of honey and royal jelly.

Compared with the control group, serum levels of renal injury products were significantly decreased in cancer patients receiving honey and royal jelly capsules. Chemotherapy is still one of the most commonly used cancer therapies which gained beneficial outcome to patients with tumor.

Chemotherapeutic agents are rapidly discovered and developed by academia and industry, however, common adverse reaction and side effects are still difficult to overcome due tothe biological and chemical nature of the chemotherapeutics.

Multiple component herbal products that have been ethno medically used for a thousand and hundreds of years have been proved their potential in reducing the side effects of chemotherapeutic agents, as summarized by our study.

However, there are some problems remaining to be solved for facilitating the clinical application of these herbal products in the management of chemotherapy-induced side effects. First, the mechanism of action remains largely unclear.

Although some experimental studies have used cellular model to explore some signaling transduction caused by herbal extract treatment, it is unlikely to be reproducible in animal and human; second, a great concern on herbal-drug interaction may be issued when the herbal products are considered to be used clinically, unfortunately, in vivo pharmacokinetic studies on both herbal products and the chemotherapeutic agents are scanty.

Third, the uncertainty in composition of herbal extracts makes it difficult to gain a consistent efficacy in clinical application.

Due to the nature and resource of herbal products, variance in quality between batch and batch products is often observed. This requires a more restricted way in quality control during production and manufacturing. Last but not least, the clinical trials related herbal products are not enough while the most experiments were still performed on the cell or animal platforms.

More and more clinical trials with strictly followed protocols and highly standard design are important for illustrating the clinical efficacy and safeness of herbal products in the treatment of chemotherapy-induced side effects.

Compared with using conventional chemical drugs to decrease the side effects induced by chemotherapy, natural herbal medicines have many advantages. Because of the interactions of chemotherapy drugs and active ingredients in herbal medicines, it can have better effects than conventional chemical drugs.

For example, CYP revulsive is used to be antidote in drug poisoning while it can reduce the efficacy of drugs at other time. Moreover, if the security of herbal medicines can be guaranteed, the natural products may help more patients to get treat, because natural products made by herbs are much cheaper than the conventional chemical drugs.

In conclusion, more and more evidence shows that compound drugs containing natural products can better reduce side effects caused by chemotherapy, thereby improving the QOL of cancer patients.

However, since the compound is not a single natural product, the interaction and basic pharmacological effects of the active ingredients in the compounds need to be further studied in order to more clearly illustrate the mechanism of reducing the side effects of chemotherapy by natural compounds.

In addition, rigorous clinical trials of drugs can provide reliable and decisive evidence, rather than just stay in cell and animal trials. YF conceived the review. H-YT, SL, and FC collected and analyzed the data. BF and NW drafted the manuscript.

All authors revised and commented on the paper and discussed the paper. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. Adachi, I. Gan To Kagaku Ryoho 16 4 Pt 2—2 , — PubMed Abstract Google Scholar.

Allemani, C. Public health surveillance of cancer survival in the United States and worldwide: the contribution of the CONCORD programme. Cancer , — doi: PubMed Abstract CrossRef Full Text Google Scholar.

Andersen, B. Psychological interventions for cancer-patients to enhance the quality-of-life. CrossRef Full Text Google Scholar. Aoyama, T. Double-blind, placebo-controlled, randomized phase II study of TJ hangeshashinto for gastric cancer chemotherapy-induced oral mucositis. Cancer Chemother.

Bai, C. Zhonghua Liu Xing Bing Xue Za Zhi 24, — Bao, Y. Clinical observation of traditional Chinese medicine in prevention of peripheral neurotoxicity induced by modified DCF chemotherapy regimen: a report of 35 cases.

Jiangsu J. Ben-Arye, E. Benichou, C. Causality assessment of adverse reactions to drugs. An original model for validation of drug causality assessment methods — case-reports with positive rechallenge.

Berger, T. Cancer 33, — Bjornsson, E. Hepatotoxicity associated with statins: reports of idiosyncratic liver injury post-marketing. Boon, H. Botanical medicine and cancer: a review of the safety and efficacy.

Expert Opin. Bossi, P. Searching for evidence to support the use of ginger in the prevention of chemotherapy-induced nausea and vomiting.

Chan, C. Oral complications in Chinese cancer patients undergoing chemotherapy. Support Care Cancer 11, 48— Google Scholar. Chan, K. The use of Chinese herbal medicine to improve quality of life in women undergoing chemotherapy for ovarian cancer: a double-blind placebo-controlled randomized trial with immunological monitoring.

Chau, T. Causality assessment of herb-induced liver injury using multi disciplinary approach and the Roussel Uclaf Causality assessment Method RUCAM. Chen Effect observation of Huangqi injection combined with chemotherapy on quality of life of postoperation patients with advanced stage colorectal carcinoma.

Hebei J. Chen, H. Effect of chinese herbal compound combining chemotherapy on recurrence breast cancer. Chinese Med. Chen, Y. A dual-fluorescent whole-well imaging approach for screening active compounds against doxorubicin-induced cardiotoxicity from natural products.

RSC Adv. Chen, L. A Study of Effect Yiliu Decocotion Combined with Chemotherapy on Quanlity of Life of Qi Deficiency and Blood Stasis Type Postperative Ovarian Carcinoma Patients. Fuzhou: Fujian University of Traditional Chinese Medicine.

Chen, S. Oral Chinese herbal medicine CHM as an adjuvant treatment during chemotherapy for non-small cell lung cancer: a systematic review. Lung Cancer 68, — Intestinal glucuronidation protects against chemotherapy-induced toxicity by irinotecan CPT Chen, W.

Adjunctive therapy of hypopharyngeal carcinoma by Qingliu Lianghou Recipe. Western Med. Chen, Y, and Wang, Y. Study of Huangqi injection plus raltitrexed combined with oxaliplatin in treatment of advanced colorectal cancer. New Drugs Clin. Choi, Y. Effect of sipjeondaebo-tang on cancer-induced anorexia and cachexia in CT tumor-bearing mice.

Cohen, Z. Selective anticancer effects and protection from chemotherapy by the botanical compound LCS implications for cancer treatment. Cowley, M. The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis.

Neuron 37, — Cui, H. Clinical observation of efficacy of Huangqi injection in prevention and treatment of neurotoxicity induced by oxaliplatin-containing chemotherapy regimen. Adverse Drug React. Dai, Z. Zhong Xi Yi Jie He Xue Bao 6, — Dambach, D.

New technologies and screening strategies for hepatotoxicity: use of in vitro models. Danan, G. RUCAM in drug and herb induced liver injury: the update. Dong, J. Shenqi fuzheng, an injection concocted from Chinese medicinal herbs, combined with platinum-based chemotherapy for advanced non-small cell lung cancer: a systematic review.

Cancer Res. du Bois, A. Evaluation of neurotoxicity induced by paclitaxel second-line chemotherapy. Care Cancer 7, — Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and year survival: an overview of the randomised trials.

Lancet , — European Medicines Agency [EMA] Assessment of Case Reports Connected to Herbal Medicinal Products Containing Cimicifugae Racemosae Rhizoma Black Cohosh, Root.

pdf [accessed December 30, ]. Fang, F. Clinical observation on the efficacy of Shenmai injection in prevention and treatment of neurotoxicity induced by oxaliplatin-containing chemotherapy regimen.

Francois, M. Increased ghrelin but low ghrelin-reactive immunoglobulins in a rat model of methotrexate chemotherapy-induced anorexia.

Frenzel, C. Herbal hepatotoxicity: clinical characteristics and listing compilation. Gao, J. The clinical research of Shenmai injection in prevention of oxaliplatin chemotherapy induced toxicity.

García-Cortés, M. Espanola Enfermed. Grothey, A. Clinical management of oxaliplatin-associated neurotoxicity. Colorectal Cancer 5 Suppl. Hao, T. Zhongguo Zhong Yao Za Zhi 40, — Harada, T. Rikkunshito for preventing chemotherapy-induced nausea and vomiting in lung cancer patients: results from 2 prospective, randomized phase 2 trials.

Hardeman, E. Acute hepatitis induced by greater celandine chelidonium majus. Acta Gastroenterol. Hatakeyama, H. Hangeshashinto improves the completion rate of chemoradiotherapy and the nutritional status in patients with head and neck cancer. ORL J. Hitomi, S. Huang, Y. Study on yiqiyangyin decoction combine with GP chemotherapy in advanced non-small cell lung cancer.

New Chin. Iijima, O. Jing, J. Traditional chinese medicine and herb-induced liver injury: comparison with drug-induced liver injury. Jorden, W. The effect of Shenfu injection in the prevention of oxaliplatin-induced peripheral neurotoxicity. West Med. Joy, J. Amelioration of cisplatin induced nephrotoxicity in Swiss albino mice by Rubia cordifolia extract.

Jun, L. Clinical research on 58 cases of advanced non-small cell lung cancer treated by the intergrated TCM and western medicine. Henan Tradit. Jung, K. Drug-induced liver injury: twenty five cases of acute hepatitis following ingestion of Polygonum multiflorum Thun.

Gut Liver 5, — Kambhampati, S. Intracellular delivery of dendrimer triamcinolone acetonide conjugates into microglial and human retinal pigment epithelial cells. Kang, S.

Clinical characteristics of cases of acute toxic hepatitis. Korean J. Kato, Y. Gosha-jinki-gan reduced oxaliplatin-induced hypersensitivity to cold sensation and its effect would be related to suppression of the expression of TRPM8 and TRPA1 in rats. Drugs 25, 39— Kawashiri, T.

Pain Kebieche, M. Evaluation of epirubicin-induced acute oxidative stress toxicity in rat liver cells and mitochondria, and the prevention of toxicity through quercetin administration. Ko, J. Target-oriented mechanisms of novel herbal therapeutics in the chemotherapy of gastrointestinal cancer and inflammation.

Koehn, F. The evolving role of natural products in drug discovery. Drug Discov. Kohguchi, M. Immuno-potentiating effects of the antler-shaped fruiting body of Ganoderma lucidum Rokkaku-Reishi.

Konmun, J. A phase II randomized double-blind placebo-controlled study of 6-gingerol as an anti-emetic in solid tumor patients receiving moderately to highly emetogenic chemotherapy. Kono, T. Goshajinkigan oxaliplatin neurotoxicity evaluation GONE : a phase 2, multicenter, randomized, doubleblind, placebocontrolled trial of goshajinkigan to prevent oxaliplatininduced neuropathy.

Multitargeted effects of hangeshashinto for treatment of chemotherapy-induced oral mucositis on inducible prostaglandin E2 production in human oral keratinocytes.

Cancer Ther. Kuriyama, A. Goshajinkigan for prevention of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Support Care Cancer 26, — Lenfers, B. Substantial activity of budesonide in patients with irinotecan CPT and 5-fluorouracil induced diarrhea and failure of loperamide treatment.

Li, D. Clinical study on Huangqi Guizhi Wuwu decoction treating the neurosensory toxicity caused by oxaliplatin. Nanjing Univ. Li, H. Zhongguo Zhong Xi Yi Jie He Za Zhi 35, — Production of Th1-and Th2-dependent cytokines induced by the Chinese medicine herb, Rhodiola algida , on human peripheral blood monocytes.

Li, M. To observe the curative effect of Yiqi Yangxue Decoction on bone marrow suppression after chemotherapy.

Shaanxi Tradit. Li, Y. Effect of Jinlong capsule on proliferation and apoptosis of human pancreatic cancer cells BxPC Lin, H. Nursing observation of huangqi guizhi wuwu decoction in preventing peripheral neurotoxicity induced by oxaliplatin.

Gen Nurs. Liu, H. Antidiarrheal activity of extract from herba kummerowiae in mice. Asian J. Liu, X. Kanglaite injection combined with chemotherapy versus chemotherapy alone in the treatment of advanced non-small cell lung carcinoma.

Liu, Y. Clinical Study on Oophoroma After Treated with Fuzhengquyu Decoction Combined with Chemotherapy. Heilongjiang: Heilongjiang University Of Chinese Medicine. Loo, W. Rhodiola algida improves chemotherapy-induced oral mucositis in breast cancer patients. Drugs 19 Suppl.

Lu, M. Effect of astragalus membranaceus in rats on peripheral nerve regeneration: in vitro and in vivo studies. Trauma 68, — Lu, Q. Clinical observation of wenshen shengbai decoction for myelosuppression induced by chemotherapy for postoperative breast cancer.

Shanghai Univers. Lu, Y. Chinese herb related molecules of cancer-cell-apoptosis: a minireview of progress between Kanglaite injection and related genes.

Marx, W. GingerMechanism of action in chemotherapy-induced nausea and vomiting: a review. Food Sci. Matsuda, C. Double-blind, placebo-controlled, randomized phase II study of TJ Hangeshashinto for infusional fluorinated-pyrimidine-based colorectal cancer chemotherapy-induced oral mucositis.

McWhirter, D. Chemotherapy induced hepatotoxicity in metastatic colorectal cancer: a review of mechanisms and outcomes. Menon, L. Effect of rasayanas in the inhibition of lung metastasis induced by B16F melanoma cells.

Mikalauskas, S. Dietary glycine protects from chemotherapy-induced hepatotoxicity. Amino Acids 40, — Molina, J. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin.

Navari, R. Management of chemotherapy-induced nausea and vomiting in pediatric patients. Drugs 19, — Nonaka, Y. Gan To Kagaku Ryoho 32, — Ogawa, K. Protective effect of the Japanese traditional medicine juzentaihoto on myelosuppression induced by the anticancer drug TS-1 and identification of a potential biomarker of this effect.

BMC Complement. Ohkawa, K. Risk factors for antituberculous chemotherapy-induced hepatotoxicity in Japanese pediatric patients. Ohnishi, S. Additive effect of rikkunshito, an herbal medicine, on chemotherapy-induced nausea, vomiting, and anorexia in uterine cervical or corpus cancer patients treated with cisplatin and paclitaxel: results of a randomized phase II study JORTC KMP Oki, E.

Preventive effect of Goshajinkigan on peripheral neurotoxicity of FOLFOX therapy GENIUS trial : a placebo-controlled, double-blind, randomized phase III study. Osama, H. Effect of honey and royal jelly against cisplatin-induced nephrotoxicity in patients with cancer.

Robles-Diaz, M. Distinct phenotype of hepatotoxicity associated with illicit use of anabolic androgenic steroids. Rojas, C. Molecular mechanisms of 5-HT3 and NK1 receptor antagonists in prevention of emesis.

Schele, E. Centrally administered ghrelin acutely influences food choice in rodents. PLoS One e Shindo, Y. Japanese traditional kampo medicine reduce toxicities with colorectal cancer chemotherapy. Reduction of oxaliplatin-related neurotoxicity by goshajinkigan, powdered processed aconite root and reduction of cptrelated diarrhea by hangeshashinto.

Shiomi, Y. Z hydrochloride ameliorates chemotherapy-induced anorexia in rodents via activation of the ghrelin receptor, GHSR1a. Steinmeyer, J. The pharmacological basis of therapeutics in osteoarthritis. Medizinische Welt. Sun, Q. Clinical observation of traditional Chinese medicine combined with chemotherapy in the treatment of advanced non small cell carcinoma.

Hainan Med. Takeda, H. Rikkunshito as a ghrelin enhancer. Methods Enzymol. Pathophysiologic basis of anorexia: focus on the interaction between ghrelin dynamics and the serotonergic system.

Rikkunshito, an herbal medicine, suppresses cisplatin-induced anorexia in rats via 5-HT2 receptor antagonism. Gastroenterology , — Teschke, R. Traditional chinese medicine induced liver injury. Herbal hepatotoxicity in traditional and modern medicine: actual key issues and new encouraging steps.

Greater Celandine hepatotoxicity: a clinical review. Proposal for a kava quality standardization code. Food Chem. Herbal traditional Chinese medicine and its evidence base in gastrointestinal disorders. World J. Review article: herbal hepatotoxicity — An update on traditional Chinese medicine preparations.

Testart-Paillet, D. Contribution of modelling chemotherapy-induced hematological toxicity for clinical practice. Trotti, A. Mucositis incidence, severity and associated outcomes in patients with head and neck cancer receiving radiotherapy with or without chemotherapy: a systematic literature review.

Vaudry, H. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. M S-Med. Vayalil, P. Protective effects of rasayanas on cyclophosphamide- and radiation-induced damage. Rasayanas: evidence for the concept of prevention of diseases. Wadler, S. Recommended guidelines for the treatment of chemotherapy-induced diarrhea.

Wai, C. Presentation of drug-induce dliver injury in Singapore. Singapore Med. Wang, B. Systematic review of beta-elemene injection as adjunctive treatment for lung cancer. Wang, C. Integrated treatment of aqueous extract of solanum nigrum-potentiated cisplatin- and doxorubicin-induced cytotoxicity in human hepatocellular carcinoma cells.

Based Complement. Wang, W. Anti-tumor activities of active ingredients in compound kushen injection. Acta Pharmacol. Wang, Q. Watcha, M. Postoperative nausea and vomiting - its etiology, treatment, and prevention. Anesthesiology 77, —

Herbs Healthy mindset spices can Exteacts so much more than enhance the flavor of food. They can Cancer-fightijg stimulate the immune system and help prevent cancer. Here are six ways to spice up your food and keep you in good health. It is also an anti-inflammatory. Tip: Mix with black pepper piperine and olive oil to activate and help with absorption. Chinese Medicine volume 14Article number: 48 Cite Efffects article. Cancer-fighhing details. Numerous natural products effecte from Chinese Increase cognitive endurance medicine exhibit anti-cancer activities, including anti-proliferative, Increase cognitive endurance, anti-metastatic, anti-angiogenic effects, as well as regulate Building immune system resilience, reverse multidrug Cancr-fighting, balance immunity, and enhance chemotherapy effets vitro and in vivo. To provide new herrbal into the estracts path ahead, we systemically reviewed the ehrbal recent advances reported since on the key compounds with anti-cancer effects derived from Chinese herbal medicine curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine in scientific databases PubMed, Web of Science, Medline, Scopus, and Clinical Trials. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.

Author: Basar

5 thoughts on “Cancer-fighting effects of herbal extracts

  1. Ich meine, dass Sie den Fehler zulassen. Geben Sie wir werden besprechen. Schreiben Sie mir in PM, wir werden reden.

  2. Entschuldigen Sie, was ich jetzt in die Diskussionen nicht teilnehmen kann - es gibt keine freie Zeit. Ich werde befreit werden - unbedingt werde ich die Meinung in dieser Frage aussprechen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com