Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology

Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology

Journal Pre-proof Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology Thomas Efferth, Franz Oesch PII: S1044-579X(19)3040...

5MB Sizes 0 Downloads 66 Views

Journal Pre-proof Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology Thomas Efferth, Franz Oesch

PII:

S1044-579X(19)30408-0

DOI:

https://doi.org/10.1016/j.semcancer.2019.12.010

Reference:

YSCBI 1734

To appear in:

Seminars in Cancer Biology

Received Date:

24 July 2019

Accepted Date:

15 December 2019

Please cite this article as: Efferth T, Oesch F, Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology, Seminars in Cancer Biology (2019), doi: https://doi.org/10.1016/j.semcancer.2019.12.010

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier.

Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology

Thomas Efferth1* and Franz Oesch2

Department of Pharmaceutical Biology, Johannes Gutenberg University, Mainz, Germany

2

Institute of Toxicology, Medical Center, Johannes Gutenberg University, Mainz, Germany

ro of

1

* Corresponding author: Staudinger Weg 5, 55128 Mainz, Germany. Tel: 49-6131-3925751; Fax: 49-

Jo

ur

na

lP

Running title: Alkaloids for cancer therapy

re

-p

611-3923752; E-mail: [email protected]

1

Abstract Drug repurposing (or repositioning) is an emerging concept to use old drugs for new treatment indications. Phytochemicals isolated from medicinal plants have been largely neglected in this context, although their pharmacological activities have been well investigated in the past, and they may have considerable potentials for repositioning. A grand number of plant alkaloids inhibit syngeneic or xenograft tumor growth in vivo. Molecular modes of action in cancer cells include induction of cell cycle arrest, intrinsic and extrinsic apoptosis, autophagy, inhibition of angiogenesis and glycolysis, stress and anti-inflammatory responses, regulation of immune functions, cellular differentiation, and inhibition of invasion and metastasis. Numerous underlying signaling processes are affected by plant alkaloids. Furthermore, plant alkaloids suppress carcinogenesis, indicating chemopreventive

ro of

properties. Some plant alkaloids reveal toxicities such as hepato-, nephro- or genotoxicity, which disqualifies them for repositioning purposes. Others even protect from hepatotoxicity or cardiotoxicity of xenobiotics and established anticancer drugs. The present survey of the published literature clearly demonstrates that plant alkaloids have the potential for repositioning in cancer therapy. Exploitation

-p

of the chemical diversity of natural alkaloids may enrich the candidate pool of compounds for cancer chemotherapy and –prevention. Their further preclinical and clinical development should follow the

re

same stringent rules as for any other synthetic drug as well. Prospective randomized, placebocontrolled clinical phase I and II trials should be initiated to unravel the full potential of plant alkaloids

lP

for drug repositioning.

Introduction

na

Key words: Natural products, Phytochemicals, Network pharmacology, Targeted chemotherapy

ur

The term drug repurposing (or repositioning, re-profiling) describes the use of already approved drugs against one disease for re-use against another disease [1]. Proof-of-principle examples are the new

Jo

applications of thalidomide against multiple myeloma and leprosy as well as sildenafil against erectile dysfunction and pulmonary hypertension. As a matter of fact, the number of newly released FDAapproved drugs decreased since the 1990s, and there is an urgent need for novel drugs against cancer as well as other diseases [2]. Old drugs have several advantages compared to the development of new entities: Their safety and toxicity profile as well as their pharmacokinetic behavior are already wellstudied in human beings, which considerably decreases the drug developmental costs [3]. Issues of intellectual property with old drugs may be overcome by developing novel derivatives based on the chemical scaffolds of old drugs [4]. New anticancer drugs are required because of the development of 2

tumor resistance to established drugs and their severe, partwise life-threatening side effects [5]. Drug resistance may occur due to the occurrence of mutations in critical genes and heterogeneic tumor subpopulations, which are non-responsive to treatment [6-9]. The quest for drug repositioning opportunities is greatly facilitated by computational approaches, which allow the identification of novel targets and signaling pathways for old drugs [10-14]. A timely overview of synthetic drugs considered for repositioning is given by Olgen and Kotra (2018) [15]. In the present review, we focus on natural compounds, i.e. alkaloids from plants. During the past years, the main spotlight of drug repositioning research was on FDA-approved synthetic drugs and on the market for other disease indications. The field is, however, much larger and the entire area of medicinal herbs and their isolated phytochemicals have been largely neglected as of

ro of

yet. However, phytochemicals offer a huge potential for being repositioned. During the past decades, several ten-thousand phytochemicals have been isolated from plants used in traditional medical system all over the world. These medicinal plants have been applied for hundreds or thousands of years for diverse disease conditions. In addition to traditional medicine, rationale phytotherapy and

-p

pharmacognosy as disciplines of academic pharmacy programs led to the development of numerous approved phytotherapeutics on the market.

re

We give here just a few examples out of many others to illustrate the suitability of isolated phytochemicals for drug repositioning, since they reveal more than one specific pharmacological activity:

Artemisinin from Artemisia annua, reveals not only anti-malarial activity, but is also active

lP



against cancer, certain viral infections, trypanosomiasis, schistosomiasis etc. [16, 17]. 

Colchicin from Colchicum autumnale kills cancer cells in vitro, but cannot be used in cancer



na

patients because it is too toxic. Instead, it is clinically applied to treat gout. Ergot alkaloids from Claviceps purpurea are (partial) dopamine receptor and serotonin

ur

receptor agonists, α-adrenoreceptor antagonist, inhibit prolactin and somatotropin release, act against migraine, and contract uterus musculature and blood vessels. The synthetic ergot derivative, bromocriptine is also active against Parkinson disease. Quinine, quinidine, and cinchonine from Cinchona officinalis are active against malaria and

Jo



cardiovascular diseases.



Tetrahydrocannabiol and cannabidiol from Cannabis sativa are banned for its addictive potential. However, they were recently reconsidered for pain treatment in patients suffering from late stage cancer or multiple sclerosis.

To our opinion, it is worth investigating, whether phytochemicals exert therapeutic potentials to treat cancer. The idea is not to promote unproven herbal preparations from complementary and alternative medicine, but to scientifically investigate chemically defined compounds from medicinal 3

plants and to take them as chemical lead scaffolds for chemical derivatization. Novel derivatives are then supplied to the pharmaceutical drug development pipeline in a comparable manner as synthetic drugs as well. The validity of this concept is substantiated by surveys of the National Cancer Institute, USA, showing that 75 % of all anticancer drugs are in the one way or another based on natural resources [18, 19]. This astonishingly high percentage emphasizes the relevance of naturally derived drugs for cancer chemotherapy. It can be envisioned that nature developed complex biosynthesis routes to produce phytochemicals during evolution of plants on earth as chemical defense against microbial attack (viruses, bacteria, protozoans) and herbivores (insects, worms, mammals). With the help of medical chemistry, these phytochemicals can serve as lead compounds to adapt and optimize them to take

ro of

advantage of their pharmacological activities and to treat diseases in patients.

There is a plethora of literature on the cytotoxic activity of natural compounds from diverse chemical classes, e.g. terpenoids, lignans, steroids, flavonoids, naphthoquinones, anthranoids, plant acids, saponins, alkaloids etc. It is beyond the scope of this review to report on all data available.

-p

Rather, we focused on one chemical class of phytochemicals, which is long known for its pharmacological activities, i.e. alkaloids.

re

We performed a systematic review by screening the PubMed database with the search terms “repositioning/repurposing + alkaloid + plant + cancer + in vivo/xenograft”. Papers written in English were included until June 20, 2019. Since there are more than 3000 papers on the cytotoxic activity of

lP

plant alkaloids towards cancer cells in general and since many compounds cytotoxic in vitro do not reveal tumor inhibition in living organisms, we restricted our search to investigations showing anticancer activity of plant alkaloids in vivo. Our search strategy was confined to alkaloids that are

na

already known for their pharmacological activity for other diseases than cancer, in order to emphasize their potential for drug repurposing. We also highlighted the toxicity of these plant alkaloids. This

ur

represents an important aspect, if it comes to clinical drug development. Only those compounds can be further considered which reveal convincing safety and tolerability. To further focus our review, we did not include publications on combination treatments of plant alkaloids with other drugs.

Jo

Furthermore, (semi)synthetic derivatives of plant alkaloids and nanotechnological preparations including plant alkaloids were also excluded. The intention of the present review was to consider, whether plant-derived alkaloids with diverse

pharmacological activities may also be further developed for cancer chemotherapy. As a result, we came up with a panel of alkaloids, which are compiled in Table 1. This table shows not only the pharmacological activities beyond cancer, but also the plants they have been isolated from. Most of these plants have been used in traditional medicine in Asia, Near East or South-America since ages. Their medical uses have also been presented. By literature mining, we further analyzed, whether these 4

plant-derived alkaloids might be appealing candidates for drug repositioning in cancer therapy. Based on the available safety and efficacy data in vivo, plant-derived alkaloids may be discussed as lead compounds for further drug development.

Inhibition of primary tumors Induction of cell cycle arrest In this review, we considered reports performed both with syngeneic murine or rat tumors transplanted to rodents as well as human xenograft tumors transplanted to nude mice. Syngeneic tumors have the advantage that the host animals have an intact immune system, which means that

ro of

immune defense mechanisms against tumors contribute to the overall anticancer activity of investigated compounds. Although xenograft tumors are transplanted to immuno-deficient mice, this model has the advantage that human tumors can be investigated in living organisms (albeit not in human patients).

An early reaction of tumor cells to toxic insults is the induction of cell cycle arrest, which allows

-p

cells either to repair damaged sites or to induce programmed cell death. Cell cycle arrest can occur at different phases of the cell cycle. This has been shown not only for synthetic drugs, but also for

re

phytochemicals [20-24]. Defined checkpoints serve the cellular control of genomic integrity. Arrest at these checkpoints enable the repair of lesions. The G0/1 and G2/M checkpoints allow the repair of

lP

DNA lesions, while the S-phase arrest enables repair during DNA replication. The cell cycle is driven and by three regulatory levels: (1) cyclins drive the cell cycle from G1 to S, G2 and M phases; (2) cyclin dependent kinases (CDKs) control the activity of cyclins; (3) CDK inhibitors control CDKs (p21, B). ATM

na

and ATR are sensor proteins which regulate the tumor suppressor p52 to control cyclins and CDKs in various cell cycle phases to induce DNA repair. Plant alkaloids induced either G0/G1 arrest or G2/M arrest. In few cases even S phase arrest was reported (Table 2). Under conditions, where DNA repair

ur

cannot be performed (e.g. growth factor deficiency, overload of the DNA repair capacity etc.), p53 can

Jo

act as sensor to induce apoptosis.

Induction of apoptosis Numerous investigations on phytochemicals describe the inhibition of tumor growth by induction of classical programmed cell death (apoptosis) in vitro [25-30]. There are much less in vivo investigations. Table 3 shows a large panel of phytochemicals from diverse alkaloid classes, which induced apoptosis in both syngeneic and xenograft tumors. A broad spectrum of different tumor entities rather than certain tumor types were inhibited, e.g. colorectal, breast, gastric, endometrial, cervical, hepatocellular, lung, and prostatic carcinoma, glioblastoma, melanoma, osteosarcoma, as well as rare tumors such as pheochromocytoma, neuroblastoma, and cholangiocarcinoma. The alkaloids analyzed 5

seem to exert a general anticancer activity. It is pleasing that also some rare tumor types have been investigated. Frequently, the treatment options available for rare tumors are not satisfactory. Therefore, the treatment of pheochromocytoma, neuroblastoma, and cholangiocarcinoma with plantderived alkaloids may offer an opportunity to improve the outcome of these diseases. The majority of investigations reported the induction of the intrinsic, mitochondrial pathway of apoptosis by plant alkaloids. Mitochondria are central for the intrinsic pathway of apoptosis. Members of the Bcl-2 family are located in the outer mitochondrial membrane. There are pro-apoptotic members (Bax, Bak etc.) and anti-apoptotic members (Bcl-2, Bcl-xL etc.), which form homo- and heterodimers. Upon suitable stimuli (oxidative stress, xenobiotics, cytotoxic drugs etc.), the formation of pro-apoptotic dimers prevail, leading to a release of cytochrome C from the inside of mitochondria

ro of

to the cytosol. Then, cytochrome C binds to Apap-1, which in turn binds to procapsapse-9. This complex has been termed apoptosome. Procaspase-9 is cleaved by the apoptosome into caspase-9 (initiator caspase). This protease cleaves procaspase-3 into the active caspase-3 (effector caspase), which executes cell death. In parallel to cytochrome c, SMACs are released from the mitochondria into the

-p

cytosol, which bind and inhibit IAPS. IAPs (cIAP-1/2, XIAP) are proteins that inhibit caspases. IAP inhibition by SMACs allows activation of caspases by autophagosomes [31]. As shown in Table 3, the

re

induction of the intrinsic mitochondrial pathway is well documented for several plant alkaloids with anticancer activity.

lP

Only a few authors described the involvement of Fas as major player of the extrinsic apoptotic pathway by plant alkaloids. The Fas ligand (or other ligands such as TNF-α) bind to the Fas receptor or other death receptors of the TNF-α receptor family. Binding of ligands to their corresponding receptors

na

lead to the formation FADD or TRADD complexes, which activates caspase-8 (initator caspase). cIAP1/2 and FLIP are negative regulators that can inhibit the extrinsic receptor-driven pathway of apoptosis [31]. Based on the available data (Table 3), it is not clear, whether or not intrinsic apoptosis is more

ur

important for plant alkaloids than the extrinsic mode of cell death for the anticancer activity of some plant alkaloids. It is possible that the extrinsic death receptor-driven pathway has been less

Jo

investigated, but is of similar importance as intrinsic apoptosis. This open question should be addressed in more detail in the future. It is a striking feature that the alkaloids induced apoptosis was independent of their chemical

structure or biological function. This indicates that apoptosis may not be the primary target of these compounds, but that the actual drug targets are more upstream from the apoptosis cascade. Most – if not all - clinically established anticancer drugs address different target structures (e.g. microtubule, DNA topoisomerases I or II, DNA biosynthetic enzymes, DNA lesions, hormones, growth factor receptors, signaling kinases etc.). However, the interaction of these anticancer drugs with their targets 6

finally leads to the induction of apoptosis further downstream in the signaling cascade. The same may be true for the diverse plant alkaloids shown in Table 3.

Induction of non-apoptotic cell death Unraveling the molecular mechanisms of apoptosis during the past three decades also brought up results showing that programmed cell death can be committed in a non-apoptotic fashion. One of the most prominent caspase-independent, non-apoptotic mechanisms is autophagic cell death. Human homologues of the atg genes from Saccharomyces cerevisiae play a major role in autophagy. mTOR and AMPK phosphorylate the Atg1 homologues ULK-1 and ULK-2. Their dephosphorylation results in activation of these two kinases, which then phosphorylate and activate the Atg6 homologue Beclin-1.

ro of

ULK-1/2 and Beclin-1 are parts of a larger protein complex, which also consists of VPS34. This protein activates further downstream events involving further Atg homologues leading to the formation of the autophagosome and activation of the LC3 protein, which enables fusion with lysosomes. Then, these vesicles release their hydrolytic enzymes that degrade cellular proteins leading to autophagic cell

-p

death [32, 33].

Autophagy in tumors by phytochemicals in general and specifically by plant alkaloids has been

re

less intensively investigated than apoptosis [34-36]. Nevertheless, quite a number of reports deliver convincing evidence that autophagy represents an important mechanism involved in the inhibition of

lP

tumor growth in vivo (Table 4). Autophagy can occur either alone or together with apoptosis upon treatment with some plant alkaloids.

In recent years, further modes of cell death have been described. Although this novel field of

na

research has not been well studied yet in the context of plant alkaloids, a few authors reported novel non-apoptotic cell death modes such as pyroptosis (inflammatory cell death) or oncosis (ischemic cell death) [37, 38]. The role of non-apoptotic forms of cell death and their contribution to the overall

ur

inhibition of tumor growth by plant alkaloids in comparison to apoptosis and autophagy deserves

Jo

further attention in the years to come.

Inhibition of tumor angiogenesis While early in tumor development small tumors maintain their supply of nutrients and oxygen by passive diffusion from the surrounding normal tissue environment, larger tumors cannot. They suffer from hypoxic areas. Tumor cells react to this challenge by a process called angiogenic switch. The transcription factor HIF-1α activates expression of proteins that stimulate the formation of new blood vessels (neoangiogenesis) growing into the tumor. One of the best known angiogenic factors is VEGF.

7

There is a balance of proteins that act in a pro-angiogenic (VEGF, bFGF, TGF-β1, IGF, G-CSF and others) or anti-angiogenic fashion (e.g. ANG, END, INFα, p53) [39, 40]. The formation of novel vessels represents a typical feature of progressive tumors. The inhibition of neoangiogenesis became an important treatment principle in the past years and several angiogenesis inhibitors were market as anticancer drugs (e.g. bevacizumab) [41, 42]. The inhibition of angiogenesis is also an important mechanisms of natural products and their derivatives [43-45]. As can be seen in Table 5, a broad range of tumor models have been used to investigate the antiangiogenic effects of plant alkaloids in vivo, including the downregulation of VEGF and its receptors, as well as signaling molecules such as AKT, SRC, FAK, ERK, p53, and transcription factors

ro of

such as NFκB and mTOR. Other typical regulators of angiogenesis have not been reported for syngeneic or xenograft tumors yet. Future investigations should, therefore, focus on other angiogenesis regulators than VEGF to explore the full mechanistic regulation of angiogenesis inhibition by plant

-p

alkaloids.

Inhibition of signal transduction

re

Cellular mechanisms such as cell death, angiogenesis, differentiation, stress response, immune response, anti-inflammatory response etc. are all driven and regulated by signal transduction

lP

pathways. Therefore, it is important to understand the signaling routes that are affected upon treatment of tumors with phytochemicals [46-50]. As can be seen from Table 6, plant alkaloids influenced cellular signaling by many different pathways. It is a general feature of most natural product

na

that they are not mono-specific, but act in multi-specific modes of action. Therefore it comes as no surprise that multiple signaling pathways are either activated or shut off by alkaloids. Numerous transcription factors were described to be affected by plant alkaloids, including

ur

NFκB, MYC, FOS, CREB, WT1, mTOR and others. The WNT/β-catenin, ERK-, JNK-, and AKT-related pathways, as well as STAT and AMPK-related pathways all have been reported to be involved in plant

Jo

alkaloid-induced cellular effects.

Other mechanisms In addition to angiogenesis, several other mechanisms play a role for the antitumor activity of plant alkaloids, albeit these mechanisms have been much less investigated as yet. In comparison to the above mentioned mechanisms, few papers report on the regulation of immune function (release of cytokines, upregulation of innate immune responses), the anti-inflammatory activity, stress response (antioxidant stress response genes, endoplasmic reticulum stress, heat shock protein HSP70), and alterations in tumor metabolism (impaired aerobic respiration and glycolysis), and tumor 8

differentiation upon treatment with plant alkaloids (Table 7). These modes of actions of plant alkaloids warrant more detailed investigation in the future to better understand their role for tumor inhibition.

Inhibition of invasion and metastasis The chapters above all deal with primary tumors. Most patients die, however, not because of their primary tumors, but because the primary tumors invade the neighboring tissues and form lymph node metastases or metastases in distant organs after spreading of tumor cells via the blood system. The metastatic process of progressive tumors with its different steps (local invasion, intravasation, circulation, extravasation and proliferation) represents, therefore, a mechanism of utmost importance for the survival prognosis of cancer patients.

ro of

Loss of contact inhibition and altered cell adhesion are important for invasion and migration of tumor cells. Migrating cells decrease the expression of E-cadherin (which regulates adhesion between epithelial cells) and upregulate the expression of N-cadherin (which mediates the interaction between tumor and stroma cells). Tumor cells degrade the extracellular matrix (ECM), which maintains tissue

-p

structures and acts as barrier for cell invasion. Among the ECM constituents are collagens, fibronectin, laminins proteoglycans and many others. Cellular receptors of ECM constituents are integrins.

re

Proteolytic enzymes of ECM are uPA and MMPs. Inhibitors of MMPs are TIMPs [51, 52]. The capability of tumor cells to migrate is increased by a phenotypic change of epithelial to mesenchymal cellular

lP

features (e.g. downregulation of cytokeratin as epithelial marker and upregulation of vimentin as mesenchymal marker) to facilitate the metastasis process. Once the metastasizing cells reached their target organ, they differentiate back to epithelial cells. This phenomenon has been termed epithelial-

na

mesenchymal transition (EMT) [53, 54]. Chemokines (e.g. CXCRs) navigate metastasizing tumor cells into certain organs and favor their nidation there – a phenomenon termed homing factor. Typical organs of metastasis are lung, liver, and brain bone marrow. Certain transcription factors, e.g. SNAIL-

ur

1/2 and SLUG, orchestrate the expression of these invasion and metastasis factors. Numerous investigations pointed to the inhibition of metastatic tumors by phytochemicals [55-

Jo

59]. We report here on papers, which describe either the inhibition of metastases by plant alkaloids in vivo or the inhibition of primary tumors in vivo and mechanisms of metastasis inhibition in vitro (Table 8). Plant alkaloids inhibited metastasis process into the lung and liver, inhibited EMT (upregulation of mesenchymal markers such as vimentin, downregulation of epithelial markers such as cytokeratin), inhibited matrix metalloproteinases (MMP-2, MMP-3, MMP-9, MMP-13), the metastasis-regulating transcription factors SNAIL-1 and SNAIL-2, uPA, cathepsin C etc. Among the adhesion molecules, the upregulation of E-cadherin and downregulation of N-cadherin was described. Several signal transduction pathways were not only inhibited in primary tumors (see chapters above), but also in metastasis by plant alkaloids, e.g. WNT/β-catenin, JAK, STAT, ERK, AMPK, and TGF-β 9

signaling etc. Mechanisms of plant alkaloid actions found in primary tumors (see Tables 2-5) were also observed in metastasis, e.g. cell cycle arrest, apoptosis, and angiogenesis inhibition. Interestingly, inflammatory markers (e.g. COX-2, PGE2) were also inhibited in metastasis by alkaloids, indicating that inflammation may not only play a role for initial tumorigenesis, but also for the metastatic process. The activation of natural killer cell activity by theophylline is a clue that plant alkaloids affect the immune system to exert anti-metastatic effects. This aspects requires more attention, and more investigations are required with more plant-derived alkaloids and more tumor types. Especially, the influence of the immune system should be compared in tumor-bearing immunocompetent and immuno-deficient mice.

ro of

Chemoprevention of carcinogenesis

All results presented so far in this review article focused on the tumor treatment, i.e. eradication of already established tumors. Another aspect that has to be taken into account is the prevention of tumor development from the beginning on. Chemoprevention is an important field in cancer research,

-p

as long as all established therapy options such as chemotherapy, radiotherapy, immunotherapy and surgery do not reveal satisfying success rates to reliably cure cancer [60, 61]. As natural products are

re

frequently considered as being well tolerated with few side effects, chemoprevention of carcinogenesis by phytochemicals may represent an attractive strategy to combat cancer [62-64].

lP

Plant alkaloids product may prevent carcinogenesis by chemicals (chemical carcinogenesis), irradiation (physical carcinogenesis) and viral infections (biological carcinogenesis). A number of reports found that the incidence of tumors induced by chemical carcinogens (BP, NKK, DEN, DMBA,

na

DMH, DSS, urethane) were significantly reduced by co-application of plant alkaloids (Table 9). Several papers describe the chemopreventive effects of plant alkaloids on UV-B light induced skin carcinogenesis as example of physical carcinogenesis (Table 9).

ur

Plant alkaloids have not been investigated yet in the context of biological carcinogenesis, i.e. virally induced tumors. However, the expression of the lymphoma-inducing Epstein-Barr protein EBNA-1 was

Jo

downregulated in nasopharyngeal xenograft tumors by berberine [65]. This indicates that plant alkaloids may bear the potential also to prevent virus-induced carcinogenesis. Another approach to investigate the chemopreventive activity of plant alkaloids was the

application of genetically manipulated animals, which spontaneously develop tumors, e.g. ApcMin/+ mice or TRAMP mice. Alternatively, non-manipulated tumor cells were transplanted in low number into animals. In all these cases, some plant alkaloids efficiently inhibited carcinogenesis (Table 9).

Toxicity of plant alkaloids 10

The use of plant alkaloids for cancer therapy depends not only on their efficacy to inhibit tumor growth, but also on their tolerability by normal tissues. Although natural products in general are frequently considered as being safe and although many natural products are indeed safe, others are not [66-72]. Therefore, the testing for toxicities exerted by plant alkaloids represents a sine qua non condition in the drug development process in the same manner, as it is performed for synthetic drugs. As shown in Table 10, organ toxicities have been observed in animals transplanted with syngeneic tumors and treated with piperine, piplartine, or oxymatrine, such as hepatotoxicity and nephrotoxicity. While these acute toxicities might be reversible after cessation of drug treatment, it is alarming that some alkaloids seem to stimulate tumor growth and metastasis or act as co-carcinogens (capsaicin, piperine). Others seem to be genotoxic (sanguinarine, harmine, caffeine). As a matter of

ro of

course, drugs applied to cure cancer have to be neither genotoxic nor carcinogenic. In addition to DNAdamaging agents which initiate tumorigenesis, caffeine has been found to stimulate mammary gland development. This result might be interpreted as a proliferation-promoting effect, which is also not desirable for antitumor drugs. These plant alkaloids are, hence, not suited as candidates for drug

-p

repurposing in cancer therapy.

Other alkaloids reveal milder side effects such as inflammation (theophylline) or no observed

re

side effects (noscapine, sinomenine, coptisine, matrine). These alkaloids might be more suitable for further repositioning in cancer therapy than the above mentioned toxic alkaloids. Interestingly, there are also authors reporting on plant alkaloids that prevented laboratory

lP

animals from toxicity caused by other agents. Berberine protected from the gastrointestinal mucosa from heavy alcohol consumption and doxorubicin-induced cardiomyopathy. Nuciferine protected from nicotine-induced liver injury (Table 10). These two alkaloids may, therefore, be beneficial partners for

na

combination chemotherapy together with other established anticancer drugs. This aspect deserves

ur

further attention in the future.

Conclusion and perspectives

Jo

There is a grand number of alkaloids derived from plants that could be considered for repositioning in cancer therapy, because they inhibited the growth of syngeneic and xenograft tumors in vivo. A lot is known about the molecular modes of action in cancer cells, e.g. induction of cell cycle arrest in G0/G1 cell cycle phases, induction of intrinsic and extrinsic apoptosis, autophagy and other modes of nonapoptotic cell death, inhibition of angiogenesis and glycolysis, stress and anti-inflammatory responses, regulation of immune functions, induction of cellular differentiation, inhibition of invasion and metastasis. All these mechanisms are regulated by specific signal transduction processes, which were affected by plant alkaloids. Furthermore, plant alkaloids suppressed carcinogenesis, indicating that they might be used as chemopreventive drugs. As all pharmacologically active drugs, some plant 11

alkaloids already at therapeutically potentially useful doses revealed toxicities such as hepato- nephroor genotoxicity, which disqualifies them for repositioning purposes. Others were described to even protect from hepatotoxicity or cardiotoxicity of xenobiotics and established anticancer drugs. The present survey of the published literature demonstrates that plant alkaloids may have the potential to be repositioned for cancer therapy. Exploitation of the chemical diversity of natural alkaloids may enrich the candidate pool of compounds for cancer chemotherapy and –prevention. Previously, natural products have sometimes been critically discussed and called dirty drugs, because they are not mono-specific, but exert multiple effects. This multi-specificity however, turned out as selection advantage during evolution of plant life, because multi-specific compounds are less

ro of

prone to resistance development than mono-specific ones [73]. During the past decades, it turned out that many synthetic drugs are also not mono-specific. This is actually the basis of the entire concept of drug repositioning and increasingly emerges as considerable advantage in the development of new strategies to combat diseases such as cancer.

-p

This review does not only show the achievements of the past years to understand the anticancer activity of plant alkaloids, it also discloses gaps and deficiencies in research. For example, less is still known on the role of stem cells or the microenvironment for the activity of plant alkaloids.

re

The mechanisms behind the influence of alkaloids on immune system, on anti-inflammatory and stress response reactions, on cancer metabolism and on tumor differentiation are not well understood.

lP

Epigenetics as new field of research in many disciplines of life sciences has been barely investigated in the context of alkaloids. How do plant alkaloids influence DNA methylation, histone acetylation or miRNA regulation? These and other questions deserve to be addressed in the future to better evaluate

na

the potential of plant alkaloid for repositioning in cancer therapy. Research on plant alkaloids is not only valuable to bring them into the clinics. Knowledge

ur

gained with investigating these compounds may also be utilized for the generation of derivatives with improved pharmacological features, i.e. improved bioavailability and pharmacokinetics, less resistance

Jo

development and less side effects etc. For instance matrine and oxymatrine are quinazoline alkaloids. Knowledge gained with this alkaloid class may be helpful to synthesize even completely novel quinazoline alkaloids for therapeutic purposes. Several synthetic quinazoline alkaloids entered the clinic during recent years, e.g. afatinib, erlotinib, gefitinib and lapatinib. The further preclinical and clinical development of plant alkaloids should follow the same stringent rules as any synthetic drug as well. What is most required at this point is the conduction of clinical studies to estimate the clinical utility of plant-derived alkaloids. Though preliminary clinical data are available [74, 75], prospective randomized, placebo-controlled clinical Phase I and II trials should 12

be initiated in the years to come to unravel the full potential of plant alkaloids in the drug repositioning process.

Funding Source There were no extramural funds to write this paper.

Jo

ur

na

lP

re

-p

ro of

Conflict of interest: The authors declare that there no conflict of interest.

13

References

Jo

ur

na

lP

re

-p

ro of

1. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov 2004;3(8):673–683. 2. Sleire L, Førde HE, Netland IA, Leiss L, Skeie BS, Enger PØ. Drug repurposing in cancer. Pharmacol Res 2017;124:74-91. 3. Yang EJ, Wu C, Liu Y, Lv J, Sup Shim J. Revisiting Non-Cancer Drugs for Cancer Therapy. Curr Top Med Chem. 2016;16(19):2144-2155. 4. Chen H, Wu J, Gao Y, Chen H, Zhou J. Scaffold Repurposing of Old Drugs Towards New Cancer Drug Discovery. Curr Top Med Chem 2016;16(19):2107-2114. 5. Fong W, To KKW. Drug repurposing to overcome resistance to various therapies for colorectal cancer. Cell Mol Life Sci 2019 [Epub ahead of print] 6. Li YY, Jones SJ. Drug repositioning for personalized medicine. Genome Med 2012;4(3):27. 7. Schmidt F, Efferth T. Tumor heterogeneity, single-cell sequencing, and drug resistance. Pharmaceuticals (Basel) 2016;9(2). pii: E33. 8. Hamdoun S, Jung P, Efferth T. Drug repurposing of the anthelmintic niclosamide to treat multidrug-resistant leukemia. Front Pharmacol 2017;8:110. 9. Seo EJ, Sugimoto Y, Greten HJ, Efferth T. Repurposing of bromocriptine for cancer therapy. Front Pharmacol 2018;9:1030. 10. Kadioglu O, Efferth T. Contributions from emerging transcriptomics technologies and computational strategies for drug discovery. Invest New Drugs 2014;32(6):1316-1319. 11. Jiao M, Liu G, Xue Y, Ding C. Computational drug repositioning for cancer therapeutics. Curr Top Med Chem 2015;15(8):767-775. 12. Würth R, Thellung S, Bajetto A, Mazzanti M, Florio T, Barbieri F. Drug-repositioning opportunities for cancer therapy: novel molecular targets for known compounds. Drug Discov Today 2016;21(1):190-199. 13. Cheng F, Hong H, Yang S, Wei Y. Individualized network-based drug repositioning infrastructure for precision oncology in the panomics era. Brief Bioinform 2017;18(4):682-697. 14. Peyvandipour A, Saberian N, Shafi A, Donato M, Draghici S. A novel computational approach for drug repurposing using systems biology. Bioinformatics 2018;34(16):2817-2825. 15. Olgen S, Kotra L. Drug repurposing in the development of anticancer agents. Curr Med Chem 2018 [Epub ahead of print]. 16. Efferth T. From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin Cancer Biol 2017;46:65-83. 17. Efferth T. Beyond malaria: The inhibition of viruses by artemisinin-type compounds. Biotechnol Adv 2018;36(6):1730-1737. 18. Newman DJ, Cragg GM. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J Nat Prod 2012;75(3):311-335. 19. Newman DJ, Cragg GM. Natural products as sources of new drugs from 1981 to 2014. J Nat Prod 2016;79(3):629-661 20. Wu CF, Klauck SM, Efferth T. Anticancer activity of cryptotanshinone on acute lymphoblastic leukemia cells. Arch Toxicol 2016;90(9):2275-2286. 21. Bi YL, Min M, Shen W, Liu Y. Genistein induced anticancer effects on pancreatic cancer cell lines involves mitochondrial apoptosis, G0/G1cell cycle arrest and regulation of STAT3 signalling pathway. Phytomedicine 2018;39:10-16 22. Özenver N, Saeed M, Demirezer LÖ, Efferth T. Aloe-emodin as drug candidate for cancer therapy. Oncotarget 2018;9(25):17770-17796. 23. Sampath S, Subramani S, Janardhanam S, Subramani P, Yuvaraj A, Chellan R. Bioactive compound 1,8-Cineole selectively induces G2/M arrest in A431 cells through the upregulation of the p53 signaling pathway and molecular docking studies. Phytomedicine 2018;46:57-68.

14

Jo

ur

na

lP

re

-p

ro of

24. Liu JS, Huo CY, Cao HH, Fan CL, Hu JY, Deng LJ, Lu ZB, Yang HY, Yu LZ, Mo ZX, Yu ZL. Aloperine induces apoptosis and G2/M cell cycle arrest in hepatocellular carcinoma cells through the PI3K/Akt signaling pathway. Phytomedicine 2019;61:152843. 25. Zhao Q, Kretschmer N, Bauer R, Efferth T. Shikonin and its derivatives inhibit the epidermal growth factor receptor signaling and synergistically kill glioblastoma cells in combination with erlotinib. Int J Cancer 2015;137(6):1446-1456. 26. Parveen A, Akash MS, Rehman K, Kyunn WW. Anticancer activities of medicinal plants: Modulation of p53 expression and induction of apoptosis. Crit Rev Eukaryot Gene Expr 2016;26(3):257-71. 27. Jin W, Zhou L, Yan B, Yan L, Liu F, Tong P, Yu W, Dong X, Xie L, Zhang J, Xu Y, Li C, Yuan Q, Shan L, Efferth T. Theabrownin triggers DNA damage to suppress human osteosarcoma U2OS cells by activating p53 signalling pathway. J Cell Mol Med 2018;22(9):4423-4436 28. Souza FO, Sorbo JM, Regasini LO, Bolzani VDS, Rosa JC, Czernys ÉDS, Valente V, Moreira TF, Navegante G, Fernandes BC, Soares CP. Nitensidine B affects proteins of the glycolytic pathway and induces apoptosis in cervical carcinoma cells immortalized by HPV16. Phytomedicine 2018;48:179-186. 29. Mbaveng AT, Bitchagno GTM, Kuete V, Tane P, Efferth T. Cytotoxicity of ungeremine towards multi-factorial drug resistant cancer cells and induction of apoptosis, ferroptosis, necroptosis and autophagy. Phytomedicine 2019:152832 30. El-Readi MZ, Eid S, Abdelghany AA, Al-Amoudi HS, Efferth T, Wink M. Resveratrol mediated cancer cell apoptosis, and modulation of multidrug resistance proteins and metabolic enzymes. Phytomedicine 2019;55:269-281. 31. Alberts B, Johnson A, Lewis J, Morgan D, Raff M, Roberts K, Walter P. Molecular Biology of the Cell (6th ed.). Garland Science. 2015. ISBN 978-0815344322. 32. Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer 2017;17(9):528-542. 33. Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol 2018;19(6):349-364. 34. Saeed M, Kadioglu O, Khalid H, Sugimoto Y, Efferth T. Activity of the dietary flavonoid, apigenin, against multidrug-resistant tumor cells as determined by pharmacogenomics and molecular docking. J Nutr Biochem 2015;26(1):44-56. 35. Hegazy MF, Abdelfatah S, Hamed AR, Mohamed TA, Elshamy AA, Saleh IA, Reda EH, AbdelAzim NS, Shams KA, Sakr M, Sugimoto Y, Paré PW, Efferth T. Cytotoxicity of 40 Egyptian plant extracts targeting mechanisms of drug-resistant cancer cells. Phytomedicine 2019;59:152771. 36. Cai CZ, Zhou HF, Yuan NN, Wu MY, Lee SM, Ren JY, Su HX, Lu JJ, Chen XP, Li M, Tan JQ, Lu JH. Natural alkaloid harmine promotes degradation of alpha-synuclein via PKA-mediated ubiquitin-proteasome system activation. Phytomedicine 2019;61:152842. 37. Chu Q, Jiang Y, Zhang W, Xu C, Du W, Tuguzbaeva G, Qin Y, Li A, Zhang L, Sun G, Cai Y, Feng Q, Li G, Li Y, Du Z, Bai Y, Yang B. Pyroptosis is involved in the pathogenesis of human hepatocellular carcinoma. Oncotarget 2016;7(51):84658-84665. 38. Sun Y, Yu J, Liu X, Zhang C, Cao J, Li G, Liu X, Chen Y, Huang H. Oncosis-like cell death is induced by berberine through ERK1/2-mediated impairment of mitochondrial aerobic respiration in gliomas. Biomed Pharmacother 2018;102:699-710. 39. Abdollahi A, Folkman J. Evading tumor evasion: current concepts and perspectives of antiangiogenic cancer therapy. Drug Resist Updat 2010;13(1-2):16-28. 40. Kuczynski EA, Vermeulen PB, Pezzella F, Kerbel RS, Reynolds AR. Vessel co-option in cancer. Nat Rev Clin Oncol 2019 [Epub ahead of print] 41. Chung AS, Lee J, Ferrara N. Targeting the tumour vasculature: insights from physiological angiogenesis. Nat Rev Cancer 2010;10(7):505-514. 42. Albini A, Tosetti F, Li VW, Noonan DM, Li WW. Cancer prevention by targeting angiogenesis. Nat Rev Clin Oncol 2012;9(9):498-509. 15

Jo

ur

na

lP

re

-p

ro of

43. Dell'Eva R, Pfeffer U, Vené R, Anfosso L, Forlani A, Albini A, Efferth T. Inhibition of angiogenesis in vivo and growth of Kaposi's sarcoma xenograft tumors by the anti-malarial artesunate. Biochem Pharmacol 2004;68(12):2359-2366. 44. Anfosso L, Efferth T, Albini A, Pfeffer U. Microarray expression profiles of angiogenesis-related genes predict tumor cell response to artemisinins. Pharmacogenomics J 2006;6(4):269-278. 45. Soomro S, Langenberg T, Mahringer A, Konkimalla VB, Horwedel C, Holenya P, Brand A, Cetin C, Fricker G, Dewerchin M, Carmeliet P, Conway EM, Jansen H, Efferth T. Design of novel artemisinin-like derivatives with cytotoxic and anti-angiogenic properties. J Cell Mol Med 2011;15(5):1122-1135. 46. Konkimalla VB, McCubrey JA, Efferth T. The role of downstream signaling pathways of the epidermal growth factor receptor for artesunate's activity in cancer cells. Curr Cancer Drug Targets 2009;9(1):72-80. 47. Wu CF, Seo EJ, Klauck SM, Efferth T. Cryptotanshinone deregulates unfolded protein response and eukaryotic initiation factor signaling in acute lymphoblastic leukemia cells. Phytomedicine 2016;23(2):174-180. 48. Kim S, You D, Jeong Y, Yu J, Kim SW, Nam SJ, Lee JE. Berberine down-regulates IL-8 expression through inhibition of the EGFR/MEK/ERK pathway in triple-negative breast cancer cells. Phytomedicine 2018;50:43-49. 49. Han B, Jiang P, Li Z, Yu Y, Huang T, Ye X, Li X. Coptisine-induced apoptosis in human colon cancer cells (HCT-116) is mediated by PI3K/Akt and mitochondrial-associated apoptotic pathway. Phytomedicine 2018;48:152-160. 50. Shi X, Zhu M, Kang Y, Yang T, Chen X, Zhang Y. Wnt/β-catenin signaling pathway is involved in regulating the migration by an effective natural compound brucine in LoVo cells. Phytomedicine 2018;46:85-92. 51. Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer 2003;3(6):453-458. 52. Seton-Rogers S. Metastasis: opposing forces in invasion. Nat Rev Cancer 2011;11(9):624-625. 53. Leber MF, Efferth T. Molecular principles of cancer invasion and metastasis (review). Int J Oncol 2009;34(4):881-895. 54. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019;20(2):69-84. 55. Rasheed SA, Efferth T, Asangani IA, Allgayer H. First evidence that the antimalarial drug artesunate inhibits invasion and in vivo metastasis in lung cancer by targeting essential extracellular proteases. Int J Cancer 2010;127(6):1475-1485. 56. Hamdoun S, Efferth T. Ginkgolic acids inhibit migration in breast cancer cells by inhibition of NEMO sumoylation and NF-κB activity. Oncotarget 2017;8(21):35103-35115. 57. Kim S, You D, Jeong Y, Yu J, Kim SW, Nam SJ, Lee JE. Berberine down-regulates IL-8 expression through inhibition of the EGFR/MEK/ERK pathway in triple-negative breast cancer cells. Phytomedicine 2018;50:43-49. 58. Allegra M, De Cicco P, Ercolano G, Attanzio A, Busà R, Cirino G, Tesoriere L, Livrea MA, Ianaro A. Indicaxanthin from Opuntia Ficus Indica (L. Mill) impairs melanoma cell proliferation, invasiveness, and tumor progression. Phytomedicine 2018;50:19-24. 59. Salehi B, Varoni EM, Sharifi-Rad M, Rajabi S, Zucca P, Iriti M, Sharifi-Rad J. Epithelialmesenchymal transition as a target for botanicals in cancer metastasis. Phytomedicine 2019;55:125-136. 60. William WN Jr, Heymach JV, Kim ES, Lippman SM. Molecular targets for cancer chemoprevention. Nat Rev Drug Discov 2009;8(3):213-225. 61. Ricciardiello L, Ahnen DJ, Lynch PM. Chemoprevention of hereditary colon cancers: time for new strategies. Nat Rev Gastroenterol Hepatol 2016;13(6):352-361. 62. Efferth T. Pharmacognosy and molecular pharmacology of small molecules of natural origin for cancer therapy and chemoprevention. Curr Drug Targets 2006;7(3):237-238. 16

Jo

ur

na

lP

re

-p

ro of

63. Abba ML, Patil N, Leupold JH, Saeed MEM, Efferth T, Allgayer H. Prevention of carcinogenesis and metastasis by artemisinin-type drugs. Cancer Lett 2018;429:11-18. 64. Efferth T. Editorial: Chemoprevention of cancer by natural products. Cancer Lett 2019;459:1314. 65. Wang C, Wang H, Zhang Y, Guo W, Long C, Wang J, Liu L, Sun X. Berberine inhibits the proliferation of human nasopharyngeal carcinoma cells via an Epstein-Barr virus nuclear antigen 1-dependent mechanism. Oncol Rep 2017;37(4):2109-2120. 66. Youns M, Hoheisel JD, Efferth T. Toxicogenomics for the prediction of toxicity related to herbs from traditional Chinese medicine. Planta Med 2010;76(17):2019-2025. 67. Efferth T, Kaina B. Toxicity of the antimalarial artemisinin and its dervatives. Crit Rev Toxicol 2010;40(5):405-421. 68. Efferth T, Kaina B. Toxicities by herbal medicines with emphasis to traditional Chinese medicine. Curr Drug Metab 2011;12(10):989-996. 69. Allard T, Wenner T, Greten HJ, Efferth T. Mechanisms of herb-induced nephrotoxicity. Curr Med Chem 2013;20(22):2812-2819. 70. Uhl M, Schwab S, Efferth T. Fatal liver and bone marrow toxicity by combination treatment of dichloroacetate and artesunate in a glioblastoma multiforme patient: Case report and review of the literature. Front Oncol 2016;6:204. 71. Efferth T, Schöttler U, Krishna S, Schmiedek P, Wenz F, Giordano FA. Hepatotoxicity by combination treatment of temozolomide, artesunate and Chinese herbs in a glioblastoma multiforme patient: case report review of the literature. Arch Toxicol 2017;91(4):1833-1846. 72. Fischer N, Seo EJ, Efferth T. Prevention from radiation damage by natural products. Phytomedicine 2018;47:192-200. 73. Efferth T, Koch E. Complex interactions between phytochemicals. The multi-target therapeutic concept of phytotherapy. Curr Drug Targets 2011;12(1):122-132. 74. Orlefors H, Sundin A, Fasth KJ, Oberg K, Långström B, Eriksson B, Bergström M. Demonstration of high monoaminoxidase-A levels in neuroendocrine gastroenteropancreatic tumors in vitro and in vivo-tumor visualization using positron emission tomography with 11C-harmine. Nucl Med Biol 2003;30(6):669-679. 75. Goller L, Bergstrom M, Nilsson S, Westerberg G, Langstrom B. Mao-a enzyme binding in bladder-cancer characterized with [C-11] harmine in frozen-section autoradiography. Oncol Rep 1995;2(5):717-721. 76. 73. Qian K, Wang G, Cao R, Liu T, Qian G, Guan X, Guo Z, Xiao Y, Wang X. Capsaicin suppresses cell proliferation, induces cell cycle arrest and ROS production in bladder cancer cells through FOXO3a-mediated pathways. Molecules 2016;21(10). pii: E1406. 77. 74. Lin S, Zhang J, Chen H, Chen K, Lai F, Luo J, Wang Z, Bu H, Zhang R, Li H, Tong H. Involvement of endoplasmic reticulum stress in capsaicin-induced apoptosis of human pancreatic cancer cells. Evid Based Complement Alternat Med. 2013;2013:629750. 78. 75. Zhang JH, Lai FJ, Chen H, Luo J, Zhang RY, Bu HQ, Wang ZH, Lin HH, Lin SZ. Involvement of the phosphoinositide 3-kinase/Akt pathway in apoptosis induced by capsaicin in the human pancreatic cancer cell line PANC-1. Oncol Lett 2013;5(1):43-48. 79. 76. Brown KC, Witte TR, Hardman WE, Luo H, Chen YC, Carpenter AB, Lau JK, Dasgupta P. Capsaicin displays anti-proliferative activity against human small cell lung cancer in cell culture and nude mice models via the E2F pathway. PLoS One 2010;5(4):e10243. 80. 77. Thoennissen NH, O'Kelly J, Lu D, Iwanski GB, La DT, Abbassi S, Leiter A, Karlan B, Mehta R, Koeffler HP. Capsaicin causes cell-cycle arrest and apoptosis in ER-positive and -negative breast cancer cells by modulating the EGFR/HER-2 pathway. Oncogene 2010;29(2):285-296. 81. 78. Bhutani M, Pathak AK, Nair AS, Kunnumakkara AB, Guha S, Sethi G, Aggarwal BB. Capsaicin is a novel blocker of constitutive and interleukin-6-inducible STAT3 activation. Clin Cancer Res 2007;13(10):3024-3032. Erratum in: Editor's Note: Capsaicin is a novel blocker of constitutive and interleukin-6-inducible STAT3 activation. [Clin Cancer Res. 2018] 17

Jo

ur

na

lP

re

-p

ro of

82. 79. Zhang H, Jiao Y, Shi C, Song X, Chang Y, Ren Y, Shi X. Berbamine suppresses cell viability and induces apoptosis in colorectal cancer via activating p53-dependent apoptotic signaling pathway. Cytotechnology 2018;70(1):321-329. 83. 80. Huang T, Xiao Y, Yi L, Li L, Wang M, Tian C, Ma H, He K, Wang Y, Han B, Ye X, Li X. Coptisine from Rhizoma Coptidis suppresses HCT-116 cells-related tumor growth in vitro and in vivo. Sci Rep 2017;7:38524. 84. 81. He X, Maimaiti M, Jiao Y, Meng X, Li H. Sinomenine induces G1-phase cell cycle arrest and apoptosis in malignant glioma cells via downregulation of sirtuin 1 and induction of p53 acetylation. Technol Cancer Res Treat 2018;17:1533034618770305. 85. 82. Li X, Wang K, Ren Y, Zhang L, Tang XJ, Zhang HM, Zhao CQ, Liu PJ, Zhang JM, He JJ. MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study. Cell Death Dis 2014;5:e1356. 86. 83. Lu XL, Zeng J, Chen YL, He PM, Wen MX, Ren MD, Hu YN, Lu GF, He SΧ. Sinomenine hydrochloride inhibits human hepatocellular carcinoma cell growth in vitro and in vivo: Involvement of cell cycle arrest and apoptosis induction. Int J Oncol 2013;42(1):229-38. 87. 84. Tian DD, Zhang RX, Wu N, Yuan W, Luo SH, Chen HQ, Liu Y, Wang Y, He BC, Deng ZL. Tetrandrine inhibits the proliferation of human osteosarcoma cells by upregulating the PTEN pathway. Oncol Rep 2017;37(5):2795-2802. 88. 85. Wang Q, Du H, Geng G, Zhou H, Xu M, Cao H, Zhang B, Song G, Hu T. Matrine inhibits proliferation and induces apoptosis via BID-mediated mitochondrial pathway in esophageal cancer cells. Mol Biol Rep 2014;41(5):3009-3020. 89. 86. Ku BM, Lee YK, Jeong JY, Ryu J, Choi J, Kim JS, Cho YW, Roh GS, Kim HJ, Cho GJ, Choi WS, Kang SS. Caffeine inhibits cell proliferation and regulates PKA/GSK3β pathways in U87MG human glioma cells. Mol Cells 2011;31(3):275-279. 90. 87. Zhao B, Shen C, Zheng Z, Wang X, Zhao W, Chen X, Peng F, Xue L, Shu M, Hou X, Wang K, Zhong C, Sun J, Wan J, Zhao S. Peiminine inhibits glioblastoma in vitro and in vivo through cell cycle arrest and autophagic flux blocking. Cell Physiol Biochem 2018;51(4):1566-1583. 91. 88. Yang H, Yin P, Shi Z, Ma Y, Zhao C, Zheng J, Chen T. Sinomenine, a COX-2 inhibitor, induces cell cycle arrest and inhibits growth of human colon carcinoma cells in vitro and in vivo. Oncol Lett 2016;11(1):411-418. 92. 89. Zhang C, Fan X, Xu X, Yang X, Wang X, Liang HP. Evodiamine induces caspase-dependent apoptosis and S phase arrest in human colon lovo cells. Anticancer Drugs 2010;21(8):766-776. 93. 90. Wang C, Wang H, Zhang Y, Guo W, Long C, Wang J, Liu L, Sun X. Berberine inhibits the proliferation of human nasopharyngeal carcinoma cells via an Epstein-Barr virus nuclear antigen 1-dependent mechanism. Oncol Rep 2017;37(4):2109-2120. 94. 91. Cai Y, Xia Q, Luo R, Huang P, Sun Y, Shi Y, Jiang W. Berberine inhibits the growth of human colorectal adenocarcinoma in vitro and in vivo. J Nat Med 2014;68(1):53-62. 95. 92. Paydar M, Kamalidehghan B, Wong YL, Wong WF, Looi CY, Mustafa MR. Evaluation of cytotoxic and chemotherapeutic properties of boldine in breast cancer using in vitro and in vivo models. Drug Des Devel Ther 2014;8:719-733. 96. 93. Yang ZR, Liu M, Peng XL, Lei XF, Zhang JX, Dong WG. Noscapine induces mitochondriamediated apoptosis in human colon cancer cells in vivo and in vitro. Biochem Biophys Res Commun 2012;421(3):627-633. 97. 94. Visnyei K, Onodera H, Damoiseaux R, Saigusa K, Petrosyan S, De Vries D, Ferrari D, Saxe J, Panosyan EH, Masterman-Smith M, Mottahedeh J, Bradley KA, Huang J, Sabatti C, Nakano I, Kornblum HI. A molecular screening approach to identify and characterize inhibitors of glioblastoma stem cells. Mol Cancer Ther 2011;10(10):1818-1828. 98. 95. Wu WS, Chien CC, Liu KH, Chen YC, Chiu WT. Evodiamine prevents glioma growth, induces glioblastoma cell apoptosis and cell cycle arrest through JNK activation. Am J Chin Med 2017;45(4):879-899. 18

Jo

ur

na

lP

re

-p

ro of

99. 96. Yang J, Cai X, Lu W, Hu C, Xu X, Yu Q, Cao P. Evodiamine inhibits STAT3 signaling by inducing phosphatase shatterproof 1 in hepatocellular carcinoma cells. Cancer Lett 2013;328(2):243251. 100. 97. Wang Y, Wang C, Jiang C, Zeng H, He X. Novel mechanism of harmaline on inducing G2/M cell cycle arrest and apoptosis by up-regulating Fas/FasL in SGC-7901 cells. Sci Rep 2015;5:18613. 101. 98. Dai F, Chen Y, Song Y, Huang L, Zhai D, Dong Y, Lai L, Zhang T, Li D, Pang X, Liu M, Yi Z. A natural small molecule harmine inhibits angiogenesis and suppresses tumour growth through activation of p53 in endothelial cells. PLoS One 2012;7(12):e52162. 102. 100. Bezerra DP, Castro FO, Alves AP, Pessoa C, Moraes MO, Silveira ER, Lima MA, Elmiro FJ, Costa-Lotufo LV. In vivo growth-inhibition of Sarcoma 180 by piplartine and piperine, two alkaloid amides from piper. Braz J Med Biol Res 2006;39(6):801-807. 103. 101. Puthdee N, Vaeteewoottacharn K, Seubwai W, Wonkchalee O, Kaewkong W, Juasook A, Pinlaor S, Pairojkul C, Wongkham C, Okada S, Boonmars T, Wongkham S. Establishment of an allo-transplantable hamster cholangiocarcinoma cell line and its application for in vivo screening of anti-cancer drugs. Korean J Parasitol 2013;51(6):711-717. 104. 102. Ho YT, Yang JS, Lu CC, Chiang JH, Li TC, Lin JJ, Lai KC, Liao CL, Lin JG, Chung JG. Berberine inhibits human tongue squamous carcinoma cancer tumor growth in a murine xenograft model. Phytomedicine 2009;16(9):887-890. 105. 103. Letasiová S, Jantová S, Múcková M, Theiszová M. Antiproliferative activity of berberine in vitro and in vivo. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2005;149(2):461-463. 106. 104. Landen JW, Hau V, Wang M, Davis T, Ciliax B, Wainer BH, Van Meir EG, Glass JD, Joshi HC, Archer DR. Noscapine crosses the blood-brain barrier and inhibits glioblastoma growth. Clin Cancer Res 2004;10(15):5187-5201. 107. 105. Landen JW, Lang R, McMahon SJ, Rusan NM, Yvon AM, Adams AW, Sorcinelli MD, Campbell R, Bonaccorsi P, Ansel JC, Archer DR, Wadsworth P, Armstrong CA, Joshi HC. Noscapine alters microtubule dynamics in living cells and inhibits the progression of melanoma. Cancer Res 2002;62(14):4109-4114. 108. 106. Wang Y, Zhang B, Liu W, Dai Y, Shi Y, Zeng Q, Wang F. Noninvasive bioluminescence imaging of the dynamics of sanguinarine induced apoptosis via activation of reactive oxygen species. Oncotarget 2016;7(16):22355-67. 109. 107. Aoki T, Shimada K, Sakamoto A, Sugimoto K, Morishita T, Kojima Y, Shimada S, Kato S, Iriyama C, Kuno S, Harada Y, Tomita A, Hayakawa F, Kiyoi H. Emetine elicits apoptosis of intractable B-cell lymphoma cells with MYC rearrangement through inhibition of glycolytic metabolism. Oncotarget 2017;8(8):13085-13098. 110. 108. Johnson RK, Jondorf WR. Some inhibitory effects of (-)-emetine on growth of Ehrlich ascites carcinoma. Biochem J 1974;140(1):87-94. 111. 109. Zhang Y, Deng X, Lei T, Yu C, Wang Y, Zhao G, Luo X, Tang K, Quan Z, Jiang D. Capsaicin inhibits proliferation and induces apoptosis in osteosarcoma cell lines via the mitogen-activated protein kinase pathway. Oncol Rep 2017;38(5):2685-2696. 112. 110. Xie L, Xiang GH, Tang T, Tang Y, Zhao LY, Liu D, Zhang YR, Tang JT, Zhou S, Wu DH. Capsaicin and dihydrocapsaicin induce apoptosis in human glioma cells via ROS and Ca2+-mediated mitochondrial pathway. Mol Med Rep 2016;14(5):4198-4208. 113. 111. Liu T, Wang G, Tao H, Yang Z, Wang Y, Meng Z, Cao R, Xiao Y, Wang X, Zhou J. Capsaicin mediates caspases activation and induces apoptosis through P38 and JNK MAPK pathways in human renal carcinoma. BMC Cancer 2016;16(1):790. 114. 112. Pramanik KC, Srivastava SK. Apoptosis signal-regulating kinase 1-thioredoxin complex dissociation by capsaicin causes pancreatic tumor growth suppression by inducing apoptosis. Antioxid Redox Signal 2012;17(10):1417-1432.

19

Jo

ur

na

lP

re

-p

ro of

115. 113. Lu HF, Chen YL, Yang JS, Yang YY, Liu JY, Hsu SC, Lai KC, Chung JG. Antitumor activity of capsaicin on human colon cancer cells in vitro and colo 205 tumor xenografts in vivo. J Agric Food Chem 2010;58(24):12999-13005. 116. 114. Zhang R, Humphreys I, Sahu RP, Shi Y, Srivastava SK. In vitro and in vivo induction of apoptosis by capsaicin in pancreatic cancer cells is mediated through ROS generation and mitochondrial death pathway. Apoptosis 2008;13(12):1465-1478. 117. 115. Sánchez AM, Sánchez MG, Malagarie-Cazenave S, Olea N, Díaz-Laviada I. Induction of apoptosis in prostate tumor PC-3 cells and inhibition of xenograft prostate tumor growth by the vanilloid capsaicin. Apoptosis 2006;11(1):89-99. 118. 116. Ito K, Nakazato T, Yamato K, Miyakawa Y, Yamada T, Hozumi N, Segawa K, Ikeda Y, Kizaki M. Induction of apoptosis in leukemic cells by homovanillic acid derivative, capsaicin, through oxidative stress: implication of phosphorylation of p53 at Ser-15 residue by reactive oxygen species. Cancer Res 2004;64(3):1071-1078. 119. 117. Yoo ES, Choo GS, Kim SH, Woo JS, Kim HJ, Park YS, Kim BS, Kim SK, Park BK, Cho SD, Nam JS, Choi CS, Che JH, Jung JY. Antitumor and apoptosis-inducing effects of piperine on human melanoma cells. Anticancer Res 2019;39(4):1883-1892. 120. 118. Zhao Y, Jing Z, Lv J, Zhang Z, Lin J, Cao X, Zhao Z, Liu P, Mao W. Berberine activates caspase-9/cytochrome c-mediated apoptosis to suppress triple-negative breast cancer cells in vitro and in vivo. Biomed Pharmacother. 2017 Nov;95:18-24. 121. 119. Tsang CM, Cheung YC, Lui VW, Yip YL, Zhang G, Lin VW, Cheung KC, Feng Y, Tsao SW. Berberine suppresses tumorigenicity and growth of nasopharyngeal carcinoma cells by inhibiting STAT3 activation induced by tumor associated fibroblasts. BMC Cancer 2013;13:619. 122. 120. Choi MS, Oh JH, Kim SM, Jung HY, Yoo HS, Lee YM, Moon DC, Han SB, Hong JT. Berberine inhibits p53-dependent cell growth through induction of apoptosis of prostate cancer cells. Int J Oncol 2009;34(5):1221-1230. 123. 121. Katiyar SK, Meeran SM, Katiyar N, Akhtar S. p53 Cooperates berberine-induced growth inhibition and apoptosis of non-small cell human lung cancer cells in vitro and tumor xenograft growth in vivo. Mol Carcinog 2009;48(1):24-37. 124. 122. Zhang H, Jiao Y, Shi C, Song X, Chang Y, Ren Y, Shi X. Berbamine suppresses cell proliferation and promotes apoptosis in ovarian cancer partially via the inhibition of Wnt/βcatenin signaling. Acta Biochim Biophys Sin (Shanghai). 2018 Jun 1;50(6):532-539. 125. 123. Zhao Y, Lv JJ, Chen J, Jin XB, Wang MW, Su ZH, Wang LY, Zhang HY. Berbamine inhibited the growth of prostate cancer cells in vivo and in vitro via triggering intrinsic pathway of apoptosis. Prostate Cancer Prostatic Dis 2016;19(4):358-366. 126. 124. Wang GY, Lv QH, Dong Q, Xu RZ, Dong QH. Berbamine induces Fas-mediated apoptosis in human hepatocellular carcinoma HepG2 cells and inhibits its tumor growth in nude mice. J Asian Nat Prod Res. 2009;11(3):219-28. 127. 125. Chai FN, Ma WY, Zhang J, Xu HS, Li YF, Zhou QD, Li XG, Ye XL. Coptisine from Rhizoma coptidis exerts an anti-cancer effect on hepatocellular carcinoma by up-regulating miR-122. Biomed Pharmacother 2018;103:1002-1011. 128. 126. Han B, Jiang P, Li Z, Yu Y, Huang T, Ye X, Li X. Coptisine-induced apoptosis in human colon cancer cells (HCT-116) is mediated by PI3K/Akt and mitochondrial-associated apoptotic pathway. Phytomedicine 2018;48:152-160. 129. 127. Xu G, Niu Z, Dong J, Zhao Y, Zhang Y, Li X. Noscapine inhibits human hepatocellular carcinoma growth through inducing apoptosis in vitro and in vivo. Neoplasma 2016;63(5):726733. 130. 128. Liu M, Luo XJ, Liao F, Lei XF, Dong WG. Noscapine induces mitochondria-mediated apoptosis in gastric cancer cells in vitro and in vivo. Cancer Chemother Pharmacol 2011;67(3):605-612. 131. 129. Jackson T, Chougule MB, Ichite N, Patlolla RR, Singh M. Antitumor activity of noscapine in human non-small cell lung cancer xenograft model. Cancer Chemother Pharmacol 2008;63(1):117-126. 20

Jo

ur

na

lP

re

-p

ro of

132. 130. Liu W, Yi DD, Guo JL, Xiang ZX, Deng LF, He L. Nuciferine, extracted from Nelumbo nucifera Gaertn, inhibits tumor-promoting effect of nicotine involving Wnt/β-catenin signaling in non-small cell lung cancer. J Ethnopharmacol 2015;165:83-93. 133. 131. Gong X, Chen Z, Han Q, Chen C, Jing L, Liu Y, Zhao L, Yao X, Sun X. Sanguinarine triggers intrinsic apoptosis to suppress colorectal cancer growth through disassociation between STRAP and MELK. BMC Cancer 2018;18(1):578. 134. 132. Sun Z, Zheng L, Liu X, Xing W, Liu X. Sinomenine inhibits the growth of melanoma by enhancement of autophagy via PI3K/AKT/mTOR inhibition. Drug Des Devel Ther 2018;12:2413-2421. 135. 133. Sun M, Lou W, Chun JY, Cho DS, Nadiminty N, Evans CP, Chen J, Yue J, Zhou Q, Gao AC. Sanguinarine suppresses prostate tumor growth and inhibits survivin expression. Genes Cancer 2010;1(3):283-292. 136. 134. Qin R, Shen H, Cao Y, Fang Y, Li H, Chen Q, Xu W. Tetrandrine induces mitochondria-mediated apoptosis in human gastric cancer BGC-823 cells. PLoS One 2013;8(10):e76486. 137. 135. Liu C, Gong K, Mao X, Li W. Tetrandrine induces apoptosis by activating reactive oxygen species and repressing Akt activity in human hepatocellular carcinoma. Int J Cancer 2011;129(6):1519-1531. 138. 136. Chen Y, Chen JC, Tseng SH. Tetrandrine suppresses tumor growth and angiogenesis of gliomas in rats. Int J Cancer 2009;124(10):2260-2269. 139. 137. Gu YY, Chen MH, May BH, Liao XZ, Liu JH, Tao LT, Man-Yuen Sze D, Zhang AL, Mo SL. Matrine induces apoptosis in multiple colorectal cancer cell lines in vitro and inhibits tumour growth with minimum side effects in vivo via Bcl-2 and caspase-3. Phytomedicine 2018;51:214-225. 140. 138. Xiao X, Ao M, Xu F, Li X, Hu J, Wang Y, Li D, Zhu X, Xin C, Shi W. Effect of matrine against breast cancer by downregulating the vascular endothelial growth factor via the Wnt/βcatenin pathway. Oncol Lett 2018;15(2):1691-1697. 141. 139. Wu J, Hu G, Dong Y, Ma R, Yu Z, Jiang S, Han Y, Yu K, Zhang S. Matrine induces Akt/mTOR signalling inhibition-mediated autophagy and apoptosis in acute myeloid leukaemia cells. J Cell Mol Med 2017;21(6):1171-1181. 142. 140. Zhang S, Zhang Y, Zhuang Y, Wang J, Ye J, Zhang S, Wu J, Yu K, Han Y. Matrine induces apoptosis in human acute myeloid leukemia cells via the mitochondrial pathway and Akt inactivation. PLoS One 2012;7(10):e46853. 143. 141. Liang CZ, Zhang JK, Shi Z, Liu B, Shen CQ, Tao HM. Matrine induces caspasedependent apoptosis in human osteosarcoma cells in vitro and in vivo through the upregulation of Bax and Fas/FasL and downregulation of Bcl-2. Cancer Chemother Pharmacol 2012;69(2):317-331. 144. 142. Liu T, Song Y, Chen H, Pan S, Sun X. Matrine inhibits proliferation and induces apoptosis of pancreatic cancer cells in vitro and in vivo. Biol Pharm Bull 2010;33(10):17401745. 145. 143. Ma L, Wen S, Zhan Y, He Y, Liu X, Jiang J. Anticancer effects of the Chinese medicine matrine on murine hepatocellular carcinoma cells. Planta Med 2008;74(3):245-251. 146. 144. Wu C, Huang W, Guo Y, Xia P, Sun X, Pan X, Hu W. Oxymatrine inhibits the proliferation of prostate cancer cells in vitro and in vivo. Mol Med Rep. 2015 Jun;11(6):41294134. 147. 145. Zhang Y, Sun S, Chen J, Ren P, Hu Y, Cao Z, Sun H, Ding Y. Oxymatrine induces mitochondria dependent apoptosis in human osteosarcoma MNNG/HOS cells through inhibition of PI3K/Akt pathway. Tumour Biol 2014;35(2):1619-1625. 148. 146. Wu XS, Yang T, Gu J, Li ML, Wu WG, Weng H, Ding Q, Mu JS, Bao RF, Shu YJ, Cao Y, Wang XA, Ding QC, Dong P, Xie SF, Liu YB. Effects of oxymatrine on the apoptosis and proliferation of gallbladder cancer cells. Anticancer Drugs 2014;25(9):1007-1015. 21

Jo

ur

na

lP

re

-p

ro of

149. 147. Ruijun W, Wenbin M, Yumin W, Ruijian Z, Puweizhong H, Yulin L. Inhibition of glioblastoma cell growth in vitro and in vivo by brucine, a component of Chinese medicine. Oncol Res 2014;22(5-6):275-281. 150. 148. Jin ZL, Yan W, Qu M, Ge CZ, Chen X, Zhang SF. Cinchonine activates endoplasmic reticulum stress-induced apoptosis in human liver cancer cells. Exp Ther Med 2018;15(6):50465050. 151. 149. Qi Y, Pradipta AR, Li M, Zhao X, Lu L, Fu X, Wei J, Hsung RP, Tanaka K, Zhou L. Cinchonine induces apoptosis of HeLa and A549 cells through targeting TRAF6. J Exp Clin Cancer Res 2017;36(1):35. 152. 150. Wu WS, Chien CC, Chen YC, Chiu WT. Protein kinase RNA-like endoplasmic reticulum kinase-mediated Bcl-2 protein phosphorylation contributes to evodiamine-induced apoptosis of human renal cell carcinoma cells. PLoS One 2016;11(8):e0160484. 153. 151. Tu YJ, Fan X, Yang X, Zhang C, Liang HP. Evodiamine activates autophagy as a cytoprotective response in murine Lewis lung carcinoma cells. Oncol Rep 2013;29(2):481-490. 154. 152. Ding Y, He J, Huang J, Yu T, Shi X, Zhang T, Yan G, Chen S, Peng C. Harmine induces anticancer activity in breast cancer cells via targeting TAZ. Int J Oncol 2019;54(6):1995-2004. 155. 153. Hai-Rong C, Xiang H, Xiao-Rong Z. Harmine suppresses bladder tumor growth by suppressing vascular endothelial growth factor receptor 2-mediated angiogenesis. Biosci Rep 2019;39(5). pii: BSR20190155. 156. 154. Yang ZH, Wang XH, Wang HP, Hu LQ, Zheng XM, Li SW. Capsaicin mediates cell death in bladder cancer T24 cells through reactive oxygen species production and mitochondrial depolarization. Urology 2010;75(3):735-741. 157. 155. Wang J, Qi Q, Feng Z, Zhang X, Huang B, Chen A, Prestegarden L, Li X, Wang J. Berberine induces autophagy in glioblastoma by targeting the AMPK/mTOR/ULK1-pathway. Oncotarget 2016;7(41):66944-66958. 158. 156. Wu G, Liu T, Li H, Li Y, Li D, Li W. c-MYC and reactive oxygen species play roles in tetrandrine-induced leukemia differentiation. Cell Death Dis 2018;9(5):473. 159. 157. Liu T, Zhang Z, Yu C, Zeng C, Xu X, Wu G, Huang Z, Li W. Tetrandrine antagonizes acute megakaryoblastic leukaemia growth by forcing autophagy-mediated differentiation. Br J Pharmacol 2017;174(23):4308-4328. 160. 158. Liu T, Men Q, Wu G, Yu C, Huang Z, Liu X, Li W. Tetrandrine induces autophagy and differentiation by activating ROS and Notch1 signaling in leukemia cells. Oncotarget 2015(10):7992-8006. 161. 159. Gong K, Chen C, Zhan Y, Chen Y, Huang Z, Li W. Autophagy-related gene 7 (ATG7) and reactive oxygen species/extracellular signal-regulated kinase regulate tetrandrineinduced autophagy in human hepatocellular carcinoma. J Biol Chem 2012;287(42):3557635588. 162. 160. Cho YR, Lee JH, Kim JH, Lee SY, Yoo S, Jung MK, Kim SJ, Yoo HJ, Pack CG, Rho JK, Son J. Matrine suppresses KRAS-driven pancreatic cancer growth by inhibiting autophagymediated energy metabolism. Mol Oncol 2018;12(7):1203-1215. 163. 161. Zhou H, Xu M, Gao Y, Deng Z, Cao H, Zhang W, Wang Q, Zhang B, Song G, Zhan Y, Hu T. Matrine induces caspase-independent program cell death in hepatocellular carcinoma through bid-mediated nuclear translocation of apoptosis inducing factor. Mol Cancer 2014;13:59. 164. 162. Jiang Y, Jiao Y, Wang Z, Li T, Liu Y, Li Y, Zhao X, Wang D. Sinomenine hydrochloride inhibits human glioblastoma cell growth through reactive oxygen species generation and autophagy-lysosome pathway activation: An in vitro and in vivo study. Int J Mol Sci 2017;18(9). pii: E1945. 165. 163. Hamsa TP, Kuttan G. Antiangiogenic activity of berberine is mediated through the downregulation of hypoxia-inducible factor-1, VEGF, and proinflammatory mediators. Drug Chem Toxicol 2012;35(1):57-70. 22

Jo

ur

na

lP

re

-p

ro of

166. 164. Pica F, Balestrieri E, Serafino A, Sorrentino R, Gaziano R, Moroni G, Moroni N, Palmieri G, Mattei M, Garaci E, Sinibaldi-Vallebona P. Antitumor effects of the benzophenanthridine alkaloid sanguinarine in a rat syngeneic model of colorectal cancer. Anticancer Drugs 2012 Jan;23(1):32-42. 167. 165. De Stefano I, Raspaglio G, Zannoni GF, Travaglia D, Prisco MG, Mosca M, Ferlini C, Scambia G, Gallo D. Antiproliferative and antiangiogenic effects of the benzophenanthridine alkaloid sanguinarine in melanoma. Biochem Pharmacol 2009;78(11):1374-1381. 168. 166. Xiao W, Jiang Y, Men Q, Yuan L, Huang Z, Liu T, Li W, Liu X. Tetrandrine induces G1/S cell cycle arrest through the ROS/Akt pathway in EOMA cells and inhibits angiogenesis in vivo. Int J Oncol 2015;46(1):360-8. 169. 167. Chen H, Zhang J, Luo J, Lai F, Wang Z, Tong H, Lu D, Bu H, Zhang R, Lin S. Antiangiogenic effects of oxymatrine on pancreatic cancer by inhibition of the NF-κB-mediated VEGF signaling pathway. Oncol Rep 2013;30(2):589-595. 170. 168. Saraswati S, Agrawal SS. Brucine, an indole alkaloid from Strychnos nux-vomica attenuates VEGF-induced angiogenesis via inhibiting VEGFR2 signaling pathway in vitro and in vivo. Cancer Lett 2013;332(1):83-93. 171. 169. Agrawal SS, Saraswati S, Mathur R, Pandey M. Cytotoxic and antitumor effects of brucine on Ehrlich ascites tumor and human cancer cell line. Life Sci 2011;89(5-6):147-158. 172. 170. Shi L, Yang F, Luo F, Liu Y, Zhang F, Zou M, Liu Q. Evodiamine exerts anti-tumor effects against hepatocellular carcinoma through inhibiting β-catenin-mediated angiogenesis. Tumour Biol 2016;37(9):12791-12803. 173. 171. Hamsa TP, Kuttan G. Harmine inhibits tumour specific neo-vessel formation by regulating VEGF, MMP, TIMP and pro-inflammatory mediators both in vivo and in vitro. Eur J Pharmacol 2010;649(1-3):64-73. 174. 172. Skopinska-Rózewska E, Janik P, Przybyszewska M, Sommer E, Bialas-Chromiec B. Inhibitory effect of theobromine on induction of angiogenesis and VEGF mRNA expression in v-raf transfectants of human urothelial cells HCV-29. Int J Mol Med 1998;2(6):649-652. 175. 173. Barcz E, Sommer E, Sokolnicka I, Gawrychowski K, Roszkowska-Purska K, Janik P, Skopinska-Rózewska E. The influence of theobromine on angiogenic activity and proangiogenic cytokines production of human ovarian cancer cells. Oncol Rep 1998;5(2):517-520. 176. 174. Menon LG, Ingle AD, Gude RP. Tumor regression of B16F10 melanoma in vivo by prevention of neovascularization: study on theophylline. Cancer Biother Radiopharm 2002;17(2):213-217. 177. 175. Mori A, Lehmann S, O'Kelly J, Kumagai T, Desmond JC, Pervan M, McBride WH, Kizaki M, Koeffler HP. Capsaicin, a component of red peppers, inhibits the growth of androgenindependent, p53 mutant prostate cancer cells. Cancer Res 2006;66(6):3222-3229. 178. 176. Ruan H, Zhan YY, Hou J, Xu B, Chen B, Tian Y, Wu D, Zhao Y, Zhang Y, Chen X, Mi P, Zhang L, Zhang S, Wang X, Cao H, Zhang W, Wang H, Li H, Su Y, Zhang XK, Hu T. Berberine binds RXRα to suppress β-catenin signaling in colon cancer cells. Oncogene 2017;36(50):69066918. 179. 177. Goto H, Kariya R, Shimamoto M, Kudo E, Taura M, Katano H, Okada S. Antitumor effect of berberine against primary effusion lymphoma via inhibition of NF-κB pathway. Cancer Sci 2012;103(4):775-781. 180. 178. Allegra M, De Cicco P, Ercolano G, Attanzio A, Busà R, Cirino G, Tesoriere L, Livrea MA, Ianaro A. Indicaxanthin from Opuntia ficus indica (L. Mill) impairs melanoma cell proliferation, invasiveness, and tumor progression. Phytomedicine 2018;50:19-24. 181. 179. Ren H, Zhao J, Fan D, Wang Z, Zhao T, Li Y, Zhao Y, Adelson D, Hao H. Alkaloids from nux vomica suppresses colon cancer cell growth through Wnt/β-catenin signaling pathway. Phytother Res 2019;33(5):1570-1578. 182. 180. Guo XX, Li XP, Zhou P, Li DY, Lyu XT, Chen Y, Lyu YW, Tian K, Yuan DZ, Ran JH, Chen DL, Jiang R, Li J. Evodiamine induces apoptosis in SMMC-7721 and HepG2 cells by suppressing NOD1 signal pathway. Int J Mol Sci 2018;19(11). pii: E3419. 23

Jo

ur

na

lP

re

-p

ro of

183. 181. Yochum ZA, Cades J, Mazzacurati L, Neumann NM, Khetarpal SK, Chatterjee S, Wang H, Attar MA, Huang EH, Chatley SN, Nugent K, Somasundaram A, Engh JA, Ewald AJ, Cho YJ, Rudin CM, Tran PT, Burns TF. A first-in-class TWIST1 inhibitor with activity in oncogenedriven lung cancer. Mol Cancer Res 2017;15(12):1764-1776. 184. 182. Yi T, Zhuang L, Song G, Zhang B, Li G, Hu T. Akt signaling is associated with the berberine-induced apoptosis of human gastric cancer cells. Nutr Cancer 2015;67(3):523-531. 185. 183. Qi Q, Li R, Li HY, Cao YB, Bai M, Fan XJ, Wang SY, Zhang B, Li S. Identification of the anti-tumor activity and mechanisms of nuciferine through a network pharmacology approach. Acta Pharmacol Sin 2016;37(7):963-972. 186. 184. Qi Y, Zhao X, Chen J, Pradipta AR, Wei J, Ruan H, Zhou L, Hsung RP, Tanaka K. In vitro and in vivo cancer cell apoptosis triggered by competitive binding of Cinchona alkaloids to the RING domain of TRAF6. Biosci Biotechnol Biochem 2019;83(6):1011-1026. 187. 185. Fan XX, Leung EL, Xie Y, Liu ZQ, Zheng YF, Yao XJ, Lu LL, Wu JL, He JX, Yuan ZW, Fu J, Wei CL, Huang J, Xiao DK, Luo LX, Jiang ZB, Zhou YL, Kam RK, Liu L. Suppression of lipogenesis via reactive oxygen species-AMPK signaling for treating malignant and proliferative diseases. Antioxid Redox Signal 2018;28(5):339-357. Erratum in: Antioxid Redox Signal 2019;30(4):710. 188. 186. Liu Q, Xu X, Zhao M, Wei Z, Li X, Zhang X, Liu Z, Gong Y, Shao C. Berberine induces senescence of human glioblastoma cells by downregulating the EGFR-MEK-ERK signaling pathway. Mol Cancer Ther 2015;14(2):355-363. 189. 187. Wang J, Peng Y, Liu Y, Yang J, Ding N, Tan W. Berberine, a natural compound, suppresses Hedgehog signaling pathway activity and cancer growth. BMC Cancer 2015;15:595. 190. 188. Wang N, Zhu M, Wang X, Tan HY, Tsao SW, Feng Y. Berberine induced tumor suppressor p53 up-regulation gets involved in the regulatory network of MIR-23a in hepatocellular carcinoma. Biochim Biophys Acta 2014;1839(9):849-857. 191. 189. Chelmicka-Schorr E, Arnason BG, Holshouser SJ. C-6 glioma growth in rats: suppression with a beta-adrenergic agonist and a phosphodiesterase inhibitor. Ann Neurol 1980;8(4):447-449. 192. 190. Chelmicka-Schorr E, Sportiello MG, Atweh SF, Arnason BG. Response of C-6 glioma to isoproterenol and papaverine in vivo depends on beta-adrenergic receptor density. Ann Neurol 1984;15(1):96-99. 193. Zhang B, Wang X, Deng J, Zheng H, Liu W, Chen S, Tian J, Wang F. p53-dependent upregulation of miR-16-2 by sanguinarine induces cell cycle arrest and apoptosis in hepatocellular carcinoma. Cancer Lett 2019;459:50-58. 194. 192. Jung YY, Shanmugam MK, Narula AS, Kim C, Lee JH, Namjoshi OA, Blough BE, Sethi G, Ahn KS. Oxymatrine attenuates tumor growth and deactivates STAT5 signaling in a lung cancer xenograft model. Cancers (Basel) 2019;11(1). pii: E49. 195. 193. Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Puttamallesh VN, Jain AP, Pinto SM, Balaji SA, Chavan S, Sahasrabuddhe NA, Mathur PP, Kumar MM, Prasad TS, Santosh V, Sukumar G, Califano JA, Rangarajan A, Sidransky D, Pandey A, Gowda H, Chatterjee A. A dual specificity kinase, DYRK1A, as a potential therapeutic target for head and neck squamous cell carcinoma. Sci Rep 2016;6:36132. Erratum in: Sci Rep 2017;7:46864. 196. 194. Kreider JW, Wade DR, Rosenthal M, Densley T. Maturation and differentiation of B16 melanoma cells induced by theophylline treatment. J Natl Cancer Inst 1975;54(6):14571467. 197. 195. Zhi D, Zhou K, Yu D, Fan X, Zhang J, Li X, Dong M. hERG1 is involved in the pathophysiological process and inhibited by berberine in SKOV3 cells. Oncol Lett 2019;17(6):5653-5661. 198. 196. Guo S, Chen Y, Pang C, Wang X, Shi S, Zhang H, An H, Zhan Y. Matrine is a novel inhibitor of the TMEM16A chloride channel with antilung adenocarcinoma effects. J Cell Physiol 2019;234(6):8698-8708. 199. 197. Li W, Yu X, Tan S, Liu W, Zhou L, Liu H. Oxymatrine inhibits non-small cell lung cancer via suppression of EGFR signaling pathway. Cancer Med 2018;7(1):208-218. 24

Jo

ur

na

lP

re

-p

ro of

200. 198. Li HL, Wu H, Zhang BB, Shi HL, Wu XJ. MAPK pathways are involved in the inhibitory effect of berberine hydrochloride on gastric cancer MGC 803 cell proliferation and IL-8 secretion in vitro and in vivo. Mol Med Rep 2016;14(2):1430-1438. 201. 199. Mandal A, Poddar MK. Long-term caffeine consumption reverses tumor-induced suppression of the innate immune response in adult mice. Planta Med 2008;74(15):1779-1784. 202. 200. Chuang TY, Min J, Wu HL, McCrary C, Layman LC, Diamond MP, Azziz R, Al-Hendy A, Chen YH. Berberine inhibits uterine leiomyoma cell proliferation via downregulation of cyclooxygenase 2 and pituitary tumor-transforming gene 1. Reprod Sci 2017;24(7):1005-1013. 203. 201. Li J, Li O, Kan M, Zhang M, Shao D, Pan Y, Zheng H, Zhang X, Chen L, Liu S. Berberine induces apoptosis by suppressing the arachidonic acid metabolic pathway in hepatocellular carcinoma. Mol Med Rep 2015;12(3):4572-4577. 204. 202. Liu NC, Hsieh PF, Hsieh MK, Zeng ZM, Cheng HL, Liao JW, Chueh PJ. Capsaicinmediated tNOX (ENOX2) up-regulation enhances cell proliferation and migration in vitro and in vivo. J Agric Food Chem 2012;60(10):2758-2765. 205. 203. Lai LH, Fu QH, Liu Y, Jiang K, Guo QM, Chen QY, Yan B, Wang QQ, Shen JG. Piperine suppresses tumor growth and metastasis in vitro and in vivo in a 4T1 murine breast cancer model. Acta Pharmacol Sin 2012;33(4):523-530. 206. 204. Hu Q, Li L, Zou X, Xu L, Yi P. Berberine attenuated proliferation, invasion and migration by targeting the AMPK/HNF4α/WNT5A pathway in gastric carcinoma. Front Pharmacol 2018;9:1150. 207. 205. Wang Y, Zhang S. Berberine suppresses growth and metastasis of endometrial cancer cells via miR-101/COX-2. Biomed Pharmacother 2018;103:1287-1293. 208. 206. Chu SC, Yu CC, Hsu LS, Chen KS, Su MY, Chen PN. Berberine reverses epithelial-tomesenchymal transition and inhibits metastasis and tumor-induced angiogenesis in human cervical cancer cells. Mol Pharmacol 2014;86(6):609-23. Erratum in: Mol Pharmacol 2017;91(2):158. 209. 207. Chu Q, Jiang Y, Zhang W, Xu C, Du W, Tuguzbaeva G, Qin Y, Li A, Zhang L, Sun G, Cai Y, Feng Q, Li G, Li Y, Du Z, Bai Y, Yang B. Pyroptosis is involved in the pathogenesis of human hepatocellular carcinoma. Oncotarget 2016;7(51):84658-84665. 210. 208. Su K, Hu P, Wang X, Kuang C, Xiang Q, Yang F, Xiang J, Zhu S, Wei L, Zhang J. Tumor suppressor berberine binds VASP to inhibit cell migration in basal-like breast cancer. Oncotarget 2016;7(29):45849-45862. 211. 209. Liu X, Ji Q, Ye N, Sui H, Zhou L, Zhu H, Fan Z, Cai J, Li Q. Berberine inhibits invasion and metastasis of colorectal cancer cells via COX-2/PGE2 mediated JAK2/STAT3 signaling pathway. PLoS One 2015;10(5):e0123478. 212. 210. Tsang CM, Cheung KC, Cheung YC, Man K, Lui VW, Tsao SW, Feng Y. Berberine suppresses Id-1 expression and inhibits the growth and development of lung metastases in hepatocellular carcinoma. Biochim Biophys Acta 2015;1852(3):541-551. 213. 211. Qi HW, Xin LY, Xu X, Ji XX, Fan LH. Epithelial-to-mesenchymal transition markers to predict response of berberine in suppressing lung cancer invasion and metastasis. J Transl Med 2014;12:22. 214. 212. Kim HS, Kim MJ, Kim EJ, Yang Y, Lee MS, Lim JS. Berberine-induced AMPK activation inhibits the metastatic potential of melanoma cells via reduction of ERK activity and COX-2 protein expression. Biochem Pharmacol 2012;83(3):385-394. 215. 213. Hamsa TP, Kuttan G. Berberine inhibits pulmonary metastasis through downregulation of MMP in metastatic B16F-10 melanoma cells. Phytother Res 2012;26(4):568-578. 216. 214. Tang F, Wang D, Duan C, Huang D, Wu Y, Chen Y, Wang W, Xie C, Meng J, Wang L, Wu B, Liu S, Tian D, Zhu F, He Z, Deng F, Cao Y. Berberine inhibits metastasis of nasopharyngeal carcinoma 5-8F cells by targeting Rho kinase-mediated Ezrin phosphorylation at threonine 567. J Biol Chem 2009;284(40):27456-27466.

25

Jo

ur

na

lP

re

-p

ro of

217. 215. Li Z, Chen Y, An T, Liu P, Zhu J, Yang H, Zhang W, Dong T, Jiang J, Zhang Y, Jiang M, Yang X. Nuciferine inhibits the progression of glioblastoma by suppressing the SOX2AKT/STAT3-Slug signaling pathway. J Exp Clin Cancer Res 2019;38(1):139. 218. 216. Barken I, Geller J, Rogosnitzky M. Prophylactic noscapine therapy inhibits human prostate cancer progression and metastasis in a mouse model. Anticancer Res 2010;30(2):399401. 219. 217. Wei G, Xu Y, Peng T, Yan J, Wang Z, Sun Z. Sanguinarine exhibits antitumor activity via up-regulation of Fas-associated factor 1 in non-small cell lung cancer. J Biochem Mol Toxicol 2017;31(8). doi: 10.1002/jbt.21914 220. 218. Song L, Liu D, Zhao Y, He J, Kang H, Dai Z, Wang X, Zhang S, Zan Y, Xue X. Sinomenine reduces growth and metastasis of breast cancer cells and improves the survival of tumor-bearing mice through suppressing the SHh pathway. Biomed Pharmacother 2018;98:687-693. 221. 219. Xie T, Ren HY, Lin HQ, Mao JP, Zhu T, Wang SD, Ye ZM. Sinomenine prevents metastasis of human osteosarcoma cells via S phase arrest and suppression of tumor-related neovascularization and osteolysis through the CXCR4-STAT3 pathway. Int J Oncol 2016;48(5):2098-2112. 222. 220. Zhang H, Xie B, Zhang Z, Sheng X, Zhang S. Tetrandrine suppresses cervical cancer growth by inducing apoptosis in vitro and in vivo. Drug Des Devel Ther 2018;13:119-127. 223. 221. Zhang Z, Liu T, Yu M, Li K, Li W. The plant alkaloid tetrandrine inhibits metastasis via autophagy-dependent Wnt/β-catenin and metastatic tumor antigen 1 signaling in human liver cancer cells. J Exp Clin Cancer Res 2018;37(1):7. 224. 222. Gao JL, Ji X, He TC, Zhang Q, He K, Zhao Y, Chen SH, Lv GY. Tetrandrine suppresses cancer angiogenesis and metastasis in 4T1 tumor bearing mice. Evid Based Complement Alternat Med 2013;2013:265061. 225. 223. Shen J, Wang B, Zhang T, Zhu N, Wang Z, Jin J, He Y, Hu M. Suppression of nonsmall cell lung cancer growth and metastasis by a novel small molecular activator of RECK. Cell Physiol Biochem 2018;45(5):1807-1817. 226. 224. Chang J, Hu S, Wang W, Li Y, Zhi W, Lu S, Shi Q, Wang Y, Yang Y. Matrine inhibits prostate cancer via activation of the unfolded protein response/endoplasmic reticulum stress signaling and reversal of epithelial to mesenchymal transition. Mol Med Rep 2018;18(1):945957. 227. 225. Wu X, Zhou J, Cai D, Li M. Matrine inhibits the metastatic properties of human cervical cancer cells via downregulating the p38 signaling pathway. Oncol Rep 2017;38(2):1312-1320. 228. 226. Huang H, Du T, Xu G, Lai Y, Fan X, Chen X, Li W, Yue F, Li Q, Liu L, Li K. Matrine suppresses invasion of castration-resistant prostate cancer cells by downregulating MMP-2/9 via NF-κB signaling pathway. Int J Oncol 2017;50(2):640-648. 229. 227. Ren H, Zhang S, Ma H, Wang Y, Liu D, Wang X, Wang Z. Matrine reduces the proliferation and invasion of colorectal cancer cells via reducing the activity of p38 signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2014;46(12):1049-1055. 230. 228. Li Y, Zhang ZN, Zhao HM, Tong ZC, Yang J, Wang H, Liang XJ. Matrine inhibits the invasive properties of human osteosarcoma cells by downregulating the ERK-NF-κB pathway. Anticancer Drugs 2014;25(9):1035-1043. 231. 229. Zhou W, Wu Y, Pan M, Liu D, Liu B. Proliferation and migration of lung cancer could be inhibited by oxymatrine through the regulation for miR-520/VEGF. Am J Chin Med 2019;47(4):865-878. 232. 230. Shi X, Zhu M, Kang Y, Yang T, Chen X, Zhang Y. Wnt/β-catenin signaling pathway is involved in regulating the migration by an effective natural compound brucine in LoVo cells. Phytomedicine 2018;46:85-92. 233. 231. Shu G, Mi X, Cai J, Zhang X, Yin W, Yang X, Li Y, Chen L, Deng X. Brucine, an alkaloid from seeds of Strychnos nux-vomica Linn., represses hepatocellular carcinoma cell migration 26

Jo

ur

na

lP

re

-p

ro of

and metastasis: the role of hypoxia inducible factor 1 pathway. Toxicol Lett 2013;222(2):91101. 234. 232. Zhu B, Zhao L, Liu Y, Jin Y, Feng J, Zhao F, Sun J, Geng R, Wei Y. Induction of phosphatase shatterproof 2 by evodiamine suppresses the proliferation and invasion of human cholangiocarcinoma. Int J Biochem Cell Biol 2019;108:98-110. 235. 233. Zhou P, Li XP, Jiang R, Chen Y, Lv XT, Guo XX, Tian K, Yuan DZ, Lv YW, Ran JH, Li J, Chen DL. Evodiamine inhibits migration and invasion by Sirt1-mediated post-translational modulations in colorectal cancer. Anticancer Drugs 2019;30(6):611-617. 236. 234. Du J, Wang XF, Zhou QM, Zhang TL, Lu YY, Zhang H, Su SB. Evodiamine induces apoptosis and inhibits metastasis in MDA-MB-231 human breast cancer cells in vitro and in vivo. Oncol Rep 2013;30(2):685-694. 237. 235. Zhang H, Sun K, Ding J, Xu H, Zhu L, Zhang K, Li X, Sun W. Harmine induces apoptosis and inhibits tumor cell proliferation, migration and invasion through downregulation of cyclooxygenase-2 expression in gastric cancer. Phytomedicine 2014;21(3):348355. 238. 236. Hamsa T, Kuttan G. Studies on anti-metastatic and anti-invasive effects of harmine using highly metastatic murine B16F-10 melanoma cells. J Environ Pathol Toxicol Oncol 2011;30(2):123-137. 239. 237. Cheng YC, Ding YM, Hueng DY, Chen JY, Chen Y. Caffeine suppresses the progression of human glioblastoma via cathepsin B and MAPK signaling pathway. J Nutr Biochem 2016;33:63-72. 240. 238. Dong S, Kong J, Kong J, Shen Q, Kong F, Sun W, Zheng L. Low concentration of caffeine inhibits the progression of the hepatocellular carcinoma via Akt signaling pathway. Anticancer Agents Med Chem 2015;15(4):484-92. 241. 239. Kang SS, Han KS, Ku BM, Lee YK, Hong J, Shin HY, Almonte AG, Woo DH, Brat DJ, Hwang EM, Yoo SH, Chung CK, Park SH, Paek SH, Roh EJ, Lee SJ, Park JY, Traynelis SF, Lee CJ. Caffeine-mediated inhibition of calcium release channel inositol 1,4,5-trisphosphate receptor subtype 3 blocks glioblastoma invasion and extends survival. Cancer Res 2010;70(3):11731183. 242. 240. Lentini A, Vidal-Vanaclocha F, Facchiano F, Caraglia M, Abbruzzese A, Beninati S. Theophylline administration markedly reduces hepatic and pulmonary implantation of B16F10 melanoma cells in mice. Melanoma Res 2000;10(5):435-443. 243. 241. Gitelman I, Abramow-Newerly W, Roder JC. NK-mediated reduction of malignancy in human melanoma cells treated with theophylline. Clin Exp Metastasis 1987;5(4):329-339. 244. 242. Anandakumar P, Kamaraj S, Jagan S, Ramakrishnan G, Asokkumar S, Naveenkumar C, Raghunandhakumar S, Devaki T. Capsaicin inhibits benzo(a)pyrene-induced lung carcinogenesis in an in vivo mouse model. Inflamm Res 2012;61(11):1169-1175. 245. 243. Teel RW, Huynh HT. Lack of the inhibitory effect of intragastrically administered capsaicin on NNK-induced lung tumor formation in the A.J mouse. In Vivo 1999;13(3):231-234. 246. 244. Jang JJ, Kim SH, Yun TK. Inhibitory effect of capsaicin on mouse lung tumor development. In Vivo 1989;3(1):49-53. 247. 245. Gunasekaran V, Elangovan K, Niranjali Devaraj S. Targeting hepatocellular carcinoma with piperine by radical-mediated mitochondrial pathway of apoptosis: An in vitro and in vivo study. Food Chem Toxicol 2017;105:106-118. 248. 246. Selvendiran K, Prince Vijeya Singh J, Sakthisekaran D. In vivo effect of piperine on serum and tissue glycoprotein levels in benzo(a)pyrene induced lung carcinogenesis in Swiss albino mice. Pulm Pharmacol Ther 2006;19(2):107-111. 249. 247. Wu K, Yang Q, Mu Y, Zhou L, Liu Y, Zhou Q, He B. Berberine inhibits the proliferation of colon cancer cells by inactivating Wnt/β-catenin signaling. Int J Oncol 2012;41(1):292-298. 250. 248. Zhao X, Zhang JJ, Wang X, Bu XY, Lou YQ, Zhang GL. Effect of berberine on hepatocyte proliferation, inducible nitric oxide synthase expression, cytochrome P450 2E1 and 27

Jo

ur

na

lP

re

-p

ro of

1A2 activities in diethylnitrosamine- and phenobarbital-treated rats. Biomed Pharmacother 2008;62(9):567-572. 251. 249. Su T, Yang X, Deng JH, Huang QJ, Huang SC, Zhang YM, Zheng HM, Wang Y, Lu LL, Liu ZQ. Evodiamine, a novel NOTCH3 methylation stimulator, significantly suppresses lung carcinogenesis in vitro and in vivo. Front Pharmacol 2018;9:434. 252. 250. Kim SH, Lee CS. The effect of caffeine on diethylnitrosamine-initiated hepatic altered foci in a mid-term induction system. In Vivo 1992;6(2):223-226. 253. 251. Ahsan H, Reagan-Shaw S, Eggert DM, Tan TC, Afaq F, Mukhtar H, Ahmad N. Protective effect of sanguinarine on ultraviolet B-mediated damages in SKH-1 hairless mouse skin: implications for prevention of skin cancer. Photochem Photobiol 2007;83(4):986-993. 254. 252. Lu YP, Lou YR, Peng QY, Nghiem P, Conney AH. Caffeine decreases phospho-Chk1 (Ser317) and increases mitotic cells with cyclin B1 and caspase 3 in tumors from UVB-treated mice. Cancer Prev Res (Phila) 2011;4(7):1118-1125. 255. 253. Lu YP, Lou YR, Peng QY, Xie JG, Nghiem P, Conney AH. Effect of caffeine on the ATR/Chk1 pathway in the epidermis of UVB-irradiated mice. Cancer Res 2008;68(7):25232529. 256. 254. Zajdela F, Latarjet R. Inhibition of skin carcinogenesis in vivo by caffeine and other agents. Natl Cancer Inst Monogr 1978;(50):133-140. 257. 255. Wang H, Guan L, Li J, Lai M, Wen X. The effects of berberine on the gut microbiota in Apc min/+ mice fed with a high fat diet. Molecules 2018;23(9). pii: E2298. 258. 256. Li D, Zhang Y, Liu K, Zhao Y, Xu B, Xu L, Tan L, Tian Y, Li C, Zhang W, Cao H, Zhan YY, Hu T. Berberine inhibits colitis-associated tumorigenesis via suppressing inflammatory responses and the consequent EGFR signaling-involved tumor cell growth. Lab Invest 2017;97(11):1343-1353. 259. 257. Wang L, Cao H, Lu N, Liu L, Wang B, Hu T, Israel DA, Peek RM Jr, Polk DB, Yan F. Berberine inhibits proliferation and down-regulates epidermal growth factor receptor through activation of Cbl in colon tumor cells. PLoS One 2013;8(2):e56666. 260. 258. James MA, Fu H, Liu Y, Chen DR, You M. Dietary administration of berberine or Phellodendron amurense extract inhibits cell cycle progression and lung tumorigenesis. Mol Carcinog 2011;50(1):1-7. 261. 259. Ma WK, Li H, Dong CL, He X, Guo CR, Zhang CF, Yu CH, Wang CZ, Yuan CS. Palmatine from Mahonia bealei attenuates gut tumorigenesis in ApcMin/+ mice via inhibition of inflammatory cytokines. Mol Med Rep 2016;14(1):491-498. 262. 261. Wu TY, Saw CL, Khor TO, Pung D, Boyanapalli SS, Kong AN. In vivo pharmacodynamics of indole-3-carbinol in the inhibition of prostate cancer in transgenic adenocarcinoma of mouse prostate (TRAMP) mice: involvement of Nrf2 and cell cycle/apoptosis signaling pathways. Mol Carcinog 2012;51(10):761-770. 263. 260. Liu H, Han D, Liu Y, Hou X, Wu J, Li H, Yang J, Shen C, Yang G, Fu C, Li X, Che H, Ai J, Zhao S. Harmine hydrochloride inhibits Akt phosphorylation and depletes the pool of cancer stem-like cells of glioblastoma. J Neurooncol 2013;112(1):39-48. 264. 264. Yang J, Li TZ, Xu GH, Luo BB, Chen YX, Zhang T. Low-concentration capsaicin promotes colorectal cancer metastasis by triggering ROS production and modulating Akt/mTOR and STAT-3 pathways. Neoplasma 2013;60(4):364-372. 265. 217. Hwang MK, Bode AM, Byun S, Song NR, Lee HJ, Lee KW, Dong Z. Cocarcinogenic effect of capsaicin involves activation of EGFR signaling but not TRPV1. Cancer Res 2010;70(17):6859-6869. 266. 218. Toth B, Gannett P. Carcinogenicity of lifelong administration of capsaicin of hot pepper in mice. In Vivo 1992;6(1):59-63. 267. 219. Chanda S, Erexson G, Riach C, Innes D, Stevenson F, Murli H, Bley K. Genotoxicity studies with pure trans-capsaicin. Mutat Res 2004;557(1):85-97.

28

Jo

ur

na

lP

re

-p

ro of

268. 220. Allameh A, Saxena M, Biswas G, Raj HG, Singh J, Srivastava N. Piperine, a plant alkaloid of the piper species, enhances the bioavailability of aflatoxin B1 in rat tissues. Cancer Lett 1992;61(3):195-199. 269. 221. Fang ZZ, Krausz KW, Li F, Cheng J, Tanaka N, Gonzalez FJ. Metabolic map and bioactivation of the anti-tumour drug noscapine. Br J Pharmacol 2012;167(6):1271-1286. 270. 222. Ansari KM, Dhawan A, Khanna SK, Das M. In vivo DNA damaging potential of sanguinarine alkaloid, isolated from argemone oil, using alkaline Comet assay in mice. Food Chem Toxicol 2005;43(1):147-153. 271. 223. Lu H, Zhang L, Gu LL, Hou BY, Du GH. Oxymatrine induces liver Injury through JNK signalling pathway mediated by TNF-α in vivo. Basic Clin Pharmacol Toxicol 2016;119(4):405411. 272. 224. Chen J, Hu W, Qu YQ, Dong J, Gu W, Gao Y, Fang Y, Fang F, Chen ZP, Cai BC. Evaluation of the pharmacodynamics and pharmacokinetics of brucine following transdermal administration. Fitoterapia 2013;86:193-201. 273. 225. Yamashita K, Ohgaki H, Wakabayashi K, Nagao M, Sugimura T. DNA adducts formed by the comutagens harman and norharman in various tissues of mice. Cancer Lett 1988;42(3):179-183. 274. 226. Mailhes JB, Young D, London SN. Cytogenetic effects of caffeine during in vivo mouse oocyte maturation. Mutagenesis 1996;11(4):395-399. 275. 227. Welsch CW, DeHoog JV, O'Connor DH. Influence of caffeine consumption on carcinomatous and normal mammary gland development in mice. Cancer Res 1988;48(8):2078-2082. 276. 228. Panigrahi GB, Rao AR. Influence of caffeine on arecoline-induced SCE in mouse bone-marrow cells in vivo. Mutat Res 1983;122(3-4):347-353. 277. 229. Frei JV, Venitt S. Chromosome damage in the bone marrow of mice treated with the methylating agents methyl methanesulphonate and N-methyl-N-nitrosourea in the presence or absence of caffeine, and its relationship with thymoma induction. Mutat Res 1975;30(1):89-96. 278. National Toxicology Program. NTP toxicology and carcinogenesis studies of theophylline (CAS No. 58-55-9) in F344/N rats and B6C3F1 mice (feed and gavage studies). Natl Toxicol Program Tech Rep Ser 1998;473:1-326. 279. 231. Wang XP, Lei F, Du F, Chai YS, Jiang JF, Wang YG, Yu X, Yan XJ, Xing DM, Du LJ. Protection of gastrointestinal mucosa from acute heavy alcohol consumption: The effect of berberine and its correlation with TLR2, 4/IL1β-TNFα signaling. PLoS One 2015;10(7):e0134044. 280. 232. Lv X, Yu X, Wang Y, Wang F, Li H, Wang Y, Lu D, Qi R, Wang H. Berberine inhibits doxorubicin-triggered cardiomyocyte apoptosis via attenuating mitochondrial dysfunction and increasing Bcl-2 expression. PLoS One 2012;7(10):e47351. 281. 233. Liu W, Yi DD, Guo JL, Xiang ZX, Deng LF, He L. Nuciferine, extracted from Nelumbo nucifera Gaertn, inhibits tumor-promoting effect of nicotine involving Wnt/β-catenin signaling in non-small cell lung cancer. J Ethnopharmacol 2015;165:83-93. 282. 234. Zhang Y, Sun S, Chen J, Ren P, Hu Y, Cao Z, Sun H, Ding Y. Oxymatrine induces mitochondria dependent apoptosis in human osteosarcoma MNNG/HOS cells through inhibition of PI3K/Akt pathway. Tumour Biol 2014;35(2):1619-1625. 283. 235. Geriyol P, Basavanneppa HB, Dhananjaya BL. Protecting effect of caffeine against vinblastine (an anticancer drug) induced genotoxicity in mice. Drug Chem Toxicol 2015;38(2):188-195. 284. 236. Zhang ZW, Xiao J, Luo W, Wang BH, Chen JM. Caffeine suppresses apoptosis of bladder cancer RT4 cells in response to ionizing radiation by inhibiting ataxia telangiectasia mutated-Chk2-p53 Axis. Chin Med J (Engl) 2015;128(21):2938-2945.

29

Table 1: Pharmacological activity of candidate plant-derived alkaloids for drug repositioning in cancer therapy.

Compound

Pharmacological activity

Protoalkaloids (with exocyclic nitrogen): Capsaicin Irritant, produces sensation of burns Ephedrine Indirect sympathomimetic, direct adrenoreceptor agonist Piperine Bioenhancer, increases secretion, antimicrobial

Source plant

Traditional use of source plants

Capsicum spec. Ephedra sinensis Piper nigrum

Spice

lP

re

-p

Alkaloids with heterocyclic nitrogen: (a) derived from aromatic amino acids Isoquinoline Berberine Antiseptic, reduces blood Coptis alkaloids glucose antiarrhythmic, reduces chinensis cholesterol and lipids antidepressive Berbamine Calcium channel blocker Berberis vulgaris Boldine α-adrenergic antagonist Peumus boldus

Antibacterial, hypoglycemic, gastric-mucous membrane protection, inhibitor of human organic cation transporters

Coptis chinensis

Antitussive, bronchodilatation

Papaver somniferum Nelumbo nucifera

na

Coptisine

ur

Noscapine

Jo

Nuciferine

Palmatine

Papaverine

Dopamine receptor blocker

Against jaundice, dysentery, hypertension, inflammation, and liver-related diseases Antispasmotic

Sanguinarine Na+/K+ ATPase inhibitor

30

TCM: against bronchospasms Spice; buddhistic folk medicine: constipation, insomnia, oral abcesses, toothaches, sunburn, appetiting and digestive activity

ro of

Class

Coptis chinensis Papaver somniferum Sanguinaria canadensis

TCM: Digestive disorders caused by bacterial infections

Liver and gallbladder disorders, jaundice, digestive disorders South-American folk medicine: anticonvulsant, liver and gallbladder disorders, gastrointestinal ailments, dyspepsia TCM: Digestive disorders caused by bacterial infections

Narcotic TCM and Ayurveda: hematemesis, epistaxis, hematuria, diarrhea, cholera, fever, hyperdipsia, diuretic, antidiabetic, antiinflammatory TCM: Digestive disorders caused by bacterial infections Narcotic South-American folk medicine: emetic, against warts, against respiratory ailments

Tetrandrine Quinazoline Matrine alkaloids

Anti-arthritis activity by improvement of Th1/Th2 imbalance Calcium channel blocker κ-opioid and μ-opioid receptor agonist

Oxymatrine

Decreases cardiac ischemia, myocardial injury, arrhythmia and improves heart failure Betaxanthin Indicaxanthin Anti-thalassemic activity alkaloids Ipecacuanha Emetine Anti-protozoal, induces alkaloids vomiting (b) derived from tryptophan Indole Brucine Anti-inflammatory and alkaloids analgesic agent

Evodiamine

Reduces fat uptake

Indole-3carbinol

Anti-malaria, anti-babesiosis

Quinine

Anti-malaria

Harmine, harmaline

MAO inhibitor, hallucinogenic

Stephania tetrandra Sophora flavescens

Antiemetic, diuretic

Sophora flavescens Opuntia ficus-indica Carapichea ipecacuanha

Competitive A2a adenosin receptor antagonist, inhibitor of cAMP degradation Theobromine Diuretic, vasodilatating, relaxing smooth heart muscles Theophylline Non-selective inhibitor of phosphodiesterases, A1 and A2 adenosin receptor antagonist, inhibitor of cAMP degradation

31

Emetic, intentional vomiting after poisoning, nauseant, expectorant, diaphoretic Ayurveda and TCM: against pain and swelling

Cinchona officinalis Tetradium ruticarpum

Peruvian traditional medicine: anti-fever, anti-malaria TCM and Kampo medicine: against abdominal pain, acid regurgitation, nausea, diarrhea, acts dysmenorrheal, antiinflammatory, anti-infective Diet

re

lP

na

Jo

(c) derived from purine Caffeine

TCM: hepatitis, viral myocarditis, gastrointestinal hemorrhage, and skin diseases TCM: hepatitis, viral myocarditis, gastrointestinal hemorrhage, and skin diseases Fruit

Strychnos nux-vomica

-p

Anti-malaria

TCM: against rheumatism and arthritis

Various cruciferous vegetables Cinchona officinalis Peganum harmala, Banisteriopsis caapi

ur

Harmane alkaloids

Cinchonine

Sinomenium acutum

ro of

Sinomenine

Coffea arabica, C. robusta Theobroma spec. Camelia sinensis, C. arabica, Theobroma spec.

Peruvian traditional medicine: anti-fever, anti-malaria Traditional Iranian medicine: carminative, anthelmintic, aphrodisiac, galactagogue, diuretic, emmenagogue, antithrombotic, phlegmatic purgative, and strengthening the vision Beverage

Beverages, Chocolate Beverages, Chocolate

Ayurveda: asthma, bronchitis tuberculosis

Jo

ur

na

lP

re

-p

ro of

Pseudoalkaloids (with exocyclic nitrogen): Steroidal Peiminine Anti-inflammatory, anti-allergic, Fritillaria alkaloids antitussive, expectorant roylei

32

Table 2: Inhibition of tumor growth in vivo and cell cycle effects by plant alkaloids.

Mode of action

Reference

Tumor growth↓, G0/G1 cell cycle arrest, oxidative stress (ROS↑, catalase↑, SOD-2↑), FOXO3a↑ Tumor growth↓, G0/G1 cell cycle arrest, ER-stress response↑ (GRP-78↑, p-ERK↑, p-EIF-2a↑, ATF4↑, GADD-153↑)

Qian et al., 2016 [76] Lin et al., 2013 [77]

Capsaicin

Tumor growth↓, G0/G1 cell cycle arrest, p-PI3K↓, p-AKT↓ Tumor growth↓, G0/G1 arrest, E2F-responsive proliferative genes↓ (cyclin E, thymidylate synthase, Cdc25A, Cdc6), E2F4↑

Zhang et al., 2013 [78] Brown et al., 2010 [79]

Xenograft PANC-1 pancreas Ca Xenograft H69 lung Ca

Capsaicin

ro of

Compound Model Tumor type G0/G1 cell cycle arrest: Capsaicin Xenograft 5637 and T24 bladder Ca Capsaicin Xenograft PANC-1 and SW1990 pancreas Ca

Othotopic MDA-MB-231 Tumor growth↓, G0/G1 cell cycle arrest (cyclin xenograft breast Ca D1↓), HER2↓, ERK↓, KIP-1/p27↑

Capsaicin

Xenograft U 266 multiple myeloma

Tumor growth↓, G0/G1 cell cycle arrest, IL-6induced STAT3 activation↓, JAK1↓, c-SRC↓, STAT3-regulated gene products↓ (cyclin D1, Bcl-2, Bcl-xL, survivin, VEGF)

Berbamine

Xenograft SW480 colorectal Ca Xenograft HCT-116 colon Ca Xenograft U87 glioblastoma Xenograft MDA-MB-231 breast Ca

Tumor growth↓, G0/G1 cell cycle arrest

Sinomenine

Xenograft Hep3B hepatocellular Ca Tetrandrine Xenograft 143 B osteosarcoma Matrine Xenograft Eca-109 esophagus Ca Caffeine Xenograft U87.MG glioblastoma Peiminine Xenograft U251 Glioblastoma S-phase arrest: Sinomenine Xenograft SW1116 colon Ca Evodiamine Xenograft LoVo colon Ca

Jo

ur

Sinomenine

re

Tumor growth↓, G0/G1 cell cycle arrest, RAS-ERK pathway↓ Tumor growth↓, G0/G1 cell cycle arrest, p53↑, p53 acetylation↑, SIRT-1↓ Tumor growth↓, G1/S cell cycle arrest, ROS↑, ATM/Chk-2- and ATR/Chk1-mediated DNA-damage response, p-ERK↑, p-JNK↑, p38 MAPK↑ Tumor growth↓, G0/G1 cell cycle arrest

lP

Sinomenine

na

Coptisine

-p

Capsaicin

Tumor growth↓, G0/G1 cell cycle arrest, PTEN↑, p-PTEN↓, p38 MAPK↑, p-p38 MAPK↑ Tumor growth↓, G0/G1 cell cycle arrest, apoptosis↑ (p53↑, p21↑, Bcl-2/Bid ratio↓) Tumor growth↓, G0/G1 cell cycle arrest (pRB↑)

Thoennissen et al., 2010 [80] Bhutani et al., 2007 [81]

Zhang et al., 2018 [82] Huang et al., 2017 [83] He et al., 2018 [84] Li et al., 2014 [85] Lu et al., 2013 [86]

Tumor growth↓, G0/G1 cell cycle arrest, p-AKT↓, p-GSK-3β↓

Tian et al., 2017 [87] Wang et al., 2014 [88] Ku et al., 2011 [89] Zhao et al., 2018 [90]

Tumor growth↓, CIP-1/p21↓, cyclin D1↓, cyclin E↓, COX-2↓, Tumor growth↓, S-phase cell cycle arrest (cyclin A↓, CDK2↓, Cdc25c↓)

Yang et al., 2016 [91] Zhang et al., 2010 [92]

G2/M cell cycle arrest: 33

Noscapine Emetine Evodiamine Evodiamine

Harmine Harmine

Xenograft GBM157 glioblastoma Xenograft U87 glioblastoma Xenograft HepG2 hepatocellular Ca

Tumor growth↓, mitotic genes↓ (MELK, ASPM, TOP-2A, and FOXM-1b) Tumor growth↓, G2/M cell cycle arrest ( tubulin polymerization↑, cyclin B1↑), p-JNK↑ Tumor growth↓, G2/M cell cycle arrest, IL-6induced STAT3 activation↓, p-JAK2↓, p-Src↓, pERK1/2↓, SHP1↑

Xenograft SGC-7901 gastric Ca Xenograft A549 lung Ca

Tumor growth↓, G2/M cell cycle arrest

Harmaline

Xenograft SGC-7901 gastric Ca Inhibition of tumor growth: Piperine, Syngeneic Sarcoma 180 piplartine

Tumor growth↓, G2/M cell cycle arrest, p53 signaling↑ Tumor growth↓, G2/M cell cycle arrest, pCdc2, p21.pp53, cyclin C↑, p-Cdc25c↓ Tumor growth↓

Syngeneic Hamster Ham- Tumor growth↓ 1 cholangio Ca

Berberine

Xenograft SCC-4 tongue squamous Ca Syngeneic B16 melanoma

Tumor growth↓ Tumor growth at high doses↓, but ↑ at high doses

Xenograft Rat C6 glioma Tumor growth↓

ur

Noscapine

na

lP

Berberine

Berberine

Syngeneic B16LS9 melanoma Sanguinarine Xenograft UM-SCC-22BcFluc head and neck Ca Emetine Xenograft PDX lymphoma Emetine Syngeneic Sarcoma 180, Ehrlich ascites tumor

Jo

Noscapine

Wang et al., 2017 [93] Cai et al., 2014 [94] Paydar et al., 2014 [95] Yang et al., 2012 [96] Visnyei et al., 2011 [97] Wu et al., 2017 [98] Yang et al., 2013 [99]

ro of

Boldine

-p

Berberine

Xenograft HONE1 Tumor growth↓, G2/M cell cycle arrest, EBNA-1↓ nasopharynx Ca Xenograft LoVo colon Ca Tumor growth↓, G2/M cell cycle arrest (cyclin B1↓, Cdc2↓, Cdc25c↓) Xenograft MDA-MB-231 Tumor growth↓, G2/M cell cycle arrest, HSP70↓ breast Ca Xenograft LoVo Colon Ca G2/M cell cycle arrest

re

Berberine

Tumor growth and progression↓ Tumor growth↓

Growth of tumors with MYC rearrangement↓ Tumor growth↓

34

Wang et al., 2015 [100] Dai et al., 2012 [101] Wang et al., 2015 [100] Bezerra et al., 2006 [102] Puthdee et al., 2013 [103] Ho et al., 2009 [104] Letasiová et al., 2005 [105] Landen et al., 2004 [106] Landen et al., 2002 [107] Wang et al., 2016 [108] Aoki et al., 2017 [109] Johnson et al., 1974 [110]

Table 3: Inhibition of tumor growth in vivo and induction of apoptosis by plant alkaloids.

Model Xenograft

Capsaicin, dehydrocapsaicin

Xenograft

Capsaicin

Xenograft

Capsaicin

Xenograft

Capsaicin

Xenograft

Capsaicin

Tumor type HOS osteosarcoma U251 glioma

Mode of action Tumor growth↓, apoptosis↑, ERK1/2↓, p38↓ Tumor growth↓, ROS↑, Ca2+↑, apoptosis↑ (mitochondrial depolarization, cytochrome c release, caspase-3↑, caspase-9↑)

Reference Zhang et al., 2017 [111] Xie et al., 2016 [112]

786-O renal Ca PANC-1 and SW1990 pancreas Ca

Tumor growth↓, apoptosis↑ (FADD↑, Bax↑, Bcl-2↓, caspases-3, -8, -9↑) Tumor growth↓, apoptosis↑

Liu et al., 2016 [113] Lin et al., 2013 [77]

PANC-1 pancreas Ca Orthotopic AsPC-1 xenograft pancreas Ca

Tumor growth↓, apoptosis↑, p-PI3K↓, pAKT↓ Tumor growth↓, thioredoxin↓, p-ASK-1↑, caspase-3↑, PARP cleavage

Zhang et al., 2013 [78] Pramanik and Srivastava, 2012 [114] Tumor growth↓, ROS↑, Ca2+↑, apoptosis↑ Lu et al., (Fas↑, Bax↑, Bcl-2↓, mitochondrial 2010 [115] membrane potential↓, cytochrome c release, caspases↑) Tumor growth↓, apoptosis↑ (PARP Thoennissen cleavage), HER2↓, ERK↓, KIP-1/p27↑ et al., 2010 [80] Tumor growth↓, ROS↑, apoptosis↑ (Bax↑, Zhang et al., Bcl-2↓, mitochondrial depolarization, 2008 [116] survivin↓, JNK activation↑, cytochrome c and AIF release, caspase-3↑)

Xenograft

Capsaicin

Orthotopic MDA-MB-231 xenograft breast Ca

Capsaicin

Xenograft

AsPC-1 pancreas Ca

Capsaicin

Xenograft

U 266 multiple Tumor growth↓, apoptosis↑ (caspase↑), myeloma IL-6-induced STAT3 activation↓, JAK1↓, cSRC↓, STAT3-regulated gene products↓ (cyclin D1, Bcl-2, Bcl-xL, survivin, VEGF)

Bhutani et al., 2007 [81]

Capsaicin

Xenograft

PC-3 prostate Ca

Tumor growth↓, ROS↑, apoptosis↑(mitochondrial membrane potential↓) NB4 leukemia Tumor growth↓, ROS↑, apoptosis↑, pp53↑ Melanoma Tumor growth↓, apoptosis↑ (Bax↑, Bcl2↓, XIAP↑, PARP cleavage, caspases-3 and 9↑), p-JNK↑, p-p38↑, p-ERK-1/2↓

Sánchez et al., 2006 [117] Ito et al., 2004 [118] Yoo et al., 2019 [119]

MDA-MB-231 and BT549 breast Ca

Zhao et al., 2017 [120]

ur

na

lP

re

-p

Capsaicin

Xenograft

Jo

Capsaicin

Colo 205 colon Ca

ro of

Compound Capsaicin

Piperine

Xenograft

Berberine

Xenograft

Berberine

Syngeneic HONE1 nasopharynx Ca

Tumor growth↓, DNA double strand breaks↑, apoptosis↑ (Bax↑, Bcl-2↓, cytochrome c release↑, caspases-3 and 9↑) Tumor growth↓, apoptosis↑, EBNA-1↓

35

Wang et al., 2017 [93]

Xenograft

Berberine

Xenograft

Berberine

Xenograft

Berbamine

Xenograft

SW480 colorectal Ca

Berbamine

Xenograft

Berbamine

Xenograft

SKOV3 ovarian Ca PC-3 prostate Ca

Tumor growth↓, β-catenin↓, caspases-3 and -9↑ Tumor growth↓, apoptosis↑ (mitochondrial dysfunction, Bax/Bcl-2 ratio↑, activation of caspases-3 and -9↑)

Zhang et al., 2018 [124] Zhao et al., 2016 [125]

Berbamine

Xenograft

Xenograft

Tumor growth↓, apoptosis↑ (Fas↑, p53↑, mitochondrial membrane potential↓, caspases-3, -8, -9↑) Tumor growth↓, apoptosis↑ (Bax↑, Bcl2↓, mitochondrial membrane potential↓, cytochrome c release, DNA fragmentation, NFκB↑, caspases-3, -7, -9↑), HSP70↓

Wang et al., 2009 [126]

Boldine

HepG2 hepatocellular Ca MDA-MB-231 breast Ca

Coptisine

Xenograft

Tumor growth↓, miR-122↑, apoptosis↑

Chai et al., 2018 [127]

Coptisine

Xenograft

HepG2 hepatocellular Ca HCT116 colorectal Ca

Coptisine

Xenograft

Noscapine

Xenograft

Noscapine

Xenograft

Noscapine

Xenograft

na

HCT-116 colon Ca HepG2 hepatocellular Ca LoVo Colon Ca BGC823 gastric Ca H460 lung Ca

ur Xenograft

Jo

Noscapine

Nuciferine

Xenograft

A549 lung Ca

Sanguinarine Orthotopic HCT116 xenograft colorectal Ca Sinomenine Syngeneic Murine B16F10 melanoma Sinomenine

Xenograft

Tumor growth↓, apoptosis↑, STAT-3↓

Tsang et al., 2013 [121] Tumor growth↓, p53-dependent Choi et al, apoptosis↑ 2009 [122] Tumor growth↓, apoptosis↑ (Bax↑, Bak↑, Katiyar et Bcl-2↓, Bcl-xL↓, caspase-3↑) al., 2009 [123] Tumor growth↓, apoptosis↑ (p53↑, Bax↑, Zhang et al., Bcl-2↓, PARP cleavage, caspases-3, -8, -9↑) 2018 [82]

U87 glioblastoma

-p

re

Paydar et al., 2014 [95]

Tumor growth↓, apoptosis↑ (Bax↑, Bad↑, Han et al., Bcl-2↓, Bxl-xL↓, XIAP↓, cytochrome c 2018 [128] release, APAF-1↑, AIF↑, caspase-3↑)

lP

C666-1 nasopharynx Ca PC-3 prostate Ca A549 and H1299 lung Ca

ro of

Berberine

Tumor growth↓, apoptosis↑ (caspases↑), RAS-ERK pathway↓ Apoptosis↑ (caspase-3↑, PARP cleavage, Bcl-2/Bax ratio↓)

Huang et al., 2017 [85] Xu et al., 2016 [129]

Apoptosis↑ (Bax↑, Bcl-2↓, cytochrome c release, survivin↓, caspases-3 and -9↑) Apoptosis↑ (Bax↑, cytochrome c release, Bcl-2↓, caspase-3↑, caspase-9↑) Apoptosis↑ (Bax↑, Bcl-2↓, PARP cleavage, caspase-3↑)

Yang et al., 2012 [96] Liu et al., 2011 [130] Jackson et al., 2008 [131] Apoptosis↑ (Bcl-2/Bax↓) Liu et al., 2015 [132] Tumor growth↓, apoptosis↑, p-STRAP↓, p- Gong et al., MELK↓ 2018 [133] Tumor growth↓, apoptosis↑ (caspase-3↑, Sun et al., Bax/Bcl-2 ratio↑), autophagy ↑ (Beclin-1↑, 2018 [134] LC3-II↑, p62↓) Tumor growth↓, apoptosis↑, p53↑, p53 acetylation↑, SIRT-1↓

36

He et al., 2018 [84]

Sinomenine

Xenograft

Hep3B hepatocellular Ca

Sanguinarine Xenograft Tetrandrine

Xenograft

Tetrandrine

Xenograft

Tetrandrine

Xenograft

DU145 prostate Ca 143 B osteosarcoma BGC-823 gastric Ca

Matrine

Xenograft

4T1 breast Ca

Matrine

Xenograft

C1498 leukemia

Matrine

Xenograft

Matrine

Xenograft

Eca-109 esophagus Ca HL-60 leukemia

Matrine

Xenograft

Tumor growth↓, apoptosis↑

Tumor growth↓, apoptosis↑ (Bax↑, Bcl2↓, caspase-3↑) Tumor growth↓, apoptosis↑, WNT/βcatenin signaling↓, VEGF↓ Tumor growth↓, apoptosis↑ (caspase-3↑, PARP cleavage) , autophagy↑ (LC3-II↑, pAKT↓, mTOR↓, p70S6K↓, 4EBP1↓) Tumor growth↓, apoptosis↑ (p53↑, p21↑, Bcl-2/Bid ratio↓) Tumor growth↓, apoptosis↑ (Bcl-2/Bax ratio↑, mitochondrial membrane potential↓, cytochrome c release, caspase3↑), p-AKT↓, p-ERK1/2↓

MNNG/HOS osteosarcoma BxPC-3 pancreas Ca

Tumor growth↓, apoptosis↑ (Bax↑, Bcl2↓, caspases-3, -8, -9↑) Xenograft Tumor growth↓ (PCNA↓), apoptosis↑ (Fas↑, Bcl-2/Bax ratio↓, caspases-3, -8, 9↑) Syngeneic Murine H22 Tumor growth↓, apoptosis↑ (Bax↑, Bclhepatocellular 2↓) Ca Xenograft PC-3 prostate Tumor growth↓, apoptosis↑ (p53↑, Bax↑, Ca Bcl-2↓) Xenograft MNNG/HOS Tumor growth↓, apoptosis↑ (Bax↑, Bclosteosarcoma 2↓, mitochondrial dysfunction, cytochrome c release, caspases-3 and -9↑), p-PI3K↓, pAKT↓

Jo

Matrine

Oxymatrine Oxymatrine

Lu et al., 2013 [86]

Sun et al., 2010 [135] Tumor growth↓, apoptosis↑, PTEN↑, pTian et al., PTEN↓, p38 MAPK↑, p-p38 MAPK↑ 2017 [87] Tumor growth↓, apoptosis↑ (Bax↑, Bak↑, Qin et al., Bad↑, Bcl-2↓, Bcl-xL↓, cytochrome c 2013 [136] release, Apaf-1↑) Tumor growth↓, apoptosis↑, AKT↑, JNK↓, Liu et al., ERK↓ 2011 [137]

ur

Matrine

na

Tetrandrine

Li et al., 2014 [85]

Tumor growth↓, apoptosis↑, survivin↓

lP

Matrine

HepG2 hepatocellular Ca Syngeneic Rat RT-2 glioma Xenograft Colorectal Ca

Tumor growth↓, ROS↑, ATM/Chk-2- and ATR/Chk1-mediated DNA-damage response, p-ERK↑, p-JNK↑, p38 MAPK↑ Tumor growth↓, apoptosis↑ (p21/WAF1/ CIP1 ↑, Bax↑, Bcl-2↓, mitochondrial membrane potential↓, cytochrome c and Omi/HtrA2 release↑, survivin↓, caspases-3, -8,-9,-10↑, PARP cleavage)

ro of

MDA-MB-231 breast Ca

-p

Xenograft

re

Sinomenine

37

Chen et al., 2009 [138] Gu et al., 2018 [139] Xiao et al., 2018 [140] Wu et al., 2017 [141] Wang et al., 2014 [88] Zhang et al., 2012 [142]

Liang et al., 2012 [143] Liu et al., 2010 [144] Ma et al., 2008 [145] Wu et al., 2015 [146] Zhang et al., 2014 [147]

Oxymatrine

Xenograft

Brucine

Xenograft

Cinchonine

Xenograft

Cinchonine

Xenograft

Evodiamine

Xenograft

Evodiamine

Xenograft

Harmine

Xenograft

A549 lung Ca

Harmaline

Xenograft

SGC-7901 gastric Ca

Caffeine

Xenograft

na

Harmine

Jo

ur

U87 glioblastoma

Wu et al., 2017 [98]

Tu et al., 2013 [153] Yang et al., 2013 [99]

Zhang et al., 2010 [92] Ding et al., 2019 [154] Hai-Rong et al., 2019 [155] Tumor growth↓, p53 signaling↑, Dai et al., apoptosis↑ 2012 [101] Tumor growth↓, p21↑, p-p53↑, cyclin C↑, Wang et al., p-Cdc25c↓, Fas/FasL↑, activation of 2015 [100] caspases-3,-8↑ Tumor growth↓, apoptosis↑ (caspase-3↑, Ku et al., PARP cleavage) 2011 [89]

lP

Harmine

Wu et al., 2016 [152]

Tumor growth↓, apoptosis↑ (caspase-3↑, PARP cleavage, p-Bcl-2↑), p-PERK↑, pJNK↑ Syngeneic Murine Lewis Tumor growth↓, apoptosis↑, autophagy↑ lung Ca (LC3-II↑, autophagosome formation↑) Xenograft HepG2 Tumor growth↓, apoptosis↑, IL-6-induced hepatoSTAT3 activation↓, p-JAK2↓, p-Src↓, pcellular Ca ERK1/2↓, SHP1↑ Xenograft LoVo colon Ca Tumor growth↓, apoptosis↑ (caspases-3, 8, -9↑) Xenograft MCF-7 breast Bcl-2↓, Bax↑ Ca Xenograft RT4 bladder Tumor growth↓, apoptosis↑ Ca

re

Evodiamine

A498 renal cell Ca

Ruijun et al., 2014 [149] Jin et al., 2018 [150] Qi et al., 2017 [151]

ro of

Evodiamine

Wu et al., 2014 [148]

-p

Evodiamine

SGC-996 Tumor growth↓, apoptosis↑ (Bax↑, Bclgallbladder Ca 2↓, mitochondrial membrane potential↓, NFκB↓, caspase-3↑) U251 glioma Tumor growth↓, apoptosis↑ (Bax↑, Bcl2↓) HepG2 liver Tumor growth↓, apoptosis↑ (PARP1 Ca cleavage, caspase-3↑) HeLa cervix Ca Tumor growth↓, apoptosis (Bax↑, Bcl-2↓, and A549 lung binding to RING domain of TRAF6) Ca U87 Tumor growth↓, apoptosis↑ (p53↑, pglioblastoma p53↑, PARP cleavage, caspase-3↑, DNA fragmentation↑), p-JNK↑

38

Table 4: Inhibition of tumor growth in vivo and induction of non-apoptotic cells death by plant alkaloids. Mode of action Tumor growth↓, ROS↑, mitochondrial membrane potential↓, non-apoptotic cell death↑ Tumor growth↓, oncosis↑ (cell swelling↑, cytoplasmic vacuoles↑, plasma membrane blebbing↑), autophagy↑ (ATP↓, mitochondrial aerobic respiration↑, p-ERK1/2↑)

Reference Yang et al., 2010 [156]

Tumor growth↓, autophagy↑, AMPK/mTOR/ULK1 pathway↓

Wang et al., 2016 [157]

Xenograft HepG2 hepatocellular Ca Tetrandrine Xenograft THP-1 leukemia Tetrandrine Xenograft AMKL leukemia Tetrandrine Xenograft PML-RARαpositive acute promyelocytic leukemia

Tumor growth↓, pyroptosis↓

Chu et al., 2016 [37]

Tumor growth↓, ROS↑, autophagy↑, differentiation↑, c-MYC↓ Tumor growth↓, ROS↑, autophagy↑, AKT and Notch1 signaling↑ Tumor growth↓, ROS↑, autophagy↑, Notch1 signaling↑

Wu et al., 2018 [158] Liu et al., 2017 [159] Liu et al., 2015 [160]

Tetrandrine Xenograft Huh7 hepatocellular Ca Matrine Xenograft PDAC pancreas Ca

Tumor growth↓, ROS↑, autophagy↑, mitochondrial dysfunction, ERK MAPK↑ Tumor growth↓, autophagy↑, mitochondrial metabolic energy function↓, lysosomal protease↓ Tumor growth↓, Bid-mediated nuclear translocation of AIF, caspase-independent cell death Tumor growth↓, apoptosis↑ (caspase-3↑, Bax/Bcl-2 ratio↑), autophagy ↑ (Beclin-1↑, LC3-II↑, p62↓) Tumor growth↓, ROS↑, autophagy↑ (LC3BII↑), AKT-mTOR pathway↓, JNK pathway↑

Gong et al., 2012 [161] Cho et al., 2018 [162]

Tumor growth↓, apoptosis↑, autophagy↑ (LC3-II↑, autophagosome formation↑) Tumor growth↓, autophagy (p-AMPK↓, p-ULK1↓), p-AKT↓

Tu et al., 2013 [153] Zhao et al., 2018 [90]

Berberine

Xenograft U87 glioblastoma

lP

Berberine

Xenograft HepG2 hepatocellular Ca Sinomenine Syngeneic Murine B16F10 melanoma Sinomenine Xenograft U87 and SF767 glioblastoma Evodiamine Syngeneic Murine Lewis lung Ca Peiminine Xenograft U251 glioblastoma

Jo

ur

na

Matrine

Sun et al., 2018 [38]

ro of

Xenograft Glioma

-p

Berberine

re

Compound Model Tumor type Capsaicin Xenograft T24 bladder Ca

39

Zhou et al., 2014 [163] Sun et al., 2018 [134] Jiang et al., 2017 [164]

Table 5: Inhibition of tumor growth in vivo and angiogenesis by plant alkaloids.

Xenograft

Sanguinarine Syngeneic

Sanguinarine Syngeneic

Sanguinarine Xenograft

Reference Hamsa and Kuttan, 2012 [165] U87 Tumor growth↓, AMPK/mTOR/ULK1 pathway↓ Wang et al., glioblastoma 2016 [157] Pica et al., DHD/K12/TRb Tumor growth↓, angiogenesis↓ 2012 [166] colorectal adeno Ca B16 4A5 Tumor growth↓, angiogenesis↓, p-p44/42 De Stefano et melanoma MAPK, p-AKT al., 2009 [167] A375 Tumor growth↓, angiogenesis↓, p-p44/42 De Stefano et melanoma MAPK, p-AKT al., 2009 [167] Huh7 hepato- Angiogenesis↓ Xiao et al., cellular Ca 2015 [168]

Tetrandrine

Xenograft

Tetrandrine

Syngeneic Rat RT-2 glioma Xenograft PANC-1 pancreas Ca Syngeneic Ehrlich ascites tumor

Oxymatrine Brucine

ro of

Berberine

Mode of action Tumor growth↓, angiogenesis↓ (VEGF↓)

Tumor growth↓, angiogenesis↓ (VEGF↓)

-p

Model Tumor type Syngeneic B16-F10 melanoma

Tumor growth↓, NFκB↓, VEGF↓ Angiogenesis↓, p-VEGFR-2 downstream signaling↓ (SRC, FAK, ERK, AKT, mTOR)

re

Compound Berberine

syngeneic Ehrlich Peritoneal angiogenesis and microvessel ascites tumor density↓, VEGF↓

Evodiamine

Xenograft H22 hepatocellular Ca Xenograft RT4 bladder Ca

Tumor growth↓, angiogenesis↓ (VEGFα↓)

Xenograft A549 lung Ca

Tumor growth↓, angiogenesis↓, p53 signaling↑ Tumor growth↓, VEGF↓, nitric oxide↓

Syngeneic B16-F10 melanoma

Tumor growth↓, VEGFR-2↓

Jo

Harmine

na

Harmine

ur

Harmine

lP

Brucine

Theobromine Xenograft HCV-29-v-raf Angiogenesis↓, VEGF↓ transfectant urothelial cells Theobromine Xenograft Primary VEGF↓ ovarian Ca from patients Theophylline Syngeneic B16-F10 Tumor growth↓, angiogenesis↓ melanoma

40

Chen et al., 2009 [169] Chen et al., 2013 [138] Saraswati and Agarwal, 2013 [170] Agarwal et al., 2011 [171] Shi et al., 2016 [172] Hai-Rong et al., 2019 [155] Dai et al., 2012 [101] Hamsa and Kuttan, 2010 [173] SkopinskaRózewska et al., 1998 [174] Barcz et al., 1998 [175] Menon et al., 2002 [176]

Table 6: Inhibition of tumor growth in vivo and signaling pathways by plant alkaloids.

Compound Model Tumor type Transcription factors: Capsaicin Xenograft 786-O renal Ca Capsaicin Xenograft 5637 and T24 bladder Ca Capsaicin Xenograft PANC-1 and SW1990 pancreas Ca

Mode of action

Reference

Tumor growth↓, c-MYC↑

Liu et al., 2016 [113] Qian et al., 2016 [76] Lin et al., 2013 [77]

Capsaicin

Xenograft H69 lung Ca

Tumor growth↓, E2F4↑

Capsaicin

Xenograft PC-3 prostate Tumor growth↓, PSA promoter/enhancer Ca activation by dihydrotestosterone↓, NFκB activation↓, degradation of IκBα↓, proteasome activity↓

Berberine

Xenograft Colon Ca

Berberine

Syngeneic B16-F10 melanoma

Berberine

Xenograft Pimary Tumor growth↓, NFκB↓, p-IKK↓, p-IκB↓ effusion lymphoma Xenograft MDA-MB-231 Tumor growth↓, NFκB↑ breast Ca Xenograft A549 lung Ca c-MYC↓

-p

Tumor growth↓, RXRα binding Tumor growth↓, NFĸB↓, c-Fos↓, HIF↓

re

Nuciferine

Xenograft THP-1 leukemia Oxymatrine Xenograft PANC-1 pancreas Ca Indicaxanthin Xenograft A375 melanoma Emetine Xenograft GBM157 glioblastoma Brucine Xenograft DLD1 colon Ca Brucine Syngeneic Ehrlich ascites tumor

Jo

ur

na

Tetrandrine

Evodiamine

Brown et al., 2010 [79] Mori et al., 2006 [177]

ro of

Tumor growth↓, p-EIF-2a↑, ATF-4↑

lP

Boldine

Harmine

Tumor growth↓, FOXO3a↑

Tumor growth↓, c-MYC↓ Tumor growth↓, NFκB↓ Tumor growth↓, NFκB pathway ↓ Tumor growth↓, FOXM-1b Tumor growth↓, c-MYC↓, Tumor growth↓, mTOR↓

Xenograft HepG2 Tumor growth↓, NFκB↓ hepatocellular Ca Xenograft Primary lung Tumor growth↓, TWIST-1 degradation, Ca, transgenic TWIST-1 pathway↓ tumors 41

Ruan et al., 2017 [178] Hamsa and Kuttan, 2012 [165] Goto et al., 2012 [179] Paydar et al., 2014 [95] Liu et al., 2015 [132] Wu et al., 2018 [158] Chen et al., 2013 [169] Allegra et al., 2018 [180] Visnyei et al., 2011 [97] Ren et al., 2019 [181] Saraswati and Agarwal, 2013 [170] Guo et al., 2018 [182] Yochum et al., 2017 [183]

Harmine

Syngeneic B16-F10 melanoma

Tumor growth↓, nitric oxide↓, NFκB↓, CREB↓, ATF-2↓

WNT/β - catenin signaling: Berberine Xenograft Colon Ca

Evodiamine Matrine

Xenograft SKOV3 ovarian Ca Xenograft H22 hepato- Tumor growth↓, β-catenin↓ cellular Ca Xenograft 4T1 breast Ca Tumor growth↓, WNT/β-catenin signaling↓

ERK/AKT/JNK signaling: Capsaicin Xenograft HOS osteosarcoma Capsaicin Xenograft 786-O renal Ca Capsaicin Xenograft PANC-1 pancreas Ca Piperine Xenograft Melanoma

Zhang et al., 2017 [111] Tumor growth↓, p38↑, JNK MAPK Liu et al., 2016 pathways↑ [113] Tumor growth↓, p-PI3K↓, p-AKT↓ Zhang et al., 2013 [78] Tumor growth↓, p-JNK↑, p-p38↑, p-ERKYoo et al., 1/2↓ 2019 [119] Glioma Tumor growth↓, glycolysis↓ Sun et al., 2018 [38] BGC-823 Tumor growth↓, AKT/mTOR/p70S6/S6 Yi et al., 2015 gastric Ca pathway↓ [184] SY5Y neuro- Tumor growth↓, PI3K-AKT pathway↓, IL-1↓ Qi et al., 2016 blastoma and [185] CT26 colon Ca Tumor growth↓, AKT-mTOR pathway↓, JNK Jiang et al., U87 and pathway↑ 2017 [164] SF767 glioblastoma MDA-MB-231 Tumor growth↓, ROS↑, ATM/Chk-2- and Li et al., 2014 breast Ca ATR/Chk1-mediated DNA-damage response, [85] p-ERK↑, p-JNK↑, p38 MAPK↑

-p

Xenograft

Berberine

Xenograft

Nuciferine

Xenograft

Sinomenine

Xenograft

Sinomenine

Xenograft

Tetrandrine

Xenograft AMKL leukemia Xenograft Huh7 hepatocellular Ca Xenograft HepG2 hepatocellular Ca Xenograft MNNG/HOS osteosarcoma Xenograft HeLa cervix Ca and A549 lung Ca Xenograft U87 glioblastoma

Oxymatrine Cinchonine

Evodiamine

lP

na

ur

Jo

Tetrandrine

Tumor growth↓, ERK1/2↓, p38↓

re

Berberine

Tetrandrine

Ruan et al., 2017 [178] Zhang et al., 2018 [82] Shi et al., 2016 [172] Xiao et al., 2018 [140]

ro of

Berbamine

Tumor growth↓, β-catenin↓ Tumor growth↓, β-catenin↓

Hamsa and Kuttan, 2010 [173]

Tumor growth↓, ROS↑, AKT and Notch1 signaling↑ Tumor growth↓, ROS↑, autophagy↑, mitochondrial dysfunction, ERK MAPK↑ Tumor growth↓, AKT↑, JNK↓, ERK↓

Liu et al., 2017 [159] Gong et al., 2012 [161] Liu et al., 2011 [137]

Tumor growth↓, p-PI3K↓, p-AKT↓

Zhang et al., 2014 [147] Qi et al., 2017 [186]

Tumor growth↓, AKT ubiquitination↓, pAKT↓ Tumor growth↓, p-JNK↑

42

Wu et al., 2017 [98]

Xenograft A498 renal cell Ca Evodiamine Xenograft HepG2 hepatocellular Ca Harmine Xenograft MCF-7 breast Ca Other signaling pathways: Capsaicin Xenograft U 266 multiple myeloma

Tumor growth↓p-PERK↑, p-JNK↑

Tumor growth↓, IL-6-induced STAT3 activation↓, JAK1↓, c-SRC↓, STAT3regulated gene products↓ (cyclin D1, Bcl-2, Bcl-xL, survivin, VEGF)

Bhutani et al., 2007 [81]

Berberine

Xenograft H1975 lung Ca

Tumor growth↓, SREBP1↓, lipidogenesis↓, ROS/AMPK pathway↑

Fan et al., 2018 [187]

Berberine

Xenograft U87 Tumor growth↓, EGFR-MEK-ERK pathway↓ glioblastoma Allograft Primary intra- Tumor growth↓, hedgehog pathway↓, cranial medu- binding to SMO lloblastoma from Ptch+/−; p53−/− mouse

Tumor growth↓,IL-6-induced STAT3 activation↓, p-JAK2↓, p-Src↓, p-ERK1/2↓, SHP1↑ Tumor growth↓, TAZ,↓ p-ERK↓, p-AKT↓

Wu et al., 2016 [152] Yang et al., 2013 [99] Ding et al., 2019 [154]

ro of

Evodiamine

Berberine Coptisine

na

Papaverine

-p

Xenograft Hepatocellular Ca Xenograft C666-1 nasopharynx Ca Xenograft HCT-116 colon Ca Syngeneic Rat C6 glioma

Tumor growth↓ miR-23a, p53-related tumor suppressive genes p21 and GADD-45α Tumor growth↓, STAT-3↓

Wang et al., 2014 [190] Tsang et al., 2013 [121] Tumor growth↓, RAS-ERK pathway↓ Huang et al., 2017 [83] Tumor growth↓, cAMP phosphodiesterase↓ ChelmickaSchorr et al., 1980; 1984 [191, 192] Tumor growth↓, p53 occupancy on miR-16-2 Zhang et al., promoter↑, mi-16 target genes↓ (Bcl-2, 2019 [193] cyclin D1)

re

Berberine

lP

Berberine

Liu et al., 2015 [188] Wang et al., 2015 [189]

ur

Sanguinarine Xenograft Hepatocellular Ca

Tumor growth↓, p-STRAP↓, p-MELK↓

Oxymatrine

Tumor growth↓, constitutive STAT-5 activation↓, JAK1/2 and c-SRC activation↓, IL-6-induced STAT-5 phosphorylation↓

Jo

Sanguinarine Othotopic HCT116 xenograft colorectal Ca Tetrandrine Xenograft 143 B osteosarcoma Tetrandrine Xenograft PML-RARαpositive acute promyelocytic leukemia Xenograft H1299 lung Ca

Tumor growth↓, PTEN↑, p-PTEN↓, p38 MAPK↑, p-p38 MAPK↑ Tumor growth↓, Notch1 signaling↑

43

Gong et al., 2018 [133] Tian et al., 2017 [87] Liu et al., 2015 [160]

Jung et al., 2019 [194]

Xenograft CAL 27 head Tumor growth↓, DYRK-1A↓ and neck squamous cell Ca

Radhakrishnan et al., 2017 [195]

Theophylline Syngeneic B16 Tumor growth↓, melanogenesis↑, cAMP↓ melanoma Other mechanisms: Capsaicin Xenograft PC-3 prostate Tumor growth↓, proteasome activity↓ Ca Berberine Xenograft SKOV3 Tumor growth↓, hERG-1↓ ovarian Ca

Kreider et al., 1975 [196]

Berberine

Wang et al., 2017 [93] Guo et al., 2019 [198] Li et al., 2018 [199] Qi et al., 2017 [151]

Oxymatrine Cinchonine

Tumor growth↓, TMEM-16A chloride channel inhibition Tumor growth↓, activity of wild-type and mutated EGFR↓ Tumor growth↓, p-TAK-1↓

Tumor growth↓, p-JNK↑, blood brain barrier Wu et al., passage 2017 [98]

Jo

ur

na

lP

re

Evodiamine

Tumor growth↓, EBNA-1↓

Mori et al., 2006 [177] Zhi et al., 2019 [197]

ro of

Matrine

Xenograft HONE1 nasopharynx Ca Xenograft LA795 lung adeno Ca Xenograft HCC827 lung Ca Syngeneic U14 squamous cell Ca, HeLa cervix Ca Xenograft U87 glioblastoma

-p

Harmine

44

Table 7: Inhibition of tumor growth in vivo by various mechanisms by plant alkaloids.

Compound Model Tumor type Regulation of the immune functions: Berberine Xenograft MGC 803 gastric Ca Berberine Syngeneic B16-F10 melanoma Brucine

Mode of action

Reference

Tumor growth↓, IL-8 secretion↓

Li et al., 2016 [200] Hamsa and Kuttan, 2012 [165] Agarwal et al., 2011 [171] Qi et al., 2019 [186]

Tumor growth↓, IL-1β↓, TNF-α↓, GM-CSF↓

Syngeneic Ehrlich ascites TNF-α↓, IL-12↑ tumor Secretion of TNF-α, IFN-γ, and IgG↑

Harmine

Tumor growth↓, proinflammatory cytokines↓, IL-2↑, NFκB↓

Syngeneic Ehrlich ascites Tumor growth↓, innate immune response↑ tumor

-p

Caffeine

HeLa cervix Ca and A549 lung Ca Syngeneic B16-F10 melanoma

ro of

Cinchonine Xenograft

Syngeneic H22 hepatocellular Ca Sinomenine Xenograft SW1116 colon Ca Brucine Xenograft U251 glioma

Tumor growth↓, AA metabolic pathway↓, cPL2↓, COX-2↓, PGE2↓ Tumor growth↓, COX-2↓

lP

Berberine

re

Anti-inflammatory response: Berberine Xenograft UtLM uterine Tumor growth↓, COX-2↓ leiomyoma

na

Stress response: Capsaicin Xenograft

Qian et al., 2016 [76] Lin et al., 2013 [77]

Orthotopic AsPC-1 xenograft pancreas Ca

Tumor growth↓, thioredoxin↓

Xenograft

Tumor growth↓, HSP70↓ Tumor growth↓, ER-stress response↑ (GRP78↑, p-ERK↑, p-EIF-2a↑)

Pramanik and Srivastava, 2012 [114] Paydar et al., 2014 [95] Jin et al., 2018 [150]

Tumor growth↓, aerobic respiration↓, glycolysis

Sun et al., 2018 [38]

Jo

Capsaicin

Boldine

Chuang et al., 2017 [202] Li et al., 2015 [203] Yang et al., 2016 [91] Ruijun et al., 2014 [149]

Tumor growth↓, oxidative stress (ROS↑, catalase↑, SOD-2↑), FOXO3a↑ Tumor growth↓, ER-stress response↑ (GRP78↑, p-ERK↑, p-EIF-2a↑, ATF-4↑, GADD153↑)

Xenograft

5637 and T24 bladder Ca PANC-1 and SW1990 pancreas Ca

ur

Capsaicin

Tumor growth↓, COX-2↓

Hamsa and Kuttan, 2010 [173] Mandal and Poddar, 2008 [201]

MDA-MB-231 breast Ca Cinchonine Xenograft HepG2 hepatocellular Ca Regulation of tumor metabolism: Berberine Xenograft Glioma

45

Berberine

Xenograft

Peiminine

Xenograft

Differentiation: Tetrandrine Xenograft Tetrandrine Xenograft

Tumor growth↓, impaired glycolysis

THP-1 leukemia AMKL leukemia

Tumor growth↓, differentiation↑, ROS↑, MYC↓ Tumor growth↓, differentiation↑, ROS↑

Tumor growth↓, p-GSK-3β↓

Tumor growth↓, differentiation↑, ROS↑ PML-RARαpositive acute promyelocytic leukemia

Wang et al., 2016 [157] Zhao et al., 2018 [90] Wu et al., 2018 [158] Liu et al., 2017 [159] Liu et al., 2015 [160]

Jo

ur

na

lP

re

-p

ro of

Tetrandrine Xenograft

U87 glioblastoma U251 glioblastoma

46

Table 8: Inhibition of invasion and metastasis by plant alkaloids.

Tumor type HCT-116 colorectal Ca Murine 4T1 breast Ca

Mode of action Primary tumor growth in vivo↓, tNOX↑, epithelial markers↓, mesenchymal markers↑ Metastasis in vivo↓, MMP-9↓, MMP-13↓, apoptosis↑ (caspase-3↑), G2/M cell cycle arrest (cyclin B1↓)

Reference Liu et al., 2012 [204] Lai et al., 2012 [205]

Piperine

Syngeneic

Berberine

Xenograft

SGC7901 gastric cancer

Hu et al., 2018 [206]

Berberine

Xenograft

Endometrial Ca

Primary tumor growth in vivo↓, migration and invasion in vitro↓ (MMP-3↓), G0/G1 cell cycle arrest (cyclin D1↓), Myc↓, Wnt5a↓, cytoplasmic β-catenin↓, HNF4α↓, AMPKHNF4α-WNT5A signaling↓ Primary tumor growth, migration, invasion, and metastasis in vivo and in vitro↓, AP-1mediated miR-101 expression↑, COX-2↓, PGE2↓, miR-101/COX-2/PGE2 signaling↓

Berberine

Berberine

Syngeneic / Xenograft Xenograft

4T1 and MDA- Primary tumor growth and lung metastasis in MB-231 breast vivo↓, TGF-β1↓, MMP-2↓, pyroptosis↑ Ca SiHa cervix Ca Primary tumor growth and metastasis in vivo↓, MMP-2↓, uPA↓, reversal of TGFβ1-induced EMT, E-cadherin↑, N-cadherin↓, SNAIL-1↓, angiogenesis↓

Berberine

Xenograft

Berberine

Chu et al., 2016 [209]

Primary tumor growth and migration in vivo↓, binds to VASP Invasion and metastasis↓, COX-2/PGE2JAK2/STAT3 signaling↓, MMP-2↓, MMP-9↓

Su et al., 2016 [210] Liu et al., 2015 [211]

Lung metastasis in vivo↓, HIF-1α/VEGF signaling↓, ID-1↓

Tsang et al., 2015 [212]

Xenograft

Primary tumor growth in vivo↓, migration and invasion in vitro↓, reversal of TGF-β1-induced EMT, E-cadherin↑, vimentin↓, SNAIL-1↓, SLUG↓ Lung metastasis in vivo↓, ERK↓, COX-2↓, AMPK↑ Primary tumor growth and metastasis in vivo↓, ERK1/2↓, NFκB↓, ATF-2↓, CREB↓ (signaling for MMPs)

Qi et al., 2014 [213]

Lymph node metastasis in vivo↓, p-EZRIN↓, filopodia formation↓

Tang et al., 2009 [216]

lP

re

Migration and invasion in vivo and in vitro↓, pyroptosis↑

Jo

Berberine

HepG2 hepato-cellular Ca Xenograft MDA-MB-231 breast Ca Xenograft SW620 and LoVo colorectal Ca Orthotopic MHCC-97L xenograft hepatocellular Ca

na

Berberine

Sun et al., 2018 [38] Chu et al., 2014 [208]

ur

Berberine

Wang et al., 2018 [207]

ro of

Model Xenograft

-p

Compound Capsaicin

Berberine

Syngeneic

Berberine

Syngeneic

Berberine

Xenograft

A549 lung Ca

B16-F10 melanoma B16-F10 melanoma 5-8F nasopharynx Ca

47

Kim et al., 2012 [214] Hamsa and Kuttan, 2012 [215]

Xenograft

U87MG and U251 glioblastoma

Primary tumor growth in vivo↓, migration, stemness, EMT in vitro↓, SOX-2↓, AKT and STAT-3 signaling↓, SLUG↓, angiogenesis, apoptosis↑, G2/M cell cycle arrest

Noscapine

Xenograft

PC3 prostate Ca

Sanguinarine Xenograft

A549 lung Ca

Primary tumor growth and metastasis in vivo↓ Barken et al., 2010 [218] Primary tumor growth in vivo↓, migration in Wei et al., vitro↓, E-cadherin↑, N-cadherin↓, 2017 [219] vimentin↓, SMAD2/3↓, SNAIL↓, p-SNAIL-2↓, FAF-1↑, apoptosis↑, S-phase arrest

Sinomenine

Xenograft

Sinomenine

Xenograft

MDA-MB-231 breast Ca HOS osteosarcoma

Lung metastasis in vivo↓, activation of NFκB and SHh signaling↓ Metastasis in vivo↓, p-CXCR-4↓, p-STAT-3↓, MMP-2↓, MMP-9↓, RANKL↓, osteoclastogenesis↓, S-phase arrest

Song et al., 2018 [220] Xie et al., 2016 [221]

Tetrandrine

Xenograft

SiHa cervix Ca

Primary tumor growth in vivo↓, invasion and migration in vitro↓, MMP-2↓, MMP-9↓

Zhang et al., 2018 [222]

Tetrandrine

Xenograft

HCCLM9 WT Metastasis in vivo↓, EMT-related proteins↓, and HCCLM9 Wnt/β-catenin pathway↓, MTA1↓ ATG7 KO hepatocellular Ca

Zhang et al., 2018 [223]

Tetrandrine

Syngeneic

Matrine

Xenograft

Matrine

Xenograft

Murine 4T1 breast Ca SK-N-PL neuroblastoma DU 145 prostate Ca

Gao et al., 2013 [224] Shen et al., 2018 [225] Chang et al., 2018 [226]

Matrine

Xenograft

-p

re

lP

na

Xenograft

Jo

Matrine

Xenograft

Lung metastasis in vivo↓, p-ERK↓, NF-κB↓, integrin β5↓, ECSM-1↓, IAM-1↓ Primary tumor growth in vivo↓, migration in vitro↓, TBR-3↑, PI3K/AKT activation↓ Proteasomal chymotrypsin-like (CT like) activity↓, reversal of EMT (vimentin↑, Ncadherin↑, E-cadherin↓), G0/G1 cell cycle arrest (c-MYC↓, cyclin B1↓, cyclin D1↓, CDK1↓), apoptosis↑ (Bcl-2↓, Bax↑)

HeLa cervix Ca Primary tumor growth in vivo↓, invasion and migration in vitro↓, MMP-2↓, MMP-9↓, p38 signaling↓, apoptosis↑

Wu et al., 2017 [227]

DU145 and PC3 prostate Ca HT29 colorectal Ca

Primary tumor growth in vivo↓, MMP-2↓, MMP-9↓, p-p65NF-κB↓ Primary tumor growth and metastasis in vivo↓, MMP-2↓, MMP-9↓, p-p38↓

Huang et al., 2017 [228] Ren et al., 2014 [229]

ur

Matrine

Li et al., 2019 [217]

ro of

Nuciferine

Matrine

Xenograft

U2OS osteosarcoma

Primary tumor growth in vivo↓, metastasis in vitro↓, MMP-2↓, MMP-9↓, p50 and p65 nucelar translocation↓, p-IκB-β↓, p-ERK1/2↓

Li et al., 2014 [230]

Oxymatrine

Xenograft

A439 lung Ca

Brucine

Xenograft

LoVo colon Ca

Primary tumor growth in vivo↓, miR-520↑, VEGF↓ Migration in vivo↓, MMP-2↓, MMP-3↓, MMP-9↓, Frizzled-8↓, Wnt5a↓, APC↓, GSNK1A1↓, AXIN-1↓, p-LRP-5/6↓, GSK-3β↓, p-βcatenin↑

Zhou et al., 2019 [231] Shi et al., 2018 [232]

48

Evodiamine

Harmine

Harmine

Caffeine Caffeine

Caffeine

Orthotopic Glioblastoma xenograft Xenograft HepG2 hepato-cellular Ca Xenograft U87 glioblastoma B16-F10 melanoma

Theophylline Xenograft

MeWo melanoma

Zhu et al, 2019 [234] Zhou et al., 2019 [235] Du et al., 2013 [236]

Zhang et al., 2014 [237]

Invasion in vivo↓, cathepsin B↓, p-FAK↓, pERK↓ Primary tumor growth in vivo↓, invasion and migration in vitro↓, AKT signaling pathway↓

Hamsa and Kuttan, 2011 [238] Cheng et al., 2016 [239] Dong et al., 2015 [240]

Survival in vivo↑, migration in vitro↓, IP3R3mediated Ca2+ release↓

Kang et al., 2010 [241]

Lung and liver metastasis in vivo↓, apoptosis↑ Lentini et al., 2000 [242] Primary tumor growth and lung metastasis in Gitelman et vivo↓, natural killer cell activity↑ al., 1987 [243]

Jo

ur

na

lP

Theophylline Syngeneic

Shu et al., 2013 [233]

ro of

Evodiamine

-p

Evodiamine

Syngeneic/ H22 and Metastasis in vivo (H22)↓, fibronectin↓, MMPXenograft HepG2 liver Ca 2↓, lysyl oxidase↓, and cathepsin C↓ in vitro (HepG2) Xenograft TFK-1 cholPrimary tumor growth in vivo↓, invasion and angio Ca migration in vitro↓, IL-6 -induced STAT3 signaling↓, SHP-2↓ Xenograft Colorectal Ca Metastasis in vivo↓, MMP-9↓, NAD+/NADH ratio↑, SIRT-1↑, acetyl-NFκB p65↓ Xenograft MDA-MB-231 Primary tumor growth and lung metastasis in breast Ca vivo↓, MMP-9↓, uPA↓, and uPAR↓, G0/G1 cell cycle arrest (cyclin D1↓, CDK6↓, p27Kip1↓), apoptosis↑ (Bcl-2↓, Bax↑) , pERK↓, p-p38 MAPK↓ Xenograft Gastric Ca Primary tumor growth in vivo↓, migration, invasion and induced apoptosis in vitro↓, COX2↓, PCNA↓, Bcl-2↓, MMP-2↓ Syngeneic B16F-10 Primary tumor growth in vivo↓, invasion and melanoma migration in vitro↓, MMP9↑, ERK↑, VEGF↑

re

Brucine

49

Table 9: Chemoprevention of cancer by plant alkaloids.

Lung carcinogenesis

Capsaicin

Lung carcinogenesis Lung carcinogenesis Hepatocarcinogenesis

Capsaicin Piperine

Piperine

Lung carcinogenesis

Berberine

Colorectal carcinogenesis Hepatocarcinogenesis Lung carcinogenesis

Berberine Evodiamine

Hepatocarcinogenesis Sanguinarine Skin carcinogenesis

Skin carcinogenesis

Jo

Caffeine

Skin carcinogenesis

ur

Caffeine

TNF-α, IL-6, COX-2 and NF-κB

Ananda-kumar et al., 2012 [244]

No statistically significant effect

Teel et al., 1999 [245]

Tumor incidence↓

Jang et al., 1989 [246] Jang et al., 1989 [246] Gunase-karan et al., 2017 [247]

BP-treated Swiss albino mice DMH- and DSStreated rats DEN- plus PBtreated rats Urethanetreated mice

Protein bound carbohydrate levels↓

Selvendiran et al., 2006 [248]

Nuclear and cytoplasmic βcatenin↓ Hepatocyte proliferation↓, iNOS↓, CYP2E1↓, CYP1A2↓ Tumor size and numbers↓, NOTCH3 signaling↓, DNMTsinduced NOTCH3 methylation↑

Wu et al., 2012 [249] Zhao et al., 2008 [250] Su et al., 2018 [251]

DEN-treated rats UVB-lightirradiated SKH1 mice

Unscheduled DNA synthesis↓, GST-P-positive hepatic foci↓ Skin edema↓, skin hyperplasia↓, leukocyte infiltration↓, ROS↓, ODC,↑ PCNA↑, Ki-67↑

Kim et al., 1992 [252] Ahsan et al., 2007 [253]

UVB-lightirradiated SKH1 mice UVB-lightirradiated p53knockout mice UV-irradiated mouse epidermis

ATR/Chk1 pathway↓ (p-Chk1↓), lethal mitosis↑ (cyclin B1↑, caspase-3↑), apoptosis↑ Apoptosis↑, ATR-mediated pChk1↓, lethal mitosis↑ (cyclin B1↑) No effect on tumor incidence

Lu et al., 2011 [254]

na

Caffeine

BP-treated Swiss albino mice Tobaccospecific NKKtreated AJ mice BP-treated female mice DMBA-treated female mice DEN-treated rats

Caffeine, Skin theophylline carcinogenesis Other carcinogenesis models: Berberine Intstestinal carcinogenesis

Tumor incidence↓

ro of

Capsaicin

Reference

Apoptosis↑, pro-oxidant effect↑

-p

chemical carcinogenesis: Capsaicin Lung carcinogenesis

Carcinogenesis Observations induction

re

Tumor entity

lP

Compound

high fat dietTumorigenesis↓, treated verrucomicrobiota in the gut↓, ApcMin/+ mice Akkermansia and short chain fatty acid-producing bacteria↓ 50

Lu et al., 2008 [255] Zajdela and Latarjet, 1978 [256] Wang et al., 2018 [257]

Berberine

Colitis-associated DSS-treated Tumorigenesis↓, EGFR-ERK tumorigenesis ApcMin/+ mice signaling↓ and colonic epithelium hyperproliferation

Li et al., 2017 [258]

Berberine

Colorectal carcinogenesis Lung carcinogenesis Colorectal carcinogenesis

EGF-stimulated EGFR activation A540 lung Ca implantation ApcMin/+ mice

Wang et al., 2013 [259] James et al., 2011 [260] Ma et al., 2018 [261]

Indole-3carbinol

Postate carcinogenesis

TRAMP mice

Harmine

Glioblastoma carcinogenesis

Tumor cell implantation

Jo

ur

na

lP

re

-p

Palmatine

Wu et al., 2012 [262]

ro of

Berberine

ubiquitin ligase CBL ↑ to downregulate EGFR G0/G1 cell cycle arrest, p-AKT↓, pCREB↓, p-MAPK↓ Tumor size and numbers↓, life span↑, dysplasia↓, IL-1α, IL1-β, IL-8, G-CSF, and GM-CSF in the gut↓ cyclin D1↓ and apoptosis↑ (PARP cleavage, p21↑, caspases-3 and 7↑), ARE-luciferase activity↑, NRF-2-mediated genes↑ (NRF-2, NQO-1) Tumorgenicity↓, self-renewal of stem-like cancer cells↓, differentiation↑, apoptosis↑, pAKT↑

51

Liu et al., 2013 [263]

Table 10: In vivo toxicity of plant alkaloids.

Reference Bezerra et al., 2006 [260] Bezerra et al., 2006 [260] Landen et al., 2004 [261]

Landen et al., 2002 [262]

No side effects

Xenograft

AsPC-1 pancreas Ca NB4 leukemia

Capsaicin Piperine

Xenograft

Melanoma

No side effects

Coptisine

Xenograft

No effects on body weight

Noscapine

Xenograft

Sinomenine

Xenograft

Hepatocellular Ca PC3 prostate Ca SW1116 colorectal Ca

Sinomenine

Xenograft

ur

Matrine

Mice

MDA-MB-231 breast Ca LA795 lung Ca

Matrine

Xenograft

Colorectal Ca

Oxymatrine

Xenograft

Jo

No side effects

lP

Xenograft

na

Capsaicin

re

-p

ro of

Compound Model Tumor type Observations Syngeneic tumor transplantation models: Piperine Syngeneic Sarcoma-180 Hepatotoxicity (hepatocyte degeneration, microvesicular steatosis) Piplartine Syngeneic Sarcoma-180 Nephrotoxicity (hydropic changes of the proximal tubular and glomerular epithelium and tubular hemorrhage) Noscapine Syngeneic Rat C6 glioma No toxicity to the duodenum, spleen, liver, and hematopoietic system; no peripheral toxicity, but vasodilatation in brain Noscapine Syngeneic B16-LS9 No toxicity to the spleen, liver, melanoma duodenum, bone marrow, and peripheral blood Quinine Syngeneic Ehrlich ascites Promotion of tumor growth tumor Xenograft tumor transplantation models: Capsaicin Xenograft SW480 Metastasis↑, EMT↑, MMP-2↑, MMPcolorectal Ca 9↑, AKT/mTOR and STAT-3 pathways↑ Capsaicin Orthotopic MDA-MB-231 No side effects xenograft breast Ca

MNNG/HOS osteosarcoma Genotoxic and carcinogenic effects: Capsaicin TRPV1/KO TPA-promoted mice skin carcinogenesis

No toxicity, but reduced body weight No body weight change at low concentrations, but reduced body weight at high concentrations No side effects No effects on body weight No side effects on physical health No effects on body weight and mortality

Castelli et al., 1996 [263] Yang et al., 2013 [264] Thoennissen et al., 2010 [80] Zhang et al., 2008 [116] Ito et al., 2004 [118] Yoo et al., 2019 [119] Chai et al., 2018 [127] Barken et al., 2010 [218] Yang et al., 2016 [91] Li et al., 2014 [85] Guo et al., 2019 [198] Gu et al., 2018 [139] Zhang et al., 2014 [147]

Cocarcinogenic effect, size and number of Hwang et al., skin tumors↑,COX-2↑, EGF activation, p- 2010 [265] EGFR↑ 52

Capsaicin

Mice

N/A

Cecum tumor induction

Transcapsaicin

Mice

N/A

Piperine

Rats

N/A

Weak mutagenicity in in vivo bone marrow micronucleus and chromosomal aberration in human peripheral blood lymphocytes Bioavailability of aflatoxin

Noscapine

Mice

N/A

Sanguinarine Mice

N/A

Glutathione levels in liver and serum biochemistry revealed no evidence of hepatotoxicity through bioactivation in mice

Caffeine

Mice

Oxymatrine

Brucine

-p

Harmine Caffeine

na N/A

ur

Theophylline Mice

N/A

lP

re

Caffeine

Allameh et al., 1992 [268] Fang et al., 2012 [269]

Ansari et al., 2005 [270] Lu et al., 2016 [271]

ro of

Caffeine

DNA damage in blood and bone marrow cells as determined by the comet assay Mice N/A Hepatotoxicity (transaminases↑, alkaline phosphatase↑, malondialdehyde↑, TNFα↑, TRADD↑, p-SAPK/JNK↑) Mice N/A No dermal toxicity upon transdermal administration Mice N/A DNA adduct formation in liver and kidney indicated genotoxicity Mice N/A No effects on mouse oocytes regarding retardation rate and aneuploidy BD2F1 and Breast Mammary gland development↑ C3H mice carcinogenesis indicative of increased tumorigenesis upon DMBA-treatment Mice N/A Sister chromatide exchange↑

Toth and Gannett, 1992 [266] Chanda et al., 2004 [267]

Protection from toxicity: Berberine Mice

Chronic inflammation of the mesenteric arteries, no evidence of carcinogenicity, incidence of breast Ca, hepatocellular adenoma and carcinoma↓

Berberine

Rats

N/A

Protection of gastrointestinal mucosa from acute heavy alcohol consumption, TNFα↓, IL-1β↓, TLR2↓, TLR4↓ Doxorubicin-induced cardiomyopathy↓, survival↑, heart stroke volume↑, myocardial injury↓, apoptosis (caspase3↓, p-AMPKα↓, p-p53↓, Bcl-2↑)

Nuciferine

Xenograft

A549 lung Ca

Nictotine-induced liver injury↓

Oxymatrine

Rats

N/A

Doxorubicin-induced cardiotoxicity↓ (catalase, malonyldialdehyde, superoxide dismutase, glutathione peroxidase and ROS)

Jo

N/A

Chromosomal damage ↑ upon MMS cotreatment

53

Chen et al., 2013 [272] Yamashita et al., 1988 [273] Mailhes et al., 1996 [274] Welsch et al., 1988 [275] Panigrahi et al., 1983 [276] Frei and Venitt, 1975 [277] National Toxicology Program, 1998 [278] Wang et al., 2015 [279]

Liu et al., 2015 [281] Zhang et al., 2017 [282]

Lv et al., 2012 [280]

Mice

N/A

Protection from vinblastine-induced chromosomal aberrations and mitotic index in bone marrow cells

Caffeine

Syngeneic

RT4 bladder Ca Radioprotection, ATM-Chk2-p53-Puma DNA damage-signaling↓, γH2AX↓, p53binding protein 1↓

Geriyol et al., 2015 [283] Zhang et al., 2014 [284]

Jo

ur

na

lP

re

-p

ro of

Caffeine

54